You are on page 1of 16

Food Bioscience 43 (2021) 101258

Contents lists available at ScienceDirect

Food Bioscience
journal homepage: www.elsevier.com/locate/fbio

Different nano-delivery systems for delivery of nutraceuticals


Ladan Rashidi
Department of Food and Agricultural Products, Food Technology and Agricultural Products Research Center, Standard Research Institute (SRI), Iranian National
Standards Organization (INSO), PO Box 31745-139, Karaj, Iran

A R T I C L E I N F O A B S T R A C T

Keywords: The nutraceutical or ‘bioceutical’ is an active compound with a pharmaceutical and standardized nutrient, which
Nanoencapsulation has physiological benefits for human health, performance, and well-being. Nanoencapsulation technology has
Nutraceuticals received increasing attention for entrapping bioactive compounds into nanocarriers for their preservation against
Bioactive compounds
undesirable conditions, including gastrointestinal digestion and cellular metabolism; controlling their release,
Polyphenols
Nanocarriers
enhancing biodistribution and bioavailability of bioactive compounds and their delivery into the target site of the
organism’s body. This paper presents an overview of and discusses the different nanocarriers (food grade and
other nano-delivery systems) used for the encapsulation of bioactive compounds or nutraceuticals concerning the
advantages and disadvantages of their application. It is well-known that the commercialization of food products
containing nanocarriers having nutraceuticals is still in the primary stages of expansion, and risk assessment and
safety factors should be considered by national bodies before their widespread usage.

1. Introduction nanoliposomes, and nanocomplexation have been developed to protect


valuable or active compounds at suitable quantities, shelf life, and
Nutraceuticals are especially natural materials or derived from nat­ controlled release (Milincic et al., 2019). It is considered that these
ural sources, including vitamins, antioxidants, prebiotics, fatty acids, nanocarriers should not modify the physicochemical and sensory spec­
minerals, polyphenols, phytosterols, probiotics, carotenoids, fibers, ifications of the nutraceuticals or bioactive compounds.
essential oils, and bioactive peptides have received considerable atten­ The physicochemical properties of nano-delivery systems, such as
tion due to their health benefits and nutrition for human consumption size, functionality, shape, charge and hydrophilicity/hydrophobicity of
(Fig. 1). These compounds can be added as ingredients to foods to bioactive compounds/nutraceuticals impress on the cellular uptake ef­
produce “functional foods” for improving human safety and health, ficiency. These features should be taken into account for the marketing
performance, and well-being (Pateiro et al., 2021). Nutraceuticals of products produced with nanotechnology. For example, nanocarriers
derived from natural sources are quickly oxidized and unstable which based on their dimensions may easily permeate biological tissues, thus
lead to limits their utilization as nutraceutical ingredients. A suitable disrupting cells’ usual function with adverse effects on human health.
tool for the increase of bioavailability, durability, and adsorption of Cellular uptake of bioactive species (such as ions and molecules) and
nutraceuticals is encapsulation using nanotechnology methods (Ramlia, nanocarriers like nanoparticles is based on passive transport and active
Alib, Hamzah, & Yatim, 2021). Nanoencapsulation systems provide transport which hydrophobic and small molecules can be penetrated in
several benefits including undesirable taste, odor, and color masking, the lipid bilayer via passive transport, while nanoparticles and polar
providing pH-triggered controlled release, improved stability, reducing molecules can be internalized by the specific receptors via active
side effects, improved shelf life, providing moisture-triggered, protec­ transport (Dima, Assadpour, Dima, & Jafari, 2020).
tion against oxidation, preservation of volatile ingredients, protection Therefore, each type of nanocarriers is specified by advantages and
against enzyme degradation, and pH before reaching the target, pro­ deficiencies, bioavailability, degree of ensured conservation and trans­
tection the ingredients from different environmental parameters fer of bioactive agents, ease of application, biodistribution, food matrix
including oxygen, heat, water and light, improved solubility of vitamins compatibility, production and usage expenses, biodegradability, regu­
(Kiss, 2020). The number of nanocarriers such as nanoemulsions, latory situation (A series of safety, environmental, and regulatory as­
biopolymer matrices, mesoporous silica nanoparticles, carbon nano­ pects which could influence human health and the environment should
tubes, dendrimers, solid liquid nanoparticles, magnetic nanoparticles, be intently attended to) and biocompatibility (Dima et al., 2020).

E-mail addresses: l.rashidi@standard.ac.ir, ladan_rsh@yahoo.com.

https://doi.org/10.1016/j.fbio.2021.101258
Received 3 February 2021; Received in revised form 6 July 2021; Accepted 18 July 2021
Available online 23 July 2021
2212-4292/© 2021 Elsevier Ltd. All rights reserved.
L. Rashidi Food Bioscience 43 (2021) 101258

chemical structure, etc.), properties of the food matrix (including the


interaction between biocomponents), food storage conditions (including
thermal treatments, freezing, pasteurization, canning, etc.), mechanical
treatments (such as washing, cutting and homogenization), chemical or
biochemical treatments (including salting, esterification, fermentation,
etc.), processing level of food (which depends on the structure and food
composition matrix and processing technique), and characteristics of
nutraceutical delivery systems (including small size, control release, the
superficial electrical charge, and diversity of carrier materials) (Dima
et al., 2020). Results of external parameters studies on nutraceuticals
oral bioavailability showed that bioavailability is the complex system of
biological processes and physicochemical properties which help to
biomolecules distribution in the body for increasing therapeutic impact.
In addition, the study of uptake mechanisms of bioactive loaded nano­
carriers is important for both improving bioavailability and under­
standing the risk assessment of nanocarriers’ toxicity and also risks of
accumulation of nanocarriers as nanoparticles in tissues and organs.

3. Encapsulation techniques, advantages, and disadvantages

Encapsulation is one of the useful processes applied for covering


various materials in different matrices of gas/gaseous, liquid, and solid
in the food industry. The major objective of the encapsulation is coating
sensitive compounds or reducing side effects of some useful compounds
applied in high concentrations, which these compounds are located in
Fig. 1. Kinds of Bioactive compounds.
the core and coated by suitable wall materials. Encapsulation techniques
protect nutraceuticals or bioactive compounds from unbalanced and
This study will describe recent nano-delivery systems used to
unfavorable conditions, including pH, light, moisture, heat, chemical
encapsulate different nutraceuticals or bioactive compounds in the food
and biological degradation, and oxygen during storage, processing, and
industry. In addition, the advantages and disadvantages of the most
utilization (McClements, 2020; Kiss, 2020; Rashidi & Khosravi-Darani,
common encapsulation techniques and nano-delivery systems used to
2011). Wall materials have a critical role in the encapsulation tech­
encapsulate nutraceuticals are discussed. Besides safety issues on the use
nique because of their important effects on target delivery, bioavail­
of nanomaterials in food industry are described.
ability, biocompatibility, and protection of bioactive compounds. Also,
these materials should not have impact on flavor, color, texture, and
2. Nutraceuticals
other properties of foods. The important properties of suitable wall
materials contain a low cost, low viscosity, film-forming capacity, high
Nowadays, natural dietary agents are considered both by the general
solubility, low hydroscopy, high stability against the media of the tar­
public and researchers because of their properties to suppress cancers,
geted, high protection, abundant, nontoxic, and compatible via food or
prevent cancer creation, and reduce the risk of cancer development.
drug formulations (Delshadi, Bahrami, Tafti, Barba, & Williams, 2020).
These phytochemicals have low water solubility and poor stability lead
Several techniques are used for encapsulating bioactive agents which
to poorly adsorb by the human body. Nutraceutical such as polyphenols,
the preferred encapsulation technique depends on the bioactive com­
some special peptides, and essential oils are water-insoluble, unstable,
pound structure and its end use (Delshadi et al., 2020). The famous
and have low oral bioavailability and biodistribution, which these
encapsulation techniques are included supercritical fluids, inclusion
characteristics limit their biological activities (Shi et al., 2021).
complexation, nanoprecipitation, freeze-drying, spray drying, emulsifi­
Bioavailability of oral administration refers to the transfer of nutra­
cation, solvent evaporation, coacervation, ionic gelation, self-assembly
ceuticals or bioactive compounds through the gastrointestinal tract
of molecules, co-crystallization, and electrospinning which are
(GIT) stages and “absorption” is the second step of bioavailability and
described as follows:
also “bioaccessibility” is the first step of bioavailability. Polyphenols
Coacervation method is based on the interaction between two
have low bioavailability which is mainly because of the low absorption
oppositely charged polyelectrolytes in water for the formation of the
of them in the human gastrointestinal (GI) tract following consumption,
coacervate which can be used to encapsulate heat-sensitive ingredients at
high biotransformation in the gut, glycosylated type of polyphenols
room temperature but this method has difficulties in scaling up, the
compounds which reduce their diffusion across obstacles in the GI tract,
particle isolation procedure and uses large amounts of organic solvents.
and fast elimination from the human body (Milincic et al., 2019). In
Also, this method has advantages including simple preparation condi­
addition, some phytochemicals cannot be absorbed from enterocytes
tions, including low agitation and non-toxic solvent, high loading ca­
because they are unstable in the stomach (the acidic condition) or the
pacity, reduced evaporation losses, low temperature, no special
small intestine (alkaline situations). They are generally metabolized in
equipment is needed, and controlled release of active material (Ozkan,
the liver which helps their rapid excretion (Milincic et al., 2019). Some
Franco, De Marco, Xiao, & Capanoglu, 2019).
of the bioactive compounds can be subjected to chemical and
In the solvent emulsification/evaporation, the lipid dissolves in the
biochemical transformations in GIT fluids. For example, curcumin de­
water-immiscible solvent as chloroform or toluene and then is emulsi­
grades in the alkaline environment and the exposure of a certain enzyme
fied in an aqueous phase before evaporation of the solvent under
(Rafiee, Nejatian, Daeihamed, & Jafari, 2019), proteins and peptides
reduced pressure. Then, lipid precipitation upon solvent evaporation,
may be hydrolyzed in acidic situations and also under the act of pro­
therefore nanocarriers are formed. Merits of this method include lab-
teolytic enzymes (Akbarbaglu et al., 2019). In addition, there are
scale acceptability, higher stability, obtaining the smallest particle size
external parameters which can influence on the bioavailability of
but its demerits are the use of toxic solvents, increase of lipid content
nutraceuticals, including physicochemical characteristics of nutraceut­
leads to the increase of polydispersity index and particle size distribution
icals (such as the solubility, the physical state, the lipophilicity, the
(Ozkan et al., 2019).

2
L. Rashidi Food Bioscience 43 (2021) 101258

Inclusion complexation method uses for encapsulation of volatile solubility, and good stability. The disadvantages of this method are its
organic molecules and defines the molecular association between wall application for lipophilic nutraceuticals and using high volumes of water
and core, attributed to as cavity-bearing substrate and ligand, respectively. for elimination from suspension, and reducing efficiency (Tiruwa,
In addition, known as the molecular inclusion technique which takes 2015).
place at molecular levels and produces nanocapsules with sizes from The supercritical fluid technique is an alternative method in which
0.001 to 0.01 μm. Inclusion complexation technique has advantages organic solvents are not applied. Supercritical CO2 is widely applied as a
including simplest formulate, reduce evaporation and stabilize flavors, supercritical fluid due to its mild critical situations, which is not toxic,
reduce hemolysis, enhance solubility, stability, and bioavailability while non-flammability, and has a low price. The advantages of this technique
its disadvantages are high cost of preparation, stability issues, difficulty are control of particle size and morphology, preventing of losing or
in incorporating into the formulation of dosage forms, reproducibility of degradation of biological activities of thermos sensitive nutraceuticals,
physicochemical properties, small dose nutraceuticals are complexed, high encapsulation efficiency. The limiting parameter of using this
and scale-up of the manufacturing process (Ozkan et al., 2019). method is the selection of the supercritical process on the basis of the
Nanoprecipitation or solvent displacement method is used for lipo­ solubility of active material to be encapsulated and polymer matrix in
philic nutraceuticals in which a preformed polymer and the nutraceu­ the supercritical fluid (Ozkan et al., 2019).
tical are dissolved in an organic solution, then organic phase is poured Ionic-gelation or polyelectrolyte complexation is based on the
into aqueous medium containing a surfactant. Nanoprecipitation is a capability of crosslinking polyelectrolytes in the presence of multivalent
versatile technique for preparation of different types of polymer nano­ ions and is applied for extrusion or emulsification/gelation which
particles with suitable controlled particle size, surface properties, and extrusion method uses for fabrication of spherical gel particles using
size distribution. A disadvantage of this method is the low concentration dripping an aqueous polymer solution through the syringe needle or a
of polymer leads to a problem in nanoparticle recovery in the final nozzle into CaCl2 having a gelling bath and emulsification/gelation
dispersion (Tiruwa, 2015). method produces an emulsion such as a hydrophobic active component
The reverse - phase evaporation method is applied for the prepara­ in the polymer solution followed by dripping into the calcium solution.
tion of liposomes in which the lipid mixture is added to the organic This technique is simple, and easy due to its low cost and does not need
solvent, then solvent under reduced pressure using rotaevaporation is special equipment, organic solvents, high encapsulation efficiency and
removed. The lipids are re-dissolved in an organic phase for formation high temperature. Also, this method uses reversible physical cross­
the reverse-phase vesicles. The limitations of this method are the pos­ linking instead of chemical crosslinking, therefore there is not possible
sibility of remaining solvent after evaporation, time-consuming, and risk toxicity of reagents (Ozkan et al., 2019).
of high temperature exposure leading to phospholipid/nutraceutical The liposomal entrapment method can be used for encapsulation of
damage, and decreasing of encapsulation and influencing carriers’ sta­ enzymes, hormones, and vitamins which uses one or more layers of
bility (Tiruwa, 2015). lipids which is not toxic. Liposomes are composed of lipid bilayers and
Freeze drying or lyophilization contains multi stage processes, easy to make and can be stored by freeze drying. Disadvantages of
including freezing, sublimation, desorption, and finally storage steps. In liposomal entrapment are scaling up, poor physical and chemical sta­
this method, ice is sublimed for production of a gas under pressure and a bility, lipid oxidation, producing a wide range of particle size distribu­
small amount of heat (low operating temperature without the existence tions, leakage and fusion of encapsulated nutraceuticals or bioactive
of air) which can be used for heat-sensitive materials for their storage for agents, also some of the phospholipids used in this method undergo
a long time but this technique is more expensive and needs a long oxidation and hydrolysis-like reaction which leading to decrease of
dehydration time. Also, this technique creates the porous structure of liposome shelf life (Ozkan et al., 2019).
freeze-dried powders because of the ice sublimation during the process Electrospinning is used for the incorporation of nutraceuticals or
and the freeze-dried powders should be crushed or ground after drying, bioactive agents within electrospun fibers which enhances functional
therefore control of particle size is another problem of using this method materials performance in the food industry. This technique can produce
(Ozkan et al., 2019). nano-scale polymer fibers. The advantages of this method are ease of
Spray drying is the oldest technique used for encapsulation of use, cost-effective of synthesizing nanofiber, easy encapsulation of
nutraceuticals related to the atomization of the liquid into dry powder bioactive compound into nanofibers, does not need heat treatment, and
by an injector as a hot drying gas stream comprised of three steps does not impact on sensory qualities of products (Castro Coelho,
including homogenization of feed liquid as a core wall and wall material Nogueiro Estevinho, et al., 2020).
with an atomizer, drying of feed solution using a hot gas carrier for Self-assembly is another encapsulation technique with advantages,
achieving the evaporation of the solvent and collection of dry particles including the production of small nanoparticles, high encapsulation ef­
by cyclones or filter. This technique is simple, rapid, economic, easy to ficiency, and high stability but its disadvantages are the problem to
scale-up, reproducible, obtain solid particles with low moisture content, control the size and shape of nanoparticles (Castro Coelho, Nogueiro
and continuous. The disadvantages of this method include high con­ Estevinho, et al., 2020).
sumption of energy, use of high temperature leads to damage sensitive The overview of applying different encapsulation techniques for
compounds, low product yield, the low water solubility of proteins and nanoencapsulation of various bioactive compounds is presented in
polysaccharides leads to limitations for applying spray drying, lack of Tables 1–5.
controlling the droplet size and shape, high cost, and time aspects
(Ozkan et al., 2019). 4. Food grade delivery systems
The salting out method is considered as a modification of the
emulsification/solvent diffusion which is based on the separation of the 4.1. Protein - based delivery systems
water miscible solvent from the aqueous medium through the salting out
impact. Salting out technique does not use of toxic organic solvents. Protein nanocarriers can be obtained from bacterial, plant, animal,
Therefore, polymer and nutraceutical are dissolved in a solvent that and fungal sources and are applicable for the delivery of both hydro­
emulsified into the aqueous solution containing the salting out agent as philic and hydrophobic nutraceuticals (bioactive agents). Gelation,
electrolytes, including sodium chloride and calcium chloride. This extrusion, emulsification, inclusion complexion, coacervation, and
method uses electrolytes for the separation of organic solvent and spray drying are useful techniques for producing proteins based on
minimizes stress to protein. The solvent is removed by filtration or nanodelivery systems such as nanoassemblies, hydrogels, micelles,
evaporation. The advantages of this technique for encapsulation are low films, hydrocolloids, and micro- and nanoparticles (Assadpour & Jafari,
cost, high particle quality with small size, good yield and rapid 2019). Gelatin, casein, whey proteins, albumin, collagen, and elastin

3
L. Rashidi Food Bioscience 43 (2021) 101258

from animal sources and soy glycinin, and wheat gliadin and zein from 4.2. Lipid-based delivery systems
plant sources are commonly applied as protein carriers (Assadpour &
Jafari, 2019). Meat proteins including gelatin and collagen, especially The use of lipid-based nanodelivery systems is an advantage for the
gelatin derived from collagen, used for encapsulation of nutraceuticals lipid-based nutraceuticals which are hydrophobic compounds including
due to their biocompatibility, high-temperature stability, biodegrad­ fatty acids (especially ω-3 and conjugated linoleic acid (CLA)), carot­
ability, ease of bridge, and safety but disadvantages of using them are enoids (lycopene, β-carotene, lutein), antioxidants (polyphenols
low mechanical strength and rapid degradation speed (Fathi, Donsi, & including oleuropein, caffeic acid, gallic acid, tocopherols, flavonoids),
McClements, 2018; Hong et al., 2020). fat-soluble vitamins (A, E, D, and K) and phytosterols (stigmasterol,
Milk proteins contain different molecular structures and functional β-sitosterol, campesterol), to be entrapped in the liquid form. Lipid-
features. Casein is poorly water-soluble and has high-temperature sta­ based nanostructures applied for bioactive compounds of foods and
bility. It has four main protein fractions including αS1, αS2, β, and nutraceuticals’ delivery include lipid nanospheres, especially lipid
κ-casein but tends to aggregate near to their isoelectric point and in the nanoparticles, nanoliposomes, nanocochleates, and archaeosomes. The
attendance of multivalent counter-ions which limited their usage in principal techniques employed to formulate a lipid-based delivery sys­
some foods. β-casein has strong amphiphilic property for encapsulation tem are emulsification, oil-in-water (O/W) nanoemulsions or micro­
of both hydrophobic and hydrophilic nutraceuticals. Whey proteins emulsions, multilayer emulsions, filled hydrogel particles, liposomes, or
possess advantages for encapsulation of nutraceuticals, including the solid lipid nanoparticles (Assadpour & Jafari, 2019). In the following,
ability of formation hydrogels, and their surface activity but in higher the three most important lipid-based nanocarriers are described in
temperatures of around 70 ◦ C undergoe thermal denaturation (are not detail. In addition, Tables 2a, 2b, and 2c present the lipid-based nano­
thermostable), therefore, it can be used to encapsulate heat-sensitive delivery systems including nanoemulsions, solid lipid nanoparticles
nutraceuticals at ambient temperature. In upper than 70 ◦ C, the pro­ (SLNs), and nanoliposomes applied for the encapsulation of
teins relatively unfold and connect together via hydrophobic attraction nutraceuticals.
and disulfide bond generation. Different fractions of whey proteins
include α-lactoglobulin (α-Lg), β-lactoglobulin (β-Lg), and bovine serum 4.2.1. Nanoemulsions
albumin (BSA) which all of them can be used for encapsulation of Nanoemulsions are heterogeneous systems consisting of dispersions
bioactive compounds (Fathi et al., 2018). β-LG uses for encapsulation of of nano-scale droplets formed by shear-induced rupturing. They contain
low molecular acid-sensitive bioactive compounds for its stability in two immiscible liquids, in which one liquid phase is dispersed as drop­
acidic media and proteolytic enzymes in the stomach. Egg proteins like lets into a continuous liquid phase and stabilized with an emulsifier
lysozyme, ovomucin, albumin, livetins, etc. can be applied for encap­ (Peng, Meng, et al., 2019). Therefore, the main three components of
sulation purposes but egg ovalbumin is further commonly used for nanoemulsions are oil, surfactant or cosurfactant, and aqueous phase, in
encapsulation due to its relatively resistance to pepsin digestion in the which oil can be of any type, such as olive oil, castor oil, corn oil, sesame
stomach. Albumin can be used for water-soluble bioactive compounds. oil, coconut oil, linseed oil, medium-chain triglyceride oil (MCT), min­
Zein proteins are found in corn and constitute of three-quarters hydro­ eral oil, peanut, or evening primrose. Nanoemulsions are distinguished
phobic and one-quarter hydrophilic amino acid residues lead to be as an O/W (oil in water) or W/O (water in oil) emulsion with mean
insoluble in water and soluble in some organic solvents like ethanol. It droplet size ranging from 20 to 200 nm which are generally recognized
can be used for encapsulation of water-insoluble bioactive compounds as safe (GRAS) by the The Food and Drug Administration (FDA), 2014
but it shows the limited digestion in the stomach leads to the use of it for and can be used for human consumption. Table 2a shows nanoemulsion
encapsulation of acid-sensitive compounds. Soy proteins isolated from carriers applied for the encapsulation of bioactive agents. In the syn­
soybeans and show surface activity and structure formation lead to thesis of nanoemulsions, the selection of emulsifier type is highly
development delivery systems and encapsulation but they should be important for controlling interfacial properties, including thickness,
carefully purified to prevent excessive protein aggregation and dena­ droplet, rheology, and charge. It was reported that using double-layer
turation due to loss of their functionality. Pea proteins consist of techniques (oil droplets coated by double-layered interfacial mem­
different globular proteins such as albumins, gluteins, and globulins (the branes) for forming W/O/W multiple emulsions could efficiently coat oil
major fraction of pea proteins consists of three types including legumin, particles during emulsification (Peng, Meng, et al., 2019). Nano­
vicillin, and convicillin) which have surface-active, and structure for­ emulsions do not sediment or cream since the Brownian motion is
mation properties. Also, a recombinant protein can be applied for greater than the small creaming rate induced by gravity. The most
encapsulation due to its higher purification than natural proteins but its common methods for the preparation of nanoemulsions include
consumer acceptability and economical production should be consid­ high-pressure homogenization and microfluidization, but other methods
ered (Fathi et al., 2018). as ultrasonication and in-situ emulsification are applied as well (Peng,
Protein-based delivery systems show advantages including low Meng, et al., 2019).
toxicity, stability, the possibility of surface modification of protein Advantages of using nanoemulsions include that they can show high
nanoparticles, and easily biodegradability. The demerits of proteins or kinetic stability against flocculation and creaming, possess somewhat
peptides used for delivery of nutraceuticals include low bioavailability, thermodynamic stability and potential application for personal and
rapid metabolism, conformational changes, non-covalent complexation health care products and transport easily through the cell membrane
with blood products, opsonization, and metabolic liability which using which leads to increase their bioavailability, biocompatibility, the sur­
different encapsulation techniques can be overcome to their application face groups can be conjugated with targeting ligands. Although, disad­
limitations as nano-delivery systems. vantages of using nanoemulsions include the high cost of production and
Table 1 presents an overview of protein-based nanocarriers used for finding a food grade surfactant and also they are almost thermody­
the encapsulation of different nutraceuticals using various encapsula­ namically unstable systems due to different factors including environ­
tion techniques. mental factors and intrinsic factors such as coalescence, creaming,
Ostwald ripening, flocculation, etc., immediate bioactive compounds

4
L. Rashidi Food Bioscience 43 (2021) 101258

Table 1
Overview of bioactive compounds encapsulated by protein - based delivery systems.
Encapsulating Materials Nutraceutical Study/Target Encapsulation Size (nm) Encapsulation References
Technique Efficiency (%)

Zein NPs./Fucoidan & Curcumin Characterization of NPs, Release in Antisolvent – 81.07 Zhang et al. (2021)
Sulfated simulated gastrointestinal digestion precipitation
polysaccharide
Zein Nanofibers Ascorbyl Characterization of nanofibers, Electrospinning – 22.5–65.5 Miri, Habibi Najafi,
palmitate (AP) Increase of antioxidant activity of AP Movaffagh, and Ghorani
(2021)
Cationic 30 mer alpha tri- Cell viability, Immunocytochemistry Condensation 43 – O’Doherty et al. (2020)
amphipathic peptide calcium staining in MG-63 cells,
(RALA) phosphate Mineralization in porcine
(α-TCP) mesenchymal stem cells (PMSCs)
Zein nanofibers: Carvacrol Increase thermal & Mechanical Electrospinning 103 & 17 40 ± 1.9 to 81 ± Altan and Cayir (2020)
Tributyl citrate & stability, low water barrier properties 1.7
Tween 80
Gelatin NPs: porcine Buriti Oil Antimicrobial activity on Emulsification 51 OPG:86.80 Castro, Passos, et al. (2020)
gelatin (OPG) & Pseudomonas aeruginosa, Klebsiella followed by Freeze OAG:7191
sodium alginate pneumonia, and Staphylococcus drying
(OAG) aureus
L-arginine (Arg)- or L- Curcumin Characterization of NPs, In vitro Self-assemble 82–110 Curcumin: Du et al. (2020)
lysine (Lys)- Egg white release in simulated gastrointestinal, 81–91,
functionalized derived peptide Oral delivery EWDP:35–74,
chitosan–casein NPs (EWDP) Arg–CS–CA;
Curcumin:
76–87,
EWDP:48–87,
Lys–CS–CA Ps
Gelatin Nanofiber Curcumin Characterization of nanofiber, Sequential 184–236 89.05–92.57 Wang, Li, Zhang, Feng, and
Release from gelatin film, antioxidant electrospinning Zhang (2020)
activity of released curcumin during
96 h
Whey Protein NPs Olive Leaf Characterization of NPs & EE% Electrospraying 232.3–659.8 36.66–83.66 Soleimanifar, Jafari, and
Extract Assadpour (2020)
Zein NPs β-carotene Characterization of NPs & EE%, Electrospray 599–906 61.37–81.76 Mahalakshmi, Leen, Jeyan,
Release in gastrointestinal Drying and Anandharamakrishnan
(2020)
Gelatin NPs (GNPs) Genistein (GEN) Stability at pH = 2, EE% Modified two-Step 273–391 90 Song et al. (2019)
& Icariin Desolvation
Nanogels & Lysozyme Tea polyphenols DPPH and ABTS-scavenging rates of Self- Assembly 420–720 89.05 ± 3.14 Liu, Zhang, Kong, Zhang, and
(Ly) & (TP) the TP-loaded NGs, Anticancer Yang (2019)
Carboxymethyl activity on HepG2 cells
Cellulose (CMC)
Albumin Micelle Curcumin Characterization of NPs/Cytotoxicity Micelle 22.1 98.2 Motevalli et al. (2019)
effects on NPs on MCF-7 cell line
The hydrophobic cores Curcumin & Characterization of NPs, Release in Anti-solvent 100–200 Curcumin: 96.2 Yan et al. (2019)
of Zein NPs & EGCG simulated gastrointestinal tract, EGCG: 77.9
Caseinate shell DPPH radical scavenging capacity of
the NPs
Zein NPs Saffron Characterization of NPs, Stability of Electrospun/ – 74–97 Alehosseini,
encapsulated extract at UV light, & Electrospray Gómez-Mascaraque, Ghorani,
different pH (2, 7.4) and López-Rubio (2019)
Casein micelles or EGCG & folic Characterization of NPs, EE%, Self-assemble,/ 66.2 85 Malekhosseini et al. (2019)
Casein NPs acid Release in acidic & alkaline media, Anti-solvent
Stability in Thermal treatment
Casein micelles Quercetin The effect of pH & Concentrations of Self-assemble 182.2–334.4 more than 97 Ghatak and Iyyaswam (2019)
casein & additives & CTAB, EE%
β-Lactoglobulin NPs Quercetin Characterization, Digestion by Anti-solvent 180–298 13.9 Chavoshpour-Natanzia and
proteases, Release in simulated Sahihi (2019)
gastrointestinal tract,& colon &, EE%,
Release Kinetic model

release, and instability upon storage (Peng, Meng, et al., 2019). lipophilic nutraceuticals containing a core matrix (including waxes,
fatty acids and acylglycerols) and are stabilized by surfactants (such as
4.2.2. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers sphingomyelins, phospholipids, bile salts and sterols). In comparison
(NLCs) with other lipid-based nanocarriers, including nanoemulsions and li­
The lipidic phase of nanostructured lipid carriers contains the solid posomes, SLNs possess advantages such as robust protection against GI
lipid (fat) and liquid lipid at room temperature. The solid lipid nano­ tract degradation, stability to labile hydrophobic compounds, and pro­
particles (SLNs) are lipid-based delivery systems able to solubilize vide controlled release profiles of a given nutraceutical (Nandvikar,

5
L. Rashidi Food Bioscience 43 (2021) 101258

Table 2a
Overview of bioactive compounds encapsulated by lipid – based systems (Nanoemulsions).
Encapsulating Materials Nutraceutical Study/Target Encapsulation Size (nm] Encapsulation References
Technique Efficiency (%]

Nanoemulsions (O/W): WPI, Resveratrol Chemical stability, Effect of oil High Pressure 540–570 98 Cheng, Zhang, Li, Duan,
Gum Arabic, Sunflower, type Homogenization and Liang (2021)
Origanum, & Peppermint Oils
Nanoemulsion (O/W): Oleic Zataria Antiparasitic effect Ultrasonic 73–82 – Yazdi, Haniloo, Ghaffari,
acid &Tween 80 multiflora Homogenization and Torabi (2020)
essential oil
Nanoemulsions (O/W): BSG, Vitamin D3 & Characterization of Ultrasonic 115–385 – Gahruie et al. (2020)
WPC, Tween 80, Canola oil Saffron petal nanoemulsion, In vitro release Homogenization
extract
Nanoemulsions (W/O/W): Mentha piperita Characterization of Homogenization 108.66, 80.5, 88.7, & 90.8 (Roshanpour et al.,.
chitosan, Alyssum extract (MPE) Nanoemulsions, EE%, Release, 70.81, & 2020)
Homolocarpum gum (AHG) Oxidative stability of soybean oil 65.18
& complex of Chitosan & contains W/O/W/MPE
AHG (CCA), Span 80,
Soybean oil
Nanoemulsions (O/W): Tween Vitamin D3 (Vit Effects of storage & Ultrasonic 119.3 – Mehmood and Ahmed
80 & Soya-lecithi-based food- D) Temperature, Oxidative Stability Homogenization (2020)
grade canola oil contains O/W/Vit D
Nanoemulsions (W/O/W): Ferula persica Characterization, EE%, The Homogenization 76.61 ± 0.3 64.1–93.3 Estakhr, Tavakoli,
Locust bean gum & Chitosan, extract (FPE) biological effect of FPE to 147.91 ± Beigmohammadi, Alaei,
Soybean oil, Span 80 0.4 and Mousavi Khaneghah
(2020)
Nanoemulsions (O/W): Hyssop extract The effect of O/W/HE on Ultrasonic 56.713 ± 88.36 ± 2.4 Savadkouhi, Ariaii, and
Lepidium perfoliatum seed (HE) oxidative stability of Soybean oil Homogenization 1.48 Charmchian Langerodi
gum & Orchis mascula, (2020)
Soybean oil,
Nanoemulsions (O/W): Tea β-Carotene (BC) In vitro digestion assay, Stability High-Pressure lower than – Meng et al. (2019)
polyphenols-β-carotene (TP- of O/W at different Homogenization 100
BC), Polysorbate 80 temperatures, In vivo absorption
Nanoemulsions (O/W): Capmul Vitamin E Narrow beam, Muscular Homogenization 38.70 ± 82.71 ± 9.14 Gaba et al. (2019)
MCM as oil, & Tween 80 coordination, Grip strength, 3.11
Forced swimming tests, &
Akinesia test in a rat model
Nanoemulsions (W/O/W): Gallic acid Stability during storage, Homogenization 98–577 & – Gharehbeglou, Jafari,
Double Emulsions Pectin- Characterization of W/O/W 100–1520 Hamishekar, Homayouni,
WCP, PGPR, Tween 80 and Mirzaei (2019)
Nanoemulsions (W/O/W): Arbutin & Creaming stability & Sensory Homogenization 355.98 ± Arbutin: 91 ± 3 Huang, Belwal, Liu,
Arbutin, Gelatin, Hydrophobic evaluation beverage, Release 10.44 Coumaric acid: Duan, and Luo (2019)
Polyglycerol Polyricinoleate Coumaric acid kinetics in simulated 82 ± 2
(PGPR) gastrointestinal
Nanoemulsions (O/W): Birch Leaf The creaming stability, Homogenization – 80–90 Pulit-Prociak, Kabat,
Cholesterol, Soy Lecithin & Extract characterization of Węgrzyn, Zielina, and
Water nanoemulsions, EE% Banach (2019)
Nanoemulsions (O/W): WPI, Curcumin The encapsulation stability, Ultrasonic PE:359 ± PE:78 Carpenter, George, &
sodium alginate, olive oil, antioxidant activity, release Homogenization 5.6 to SE:100 Saharan. (2019)
Primary emulsion (PE) & properties of curcumin, SE:841 ±
secondary emulsion (SE) Characterization 13.4
Nanoemulsions (W/O/W): Basil Rosemary Antioxidant properties, Homogenization 154.69, 58.71, 66.39, & Rashidaie Abandansarie,
seed gum, Soy-bean protein extract (RE) Antibacterial activity, The 126.48, & 61.35 Ariaii, and Langerodi
isolate & combination of amount of phenolic compounds 139.52 (2019)
them of W/O/W/RE
Nanoemulsions (O/W): Gum Fish oil Effect of rosemary extract Ultrasonic 36.67–82 87.17 Moghadam, Pourahmad,
Arabic, Tween 80 encapsulated nanoemulsion on Homogenization Mortazavi, Davoodi, and
antimicrobial and antioxidant Azizinezhad (2019)
properties of rosemary and shelf
life of meat
Nanoemulsions (O/W): CaCl2, Resveratrol Effect of protein stabilized O/W High-pressure 130–550 63–74 Zhang, Fan, Li, Chen, and
WPI, Gum Arabic, Sunflower, on oxidative stability of homogenization Liang (2019)
Tween 80 sunflower oil

6
L. Rashidi Food Bioscience 43 (2021) 101258

Table 2b
Overview of bioactive compounds encapsulated by lipid – based systems (SLNs, & NLCs).
Encapsulating Materials Nutraceutical Study/Target Encapsulation Size (nm) Encapsulation References
Technique Efficiency (%)

NLCs:Precirol ATO 5, Olive oil, Oleuropein Improve antioxidant Hot Melt 150 99.12 Huguet-Casquero,
Tween 80 efficiency, In vitro assay on Homogenization Moreno-Sastre,
A549 & CuFi1 Lung López-Méndez, Gainza,
Epithelial Cell Lines and Pedraz (2020)
SLNs: Precirol ATO, Pluronic Curcumin Curcumin bioavailability, In Precipitation 159–302.5 68.62 Minafra et al. (2019)
F68, & Dimethyldioctadecyl- vitro Cytotoxicity effect on
Ammonium Bromide breast cancer cell lines
NLCs: Glyceryl Monooleate, Curcumin Release pattern, photo- High Shear Hot 101.80 ± 47.11 & 51.55 Kamel, Fadel, and Louis
Geleol, Glyceryl Dibehenate, cytotoxic anticancer effect Homogenization 1.41 to (2019)
Glyceryl Distearate, Tween 80, on breast cancer cells 265.70 ±
& Olive oil 19.9
SLNs: Trilaurin, Tristearin, Curcumin Characterization of SLN, EE Cold Dilution of 169.8 ± 4.7 78 ± 1.3 to Chirio et al. (2019)
Myristic acid, Glyceryl %, Cytotoxicity effects on Microemulsion to 215.3 ± 70.±2.1
Monostearate, Ethyl Acetate, pancreatic adenocarcinoma 5.5
Benzyl Alcohol, Triacetin, cell lines, In vivo study on
Butyl lactate, & polysorbate Wistar healthy rats
SLNs: Cocoa Butter, Stearoyl-2- Epigallocatechingallate Degradation during storage Hot 108–122 68.5 Shtay, Keppler,
Lactylate (SSL) & Mono- & & digestion under simulated Homogenization Schrader, and Schwarz
Diglycerides, Lecithin gastrointestinal pH (2019)
(Phospholipon® 90G) conditions

Lala, & Shinde, 2019) . Disadvantages of using SLNs in the food industry NLCs have advantages for overcoming the shortcomings of SLNs, which
include loss of bioactivity of heat-labile compounds during their prep­ include high loading capacity of bioactive compounds, preventing par­
aration, low encapsulation load, the possibility of explosion during ticle aggregation and also happening of leakage via crystal trans­
storage. The NLC produces by mixing various lipid molecules including formation during storage, preparation, and processing (Nandvikar et al.,
solid lipids with liquid lipids on the basis of preparation methods applied 2019).
for SLN. In NLC, the encapsulation of nutraceuticals increases and
expulsion phenomena during storage is limited by inhibition of perfect 4.2.3. Liposomes
crystals formation. Advantages of NLCs include possibility of targeted Liposomes are ideal carriers consisting of an aqueous core sur­
and sustained nutraceutical release, improved physical stability, easier rounded by natural or synthetic phospholipids applied for hydrophobic
to validation, the possibility free of using solvent in the process, appli­ and hydrophilic nutraceuticals. Hydrophobic nutraceuticals are gener­
cable for both hydrophilic and hydrophobic nutraceuticals, increase of ally entrapped within lipid bilayer, and hydrophilic nutraceuticals are
loading capacity, biodegradability, and biocompatibility of used lipids likely to be entrapped within the core. Due to the low stability of lipo­
in NLCs. Disadvantages of NLC include the burst release and low sta­ somes during administration and storage, liposomes with modified
bility against aggregation, irritation and sanitizing action of some sur­ surfaces by several molecules, including surfactants, have been devel­
factants. In Table 2b, recent studies in which solid lipid nanoparticles oped and applied for both food and pharmaceutical industries (Zhou
(SLNs) and nanostructured lipid carriers (NLCs) were applied for the et al., 2021).
encapsulation of nutraceuticals are presented. Both SLNs and NLCs are Emulsification, pressure extrusion, thin-film dispersion, pH gradient,
lipid-based nanocarriers, which are very suitable for food applications high-pressure homogenization, centrifugation, reverse evaporation,
since no organic solvent is used in their making (Chirio et al., 2019). melting, and freeze-drying technologies are applied for the preparation
There are three types of NLC structures based on the formulation of liposomes. Liposomes are biocompatible and bioavailable carriers,
composition and the production parameters including imperfect, and external factors, including temperature, light, enzymes’ presence,
amorphous, and multiple types. Methods for NLCs or SLNs production and pH can liberate the sudden release of their content by interfering
include microemulsion technique, hot and cold homogenization with membrane stability, causing lipid layer disruption and quick drug
method, emulsification solvent diffusion method, solvent release, non-toxic, encapsulation both of hydrophilic and hydrophobic
emulsification-evaporation method, solvent injection method or solvent bioactive compounds, increase solubility of nutraceuticals. Disadvan­
displacement method, phase inversion technique, membrane contractor tages of liposome application include low encapsulation yield and the
technique, and multiple emulsion method (Chirio et al., 2019). physical and chemical instability, low nutraceutical transport rate, dif­
Advantages of the use of SLNs include biocompatibility, bioavail­ ficulty of binding ligands to the surface, rapid clearance of mononuclear
ability, ease of scale-up, good tolerability, avoidance of the organic phagocytic system (Zorkina et al., 2020). Nanoliposomes are bioavail­
solvents usage during preparation, high loading capacity, controlled able and biocompatible and can be used for both hydrophobic and hy­
release, and physical stability. Disadvantages of SLNs are their tendency drophilic compounds and protected them from enzymatic destruction in
for particle aggregation, easy clearance by the reticuloendothelial, the body. Disadvantages of nanoliposomes are included expensive lipid
bioactive components leakage after polymorphic conversion during components and instability of lipid components in acidic environments
storage, uncertain diffusion of the bioactive agents in the lipid matrix of (Zorkina et al., 2020). Table 2c presents an overview of nutraceuticals
the vector, unpredictable gelation tendency, and low loading capacity. encapsulated by the liposome carriers.

7
L. Rashidi Food Bioscience 43 (2021) 101258

Table 2c
Overview of bioactive compounds encapsulated by lipid – based deliver systems (liposomes).
Encapsulating Materials Nutraceutical Study/Target Encapsulation Size (nm) Encapsulation References
Technique Efficiency (%)

Liposomes: Rhamnolipid, Curcumin Study release & stability electrostatic 412.9 to – Zhou et al. (2021)
phospholipids, chitosan interaction 639.8
Liposome: Phospjatidylcholine, Stingless bees Physicochemical Thin-film hydration 275.5 66.9 Ramlia, Alib,
Cholesterol Propolis extract characterization of Hamzah, & Yatim,
liposome (2021).
Liposomes decorated by chitosan: Egg Cinnamaldehyde Antibacterial efficiency Ethanol injection 400–600 38–50 Wang et al. (2021)
yolk phospholipid, Peptone, Beef
extract, Tween 80
Liposomes, Transfersomes & Sambucus Ebulus Characterization of Thin-Film 49 ± 1.52 to 75–85 Păvăloiu et al.
Ethosomes: Phosphatidylcholine, leaves extract liposomes, EE%, In vitro Hydration, 190 ± 2.53 (2020)
Cholesterol, & Sodium cholate (SELE) release Ultrasound
Treatment &
Extrusion; & Cold
Method
Liposome: WPC, Pectin, & Sodium Trifolium Characterizations of six Thin Film Hydration 282.5–491.2 85–91 Sayyed-Alangi and
Azide Resupinatum Sprout formulations of Nematzadeh (2019)
Methanolic Extract liposomes
Lipoid S100-liposomes & Cholestrol Estragole, Characterization of Ethanol injection – More than 90 % (Hammoud,
eucalyptol, liposomes, EE%, Gharib,
isoeugenol, Release, Stability Fourmentin,
pulegone, terpineol, Elaissari, &
& thymol Greige-Gerges,
2019)
Liposome: Chitosan coated Liposomal: Vitamin D3 & Enhance Microfluidic 90 and 250 88.4 & 94.7 (Dalmoro et al.,
Phosphatidylcholine, Choline Basis, vitamin k2 Mucoadhesiveness & 2019)
Cholesterol stability of liposmes
Liposome: Lecithin, Phenolic- Pistachio Hulls The phytochemical Thin Film Hydration 96.3 ± 7.18 38.4 ± 2.67 Oskoueian et al.
Enriched Fraction analysis (2019)
Liposome: Phospholipids from Rice Quercetin Characterization, EE%, Thin Film & 157.33 ± 84.92 ± 0.78 (Rodriguez,
Bran, Soy Lecithin & Egg Yolk Storage stability, Sonication 23.78 Almeda, Vidallon,
Release in simulated & Reyes, 2019)
gastrointestinal fluid
Liposome: Cholesterol, Soybean Ascorbic acid In vivo efficiency, Drug Thin Film Hydration 161 ± 1 to 17.8 ± 0.6 to 58.1 (Maione-Silva et al.,
Phosphatidylcholine, 1,2-Dioleoyl- flux, Stability 190 ± 3 ± 4.0 2019)
3-Trimethylammoniopropane &
1,2-Distearoyl-sn-glycero-3-
phospho-(1′ -rac-glycerol)
Liposome: β-Lactoglobin-Liposome Vitamin A In vivio bioavailability Thin Film Dispersion – With β-Lg: 50.17 (Rovoli, Pappas,
Complex: in mice, EE%, & Freeze Drying Without β-Lg: Lalas, Gortzi, &
Phatidylcholine (PC from egg yolk)/ characterization of 50.64 Kontopidis, 2019)
Cholesterol (CH) with the molar liposomes
ratios: PC/CH
Liposome: Lecithin, Cholesterol, & EGCG & quercetin Characterization & EE% Thin Film Hydration 111.10 ± 64.05 ± 1.56 Chen et al. (2019)
Tween 80 0.52
Liposome: Lipids- Curcumin Sustained & prolonged Thin Film Hydration 203.02 ± 4.1 76.48 ± 1.2 to Choudhary,
Phosphatidylcholine, & Cholesterol curcumin release at pH to 395.55 ± 84.92 ± 0.57 Shivakumar, and
7.4, in vivo formulation 3.2 Ojha (2019)
skin penetration
Liposome: Soy Phosphatidylcholine & Sea Fennel Aqueous Characterization, EE%, Freeze-Dried 89.5 to 137.1 65.6 and 49.1 for Alemán, Marín,
Cholesterol & Ethanolic Extract The liposomal & 96.3 to aqueous and Taladrid, Montero,
antioxidant activity & 150.1 ethanolic extract, and Carmen
Total phenolic content respectively Gómez-Guillén
(2019)
Liposome: Sunflower Lecithin Curcumin, Impact of pH, Solubility High- Pressure – 100, 54 & 93 Peng, Zou, et al.
(Sunlipon 65) & Cholesterol Quercetin, & & Chemical stability, EE Homogenization (2019)
Resveratrol %,

4.3. Carbohydrate-based delivery systems high-temperature stability in comparison with lipid or protein-based
delivery systems due to the possibility of their denaturation or
Carbohydrates consist of polysaccharide, oligosaccharide and melting. They are classified by their isolation methods, physical,
monosaccharide are natural biocompatible, bioavailable, the suitable chemical, or biochemical modifications, and biological origins including
shell under high temperature process, inexpensive food ingredients, and plant (e.g. pectin, cellulose, starch, and guar gum), animal (e.g. chito­
biodegradable materials, which can interact with different kinds of san), microbial (e.g. xanthan, cyclodextrins, and dextran), and algal
bioactive compounds via their functional groups, thus entrapping wide origin (e.g. carrageenan and alginate). Also, different methods including
varieties of hydrophilic and hydrophobic bioactive compounds’ in­ coacervation, electrospinning, electrospray, supercritical fluid, spray
gredients (Kim, Lee, & Lee, IN PRESS). Therefore, different kinds of drying, emulsion-diffusion, emulsion-droplet coalescence, reverse
carbohydrates and their combinations or chemically modified forms micelle, emulsification/solvent evaporation, salting out, high-pressure
have been applied for assembling nanodelivery systems, including cel­ homogenization, and ultra-sonication are applied for assembling
lulose, chitosan, alginate, native gums, cyclodextrins, pectin, guar gum, carbohydrate-based nanocarriers (Assadpour & Jafari, 2019).
dextrin, and starch. In addition, they can be used as a suitable shell for Natural starch is the famous polysaccharide in plants, consisting of
the encapsulation of drugs or nutraceuticals due to their glucose units linked to α-d-(1 → 4) and/or α-d-(1 → 6) linkages, which is

8
L. Rashidi Food Bioscience 43 (2021) 101258

Table 3
Overview of Bioactive Compounds Encapsulated by Carbohydrate- Based delivery systems.
Encapsulating Materials Nutraceutical Study/Target Encapsulation Size (nm) Encapsulation References
Technique Efficiency (%)

Quinoa & maize starch NPs Rutin Characterization, EE%, Simulated in Ultrasonication 107 and 222 67.4 & 26.6 Remanan and Zhu
vitro digestion (2021)
Chitosan NPs: Chitosan, & Red Ginseng Extract Antithrombotic activities, Ex-vivo & Ionic Gelation 286 ± 36.6 20.78 ± 0.20 Kim et al. (IN PRESS)
TPP Solution In vivo antiplatelet activities, In vivo
carrageenan-induced mouse tail,
Arteriovenous shunt rat thrombosis
models
Gum Arabic (GA)-based Probiotic: Viability of GA/PUL-encapsulated Electrospinning 283.15 ± – Ma et al. (2020)
nanofibers in combination Lactobacillus Lactobacillus & Storage stability 68.44 to
with Pullulan 61.05 ±
16.67
Chitosan/Gum Arabic The Antioxidant In vitro digestion, EE% Ionic Gelation CSGA: 273.2 41 to 45.3 Jeong, Lee, and Lee
(CSGA) & Chitosan/ Combination ± 23.3 to (2019)
Carrageenan (CSCR) NPs (Blueberry & 327.2 ± 23.8
Cabbage CSCR: 194.8
Concentrates) ± 14.9 to
468.9 ± 42.0
Azivash (Corchorus olitorius. Catechin Simulated gastric fluid & Simulated Electrospinning 500–1000 98.98–84.48 Hoseyni et al. (2021)
L) gum-polyvinyl alcohol intestinal fluid, EE% Process
Maltodextrein NPs Green Coffee Characterization NPs, In vitro Spray Drying 40-70desai 82.34 Desai et al. (2020)
bioactivity of NPS, Storage stability
Chitosan NPs: Chitosan, & Curcumin Oral bioavailability/Antiviral Complexation 332.4 ± 9.4 77.2 ± 3.6 Ng et al. (2020)
TPP Solution effects, Cytotoxicity effect on
Crandell-Rees feline kidney (CrFK)
cells
Nanoparticles: chitosan (CS) Saffron Bioactive Characterization NPs, EE%, Release Ionic Gelation 183–295 29.12 to 52.34 Rajabi, Jafari,
& Gum Arabic (GA) Components of Saffron in acidic and natural (IG) Rajabzadeh, Sarfarazi,
media and Sedaghati (2020)
Nanofibers: Poly (ethylene Aloe Vera skin Thermal stability & antioxidant Electrospinning 185 ± 25 to 76–105 Solaberrieta, Jiménez,
oxide) solutions/Mucilage extract activity, EE%, 250 ± 30 Cacciotti, and
Nanofibers Garrigós (2020)
Nanofibers: Basil Seed Hesperetin (HSP) Characterization of nanofibers, Electrospinning 82–320 89–96 Kurd, Fathi, and
Mucilage/ Release models, EE% Shekarchizadeh
Polyvinylalcohol (2019)
Chitosan NPs: Chitosan, & Java Tea Anti-hyperglycemic, Spray Dryer 217.7 – Naibaho, Safithri, and
TPP Solution (Orthosiphon Malondialdehyde level analysis in Wijaya (2019)
Aristatus BlMiq) vitro
Nanoparticles: Sodium Clove Essential Oil EE%, Antimicrobial Assessment, in Homogenization – 83.60–92.2 Radünz et al. (2019)
Alginate, Glycerol vitro inhibitory & bactericidal
Monostearate or Poly activity
Oxyethylene Sorbitan
Monolaurate
Chitosan NPs: Chitosan, & Physalis Alkekengi-L Antioxidant capacity & Anticancer Ionic Gelation 196 ± 20 95 ± 3.2 Mahmoudi et al.
TPP Solution Extract activity, EE% (2019)
Maltodextrin NPs Grape Pomace The effect of NPs/GPE on Spray Dryer – 82.81 ± 1.15 Dordoni et al. (2019)
Extract (GPE) antioxidant properties & Sensory
acceptability of whole wheat cocoa
biscuits, EE%
Starch NPs Catechin Hydrate Characterization of NPs, Release in Sonication & 322.7, 559.2 59.09, 48.30, & Ahmad et al. (2019)
simulated in vitro digestion, EE% Freeze Dryer & 615.6 55.00
NPs: Balangu (Lallemantia Mentha Longifolia L. Increase their utility as flavoring and Electrospray 96.53 ± 3.41 81.54–87.82 Rezaeinia, Ghorani,
Royleana) Seed Gum Essential Oil bioactive agents in foods and Emadzadeh, and
Nanocapsules (Balangu beverages, Release models, EE% Tucker (2019)
Seed Gum & Polyvinyl
Alcohol

biodegradable, digestible polymer and biocompatible, consist of connected α-d-(1 → 4) galacturonic acid residues, which is resistant to
amylose and amylopectin and is hydrophilic, which limits its application enzymatic degradation in the stomach and mouth but it is digestible by
for hydrophobic nutraceuticals but its hydrophobic derivatives such as the microflora and can be suitable for delivery of the acid-sensitive food
dialdehyde starch and propyl starch developed for encapsulation of (Assadpour & Jafari, 2019).
lipophilic bioactive compounds. The main limitation of starch for Guar gum is derived from seeds of Cyamopsis tetragonolobus and is
encapsulation of bioactive compounds is its sensitivity to acid media and water-soluble consists of linear chains of β-d-(1 → 4) mannopyranosyl
amylase hydrolysis. Cellulose is another abundant polysaccharide units with α-d-galactopyranosyl units connected by (1 → 6) linkages
composed of glucose linked with β-d (1 → 4) linkages, which has low- which is resistant to enzymatic degradation in the small intestine,
water solubility and large dimensions but using cellulose-based nano­ stomach and mouth and degrades in the colon. Its highly viscous solu­
carriers due to their non-digestible by the colon or microbial enzymes tions limit its usage which its modification should be considered. Also,
should be taken into account for their absorption and potential toxic poor hydration and low thermal stability are other limitations of using
impacts (Assadpour & Jafari, 2019). guar gum as nanocarriers (Ng, Selvarajah, Hussein, Yeap, & Omar,
Pectin as a linear anionic polysaccharide consists of linearly 2020).

9
L. Rashidi Food Bioscience 43 (2021) 101258

Chitosan is a biodegradable, cationic, and biocompatible polymer methylcellulose (FA/HPMC) using spray drying method, and encapsu­
which is obtained from alkaline deacetylation of chitin, has antimicro­ lation efficiency percent (EE%) 80.12 ± 1.57, and 85.56 ± 6.01 (Yu,
bial and antioxidant activities, possible prolonging resistance time, Roh, & Park, 2021); curcumin by a complex of an insect protein as
possibility of physical or chemical modifications to extend biopolymer mealworm protein and chitosan (particle size: 143–178 nm) using ho­
functional characteristics and enhancing sustained release in gastroin­ mogenization followed by freeze dried methods and 30 to 47 EE%
testinal media. Alginate is obtained from brown sea algae and consists of (Okagu et al., 2020); curcumin by a complex of ovalbumin (OVA) and
alternating blocks of 1 → 4 connected to β-D-mannuronic acid and α-L- sodium alginate (particle size: 300–330 nm) using the homogenization
guluronic which can be applied for encapsulation of hydrophilic technique with 70–80 EE% (Feng et al., 2019); Epigallocatechin gallate
bioactive compounds (Ng et al., 2020). Dextran consists of chains of by a complex of piperine into a zein nanocarrier (particle size: 34.03 ±
glucose α-(1 → 6) and the small amount of α-(1 → 3) linkages containing 3.1 nm to 83.49 ± 7.2 nm) using anti-solvent precipitation technique
hydroxyl groups, which can be applied for the covalent attachment of (Dahiya, Rani, Dhingra, Kumar, & Dilbaghi, 2018); folic acid by a
different organic functional groups. It can be applied for encapsulation complex of 7 S and 11 S globulins and folic acid (FA) using the
of bioactive compounds with various hydrophobicities (Desai, Stanley, complexation technique (particle size: 19.57 ± 1.2 to 51.33 ± 1.3 nm)
& Murthy, 2020). Cyclodextrins consist of 6, 7, or 8 (α, β or γ-CDs) (Ochnio et al., 2018).
glucose units liked by α-(1 → 4) glucoside bonds, which central cavity is
hydrophobic and the external surface is hydrophilic. CDs can be useful 5. Non-food grade delivery systems
for the encapsulation of temperature-sensitive, poorly soluble and
chemically labile bioactive compounds (Desai et al., 2020).. 5.1. Polymeric nanocarriers
Table 3 presents an overview of bioactive agents or nutraceuticals
encapsulated by carbohydrate-based nanocarriers. Polymeric carriers (including poly (D, L-lactide), poly (lactic acid),
PLA, poly (D, L-glycolide), PLG, poly (lactide-co-glycolide), PLGA, and
4.4. Nanocomplexation poly (cyanoacrylate), PCA) can be used to entrap both hydrophilic and
hydrophobic nutraceuticals. They should have proper biocompatibility,
The complexation method is a proper nanoencapsulation system for biodegradability, mechanical strength, The Food and Drug Administra­
the conservation of bioactive food ingredients or nutraceuticals, which tion (FDA), 2014 (FDA) approval, and low synthesis complexity. The
traps the core materials in an assembly of two biopolymers (Desai et al., poly (lactic-co-glycolic acid) (PLGA) which is a copolymer of the poly
2020). Generally, biopolymers are organized by linking proteins and (lactic acid) (PLA) and poly (glycolic acid) (PGA) is the most attractive
polysaccharides through electrostatic absorptions that contain better one (Zorkina et al., 2020). Nanocarriers based on polymeric micelles
chemical and colloidal conservation. Therefore, the net positive charge have certain advantages, including low toxicity, small size, potential
of protein interplays with the anionic groups upon the polysaccharide at application for the treatment of various cancers, greater mechanical and
pH values less than the protein’s isoelectric point (pI). The biopolymer chemical stability, biocompatibility, ease of production, sustained
complexation is commenced using the layer-by-layer method by the release of nutraceuticals, ease of modification, and increase of repro­
adsorption of ionic polysaccharides onto pre-organized protein nano­ ducibility. Also, polymeric nanoparticles as the drug or bioactive agent
particles (Rajabi et al., 2020). The stability of nanocomplexation carriers delivery system can be used for the localized or targeted delivery sys­
depends on several factors, including ionic strength, pH value, confor­ tems of a drug or nutraceutical to specific tissues or organ sites with an
mation, charge density, the ratio of the biopolymers, and the concen­ optimal release rate (Zorkina et al., 2020). They are synthesized by
tration of both polysaccharides and proteins (Okagu, Verma, different techniques, including solvent evaporation, salting out, dialysis,
McClements, & Udenigwe, 2020). Moreover, the complexation of car­ and supercritical fluids. Disadvantages of synthetic polymers are the
bohydrates or proteins with the phenol compounds has widely been possibility of their uncertain potential toxicity and some of them are
studied for improved stability and delivery of those compounds, non-degradable or have slow degradability, coupled with a complex and
particularly those with low solubility and bioavailability. Advantages of costly production of them. In addition, the efficiency delivering of
using these complexes include the biodegradability, lower toxicity, and nutraceuticals by polymeric micelles across the intact BBB still needs
biocompatibility with cells. In recent years, numerous studies were more investigation. Table 4 presents the nutraceuticals encapsulated by
conducted for the encapsulation of nutraceuticals, including ferulic acid polymeric nanocarriers (Zorkina et al., 2020).
by the complexes of maltodextrin (FA/MD) and hydroxypropyl

Table 4
Overview of bioactive compounds encapsulated by polymer-based delivery systems.
Encapsulating material Nutraceutical Study/Target Encapsulation Size (nm) Encapsulation References
technique efficiency (%)

PEGylated poly (lactide-co- Sophorolipid In vitro & In vivo model Nanoprecipitation PLGA: 196.5 ± 68.8 ± 6 in PLGA to (Haggag et al.,
glycolide), Resomer® RGPd tumor cell lines (CT26 20.9117.5 ± 92.18 % ± 1.8 % in 2020)
50,105 NPs murine colon carcinoma) & 11.5 in 10 % 10 % PEG-PLGA
the effect of peripheral PEG-PLGA
hydrophilicity
Poly (lactic-co-glycolic acid) 3, 3′ -Diindolylmethane Cytotoxicity effect on Double Emulsion/ 180–210 80 (Mousa, El-Far,
(PLGA) & Poly (ethylene (DIM) & Ellagic Acid pancreatic cancer cell Solvent Evaporation Saddiq, Sudha,
glycol) (PEG) [PLGA-PEG] (EA) (suit2) implant model & Mousa, 2020)
NPs
Poly (lactic-co-glycolic acid) Callistemon Citrinus & Cytotoxicity effects on Nanoprecipitation 200–250 80 (Ahmed et al.,
NPs Berberine Extract MDA-MB231, MCF-10 A, 2019)
MCF-7breast cancers
Curcuminoids NPs: Curcuminoids In vitro modulation Nanoprecipitation Smaller than – (Santos et al.,
(Polyvinylpyrrolidone (PVP)) Extracted From capacity on the enzymes, 450 2019)
Curcuma Longa L. Cytotoxicity effect on tumor
& non-tumor cells

10
L. Rashidi Food Bioscience 43 (2021) 101258

Table 5
Overview of bioactive compounds encapsulated by silica –based delivery systems.
Encapsulating materials Nutraceutical Study/Target Size (nm) Encapsulation References
efficiency (%)

Pegylated lipid bilayer coated MSNs Curcumin Characterization of NPs, 115 ± 15 30.70 ± 3.56 Lin et al. (2018)
Cytotoxicity effect on CHO
FA-MSN-N–
–C (folic acid conjugated to Curcumin Stability under physiological 136 ± 4 to 165 49.6 ± 1.9 Chen, Sun, Wang, Wang, and
MSN-NH2) conditions, internalization into ±4 Wang (2018)
MCF-7 cells, & HEK-293 T
normal cell
Polyethyleneimine coated MCM-41 Curcumin Characterization of mcm-41, 100–200 – (Harini et al., 2017)
Anticancer property on MCF-7
cells, Release of curcumin at pH
3
MCM-48 particles Resveratrol Cytotoxicity effect on human 90, 150 & 300 – Juère et al. (2017)
colon carcinoma cell
monolayer (Caco-2), Anti-
inflammatory
Carboxyl Group-MSNs (PSNs-C) Vitamin B12 & Cisplatin Characterization of PSNs-C, 316 ± 6 – Thepphankulngarm,
Release at pH 5.5 Wonganan, Sapcharoenkun,
Tuntulani, and Leeladee
(2017)
Amino Functionalized MSNs Vitamin E (VE) characterization of MSNs, LMSN:282 VE@NH2-MSN: Zhuoxian et al. (2017)
LMSN,NH2-LMSN & VE@NH2-LMSN Release at pH 7.4, In vitro cell NH2-LMSN: 325 37.7 ± 2.5
tests and hemolytic
experiments
Fluorescein isothiocyanate-dyed MSNs Gallic acid Characterization of MSNs, 100 ± 10 for – (Rashidi et al., 2017)
(FITC-MSNs) and its Functionalized by cytotoxicity effect on Caco-2 FITC-MSNs &
3-Aminopropyltriethoxysilane (APTES) cells 130 ± 15 for AP-
(APFITC-MSNs] FITC-MSNs
MCM-41 (TMABr as surfactant and sodium α-Tocopherol Characterization of MCM-41, – – (Sun, Lu, Qiua, & Tang,
silicate) tensile strength, oxygen and 2017)
water barrier properties,
Antioxidant activity
Colloidal MSNs (MCM-48) Resveratrol (RES) saturated solubility of RES, 191 & 283 – (Summerlin et al., 2016)
cytotoxicity effects on HT-29 &
LS147T colon cancer cell lines,
anti-inflammatory on
RAW264.7 cells
Mesoporous silica nanoparticles (MSNs) Curcumin & Chrysin characterization of NPs, Curcumin: Curcumin & (Lungare, Hallam, &
cytotoxicity effect on olfactory 216.8 ± 2.1 to Chrysin: 12.34 ± Badhan, 2016)
neuroblastoma cells OBGF400 263.51 ± 8.3 1.28
Chrysin: 216.8
± 2.1 to 283.5 ±
8.3
MSNPs, AMSNPs and AMSNPs-MO, 3- Morin (2 ́ ,3,4 ́ ,5,7- Characterization of NPs, MSNP: 105 – (Arriagada et al., 2016)
Aminopropyltriethoxysilane (APTES) pentahydroxyflavone) adsorption kinetics, Isotherm & AMSNPs: 125,
Antioxidant capacity AMSNPs-MOR:
155
MCM-41nanoparticles Folic acid (FA), vitamin Characterization of NPs, pH 250–300 – Popescu et al. (2016)
MCM-41/Fe3O4 dependent release
Mesoporous-silica-coated up conversion Vitamin B12 Characterization of NPs, 100 – Xu, Ding, et al. (2016)
nanoparticles Release behavior in different
pH
Lipid bilayer coated-curcumin loaded Curcumin Characterization of NPs, 200 – (Datz, Engelke, Schirnding,
colloidal periodic mesoporous Cytotoxicity effect on Hela cells Nguyen, & Bein, 2016)
organosilica nanoparticles
Self-Fluorescent & Stimuli-Responsive Curcumin Characterization of NPs, 117 – Xu, Lü, et al. (2016)
MSNs Release behavior in different
pHs 7.4 & 5.5,
Silica-based mesoporous materials (MSU- Curcumin Charcterization of NPs, 5.9–38 – Bollu et al. (2016)
2 & MCM-41) cytotoxicity effect on CHO &
MCF-7, A549, Release at pH 7
The outer of the MSN decorated with poly Topotecan (TPT) & Charcterization of NPs, 65–75 TPT: 1.8 (Murugan et al., 2016)
(acrylic acid)-chitosan, grafting of Quercetin (QT) Cytotoxicity & ROS generation QT: 1.2
arginine-glycine-aspartic acid effect on MDA-MB-231 & MCF-
7, A549, Release at pH 7.4

5.2. Mesoporous silica nanoparticles active surface which could be functionalized to modify surface proper­
ties and link drug or bioactive molecules. MSNs can be synthesized by
Mesoporous silica nanoparticles (MSNs) are suitable nanocarriers for sol-gel or spray drying methods and applied for loading and targeted
drugs and bioactive compounds due to their unique properties, delivery of hydrophobic, hydrophilic, and negatively or positively
including surface functionality, good biocompatibility, stability, high charged drugs or bioactive compounds (Rashidi, Ganji, &
drug loading capacity, sustained release and targeted delivery of a va­ Vasheghani-Farahani, 2017). MSNs have been applied in a wide variety
riety of drug molecules, unique the mesoporous structure and chemical of industries, including feed, cosmetics, and pharmaceutical products,
resistance of the structure. They have a honeycomb-like structure and for many decades. Based on extensive physicochemical, ecotoxicology,

11
L. Rashidi Food Bioscience 43 (2021) 101258

toxicology, safety, and epidemiology data, no environmental or health methods are reported for the formation of carbon nanotubes including
risks have been associated with these materials. They do not exert any arc discharge, carbon monoxide disproportionation, laser ablation,
toxicological effects on medicines, and are approved to be GRAS chemical vapor deposition (CVD), and hydrothermal method (Kumari
(Rashidi et al., 2017). The limitations of these nanocarriers include et al., 2014). The advantages of carbon nanotubes are low-cost, high
potential toxicity in high concentrations, contribution to the level in­ aspect ratios, biocompatibility, surface area, high thermal and electrical
crease of reactive species, and inducing the elevated malondialdehyde, conductivity, light-weight, distinct optical characteristics, efficient
and decreasing glutathione level (Zorkina et al., 2020). Table 5 indicates loading of multiple molecules and high mechanical strength. Carbon
an overview of nutraceuticals encapsulated by mesoporous silica nanotubes are part of the fullerene family which fullerenes are made up
nanoparticle carries. of graphene sheets of linked pentagonal and hexagonal rings leading to
them their curved structure (Kumari et al., 2014). Limitations of using
5.3. Mesoporous silica nanoparticles carbon nanotubes include potential toxicity, similarity in carcinogenic
potential between carbon nanotubes and asbestos, cannot be function­
Magnetic nanoparticles (MNPs) possess general characteristics of ally integrated into the biological systems unless surface functionaliza­
nanoparticles and magnetic properties. These nanostructures are tion, enable cause necrosis or apoptosis of macrophage cell lines and
considered as particles with the size of lower than 100 nm, which their modifies cell morphology and contribute to the level increase of reactive
magnetic properties are determined by many factors such as the degree oxygen species. A single-walled carbon nanotube (SWCNT) embedded in
of deficiency of the crystal network, the morphology, the chemical a 1-palmitoyl-2-oleoylphosphatidylcholine (POPC) lipid bilayer con­
composition, the interplay of the particle with the neighboring matrix, taining curcumin was synthesized by layer-by-layer technique with
the type, and the particle shape and size, and the adjacent particles. The particle size of 2.12–2.7 (Sahoo, Kanchi, Mandal, Dasgupta, & Maiti,
great properties of MNPs are including excellent reactivity, larger 2018). The biocompatible nanotubes constituted of Poly-d-lysine (PDL)
surface-to-volume ratio, and unique magnetic response compared to and BSA and, Polycarbonate (Bio nanotube (BNTs)) were prepared for
their bulk materials (Hajba & Guttman, 2016). These nanoparticles are encapsulation of curcumin as a bioactive model using layer-by-layer
mainly categorized to the metal nanoparticles (Fe, Co, Ni, etc.), metal which the particle size was 400 nm and encapsulation efficiency was
oxide (Fe3O4, γ-Fe2O3, CoFe2O4, etc.), and metal alloy nanoparticles obtained 45 % (Sadeghi et al., 2013).
(FeCo, alnico, permalloy, etc.). The common nanoparticles are iron,
cobalt, silver, gold, and nickel. Metal nanoparticles have a higher 5.5. Dendrimers
saturation magnetization but they are toxic and not suitable for drug/­
gene delivery or clinical use (Vittorio et al., 2014), therefore, they are A dendrimer is a macromolecule, monodispersive, highly branched,
coated using natural materials. There are different methods for synthe­ spherical and symmetric. The dendrimer structure is three-dimensional
sizing these nanoparticles, including deposition method, highly branched, and also radially symmetrical. The dendrimer core is
co-precipitation, microemulsion, thermal decomposition, ball milling, available for the encapsulation of drugs, bioactive agents, or nutra­
polyol, spray pyrolysis, and hydrothermal or solvothermal. Nowadays, ceuticals and the peripheral functional groups are accessible for the
several studies have been reported the application of MNPs as the car­ modification of nutraceutical molecules through covalent bonds and for
riers for the delivery of polyphenols in order to the treatment of tumors. the formation of the complex. Dendrimers are generally synthesized
These carriers are easily biodegradable, nontoxic, and have chemical using the divergent method or the convergent one, in which the den­
stability in physiological situations, and possibility of chemical modifi­ drimer grows external from the multifunctional core molecule. The core
cation using the coating of magnetic nanoparticles but these nano­ molecule of dendrimer reacts with monomer molecules owning two
carriers tend to aggregate into larger clusters and potentially toxic in dormant groups, and one reactive which giving the first-generation
high concentrations. In a study carboxymethyl cellulose -Magnetic dendrimer (Abbasi et al., 2014). The merits of using these nano­
nanoparticles applied for encapsulation of galbanic acid using precipi­ carriers are the situation of core is suitable for entrapping bioactive
tation method for studding its cytotoxicity effect on Prostate Cancer compounds, the existence of numerous surface groups lead to increase
Cells which particle size and encapsulation efficiency were obtained loading percentage of nutraceuticals, and potential conjugation of sur­
9.39 nm and 40.36 %, respectively (Mohtashami, Ghows, face groups with target ligands, but its demerit is potential toxicity.
Tayarani-Najaran, & Iranshahi, 2019). Also, Iron oxide, silica-coated Curcumin encapsulated in Polyamidoamine (PAMAM)
iron oxide (SIOMNPs), chitosan-coated iron oxide (CIOMNPs) were dendrimer-plamitic acid core-shell nanoparticles were prepared by hy­
synthesize by alkaline hydrolysis technique for encapsulation of cellu­ drophobic and hydrophilic interaction via hydrogen bonding and par­
lase with particle sizes (45–60, 80–150 & 30–50 nm) and encapsulation ticle size and encapsulation efficiency were 257.9 nm and 80.8 %,
efficiencies (82, 97, & 67 %) (Kumar et al., 2019). Magnetic nano­ respectively (Tripathi et al., 2020). PAMAM G4 dendrimers conjugated
particles coated by dextran were synthesized for encapsulation of cate­ PEG were synthesized for encapsulation of curcumin via hydrogen
chin by precipitation method and used for investigating its therapeutic bonding method which the particle size and encapsulation efficiency
effect for pancreatic tumor cells with the particles size of 91 nm (Vittorio were 506.3–547.8 nm and 69.81–90.5%, respectively (Elfiyani, Srifiana,
et al., 2014). & Al Rasyied, 2018). Also, PAMAM dendrimers were prepared for
encapsulation of resveratrol, genistein and curcumin by hydrophobic
5.4. Carbon nanotubes and hydrophilic interaction via hydrogen bonding with the particle size
of 10 nm and encapsulation efficiency of 30–55 % (Chanphai &
The carbon nanotube is similar to a graphite sheet rolled into a Tajmir-Riahi, 2016). In addition, PAMAM dendrimers were prepared for
carbon cylinder composed of the benzene ring with specific hexagonal encapsulation of berberine (BBR) by hydrophobic and hydrophilic
latticework that constitute the sheet. These nanostructures have been interaction via hydrogen bonding with the particle size of 100–200 nm
used in biology as sensors for detection of DNA and protein, or as and encapsulation efficiency 29.9–37.5 % (Gupta et al., 2017). In
diagnostic devices for distinction of various proteins from serum sam­ another study, PAMAM dendrimers were synthesized for encapsulation
ples, and also as carriers to deliver vaccine, polyphenol, protein, or drug of quercetin by hydrophobic and hydrophilic interaction via hydrogen
(Kumari, Singla, Guliani, & Yadav, 2014). There are two main kinds of bonding with a particle size lower than 100 nm (Madaan, Lather, &
CNTs, including single-walled nanotubes (SWCNTs) composed of a sin­ Pandita, 2016).
gle graphite sheet seamlessly wrapped into the cylindrical tube and
multi-walled nanotubes (MWCNTs) consist of the array of nanotubes one
concentrically located inside another like rings of a tree trunk. Various

12
L. Rashidi Food Bioscience 43 (2021) 101258

6. Safety issues of using nanomaterials in the food industry applications in the food industry. This should be conducted by proper in
vivo models, which will lead to the establishment of legal regulations to
Recently, the increased application of nanotechnology and the high defend consumers safety in which industry and society can receive
number of its derived commercial applications has led to concerns benefit from new nanotechnology applications in food production,
regarding their potential risk for human health. Some reports are defending the safety, the environment and the health. In addition,
focusing on the evaluation of nanocarriers’ or nanomaterials’ toxicity further research should be conducted for the development of nano-
when they form part of a food matrix. Materials at the nanoscale can delivery systems that are commercially viable.
easily penetrate and accumulate within the human body, sub-cellular
boundaries, and breaching the cellular walls, which have an unpre­
dictable impact on the human body, plants, animals, and the environ­ Declaration of competing interest
ment because of potential toxic effects (Yu et al., 2018). The safety of
nanomaterials and their hazards of using them remain unexamined, and The authors declare that they have no known competing financial
require a main risk assessment. The study of direct and indirect impacts interests or personal relationships that could have appeared to influence
of nanomaterials on human health, including the biological fate of the work reported in this paper.
nanomaterials after digestion and their behavior within the gastroin­
testinal tract (GIT), and also their possible interactions with biological References
systems must be studied. Moreover, it is essential to develop regulatory
controls for the protection of public health from potentially adverse Abbasi, E., Fekri Aval, S., Akbarzadeh, A., Milani, M., Nasrabadi, H. T., Joo, S. W., et al.
effects of nanomaterials (Yu et al., 2018). Several expert bodies are (2014). Dendrimers: Synthesis, applications, and properties. Nanoscale Research
Letter, 9(247), 1–10. https://nanoscalereslett.springeropen.
active in this domain, including the International Standard Organization com/articles/10.1186/1556-276X-9-247#citeas.
(ISO), the US Food and Drug Administration (FDA), the Organization for Ahmad, M., Mudgil, P., Gani, A., Hamed, F., Masoodi, F. A., & Maqsood, S. (2019). Nano-
Economic Cooperation and Development (OECD), and EU Scientific encapsulation of catechin in starch nanoparticles: Characterization, release behavior
and bioactivity retention during simulated in-vitro digestion. Food Chemistry, 270(1),
Committees and Agencies. In addition, the scientific opinion as a
95–104. https://www.sciencedirect.com/science/article/abs/pii/S0308814
guideline for the nanoscience and nanotechnology risk evaluation in 618311634.
feed/food applications has been published by the European Food Safety Ahmed, R., Tariq, M., Ahmad, I. S., Fouly, H., Abbas, F., Hasan, A., et al. (2019). Poly
(lactic-co-glycolic acid) nanoparticles loaded with callistemon citrinus phenolics
Authority, 2021 (EFSA, 2021). In the EFSA, nanoparticles are defined as
exhibited anticancer properties against three breast cancer cell lines. Journal of Food
engineered nanomaterials (ENMs) which have a minimum of one Quality. https://doi.org/10.1155/2019/2638481. Article ID 2638481.
dimension within the range of 1–100 nm. If these nanomaterials are Akbarbaglu, Z., Jafari, M. S., Sarabandi, K., Mohammadi, M., Khakbaz Heshmati, M., &
present in a food product, they must go through a precise risk evaluation Pezeshki, A. (2019). Influence of spray drying encapsulation on the retention of
antioxidant properties and microstructure of flaxseed protein hydrolysates. Colloids
to prove that they are not hazardous for human consumption. Also, the and Surfaces B: Biointerfaces, 178, 421–429. https://doi.org/10.1016/j.
US FDA published a draft guideline for using nanomaterials in animal colsurfb.2019.03.038
feed (The Food and Drug Administration (FDA), 2014). A recent EU Alehosseini, A., Gómez-Mascaraque, L. G., Ghorani, B., & López-Rubio, A. (2019).
Stabilization of a saffron extract through its encapsulation within electrospun/
legislation (No. 1169/2011) has necessitated the labeling of nano­ electrosprayed zein structures. LWT -Food Science and Technology, 113, 108280 htt
materials on the packaging of novel foods (European Parliament, 2011). ps://www.sciencedirect.com/science/article/abs/pii/S0023643819306206.
By observing modern food regulations and any new nanotechnology Alemán, A., Marín, D., Taladrid, D., Montero, P., & Carmen Gómez-Guillén, M. (2019).
Encapsulation of antioxidant sea fennel (Crithmum maritimum) aqueous and
regulation for providing the public with information, the safety of ethanolic extracts in freeze-dried soy phosphatidylcholine liposomes. Food Research
nanomaterials in the food and feed industry can be ensured. Nowadays, International, 119, 665–674. https://www.sciencedirect.com/science/article/abs/pii
there are many bioactive compounds encapsulated into nano-delivery /S0963996918308305.
Altan, A., & Çayır, Ö. (2020). Encapsulation of carvacrol into ultrafine fibrous zein films
systems which are commercially available in the market, including
via electrospinning for active packaging. Food Packaging and Shelf Life, 26, 100581.
Nanotea (company Qinhuangdao Taiji Ring Nano), OilFresh (company: https://doi.org/10.1016/j.fpsl.2020.100581
OIlFresh corporation, USA), LifePak and NanoCoQ10 (company: Phar­ Arriagada, F., Correa, O., Guther, G., Nonell, S., Mura, F., Olea-Azar, C., et al. (2016).
Morin flavonoid adsorbed on mesoporous silica, a novel antioxidant nanomaterial.
manex), Nano C (company: Neurvana), Nano Resveratrol and Nano
PLOS One, 11, e0164507. https://www.ncbi.nlm.nih.gov/pmc/articles/PM
Curcumin (company: Neurvana), ACZnano Advanced Cellular (com­ C5094702/.
pany: Vitality Products, USA). Assadpour, E., & Jafari, S. M. (2019). A systematic review on nanoencapsulation of food
bioactive ingredients and nutraceuticals by various nanocarriers. Critical Reviews in
Food Science and Nutrition, 59(19), 3129–3151. https://doi.org/10.1080/
7. Conclusions 10408398.2018.1484687
Bollu, V. S., Barui, A. K., Mondal, S. K., Prashar, S., Fajardo, M., Briones, D., et al. (2016).
Considerable attention has been paid to the development of nano- Curcumin-loaded silica-based mesoporous materials: Synthesis, characterization and
cytotoxic properties against cancer cells. Materials Science and Engineering: C, 63,
scale delivery systems for the protection of nutraceuticals against un­ 393–410. https://www.sciencedirect.com/science/article/abs/pii/S09284931163
balanced and unfavorable conditions. In addition, the functional food 01801.
market developed over the decades because of increasing consumer Carpenter, J., George, S., & Saharan, V. K. (2019). Curcumin encapsulation in multilayer
oil-in-water emulsion: Synthesis using ultrasonication and studies on stability and
demand for nutraceuticals, nutritional and healthy food products. Also, antioxidant and release activities. Langmuir, 35(33), 10866–10876. https://doi.org/
there are numerous demands for reducing the amounts of applying 10.1021/acs.langmuir.9b01523
synthetic preservatives and ingredients in the food industry. In this re­ Castro Coelho, S., Nogueiro Estevinho, B., & Rocha, F. (2020). Encapsulation in food
industry with emerging electrohydrodynamic techniques: Electrospinning and
view, different nano-delivery systems employed for the protection and Electrospraying - a review. Food Chemistry, No. 127850. https://doi.org/10.1016/j.
delivery of nutraceuticals were investigated. Based on the researches, foodchem.2020.127850
non-food grade delivery systems possess more stability than food grade Castro, G. M. M. A., Passos, T. S., Nascimento, S. S.d, Medeiros, I., Araujo, N. K.,
Maciel, B. B. B., et al. (2020). Gelatin nanoparticles enable water dispersibility and
delivery systems but some of them are toxic in high concentrations potentialize the antimicrobial activity of Buriti (Mauritia flexuosa) oil. BMC
which sometimes for reduction of toxicity, their surface should be Biotechnololgy, 55(20). https://doi.org/10.1186/s12896-020-00649-4, 2020.
functionalized by nontoxic material. It is necessary to characterize the Chanphai, P., & Tajmir-Riahi, H. A. (2016). Binding analysis of antioxidant polyphenols
with PAMAM Nanoparticles. Journal of Biomolecular Structure and Dynamics, 36(13),
distribution, absorption, elimination, and metabolism behavior of nano-
3487–3495. https://pubmed.ncbi.nlm.nih.gov/29019428/.
delivery systems carrying the bioactive agents or nutraceuticals in order Chavoshpour-Natanzia, Z., & Sahihi, M. (2019). Encapsulation of quercetin-loaded
to potentiate therapeutic applications and increase the efficacy of β-lactoglobulin for drug delivery usingmodified anti-solvent method. Food
nutraceuticals in functional foods. In the future, it is important to Hydrocolloids, 96, 493–502. https://www.sciencedirect.com/science/article/abs/
pii/S0268005X18325402?via%3Dihub.
determine the acceptable daily intakes of the nanoencapsulated nutra­ Cheng, H., Zhang, H., Li, D., Duan, H., & Liang, L. (2021). Impact of oil type on the
ceuticals and to evaluate the safety of nano-delivery systems for suitable location, partition and chemical stability of resveratrol in oil-in-water emulsions

13
L. Rashidi Food Bioscience 43 (2021) 101258

stabilized by whey protein isolate plus gum Arabic. Food Hydrocolloids, 109 of Food Processing and Preservation, 1–10, 00:e14629 https://doi.org/10.1111/j
(106119). https://doi.org/10.1016/j.foodhyd.2020.106119 fpp.14629.
Chen, C., Sun, W., Wang, X., Wang, Y., & Wang, P. (2018). Rational design of curcumin Gharehbeglou, P., Jafari, S. M., Hamishekar, H., Homayouni, A., & Mirzaei, H. (2019).
loaded multifunctional mesoporous silica nanoparticles to enhance the cytotoxicity Pectin-whey protein complexes vs. small molecule surfactants for stabilization of
for targeted and controlled drug release. Materials Science and Engineering: C, 85, double nano-emulsions as novel bioactive delivery systems. Journal of Food
88–96. https://www.sciencedirect.com/science/article/abs/pii/S0928493117323 Engineering, 245, 139–148. https://www.sciencedirect.com/science/article/abs/pii
093. /S0260877418304515.
Chen, W., Zou, M., Ma, X., Lv, R., Ding, T., & Liu, D. (2019). Co-Encapsulation of EGCG Ghatak, D., & Iyyaswam, R. (2019). Selective encapsulation of quercetin from dry onion
and quercetin in liposomes for optimum antioxidant activity. Journal of Food Science, peel crude extract in reassembled casein particles. Food and Bioproducts Processing,
84, 111–121. https://pubmed.ncbi.nlm.nih.gov/30548488/. 115, 100–109. https://doi.org/10.1016/j.fbp.2019.03.003
Chirio, D., Peira, E., Dianzani, C., Muntoni, E., Gigliotti, C. L., Ferrara, B., et al. (2019). Gupta, L., Sharma, A. K., Gothwal, A., Khan, M. S., Khinchi, M. P., Qayum, A., et al.
Development of solid lipid nanoparticles by cold dilution of microemulsions: (2017). Dendrimer encapsulated and conjugated delivery of berberine: A novel
Curcumin loading, preliminary in vitro studies, and biodistribution. Nanomaterials, 9 approach mitigating toxicity and improving in vivo pharmacokinetics. International
(230), 1–17. https://doi.org/10.3390/nano9020230 Journal of Pharmaceutical, 528(1–2), 88–99. https://www.sciencedirect.com/scienc
Choudhary, V., Shivakumar, H., & Ojha, H. (2019). Curcumin-loaded liposomes for e/article/pii/S0378517317303848.
wound healing: Preparation, optimization, in-vivo skin permeation and Haggag, Y., Elshikh, M., El-Tanani, M., Bannat, I. M., McCarron, P., & Tambuwala, M. M.
bioevaluation. Journal of Drug Delivery Science and Technology, 49, 683–691. https:// (2020). Nanoencapsulation of sophorolipids in PEGylated poly(lactide-co-glycolide)
doi.org/10.1016/j.jddst.2018.12.008 as a novel approach to target colon carcinoma in the murine model. Drug Delivery and
Dahiya, S., Rani, R., Dhingra, D., Kumar, S., & Dilbaghi, N. (2018). Conjugation of Translational Research, 10, 1353–1366. https://doi.org/10.1007/s13346-020-00750-
epigallocatechin gallate and piperine into a zein nanocarrier: Implication on 3
antioxidant and anticancer potential. Advances in Natural Sciences: Nanoscience and Hajba, L., & Guttman, A. (2016). The use of magnetic nanoparticles in cancer
Nanotechnology, 9, 1–16, 035011 https://iopscience.iop.org/article/10.1088/2 theranostics: Toward handheld diagnostic devices,. Biotechnology Advances, 34,
043-6254/aad5c1/pdf. 354–361. https://www.sciencedirect.com/science/article/abs/pii/S0734975
Dalmoro, A., Bochicchio, S., Lamberti, G., Bertoncin, P., Janssens, B., & Barba, A. A. 016300088.
(2019). Micronutrients encapsulation in enhanced nanoliposomal carriers by a novel Hammoud, Z., Gharib, R., Fourmentin, S., Elaissari, A., & Greige-Gerges. (2019). 100 and
preparative technology. RSC Advances, 9(34), 19800–19812. https://doi.org/ cholesterol. International Journal of Pharmaceutics, 561, 161–170. https://doi.org/
10.1039/c9ra03022k 10.1016/j.ijpharm.2019.02.022
Datz, S., Engelke, H., Schirnding, C. V., Nguyen, L., & Bein, T. (2016). Lipid bilayer- Harini, L., Karthikeyan, B., Srivastava, S., Suresh, S. B., Ross, C., Gnanakumar, G., et al.
coated curcumin-based mesoporous organosilica nanoparticles for cellular delivery. (2017). Polyethylenimine-modified curcumin-loaded mesoporus silica nanoparticle
Microporous and Mesoporous Materials, 225, 371–377. https://www.sciencedirect. (MCM-41) induces cell death in MCF-7 cell line. IET Nanobiotechnology, 11, 57–61.
com/science/article/abs/pii/S1387181115006642. https://pubmed.ncbi.nlm.nih.gov/28476962/.
Delshadi, R., Bahrami, A., Tafti, A. G., Barba, F. J., & Williams, L. L. (2020). Micro and Hong, S., Choi, D. W., Kim, H. M., Park, C. H., Lee, W., & Park, H. H. (2020). Protein-
nano-encapsulation of vegetable and essential oils to develop functional food based nanoparticles as drug delivery systems. Pharmaceutics, 12(7), 604–633. http
products with improved nutritional profiles. Trends in Food Science & Technology, s://www.ncbi.nlm.nih.gov/pmc/articles/PMC7407889/pdf/pharmaceutics-12-
104, 72–83. https://doi.org/10.1016/j.tifs.2020.07.004 00604.pdf.
Desai, N. M., Stanley, J. G., & Murthy, P. S. (2020). Green coffee nanoparticles: Hoseyni, S. Z., Jafari, S. M., Shahiri Tabarestani, H., Ghorbani, M., Assadpour, E., &
Optimization, in vitro bioactivity and bio-release property. Journal of Sabaghi, M. (2021). Release of catechin from Azivash gum-polyvinyl alcohol
Microencapsulation, 37(1), 52–64. https://www.tandfonline.com/doi/abs/10.1080/ electrospun nanofibers in simulated food and digestion media. Food Hydrocolloids,
02652048.2019.1692946. 112(106366), 1–8. https://doi.org/10.1016/j.foodhyd.2020.106366
Dima, C., Assadpour, E., Dima, S., & Jafari, S. M. (2020). Nutraceutical nanodelivery; an Huang, H., Belwal, T., Liu, S., Duan, Z., & Luo, Z. (2019). Novel multi-phase nano-
insight into the bioaccessibility/bioavailability of different bioactive compounds emulsion preparation for co-loading hydrophilic arbutin and hydrophobic coumaric
loaded within nanocarriers. Critical Reviews in Food Science and Nutrition, 1–35. acid using hydrocolloids. Food Hydrocolloids, 93, 92–101. https://www.sciencedir
Article ID 32691612 https://doi.org/10.1080/10408398.2020.1792409. ect.com/science/article/abs/pii/S0268005X18314401.
Dordoni, R., Garrido, G. D., Marinoni, L., Torri, L., Piochi, M., & Spigno, G. (2019). Huguet-Casquero, A., Moreno-Sastre, M., López-Méndez, T. B., Gainza, E., & Pedraz, J. L.
Enrichment of whole wheat cocoa biscuits with encapsulated grape skin extract. (2020). Encapsulation of oleuropein in nanostructured lipid carriers:
International Journal of Food Science, 1–11. Article ID 9161840 https://www. Biocompatibility and antioxidant efficacy in lung epithelial cells. Pharmaceutics, 12,
hindawi.com/journals/ijfs/2019/9161840/, 2019. 429,. https://doi.org/10.3390/pharmaceutics12050429
Du, Z., Liu, J., Zhang, H., Chen, Y., Wu, X., Zhang, Y., et al. (2020). L-Arginine/l-lysine Jeong, S. J., Lee, J., & Lee, H. G. (2019). Nanoencapsulation of synergistic antioxidant
functionalized chitosan–casein core–shell and pH-responsive nanoparticles’: fruit and vegetable concentrates and their stability during in vitro digestion. Journal
Fabrication, characterization and bioavailability enhancement of hydrophobic and of the Science of Food and Agriculture, 100(3), 1056–1063. https://doi.org/10.1002/
hydrophilic bioactive compounds. Food and Function Journal, 11, 4638–4647. jsfa.10110
https://pubs.rsc.org/en/content/articlelanding/2020/fo/d0fo00005a#!divAbstract. Juère, E., Florek, J., Bouchoucha, M., Jambhrunkar, S., Wong, K. Y., Popat, A., et al.
Elfiyani, R., Srifiana, Y., & Al Rasyied, F. D. (2018). Curcumin encapsulation using (2017). Vitro dissolution, cellular membrane permeability, and anti-inflammatory
dendrimer PAMAM G4 conjugated with polyethylene glycol to improve the response of resveratrol-encapsulated mesoporous silica nanoparticles. Molecular
properties of gel dossage form. Pharmacia, 8, 107–118. https://doi.org/10.12928/ Pharmaceutics, 14, 4431–4441. https://pubs.acs.org/doi/10.1021/acs.molphar
pharmaciana.v8i1.8940 maceut.7b00529.
Estakhr, P., Tavakoli, J., Beigmohammadi, F., Alaei, S., & Mousavi Khaneghah, A. Kamel, A. E., Fadel, M., & Louis, D. (2019). Curcumin-loaded nanostructured lipid
(2020). Incorporation of the nanoencapsulated polyphenolic extract of Ferula carriers prepared using Peceol™ and olive oil in photodynamic therapy:
persica into soybean oil: Assessment of oil oxidative stability. Food Sciences and Development and application in breast cancer cell line. International Journal of
Nutrition, 8(6), 2817–2826. https://www.ncbi.nlm.nih.gov/pmc/articles/PM Nanomedicine, 14, 5073–5085. https://doi.org/10.2147/IJN.S210484
C7300055/. Kim, E.S., Lee, J-S., & Lee, H.G. (n.d.). Improvement of antithrombotic activity of red
European Food Safety Authority (Efsa). (2011). Guidance on the risk assessment of the ginseng extract by nanoencapsulation using chitosan and antithrombotic cross-
application of nanoscience and nanotechnologies in the food and feed chain. EFSA linkers: polyglutamic acid and fucodian. Journal of Ginseng Research. https://www.
Journal, 9, 2140–2176. https://efsa.onlinelibrary.wiley.com/doi/abs/10.2903/j.ef sciencedirect.com/science/article/pii/S1226845320300828#!. In press.
sa.2011.2140. Kiss, E. (2020). Nanotechnology in food systems: A review. Acta Alimentaria, 49(4),
European Parliament, Council of the EU. (2011). Regulation (EU) No. 1169/2011 of the 460–474. https://doi.org/10.1556/066.2020.49.4.12
European Parliament and of the Council of 25 October 2011 on the provision of food Kumari, A., Singla, R., Guliani, A., & Yadav, S. K. (2014). Nanoencapsulation for drug
information to consumers, amending regulations (EC) No. 1924/2006 and (EC) No. delivery. Excli Journal, 13, 265–286. https://www.ncbi.nlm.nih.gov/pmc/artic
1925/2006 of the European Parliament and of the Council. https://eur-lex.europa. les/PMC4464443/.
eu/legal-content/EN/ALL/?uri=CELEX%3A32011R1169. Kumar, S., Morya, V., Gadhavi, J., Vishnoi, A., Singh, J., & Datta, B. (2019). Investigation
Fathi, M., Donsi, F., & McClements, D. J. (2018). Protein-based delivery systems for the of nanoparticle immobilized cellulase: Nanoparticle identity, linker length and
nanoencapsulation of food ingredients. Comprehensive Reviews in Food Science and polyphenol hydrolysis. Heliyon, 5, e01702. https://www.sciencedirect.com/science/
Food Safety, 17, 920–936. https://doi.org/10.1111/1541-4337.12360 article/pii/S2405844019314835.
Feng, J., Xu, H., Zhang, L., Wang, H., Liu, S., Liu, Y., et al. (2019). Development of Kurd, F., Fathi, M., & Shekarchizadeh, H. (2019). Nanoencapsulation of hesperetin using
nanocomplexes for curcumin vehiculization using ovalbumin and sodium alginate as basil seed mucilage nanofibers: Characterization and release modeling. Food
building blocks: Improved stability, bioaccessibility, and antioxidant activity. Bioscience, 32(100475). https://www.sciencedirect.com/science/article/abs/pii
Journal of Agricultural and Food Chemistry, 67, 379–390. https://pubs.acs.org/doi/10 /S2212429217310301.
.1021/acs.jafc.8b02567. Lin, J., Cai, Q., Tang, Y., Xu, Y., Wang, Q., Li, T., et al. (2018). PEGylated Lipid bilayer
Gaba, B., Khan, T., Haider, M. F., Alam, T., Baboota, S., Parvez, S., et al. (2019). Vitamin coated mesoporous silica nanoparticles for co-delivery of paclitaxel and curcumin:
E loaded naringenin nanoemulsion via intranasal delivery for the management of Design, characterization and its cytotoxic effect. International Journal of
oxidative stress in a 6-OHDA Parkinson’s Disease Model. BioMed Research Pharmaceutics, 536, 272–282. https://www.sciencedirect.com/science/article/pii/
International, 1–20. Article ID 2382563 https://www.hindawi.com/journals S0378517317310232.
/bmri/2019/2382563/. Liu, C., Zhang, Z., Kong, Q., Zhang, R., & Yang, X. (2019). Enhancing the antitumor
Gahruie, H. H., Niakousari, M., Parastouei, K., Mokhtarian, M., Eş, I., & Mousavi activity of tea polyphenols encapsulated in biodegradable nanogels by
Khaneghah, A. (2020). Co-encapsulation of vitamin D3 and saffron petals’ bioactive macromolecular self-assembly. RSC Advances, 9, 10004–10016. https://pubs.rsc.
compounds in nanoemulsions: Effects of emulsifier and homogenizer types. Journal org/en/content/articlelanding/2019/ra/c8ra07783e#!divAbstract.

14
L. Rashidi Food Bioscience 43 (2021) 101258

Lungare, S., Hallam, K., & Badhan, R. K. S. (2016). Phytochemical-loaded mesoporous O’Doherty, M., Mulholland, E. J., Chambers, P., Pentlavalli, S., Ziminska, M.,
silica nanoparticles for nose-to-brain olfactory drug delivery. International Journal of Chalanqui, M. J., et al. (2020). Improving the intercellular uptake and osteogenic
Pharmaceutics, 513, 280–293. https://www.sciencedirect.com/science/article/pii/ potency of calcium phosphate via nanocomplexation with the RALA peptide.
S0378517316308729. Nanomaterials, 10(2442). https://doi.org/10.3390/nano10122442
Madaan, K., Lather, V., & Pandita, D. (2016). Evaluation of polyamidoamine dendrimers Okagu, O. D., Verma, O., McClements, D. J., & Udenigwe, C. C. (2020). Utilization of
as potential carriers for quercetin, a versatile flavonoid. Drug Delivery, 23(1), insect proteins to formulate nutraceutical delivery systems: Encapsulation and
254–262. https://www.tandfonline.com/doi/pdf/10.3109/10717544.2014.910564. release of curcumin using mealworm protein-chitosan nano-complexes. International
Mahalakshmi, L., Leen, M. M., Jeyan, M., & Anandharamakrishnan, C. (2020). Micro and Journal of Biological Macromolecules, 151, 333–343. https://doi.org/10.1016/j.
nano encapsulation of β-carotene in zein protein: Size dependent release and ijbiomac.2020.02.198
absorption behavior. Food & Function, 11, 1647–1660. https://pubs.rsc. Oskoueian, E., Karimi, E., Noura, R., Ebrahimi, M., Shafaei, N., & Karimi, E. (2019).
org/en/content/articlelanding/2020/fo/c9fo02088h#!divAbstract. Nanoliposomes encapsulation of enriched-phenolic fraction from pistachio hulls and
Mahmoudi, R., Ardakani, M. T., Hajipour, B., Bagheri, A., Mohammad-Beigi, H., its antioxidant, anti-inflammatory and anti-melanogenic activities. Journal of
Aliakbari, F., et al. (2019). Chitosan nanoparticles containing physalis alkekengi-L Microencapsulation, 37(1), 1–34. https://doi.org/10.1080/02652048.2019.1692941
extract: Preparation, optimization and their antioxidant activity. Bulletin of Materials Ozkan, G., Franco, P., De Marco, I., Xiao, J., & Capanoglu, E. (2019). A review of
Science, 42, 131–137. https://www.ias.ac.in/article/fulltext/boms/042/03/0131. microencapsulation methods for food antioxidants: Principles, advantages,
Maione-Silva, L., Gava de Castro, E., Nascimento, T. L., Cintra, E. R., Moreira, L. C., drawbacks and applications. Food Chemistry, 272, 496–506. https://doi.org/
Santana, B. A., et al. (2019). Ascorbic acid encapsulated into negatively charged 10.1016/j.foodchem.2018.07.205
liposomes exhibits increased skin permeation, retention and enhances collagen Pateiro, M., Gómez, B., Munekata, P. E. S., Barba, F. J., Putnik, P., Kovačević, D. B., et al.
synthesis by fibroblasts. Scientific Reports, 9, 522–529. https://www.nature.co (2021). Nanoencapsulation of promising bioactive compounds toImprove their
m/articles/s41598-018-36682-9. absorption, stability, functionality and theAppearance of the final food products.
Malekhosseini, P., Alami, M., Khomeiri, M., Esteghlal, S., Nekoei, A.-R., & Hashem Molecules, 26, 1547–1573. https://europepmc.org/backend/ptpmcrender.fcgi?acc
Hosseini, S. M. (2019). Development of casein-based nanoencapsulation systems for id=PMC7999092&blobtype=pdf.
delivery of epigallocatechin gallate and folic acid. Food Sciences and Nutrition, 7, Peng, Y., Meng, Q., Zhou, J., Chen, B., Xi, J., Long, P., et al. (2019). Nanoemulsion
519–527. https://onlinelibrary.wiley.com/doi/full/10.1002/fsn3.827. delivery system of tea polyphenols enhanced the bioavailability of catechins in rats.
Ma, J., Xu, C., Yu, H., Feng, Z., Yu, W., Gu, L., et al. (2020). Electro-encapsulation of Food Chemistry, 242, 527–532. https://www.sciencedirect.com/science/artic
probiotics in gum Arabic-pullulan blend nanofibres using electrospinning le/abs/pii/S0308814617315698.
technology. Food Hydrocolloids, 106381. https://doi.org/10.1016/j. Peng, S., Zou, L., Zhou, W., Liu, W., Liu, C., & McClements, D. J. (2019b). Encapsulation
foodhyd.2020.106381 of lipophilic polyphenols into nanoliposomes using pH-driven method: Advantages
McClements, D. J. (2020). Advances in nanoparticle and microparticle delivery systems and disadvantages. Journal of Agricultural and Food Chemistry, 67, 7506–7511. https
for increasing the dispersibility, stability, and bioactivity of phytochemicals. ://pubs.acs.org/doi/full/10.1021/acs.jafc.9b01602.
Biotechnology Advances, 38(107287). https://doi.org/10.1016/j. Popescu, S., Ardelean, I. L., Gudovan, D., Rădulescu, M., Ficai, D., Ficai, A., et al. (2016).
biotechadv.2018.08.004 Multifunctional materials such as MCM-41÷Fe3O4÷folic acid as drug delivery
Mehmood, T., & Ahmed, A. (2020). Tween 80 and soya-lecithin-based food-grade system. Romanian Journal of Morphology and Embryology, 57, 483–452 https://europ
nanoemulsions for the effective delivery of vitamin D. Langmuir, 36(11), 2886–2892. epmc.org/article/med/27516022.
https://pubs.acs.org/doi/pdf/10.1021/acs.langmuir.9b03944. Pulit-Prociak, J., Kabat, M., Węgrzyn, E., Zielina, M., & Banach, M. (2019). Encapsulation
Meng, Q., Long, P., Zhou, J., Ho, C.-T., Zou, X., Chen, B., et al. (2019). Improved of antioxidant compounds biopolymer micelles. Journal of Chemical Engineering
absorption of β-carotene by encapsulation in an oil-in-water nanoemulsion Communications, 1, 1–20. https://www.tandfonline.com/doi/abs/10.1080/0098
containing tea polyphenols in the aqueous phase. Food Research International, 116, 6445.2019.1602526?journalCode=gcec20.
731–736. https://www.sciencedirect.com/science/article/abs/pii/S0963996 Păvăloiu, R.-D., Sha’at, F., Bubueanu, C., Deaconu, M., Neagu, G., Sha’at, M., et al.
918307269. (2020). Polyphenolic extract from Sambucus ebulus L. Leaves free and loaded into
Milincic, D. D., Popovic, D. A., Levic, S. M., Kostic, A. Z., Tešic, Z. L., Nedovic, V. A., et al. lipid vesicles. Nanomaterials, 10, 56,. https://doi.org/10.3390/nano10010056
(2019). Application of polyphenol-loaded nanoparticles in food industry. Radünz, M., Trindade, M. L. M., Camargo, T. M., Radünz, A. L., Borges, C. D.,
Nanomaterials, 9(1629), 1–21. https://doi.org/10.1186/s12896-020-00649-4 Gandra, E. A., et al. (2019). Antimicrobial and antioxidant activity of
Minafra, L., Porcino, N., Bravatà, V., Gaglio, D., Bonanomi, M., Amore, E., et al. (2019). unencapsulated and encapsulated clove (Syzygium aromaticum, L.) essential oil.
Radiosensitizing effect of curcumin-loaded lipid nanoparticles in breast cancer cells. Food Chemistry, 276, 180–186. https://www.sciencedirect.com/science/artic
Scientific Reports, 9, 11134–11150. https://www.nature.com/articles/s41598-01 le/abs/pii/S0308814618317588.
9-47553-2. Rafiee, Z., Nejatian, M., Daeihamed, M., & Jafari, S. M. (2019). Application of curcumin-
Miri, M. A., Habibi Najafi, M. B., Movaffagh, J., & Ghorani, B. (2021). Encapsulation of loaded nanocarriers for food, drug and cosmetic purposes. Trends in Food Science &
ascorbyl palmitate in zein by electrospinning technique. Journal of Polymers and the Technology, 88, 445–458. https://doi.org/10.1016/j.tifs.2019.04.017
Environment, 29, 1089–1098. https://doi.org/10.1007/s10924-020-01954-x Rajabi, H., Jafari, S. M., Rajabzadeh, G., Sarfarazi, M., & Sedaghati, S. (2020). Chitosan-
Moghadam, F. V., Pourahmad, R., Mortazavi, A., Davoodi, D., & Azizinezhad, R. (2019). gum Arabic complex nanocarriers for encapsulation of saffron bioactive components.
Use of fish oil nanoencapsulated with gum Arabic carrier in low fat probiotic Colloids and Surfaces A: Physicochemical and Engineering Aspects, 578(123644). htt
fermented milk. Food Science of Animal Resource, 39(2), 309–323. https://pubmed. ps://www.sciencedirect.com/science/article/abs/pii/S092777571930620X.
ncbi.nlm.nih.gov/31149672/. Ramlia, N. A., Alib, N., Hamzah, S., & Yatim, N. A. (2021). Physicochemical
Mohtashami, L., Ghows, N., Tayarani-Najaran, Z., & Iranshahi, M. (2019). Galbanic acid- characteristics of liposome encapsulation of stingless bees’ propolis. Heliyon, 7(4).
coated Fe3O4 magnetic nanoparticles with enhanced cytotoxicity to prostate cancer e06649 https://europepmc.org/article/PMC/PMC8060604.
cells. Planta Medica, 85, 169–178. https://reference.medscape.com/medline/abst Rashidaie Abandansarie, S. S., Ariaii, P., & Langerodi, M. (2019). Effects of encapsulated
ract/30180257. rosemary extract on oxidative and microbiological stability of beef meat during
Motevalli, S. M., Eltahan, A. S., Liu, L., Magrini, A., Rosato, N., Guo, W., et al. (2019). Co- refrigerated storage. Food Science and Nutrition. 7(12), 3969–3978. https://doi.org/
encapsulation of curcumin and doxorubicin in albumin nanoparticles blocks the 10.1002/fsn3.1258, 11.
adaptive treatment tolerance of cancer cells. Biophysics Reports, 5, 19–30. https://li Rashidi, L., Ganji, F., & Vasheghani-Farahani, E. (2017). Fluorescein isothiocyanate-dyed
nk.springer.com/article/10.1007/s41048-018-0079-6. mesoporous silica nanoparticles for tracking antioxidant delivery. IET
Mousa, D. S., El-Far, A. H., Saddiq, A. A., Sudha, T., & Mousa, S. A. (2020). Nanobiotechnology, 11,454–462. https://pubmed.ncbi.nlm.nih.gov/28530196/.
Nanoformulated bioactive compounds derived from different natural products Rashidi, L., & Khosravi-Darani, K. (2011). The applications of nanotechnology in food
combat pancreatic cancer cell proliferation. International Journal of Nanomedicine, industry. Critical Reviews in Food Science and Nutrition, 51, 723–730. https://pubmed.
15, 2259–2268. https://doi.org/10.2147/IJN.S238256 ncbi.nlm.nih.gov/21838555/.
Murugan, C., Rayappan, K., Thangam, R., Bhanumathi, R., Shanthi, K., Vivek, R., et al. Remanan, M. K., & Zhu, F. (2021). Encapsulation of rutin using quinoa and maize starch
(2016). Combinatorial nanocarrier based drug delivery approach for amalgamation nanoparticles, 2021 Food Chemistry, 128534 (in press) https://www.sciencedirect.
of anti-tumor agents in breast cancer cells: An improved nanomedicine strategy. com/science/article/abs/pii/S0308814620323967.
Scientific Reports, 6(34053). https://www.nature.com/articles/srep34053. Rezaeinia, H., Ghorani, B., Emadzadeh, B., & Tucker, N. (2019). Electrohydrodynamic
Naibaho, J., Safithri, M., & Wijaya, C. H. (2019). Anti-hyperglycemic activity of atomization of Balangu (Lallemantia royleana) seed gum for the fast-release of
encapsulated Javaea-based drink on malondialdehyde formation. Journal of Applied Mentha longifolia L. essential oil: Characterization of nano-capsules and modeling
Pharmaceutical Science, 9, 088–095 https://www.japsonline.com/admin/php/uploa the kinetics of release. Food Hydrocolloids, 93, 374–385. https://www.sciencedirect.
ds/2895_pdf.pdf. com/science/article/abs/pii/S0268005X18321118.
Nandvikar, N. Y., Lala, R. R., & Shinde, A. S. (2019). Nanostructured lipid carrier: The Rodriguez, E. B., Almeda, R. A., Vidallon, M. L. P., & Reyes, C. T. (2019). Enhanced
advanced lipid carriers. International Journal of Pharmaceutical Sciences and Research, bioactivity and efficient delivery of quercetin through nanoliposomal encapsulation
3, 5252–5265. https://doi.org/10.13040/IJPSR.0975-8232.10(12).5252-65 using rice bran phospholipids. Journal of the Science of Food and Agriculture, 99,
Ng, S. W., Selvarajah, G. T., Hussein, M. Z., Yeap, S. K., & Omar, A. R. (2020). In Vitro 1980–1989. https://onlinelibrary.wiley.com/doi/abs/10.1002/jsfa.9396.
Evaluation of Curcumin-Encapsulated Chitosan Nanoparticles against Feline Roshanpour, S., Tavakoli, J., Beigmohammadi, F., & Alaei, S. (2020). Improving
Infectious Peritonitis Virus and Pharmacokinetics Study in Cats. Hindawi BioMed antioxidant effect of phenolic extract of Mentha piperita using nanoencapsulation
Research International, 2020, 1–18. Article ID 3012198 https://www.hindawi.com/jo process. Journal of Food Measurement and Characterization, 15, 23–32. https://link.
urnals/bmri/2020/3012198/. springer.com/article/10.1007/s11694-020-00606-x.
Ochnio, M. E., Martínez, J. H., Allievi, M. C., Palavecino, M., Martínez, K. D., & Rovoli, M., Pappas, I., Lalas, S., Gortzi, O., & Kontopidis, G. (2019). In vitro and in vivo
Pérez, O. E. (2018). Proteins as nano-carriers for bioactive compounds. The case of assessment of vitamin A encapsulation in a liposome-protein delivery system. Journal
7S and 11S soy globulins and folic acid complexation. Polymers, 10, 149–170. of Liposome Research, 29, 142–152. https://doi.org/10.1080/
https://core.ac.uk/display/161411528. 08982104.2018.1502314

15
L. Rashidi Food Bioscience 43 (2021) 101258

Sadeghi, R., Kalbasi, A., Emam-jomeh, Z., Razavi, S. H., Kokini, J., & Moosavi- Tripathi, P. K., Gupta, S., Rai, S., Shrivatava, A., Tripathi, S., Singh, S., et al. (2020).
Movahedi, A. A. (2013). Biocompatible nanotubes as potential carrier for curcumin Curcumin loaded poly (amidoamine) dendrimer-plamitic acid core-shell
as a model bioactive compound. Journal of Nanoparticle Research, 11, 1–11. https://li nanoparticles as anti-stress therapeutics. Journal Drug Development and Industrial
nk.springer.com/article/10.1007/s11051-013-1931-8. Pharmacy, 46(2020). https://doi.org/10.1080/03639045.2020.1724132
Sahoo, A. K., Kanchi, S., Mandal, T., Dasgupta, C., & Maiti, P. K. (2018). Translocation of Vittorio, O., Voliani, V., Faraci, P., Karmakar, B., Iemma, F., Hampel, S., et al. (2014).
bioactive molecules through carbon nanotubes embedded in lipid membrane. Magnetic catechin-dextran conjugate as targeted therapeutic for pancreatic tumour
Applied Materials and Interfaces, 1–30. https://pubs.acs.org/doi/full/10.1021/acsam cells. Journal of Drug Targeting, 22, 408–415. https://doi.org/10.3109/
i.7b18498. 1061186X.2013.878941
Santos, P. D. F., Francisco, C. R. L., Coqueiro, A., Leimann, F. V., Pinela, J., Wang, X., Cheng, F., Wang, X., Feng, T., Xia, S., & Zhang, X. (2021). Chitosan decoration
Calhelha, R. C., et al. (2019). The nanoencapsulation of curcuminoids extracted from improves the rapid and long-term antibacterial activities of cinnamaldehyde-loaded
Curcuma longa L. and an evaluation of their cytotoxic, enzymatic, antioxidant and liposomes. International Journal of Biological Macromolecules, 168, 59–66. https://doi.
anti-inflammatory activities. Food and Function, 10, 573–582. https://bibliotecadigit org/10.1016/j.ijbiomac.2020.12.003
al.ipb.pt/bitstream/10198/20072/1/139.pdf. Wang, P., Li, Y., Zhang, C., Feng, F., & Zhang, H. (2020). Sequential electrospinning of
Savadkouhi, N. R., Ariaii, P., & Charmchian Langerodi, M. (2020). The effect of multilayer ethylcellulose/gelatin/ethylcellulose nanofibrous film for sustained
encapsulated plant extract of hyssop (Hyssopus officinalis L.) in biopolymer release of curcumin. Food Chemistry, 308, 125599–125608. https://www.sciencedir
nanoemulsions of Lepidium perfoliatum and Orchis mascula on controlling oxidative ect.com/science/article/abs/pii/S0308814619317236.
stability of soybean oil. Food science and Nutrition, 8(2), 1264–1271. https://www.nc Xu, F., Ding, L., Tao, W., Yang, X.-Z., Qian, H.-S., & Yao, R.-S. (2016a). Mesoporous-
bi.nlm.nih.gov/pmc/articles/PMC7020334/. silica-coated upconversion nanoparticles loaded with vitamin B12 for near-infrared-
Sayyed-Alangi, S. Z., & Nematzadeh, M. (2019). Formulation, development and light mediated photodynamic therapy. Materials Letters, 167, 205–208. https://www.
evaluation of bifunctionalized nanoliposomes containing Trifolium resupinatum sciencedirect.com/science/article/abs/pii/S0167577X15310600.
sprout methanolic extract: As effective natural antioxidants on the oxidative stability Xu, X., Lü, S., Gao, C., Feng, C., Wu, C., Bai, X., et al. (2016b). Self-fluorescent and
of soybean oil. BMC Chemistry, 13(1), 77. https://doi.org/10.1186/s13065-019- stimuli-responsive mesoporous silica nanoparticles using a double-role curcumin
0594-7 gatekeeper for drug delivery. Chemical Engineering Journal, 300, 185–192. htt
Shi, Y., Zhou, S., Fan, S., Ma, Y., Li, D., Tao, Y., et al. (2021). Encapsulation of bioactive ps://www.sciencedirect.com/science/article/abs/pii/S138589471630537X.
polyphenols by starch and their impacts on gut microbiota. Current Opinion in Food Yan, X., Zhang, X., McClements, D. J., Zou, L., Liu, X., & Liu, F. (2019). Co-encapsulation
Science, 38, 102–111. https://doi.org/10.1016/j.cofs.2020.11.001 of epigallocatechin gallate (EGCG) and curcumin by two proteins-based
Shtay, R., Keppler, J. K., Schrader, K., & Schwarz, K. (2019). Encapsulation of nanoparticles: Role of EGCG. Journal of Agricultural and Food Chemistry, 67(48),
(─)-epigallocatechin-3-gallate (EGCG) in solid lipid nanoparticles for food 13228–13236. https://pubs.acs.org/doi/10.1021/acs.jafc.9b04415.
applications. Journal of Food Engineering, 244, 91–100. https://www.sciencedirect. Yazdi, M. K., Haniloo, A., Ghaffari, A., & Torabi, N. (2020). Antiparasitic effects of
com/science/article/abs/pii/S0260877418303960. Zataria multiflora essential oil nanoemulsion on larval stages of Echinococcus
Solaberrieta, I., Jiménez, A., Cacciotti, I., & Garrigós, M. C. (2020). Encapsulation of granulosus. Journal of Parasitic Diseases, 44, 429–435. https://doi.org/10.1007/
bioactive compounds from aloe vera agrowastes in electrospun poly (ethylene oxide) s12639-020-01193-w
nanofibers. Polymers, 12(6), 1323. https://doi.org/10.3390/polym12061323 Yu, H., Park, J.-Y., Kwon, C. W., Hong, S.-C., Park, K.-M., & Chang, P.-S. (2018). An
Soleimanifar, M., Jafari, S. M., & Assadpour, E. (2020). Encapsulation of olive leaf overview of nanotechnology in food science: Preparative methods, practical
phenolics within electrosprayed whey protein nanoparticles; production and applications, and safety. Journal of Chemistry, 1, 1–10. https://www.hindawi.com/
characterization. Food Hydrocolloids, 101(105572). https://www.sciencedirect. journals/jchem/2018/5427978/.
com/science/article/abs/pii/S0268005X19317898. Yu, J. Y., Roh, S. H., & Park, H. J. (2021). Characterization of ferulic acid encapsulation
Song, X., Gan, K., Qin, S., Chen, L., Liu, X., Chen, T., et al. (2019). Preparation and complexes with maltodextrin and hydroxypropyl methylcellulose. Food
characterization of general-purpose gelatin-based co-loading flavonoids nano-core Hydrocolloids, 106390. https://doi.org/10.1016/j.foodhyd.2020.106390
structure. Scientific Reports, 9, 6365–6376. https://pubmed.ncbi.nlm.nih.gov/ Zhang, H., Fan, Q., Li, D., Chen, X., & Liang, L. (2019). Impact of gum Arabic on the
31019215/. partition and stability of resveratrol insunflower oil emulsions stabilized by whey
Summerlin, N., Qu, Z., Pujara, N., Sheng, Y., Jambhrunkar, S., McGuckin, M., et al. protein isolate. Colloids and Surfaces B: Biointerfaces, 181, 749–755. https://doi.org/
(2016). Colloidal mesoporous silica nanoparticles enhance the biological activity of 10.1016/j.colsurfb.2019.06.034
resveratrol. Colloids and Surfaces B: Biointerfaces, 144, 1–7. https://pubmed.ncbi.nlm Zhang, H., Jiang, L., Tong, M., Lu, Y., Ouyang, X.-K., & Ling, J. (2021). Encapsulation of
.nih.gov/27060664/. curcumin using fucoidan stabilized zein nanoparticles: Preparation,
Sun, L., Lu, L., Qiua, X., & Tang, Y. (2017). Development of low-density polyethylene characterization, and in vitro release performance. Journal of Molecular Liquids, 329
antioxidant active films containing α-tocopherol loaded with MCM-41(Mobil (115586). https://doi.org/10.1016/j.molliq.2021.115586
Composition of Matter No. 41) mesoporous silica. Food Control, 71, 193–199. htt Zhou, W., Cheng, C., Ma, L., Zou, L., Liu, W., Li, R., et al. (2021). the formation of
ps://www.sciencedirect.com/science/article/abs/pii/S0956713516303322. chitosan-coated rhamnolipid liposomes containing curcumin: Stability and in vitro
The Food and Drug Administration (FDA). (2014). Final guidance for industry - assessing digestion. Molecules, 26(3). https://doi.org/10.3390/molecules26030560, 560.
the effects of significant manufacturing process changes, including emerging Zhuoxian, M., Jiali, C., Yang, H., Fang, L., Bin, F., Hongwu, Z., et al. (2017). Novel
technologies, on the safety and regulatory status of food ingredients and food contact functional mesoporous silica nanoparticles loaded with Vitamin E acetate as smart
substances. Including Food Ingredients that are Color Additives, 36533–36534. FDA- platforms for pH responsive delivery with high bioactivity. Journal of Colloid and
2011-D-0490. Interface Science, 508, 184–195. https://www.sciencedirect.com/science/artic
Thepphankulngarm, N., Wonganan, P., Sapcharoenkun, C., Tuntulani, T., & Leeladee, P. le/abs/pii/S0021979717307944.
(2017). Combining vitamin B12 and cisplatin-loaded porous silica nanoparticles via Zorkina, Y., Abramova, O., Ushakova, V., Morozova, A., Zubkov, E., Valikhov, M., et al.
coordination: A facile approach to prepare a targeted drug delivery system. New (2020). Nano carrier drug delivery systems for the treatment of neuropsychiatric
Journal of Chemistry, 167(22), 205–208. https://pubs.rsc.org/en/content/arti disorders: Advantages and limitations. Molecules, 25(22), 5294–5348. https://doi.
clelanding/2017/NJ/c7nj02754k#!divAbstract. org/10.3390/molecules25225294
Tiruwa, R. (2015). A review on nanoparticles –preparation and evaluation parameters.
Indian Journal of Pharmaceutical and Biological Research, 4(2), 27–31. www.ijpbr.in.

16

You might also like