Skip to main content

REVIEW article

Front. Nutr., 05 December 2022
Sec. Nutrition and Food Science Technology
This article is part of the Research Topic Nanostructured Steady-state Systems for Nutrient and Bioactive Compounds Delivery View all 5 articles

Nanocarrier system: An emerging strategy for bioactive peptide delivery

\nXu ZhangXu ZhangXinshe LiXinshe LiYunhao ZhaoYunhao ZhaoQing ZhengQing ZhengQiang Wu
Qiang Wu*Yougui Yu
Yougui Yu*
  • College of Food and Chemical Engineering, Shaoyang University, Shaoyang, China

Compared with small-molecule synthetic drugs, bioactive peptides have desirable advantages in efficiency, selectivity, safety, tolerance, and side effects, which are accepted by attracting extensive attention from researchers in food, medicine, and other fields. However, unacceptable barriers, including mucus barrier, digestive enzyme barrier, and epithelial barrier, cause the weakening or the loss of bioavailability and biostability of bioactive peptides. The nanocarrier system for bioactive peptide delivery needs to be further probed. We provide a comprehensive update on the application of versatile delivery systems for embedding bioactive peptides, including liposomes, polymer nanoparticles, polysaccharides, hydrogels, and self-emulsifying delivery systems, and further clarify their structural characterization, advantages, and disadvantages as delivery systems. It aims to provide a reference for the maximum utilization of bioactive peptides. It is expected to be an effective strategy for improving the bioavailability and biostability of bioactive peptides.

Introduction

Recently, researchers have focused on bioactive peptides from various origins, including animal (1), plant (2, 3), marine organisms (4, 5), bean products (6), milk (7), and fermented products (8). They are generally composed of 2–50 amino acid residues in different combinations and arrangements forming linear or cyclic structures (9). In addition, bioactive peptides have a variety of human body metabolism and physiological regulation functions, which are easy to digest and absorb, and have the effects of promoting immunity, hormone regulation, antibacterial, antiviral, decreasing blood pressure, and reducing blood lipid (911). They are widely used in food, medicine, and other fields. Particularly, it is the most popular research topic and functional factor with great development prospects in the current international food industry.

Bioactive peptides play an important role in cancer treatment, new drug screening, vaccine development, and nutritional drug delivery and are considered desired substitutes for drugs. Compared with macromolecular proteins, bioactive peptides have the advantages of high activity, tissue permeability, and weak immunogenicity under the same unit mass conditions. Compared with small molecular substances, bioactive peptides have stronger potency, selectivity, specificity, and lower cytotoxicity and drug interactions (12). Oral administration is a direct and effective way of bioactive peptide administration. However, after oral administration, bioactive peptides may be degraded by gastric acidic secretion, digestive enzymes, intestinal brush border membrane, and intestinal epithelial cell endopeptidase in the gastrointestinal tract (13). During oral administration, it is possible that bioactive peptides are hydrolyzed by 40 kinds of enzymes at least, causing subsequent loss of bioactivity before reaching systemic circulation. It was reported that most food protein-derived bioactive peptides containing more than two–three amino acid residues do not withstand simulated gastrointestinal enzymatic digestion (14). Although dipeptides and tripeptides can be fully absorbed through the synergistic transport mechanism of peptide transporters (PepT1 and PepT2), and the brush border membrane Na+/ H+ exchange transport system, or enter cells through endocytosis and cell bypass (15) as shown in Figure 1, it is still degraded into free amino acids by peptidases in blood and lose bioactivity (16). We studied the gastrointestinal digestion stability of bioactive peptides and found that the peptide composed of amino acid residues such as Phe, Tyr, Val, Leu, Arg, and Lys were not tolerant to gastrointestinal fluid digestion (5, 17, 18). It was reported that after intravenous injection of angiotensin-converting enzyme (ACE) inhibitory peptide Ile-Val-Tyr in the spontaneously hypertensive rat (SHR), it can be degraded into dipeptide Val-Tyr by the aminopeptidase existing in the blood. After intravenous injection, Ile-Val-Tyr had a better blood pressure reduction effect and could maintain for 15 min. Val-Tyr alone had an acute hypotension effect, but the blood pressure returned to normal after 5 min of injection (19). Therefore, the bioavailability of bioactive peptides depends directly on their amino acid sequence. When entering the body to perform an anti-hypertensive effect, it is likely to be degraded and weakened or lost activity.

FIGURE 1
www.frontiersin.org

Figure 1. Three barriers to bioactive peptide absorption.

The undesirable bioavailability of bioactive peptides has prompted researchers to explore efficient methods for complete delivery. Encapsulation of bioactive peptides in nanoparticles is an effective way to prevent their degradation and enhance their bioactivity (20). Many nanocarrier systems have been developed, including lipid particles, polysaccharide particles, biopolymer nanoparticles, and colloidal particles, each of which has its own advantages and limitations (21). There are various administration ways including subcutaneous injection, intravenous injection, and oral administration. In consideration of bioactive peptides derived from food resources, in general, used as functional foods and nutritional supplements, oral administration is preferential (22). Therefore, this study reviewed oral delivery systems of bioactive peptides. Furthermore, the structural characterization and application status of these delivery systems were summarized, and their advantages and disadvantages were analyzed, in order to provide a reference for the application of bioactive peptides.

Liposome carrier applied in bioactive peptide delivery

Structural characterization of the liposome delivery system

Liposome, as an efficient organic carrier, is a kind of closed vesicle similar to a biofilm structure formed by the inclusion of phospholipid and cholesterol. When the liposome is dispersed in the aqueous phase, the hydrophobic groups will spontaneously aggregate together under the hydrophobic interaction, and the hydrophilic groups will also aggregate with each other. When the system is stable, the closed ring multilayer structure of “head to head, tail to tail” will be formed (Figure 2A). The interactions between peptide and liposome depend on the hydrophobicity of the peptide, and the structure of the interfacial head-group region of the lipid bilayers of liposome was also important (23). Phospholipid-forming liposome is eliminated slowly in the blood, so drugs loaded on liposome remain in the blood circulation system for a long time. In addition, liposomes can dissolve in the cell membrane of the target organs or tissue to fulfill intracellular transportation of drugs and, thus, improve the bioavailability and stability of drugs. Liposomes contain polar, nonpolar, and amphiphilic regions in the same colloidal particles, so they are particularly suitable for the encapsulation of active peptides (24). The liposome has an aqueous solution core surrounded by a hydrophobic membrane in the form of a lipid bilayer. Hydrophilic solutes dissolved in the nucleus are not easily associated with the bilayer by hydrophobic chemicals. Therefore, liposomes can load hydrophobic and/or hydrophilic molecules. Moreover, amphiphilic peptides locate on the interface between the shell and core of the liposome structure, which would interact with the hydrophobic and hydrophilic amino acid residues, respectively. The liposome is similar to cell membranes and is favorable for the delivery of bioactive compounds, which can otherwise be degraded by the physiological environment (25).

FIGURE 2
www.frontiersin.org

Figure 2. Typical structure of bioactive peptide carriers. (A) Liposome. (B) Polymeric nanoparticles. (C) Polysaccharides nanomaterials. (D) Hydrogels.

Practical application of the liposome delivery system

The liposome delivery system has been used to package, protect, and deliver bioactive peptides for the prevention or treatment of diseases in the pharmaceutical industry (26). KRX29 is a small cyclic peptide with potential pathological effects in the treatment of heart failure, which is an effective and selective G protein-coupled receptor kinase 2 (GRK2) activity inhibitor. Although KRX29 presents a small molecular weight (eight amino acids) and can be performed in oral and intravenous administration, it has certain disadvantages including low absorption, unstable physiological environment, and short half-life. It needs to be wrapped in suitable carriers to overcome these limitations. It was found that the bioavailability and stability of drugs could be improved by wrapping into the nano-sized liposomes by thin film hydration and then by ultrasonic-assisted size reduction process (27). In addition, the encapsulated Arg-Leu-Ser-Phe-Asn-Pro (RLSFNP), a hexapeptide milk-derived angiotensin-converting enzyme (ACE) inhibitory peptide, into liposomes with remarkable sustained release and storage capacity can be directly absorbed by Caco-2 cells (28). It has also been found that muramyl peptides encapsulated in liposomes can kill macrophages more effectively than unencapsulated ones (29). Rezaei et al. found that the mutant N-terminal peptide with a zinc binding ring replaced by a disulfide bond ring (ES-SS peptide) has anti-angiogenesis and antitumor properties compared with natural peptides. In order to improve the stability and plasma half-life of ES-SS peptide, ES-SS peptide nanoliposomes with different phosphatidylcholine (PC) ratios were synthesized. ES-SS peptide was gradually released by liposomes. Compared with free peptides, liposome peptide preparation has a higher cell survival rate (30). Heuts et al. (31) studied the possibility of carrying synthetic long peptide (SLP) antigen on liposomes based on 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) and further verified the potential of DOTAP liposomes as a vaccine delivery platform for therapeutic cancer vaccines. Another study also found that P5HER-2/neu derived peptide (P5) in DOTAP-cholesterol drug liposomes injected with CpG-ODN significantly enhanced the response of cytotoxic T cells and highly inhibited the progression of tumors (32). The major problems limiting the widespread use of liposomes are stability, encapsulation efficiency, and lysosomal degradation. Chemical instability may be caused by the hydrolysis of ester bonds and/or oxidation of unsaturated acyl chains of lipids. Liposomal drug formulation could only be affirmative if the encapsulation efficiency is such that therapeutic doses could be delivered in a reasonable amount of lipid, since lipids in high doses may be toxic and also cause non-linear (saturable) pharmacokinetics. Liposomes may deliver the drug to the cells selectively, but the pharmacological activity depends on the ability of the intact drug against degradation to diffuse into the cytoplasm from the endosomes in sufficient amounts (33, 34).

Micelles are unilamellar vesicles made up of amphiphilic polymeric molecules with a hydrophobic core hidden inside the structure and a hydrophilic shell directed outwards with a size range of 10–100 nm (35). Drugs are loaded into micelles either through chemical covalent bonding or through physical encapsulation. Micelles have many advantages as drug delivery carriers, including ease of preparation, stability under physiological conditions, prolonged shelf life, extended circulation time in blood and biological fluids, increased bioavailability, and enhanced pharmacokinetics and biodistribution properties (36, 37). The peptide amphiphile micelles are small spherical particles able to stably associate with cells, making them attractive for the targeted delivery via the vasculature, and induce significant peptide-based toxicity in human lymphoma cells (38). Peptide amphiphile micelles (PAMs) are attractive vehicles for the delivery of a variety of therapeutic and prophylactic peptides (39). Furthermore, one cleavage-inducing target of interest could be glutathione, the primary antioxidant in the body, which is present in low concentrations in the blood, but much higher concentrations within cells (4042). In the food industry, liposomal delivery of proteins, enzymes, vitamins, antioxidants, and seasonings has been studied (43). It was reported that encapsulation of ghrelin (a bioactive polypeptide) into liposomes can improve its chemical stability and increase the retention time of this appetite-suppressing hormone in the blood (44). Liposomes are also used to encapsulate bioactive peptides extracted from hydrolyzed fish protein (45). Liposome encapsulation has been proven to be an appropriate method to reduce the bitterness of bioactive peptides (46). Montero et al. wrapped the shrimp polypeptide component (ST1) with biological activity, including ABTS free radical scavenging ability, angiotensin-converting enzyme activity, and dipeptidyl polypeptidase-IV inhibitory activity, into partially purified soybean phospholipid nanoliposome (L-ST1). The soybean-derived phospholipid-forming liposomes can be bound to the edible membrane of sodium caseinate without significantly losing the integrity of the vesicle structure, thereby increasing the water solubility of the liposome delivery system. Furthermore, the increase in membrane humidity and the presence of liposomes are beneficial to the adhesion of the membrane, especially when they are loaded with polypeptides (47). It has shown that the inclusion of bacteriocins in liposomes can control food spoilage and pathogens and increase the effective safety and shelf life of some foods, as antimicrobial agents can coexist in the aqueous and lipid phases of liposomes (48). The large-scale production of liposome carry is a distinct advantage compared with other carrier systems. Liposomes adapted from the pharmaceutical industry have certain shortcomings in functional food applications. Particularly, the thermal instability of liposome-encapsulated food-derived peptides beyond the phase transition temperature of the phospholipid can limit their incorporation in thermally processed food (49).

The characteristics for applications of liposomes are shown in Table 1. Compared with other embedding techniques, the dominant advantage of the liposome delivery system is that they endow the substances with water-soluble and stability in high water activity (50). Liposomes can also protect the drugs against degradation and integrally release them into target organs or tissues that need to be treated so that the drug concentration in these target organs or tissues is increased, and the therapeutic effect of drugs can be improved. In addition, a drug-loaded liposome delivery system presents resistance to metabolism and degradation by intracellular enzymes, less adverse reaction, and benefits on biocompatibility, targeting ability, and controlled release of the drug. Liposomes have been shown to be stable in embedding materials free from a range of environmental and chemical changes, including enzyme and chemical modifications, as well as buffers against extreme pH and temperature (51). The phospholipids making up liposomes can be commonly found in nature, such as in soybean and egg yolks. However, these are difficult to use in clinical settings due to stability and contamination risks (52).

TABLE 1
www.frontiersin.org

Table 1. Application and characterization of liposomes.

Solid lipid nanoparticles applied in bioactive peptide delivery

Structural characterization of solid lipid nanoparticle

Solid lipid nanoparticle (SLN) is made of natural, semi-synthetic, or synthetic lipids, which contain triglycerides, fatty acids, phospholipids, and steroids, and is widely considered a safe and biodegradable carrier (53). SLNS is a promising technology for the targeted delivery of anticancer drugs (54). The main disadvantage of SLNS is that the encapsulation efficiency is affected by the polymorph structure of solid lipids. If SLN is composed of high-purity single lipids, a perfect lattice may be formed during storage, which may reduce the solubility of drugs in the lipid matrix and eventually lead to the expulsion of encapsulated drugs. When SLN forms, lipids crystallize in disordered α and β crystal structures. During storage, lipids are gradually arranged in a more stable and orderly manner, resulting in β′ and β crystalline forms from which encapsulated drugs can be discharged (55). Drug-loaded SLN combines the best characteristics of polymer nanocarrier and liposome delivery system. It maximizes drug targeting and creates uniform particle distribution, thus providing low toxicity and good histocompatibility, but it is easy to be degraded. Therefore, the utilization efficiency is poor, and it cannot guarantee that the embedded SLN can reach the target organ and tissue and then release it (Table 2).

TABLE 2
www.frontiersin.org

Table 2. Application and characterization of solid lipid nanoparticles.

Practical application of solid lipid nanoparticle

Dumont et al. found that SLN is a promising system to overcome the inherent barriers of stability and bioavailability of the peptide by oral administration. It has been proved that SLN can encapsulate peptides after lipidation without peptide denaturation (56). Christophersen et al. (57) studied the degradation mechanisms of peptide-loaded SLNs containing different types of lipid excipients and found that the release rate became faster after lysozyme was added. However, the hydrophobicity of SLN limits the encapsulation of hydrophilic peptides. Su et al. (58) found that SLN could effectively maintain the stability of bioactive peptides in gastrointestinal digestion. In addition, the macromolecular peptide could be successfully wrapped in SLN and nanocrystallized, thereby improving their physical and chemical stability. In particular, the encapsulation in SLN protects the secondary hydrolysis of bioactive peptides in simulated gastrointestinal fluid, which helps maintain their dipeptidyl peptidase IV (DPP4) inhibitory function. It has encapsulated water-soluble peptides in biodegradable and biocompatible SLN, but the encapsulated peptides are easily released in simulated gastrointestinal fluid in the presence of proteases (59). SLN is a potential carrier system for transporting protein-type antigen vaccines while avoiding the use of organic solvents (60). Kadari et al. (61) found that angiopep-2-modified SLN was an excellently targeted drug delivery system for glioma therapy. SLN could be used to transport antioxidant glutathione (GSH) to immunocompetent fish cells, and the antioxidant activity of GSH in vitro is stable in the transport process (62).

Solid lipid nanoparticle-engineered nanostructured lipid carrier is formulated. Insulin and insulin analog—glargine insulin—are entrapped in the lipid matrix through hydrophobic ion pairing. Studies in vitro and in vivo were carried out by employing fluorescently labeled peptides. The superiority of glargine insulin-loaded nanostructured lipid carrier demonstrated significantly higher permeation (till 30% dose/ml) compared to the free peptide (63). Xu et al. (64) developed functional nanocarriers for efficient oral administration of biological macromolecules, and the developed SLN was proved to increase serum insulin concentration and produced good hypoglycemic reaction. In addition, the prepared insulin-loaded cationic solid lipid nanoparticle (CSLN) by water-in-oil emulsion technology can protect the encapsulated insulin from the degradation of pepsin and trypsin and improve its biostability (65). Hydrophobic ion pairing of leuprolide and insulin, chosen as model peptides, allowed the encapsulation of these molecules in SLN produced through the coacervation technique, nanoparticles in vitro can be used as a polypeptide sustained release system, maintaining the integrity of the peptide (66). SLN prepared by thermo-high pressure homogenization method has the advantages of good physical stability, protection of mixed unstable active substances, and controlled release, which is very suitable as a carrier for transporting active components to the skin. Furthermore, SLN technology allows polypeptides to enter the skin to play a protective role (67).

Since the basic template of SLNs is a solid lipid, lipophilic drugs have more affinity toward SLNs than hydrophilic ones. For the preparation of SLNs with lipophilic drugs, oil-in-water (O/W) emulsions are preferred. For the encapsulation of hydrophilic drugs, multiple emulsion systems such as water/oil/water (W/O/W) emulsion with or without the use of organic solvents are reported (68). Furthermore, water-in-oil high internal phase emulsions (W/O HIPEs) could be successfully used to encapsulate bitter peptides, which reduced their bitterness and improved their gastric stability (69). Double emulsion of the water-in-oil-in-water (W1/O/W2) type is one of the carrier systems used for encapsulation, protection, and delivery of both hydrophilic and hydrophobic active components (70). Recently, double emulsions have been studied in the delivery of hydrophilic bioactive compounds in the food and pharmaceutical industries (71, 72). Among the different bioactive peptide concentrations (1%−20%), 5% of bioactive peptides was determined to be the amount to encapsulate in the W1 phase in the double emulsion that would ensure emulsion droplet stability (72).

The self-emulsifying carrier applied in bioactive peptide delivery

Structural characterization of self-emulsifying drug delivery system

Self-emulsifying drug delivery system (SEDDS) is a lipid carrier, which is spontaneously formed by the isotropic mixture of oil, surfactant, and cosurfactant through mixing with water (73). SEDDS has high biocompatibility, which can protect polypeptides from degradation and increase membrane permeability. It has been widely used to improve the oral bioavailability of insoluble drugs, which can help overcome the obstacles encountered in the oral administration of hydrophilic macromolecules, namely, intestinal protease degradation, poor mucus permeability, and low cellular uptake (74). It has been proved that pepsin, trypsin, chymotrypsin, elastase, and other hydrolases are not soluble in oily SEDDS droplets. Therefore, due to the hydrophilicity of peptidases and proteolytic enzymes, they cannot enter the lipid droplets of SEDDS containing bioactive peptides, which can protect them against enzymatic degradation (75, 76). Compared with other nanocarriers, SEDDS preparation is simpler and more cost-effective. At present, the self-emulsifying method has been successfully applied to the transportation of anticancer drugs, antiviral drugs, antibacterial drugs, immunosuppressants, and natural products such as antioxidants and alkaloids (Table 3).

TABLE 3
www.frontiersin.org

Table 3. Application and characterization of self-emulsifying drug delivery systems.

Practical application of self-emulsifying drug delivery system

Self-emulsifying drug delivery system is discovered for the oral administration of hydrophilic drugs such as peptides, proteins, polysaccharides, and pDNA (77). Due to hydrophobic ion pairing (HIP) with oppositely charged lipophilic auxiliary agents, the resulting complexes can be incorporated into the lipophilic phase of SEDDS. Depending on the solubility of the complex in the pre-concentrate SEDDS, drug release can be adjusted on purpose by choosing more or less lipophilic auxiliary agents in appropriate quantities for the HIP. Within the oily droplets formed in the gastrointestinal tract, drugs are protected from degradation by proteases and nucleases and thiol-disulfide exchange reactions with dietary proteins. The oily droplets can be made mucoadhesive or highly mucus permeating depending on their target site. Furthermore, it can be tuned by adjusting their zeta potential or decorating them with cell-penetrating peptides (74).

Sustained release of bioactive peptides from SEDDS seems essential to avoid their degradation in the gastrointestinal tract. SEDDS has been shown to penetrate effectively into the intestinal mucus gel layer, making peptides or protein drugs closely in contact with the absorption membrane (73). Compared with other drug delivery systems, SEDDS is the most effective mucus permeation system. In vitro studies showed that the penetration of exenatide containing SEDDS was 2.7 times higher than that of drugs without SEDDS. In vivo evaluation experiment showed that compared with the control group, the area under plasma concentration curves (AUC) of blood glucose in the SEDDS group was decreased by 20.6% (78). Zupančič et al. (79) found that SEDDS showed mucus permeability and had protective effects on the degradation of trypsin, α-chymotrypsin, elastase, simulated gastric juice, and simulated intestinal juice. Mahmood et al. added hydrophilic macromolecular drugs into SEDDS, which could protect them from the enzyme and thiol barriers in the gastrointestinal tract. In addition, SEDDS can penetrate the mucus gel barrier in a relatively effective way to promote the transport of the combined hydrophilic macromolecular drugs to the underlying epithelial cells (74).

Polymeric nanoparticles applied in bioactive peptide delivery

Structural characterization of polymeric nanoparticle

Polymeric nanoparticle (NP) refers to a solid particle with a size range from 10 to 1,000 nm, which can encapsulate drugs in the polymer matrix to prevent them from being degraded by enzymes (Figure 2B). Compared with liposome and SLN, NP also showed higher stability in biological fluids (Table 4). In addition, it is a promising delivery system for proteins and peptides because of the versatility, sustained release, protection of encapsulated proteins, and peptides from enzymatic degradation and biocompatibility of nanoparticles (80). Polymers have great functionalization ability and can be used to couple targeted ligands to improve biological distribution (81). NP can also compensate for the disadvantages of peptide treatment by increasing bioavailability, exhibiting controlled and sustained release, and enhancing cellular uptake (82). In addition, compared with other particles, NPs can effectively transfer across the epithelial surface (83). The polymer matrix can protect peptides from degradation and provide a desirable controlled release of peptides (84).

TABLE 4
www.frontiersin.org

Table 4. Application and characterization of polymeric nanoparticles.

Polylactic-glycolic acid copolymer (PLGA) is a very important synthetic polymer material for preparing polymeric nanoparticles, which can achieve safe and effective delivery of drugs without causing tissue damage (85, 86). It has been confirmed that PLGA-based nanoparticles (PLGANPs) have a smaller particle size, better environmental adaptability, and biocompatibility than hydrogels, alginates, and chitosan (CS) polymer-coated particles (87). In addition, the adhesion and cell uptake of PLGANPs can be significantly improved by functionalizing positively charged polymers or proteins on the surface of particles (8890).

Rastogi et al. used a modified s/o/w method to prepare insulin-loaded polycaprolactone-polymer poly (ethylene glycol)-polycaprolactone (PCL-PEG-PCL) nanoparticles in order to obtain higher drug loading. Insulin was hydrophobically modified by sodium deoxycholate. After pairing with sodium deoxycholate, the lipophilicity of insulin was increased by five times, and the encapsulation efficiency was increased by 10%−50% (91). In addition, polypeptide encapsulated with nanoparticles based on polymer poly (ethylene glycol)-block-polycaprolactone (PEG-b-PCL) presented the cytotoxicity induced by polyglutamine (80). Furthermore, Kelly et al. (92) screened and developed porous silicon nanoparticle (PSNP) composites for micRNA inhibition of intracellular delivery of peptide nucleic acid (PNA).

Practical application of polymeric nanoparticle

A recent study has shown that polyethylene glycol (PEG) embedding can be used as a protective method or a ligand competition for antimicrobial peptide BP100 transposition (93). It is suggested the feasibility of applying antimicrobial peptides carried on PEG-capped NPs for possible treatments of lung diseases. The anticancer peptide NuBCP-9 was encapsulated in NPs to enhance efficacy and avoid drug resistance, which was enhanced by increasing the PEG chain length in the block copolymers (94). Hoshino et al. confirmed that NPs accelerated the clearance of toxic peptides, which is a 26 amino acid cytolytic peptide isolated from bee venom, and finally accumulated in liver macrophages. PEG hydrophilic polymer particles also help to improve the biocompatibility of synthetic NPs and enhance their stability in plasma, revealing the potential and possible limitations of using synthetic ones as a plastic antidote (95). Cellulose nanocrystal (CNC) is a natural, non-toxic, and neutral material with a rod-like or spherical construction. It has been applied in various fields such as pharmaceuticals, food industry, paper industry, composites, and polymer composites (96). Cellulose-based composite macrogels made by cellulose fiber/cellulose nanofiber (CCNM) are used as an intestine delivery vehicle for probiotics (97).

Polysaccharide nanomaterial applied in bioactive peptide delivery

Structural characterization of polysaccharides nanomaterial

Polymer-based nanocarriers can be divided into natural polymers and synthetic polymers. Chitosan (CS) is a kind of natural polysaccharide isolated from the excrement of crustaceans and fungi (98). Most polysaccharides contain hydrophilic groups such as hydroxyl, carboxyl, and amino, which can form noncovalent bonds with intestinal mucus and promote the absorption of bioactive peptides (Figure 2C). As bioactive peptides contain amphipathic substances (that harbor both negatively and positively charged molecules) similar to proteins, they can also electrostatically interact with ionic polysaccharides with positive or negative charge to form associative complexes (99). Due to their effective biological activity, non-toxic, hydrophilic, good biocompatibility and biodegradability, and extensive diversity in structure and properties (100), polysaccharide-based nanomaterials are widely used in intracellular drug delivery as one of the most practical materials in nanomedicine field (Table 5).

TABLE 5
www.frontiersin.org

Table 5. Application and characterization of polysaccharide nanomaterials.

Practical application of polysaccharides nanomaterial

The CS-based polysaccharide nanomaterial is prepared by the interaction between positively charged CS and negatively charged chlorinated polyethylene (PEC) through charges. It is considered to be a mild drug delivery method because no organic solvent or ultrasonic treatment is required (101). Yu et al. (102) developed a new type of CS-protamine nanoparticles for the anticancer effect of nuclear-targeted delivery of 5-fluorouracil (5-FU). Compared with free 5-FU, 5-FU-CS-protamine nanoparticles had the strongest cytotoxicity on tumor A549 cells and HeLa cells. In addition, an increase in apoptosis rate was also observed. It is worth noting that 5-FU-CS-protamine NPS can significantly reduce the tumor volume of BALB/c HeLa cell xenograft mice. Gao et al. (103) studied the combined delivery of irinotecan (IRN) and 5-FU by chitosan-hyaluronic acid (CS-HA) hybrid nanoparticles for the targeted treatment of gastric cancer. IRN/5-FU-CS-HA-NPs enhanced the killing effect on human gastric cancer cells in vitro, enhanced the antitumor activity in vivo of tumor-bearing mice, and had good antitumor activity. On the other hand, under extreme conditions, such as high temperature, the polysaccharide wall is susceptible to reacting with the peptide to form complex products (e.g., Maillard reaction products), which can be potentially toxic and also deplete the bioactive peptides. In order to circumvent this challenge, the reactive functional groups of polysaccharides have been modified such as carboxymethylation to produce relatively inert carriers. The molecular structure of polysaccharides contributes to their stability as carriers during the production and processing of encapsulated products (104, 105). Nasri et.al encapsulate goby fish protein hydrolysate (GPH), endowed with antioxidant activity, through an ionic gelation process using blue crab chitosan and tripolyphosphate anions and to evaluate the structural, thermal, and antioxidant properties of the elaborated microparticles. The increase in loaded GPH concentration led to an increase in encapsulation efficiency and a reduction in particle size. Furthermore, thermogravimetric analysis (TGA) profiles revealed the enhanced thermal stability of encapsulated biopeptides compared to the free ones (106).

Chitosan is a cationic polymer that allows electrostatic interaction with negatively charged sialic acid residues on the mucosal surface (107). Based on this principle, PLGA and FA-modified CS (FA-CS) composite nanocarriers were prepared by the electrostatic self-assembly method. The system can protect insulin against digestive enzymes and reduce the toxicity to HT-29 cells. In vivo experiments further showed that the system had an obvious hypoglycemic effect on diabetic mice, and the prepared composite nanocarriers were expected to become drug carriers for oral insulin and other biological macromolecules (108). Fish-purified antioxidant peptide (AOP)-loaded chitosan nanoparticles (CSNPs) were synthesized for Fourier transform infra-red (FT-IR) analysis, which demonstrated that the electrostatic interactions between biomolecule and polymer were considered as the driving force for peptide-loaded NPs formation (109). Similarly, sulfhydryl-modified low-molecular-weight chitosan was anchored to the lysine residue of insulin. These protein/chitosan conjugates have also proved to protect proteins against proteolytic enzymes, resulting in increased oral bioavailability of protein (110). PLGA covered with glycol-chitosan was used to encapsulate an HIV fusion inhibitor peptide (E2). Furthermore, the assessments ex vivo and in vivo were carried out to examine the ability of NPs to reach the vaginal epithelium. The penetration rate was measured in a swine model using confocal microscopy. E2-loaded NPs have interesting physicochemical and morphological features that provide them with mobility across the mucus, to reach the vaginal epithelium and release E2 (111). The W9-peptide, a tumor necrosis factor (TNF), was used to study the feasibility of cholesterol-bearing pullulan (CHP) nanogel as the drug delivery system. It found CHP nanogel could form a complex with the W9-peptide and prevents its aggregation in vitro (112). It suggested that the CHP nanogel worked as a suitable carrier for the W9 peptide. Santos et al. found that antimicrobial peptide (AMP) nisin has a wide range of action against pathogens and can treat diabetic foot ulcers (DFU) in patients with diabetes. However, AMP will be degraded or inactivated before reaching the target at the treatment concentration. Natural polysaccharide guar gum gel as the delivery system loading AMP can be used to treat systemic bacterial skin infections, especially those promoted by pathogens so that the sensitivity of bacteria to current antibiotics is reduced (113).

Hydrogel delivery system

Structural characterization of hydrogel delivery system

Hydrogel has three-dimensional networks of water swelling made of polymers, proteins, small molecules, or colloids (Figure 2D), and the obtained hydrogels show ultra-stable properties in highly acidic or alkaline aqueous solutions (114). The network of a hydrogel is established via covalent bonds or noncovalent interactions. Noncovalent interactions mainly include physical entanglements, hydrogen bonds, hydrophobic interactions, supramolecular interactions, electrostatic interactions, and coordination interactions (115). Hydrogel has adjustable physical properties. It prevents the degradation of unstable drugs as an effective drug delivery system. The versatility and diversity of hydrogels extend their applications to targeted drug delivery, wound dressings, contact lenses, and tissue engineering (Table 6). They contain 90% water and are highly porous to accommodate drugs for delivery and promote the controlled release (116).

TABLE 6
www.frontiersin.org

Table 6. Application and characterization of hydrogels.

Practical application of hydrogel delivery system

They developed a novel melittin-RADA28 (MR) hydrogel, composed of RADA28 and melittin, in order to promote the membrane permeability of tumor cells with the membrane-disrupting ability of melittin. It demonstrated that the formed melittin-RADA28-Lys-Leu-Ala-Lys-Leu-Ala-Lys-Lys-Leu-Ala-Lys-Leu-Ala-Lys (KLA) peptide (MRP) hydrogel has a nanofiber structure, sustained release, and attenuated hemolysis effects. Compared with free KLA, the MRP hydrogel markedly increased the cellular accumulation of KLA, produced the highest ratio of depolarized mitochondrial membrane, and decreased cell viability in vitro (117). The antibacterial effect of different concentrations of RADA16 on Staphylococcus aureus, Candida albicans, and Escherichia coli were compared. It was found that 0.25% of peptide-loaded RADA16 hydrogel could play a good antibacterial activity in a short time (118). The stable structure formed by RADA16 hydrogel provided an important carrier platform for the continuous release of antimicrobial peptides, which not only inhibited the reproduction of S. aureus, but also promoted the proliferation of bone marrow mesenchymal stem cells to a certain extent, and enhanced their osteogenic ability (119).

Dextran nanogels can be exploited as carriers for sustained/controlled release of bioactive molecules. Molinos et al. prepared a new bidimensional composite hydrogel, made of oxidized dextrin incorporating dextrin nanogel (oDex nanogel). It provides continuous delivery of protein and prevents problems of cyclic variations in the protein concentration in the blood with time. In addition, it offers maximum pharmacological efficiency at a minimum drug dose, reducing administration frequency and improving patient compliance with the therapy (120). Hirakura et al. reported a novel injectable hybrid hyaluronan (HA) hydrogel system of physically encapsulated CHP nanogels for sustained delivery of protein. Furthermore, a new hybrid hydrogel composed of CHP nanogels presented chaperone-like protection for fragile protein drugs and chemically cross-linked HA as the biocompatible controlled release matrix. CHP/protein complex nanogels were released from the hybrid hydrogels in a sustained manner in vitro and in vivo (121). The main challenge is to design a hydrogel system capable of providing drug release for several months and complete clearance at the end of drug release or in a relatively short period of time to avoid matrix accumulation (122).

Concluding remarks and future perspectives

The application of nanotechnology in biomedicine and functional food to improve human health is indispensable. The properties of nanocarriers with different materials can meet the different needs of bioactive peptide delivery. It shows great advantages in solving targeting, stability, loading, release rate, biodegradability, and biocompatibility of bioactive peptides. Certain problems to be solved urgently in the nanotechnology of bioactive peptides are as follows: (1) Due to the unique structure, it is difficult to separate and purify peptide drugs, and the effects of various modification methods and solvents may change the molecular structure of bioactive peptides, resulting in the loss of biological functions. (2) Although many nanocarriers have been shown to be low-toxic or non-toxic in vitro, it is still unclear whether they will affect organisms or even humans, and the preclinical acute and chronic toxicity research must be paid enough attention by researchers. (3) It is worth pondering whether bioactive peptides can effectively cross multiple biological barriers in vivo due to their complex forms of polymorphism and conformation. (4) After most bioactive peptides enter cells, they need to undergo intracellular transport and intracellular release to exert physiological and pharmacological effects depending on delivery efficiency. (5) It is another important problem that how nanocarriers adapt to individual differences. In summary, in the future, efforts should be made to further verify the physiological conditions such as pH value, protease level, mucus, epithelial barrier, and intestinal transport pathway, which can guide the formulation optimization and design of new delivery systems to improve stability and bioavailability of bioactive peptides after oral administration.

Author contributions

XZ composed the outline and drafted the manuscript. QW reviewed and approved the submitted version. XL, YZ, QZ, and YY read and approved the final version. All authors had full access to data and revised and approved the manuscript for publication.

Funding

We gratefully acknowledge the financial support from the National Natural Scientific Foundation of China (Grant Number 32202573) and the Natural Science Foundation of Hunan Province (Grant Number 2022JJ50028).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Bechaux J, Gatellier P, Le Page JF, Drillet Y, Sante-Lhoutellier V. A comprehensive review of bioactive peptides obtained from animal byproducts and their applications. Food Funct. (2019) 10:6244–66. doi: 10.1039/C9FO01546A

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Zheng X, Li DS, Ding K. Purification and identification of angiotensin I-converting enzyme inhibitory peptides from fermented walnut residues. Int J Food Prop. (2017) 20:S3326–33. doi: 10.1080/10942912.2016.1258574

CrossRef Full Text | Google Scholar

3. Wu Q, Li Y, Peng K, Wang XL, Ding Z, Liu L, et al. Isolation and characterization of three antihypertension peptides from the mycelia of Ganoderma lucidum (Agaricomycetes). J Agr Food Chem. (2019) 67:8149–59. doi: 10.1021/acs.jafc.9b02276

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Kang HK, Lee HH, Seo CH, Park Y. Antimicrobial and immunomodulatory properties and applications of marine-derived proteins and peptides. Mar Drugs. (2019) 17:350. doi: 10.3390/md17060350

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Wu Q, Cai QF, Tao ZP, Sun LC, Shen JD, Zhang LJ, et al. Purification and characterization of a novel angiotensin I-converting enzyme inhibitory peptide derived from abalone (Haliotis discus hannai Ino) gonads. Eur Food Res Technol. (2015) 240:137–45. doi: 10.1007/s00217-014-2315-8

CrossRef Full Text | Google Scholar

6. Tamam B, Syah D, Suhartono MT, Kusuma WA, Tachibana S, Lioe HN, et al. Proteomic study of bioactive peptides from tempe. J Biosci Bioeng. (2019) 128:241–8. doi: 10.1016/j.jbiosc.2019.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Shivanna SK, Nataraj BH. Revisiting therapeutic and toxicological fingerprints of milk-derived bioactive peptides: an overview. Food Biosci. (2020) 38:100771. doi: 10.1016/j.fbio.2020.100771

CrossRef Full Text | Google Scholar

8. Wong FC, Xiao J, Wang S, Ee KY, Chai TT. Advances on the antioxidant peptides from edible plant sources. Trends Food Sci Technol. (2020) 99:44–57. doi: 10.1016/j.tifs.2020.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Karami Z, Akbari-Adergani B. Bioactive food derived peptides: a review on correlation between structure of bioactive peptides and their functional properties. J Food Sci Tech. (2019) 56:535–47. doi: 10.1007/s13197-018-3549-4

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kapila R, Kapila S, Vij R. Efficacy of milk-derived bioactive peptides on health by cellular and animal models. In: Ronald Ross W, Robert JC, Victor R, editors. Preedy Nutrients in Dairy and their Implications on Health and Disease. Amsterdam: Elsevier (2017), p. 303–11. doi: 10.1016/B978-0-12-809762-5.00023-1

CrossRef Full Text | Google Scholar

11. Acevedo-Juárez S, Guajardo-Flores D, Heredia-Olea E, Antunes-Ricardo M. Bioactive peptides from nuts: a review. Int J Food Sci Tech. (2022) 57:2226–34. doi: 10.1111/ijfs.15543

CrossRef Full Text | Google Scholar

12. Waseem M, Kumar S, Kumar A. Bioactive Peptides. Hauppauge, NY: Nova Science Publisher Inc. (2018).

Google Scholar

13. Martínez-Maqueda D, Miralles B, Recio I, Hernández-Ledesma B. Antihypertensive peptides from food proteins: a review. Food Funct. (2012) 3:350–61. doi: 10.1039/c2fo10192k

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Peng S, Song H, Chen Y, Li S, Guan X. Oral Delivery of food-derived bioactive peptides: challenges and strategies. Food Rev Int. (2022) 4:1–29. doi: 10.1080/87559129.2022.2062772

CrossRef Full Text | Google Scholar

15. Gallego M, Grootaert C, Mora L, Aristoy MC, Van Camp J, Toldrá F, et al. Transepithelial transport of dry-cured ham peptides with ACE inhibitory activity through a Caco-2 cell monolayer. J Funct Foods. (2016) 21:388–95. doi: 10.1016/j.jff.2015.11.046

CrossRef Full Text | Google Scholar

16. Moskowitz DW. Is somatic angiotensin I-converting enzyme a mechanosensor. Diabetes Technol Ther. (2002) 4:841–58. doi: 10.1089/152091502321118847

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Wu Q, Cai QF, Yoshida A, Sun LC, Liu YX, Liu GM, et al. Purification and characterization of two novel angiotensin I-converting enzyme inhibitory peptides derived from R-phycoerythrin of red algae (Bangia fusco-purpurea). Eur Food Res Technol. (2017) 243:779–89. doi: 10.1007/s00217-016-2792-z

CrossRef Full Text | Google Scholar

18. Wu Q, Fu XP, Sun LC, Zhang Q, Liu GM, Cao MJ, et al. Effects of physicochemical factors and in vitro gastrointestinal digestion on antioxidant activity of R-phycoerythrin from red algae Bangia fusco-purpurea. Int J Food Sci Tech. (2015) 50:1445–51. doi: 10.1111/ijfs.12775

CrossRef Full Text | Google Scholar

19. Iroyukifujita H, Eiichiyokoyama K, Yoshikawa M. Classification and antihypertensive activity of angiotensin I-converting enzyme inhibitory peptides derived from food proteins. J Food Sci. (2000) 65:564–9. doi: 10.1111/j.1365-2621.2000.tb16049.x

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Chen SY, Xu XX, Li X, Yi NB, Li SZ, Xiang XC, et al. Recent advances in the intracellular delivery of macromolecule therapeutics. Biomater Sci. (2022) 4:1–29. doi: 10.1039/D2BM01348G

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Hosseini SF, Ramezanzade L, McClements DJ. Recent advances in nanoencapsulation of hydrophobic marine bioactives: bioavailability, safety, and sensory attributes of nano-fortified functional foods. Trends Food Sci Tech. (2021) 109:322–39. doi: 10.1016/j.tifs.2021.01.045

CrossRef Full Text | Google Scholar

22. Vigani B, Rossi S, Sandri G, Bonferoni MC, Caramella CM, Ferrari F, et al. Recent advances in the development of in situ gelling drug delivery systems for non-parenteral administration routes. Pharmaceutics. (2020) 12:859. doi: 10.3390/pharmaceutics12090859

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Alavi, Mehran, Hamidi, Mehrdad. Passive and active targeting in cancer therapy by liposomes and lipid nanoparticles. Drug Metab Pers Ther. (2019) 34:20180032. doi: 10.1515/dmpt-2018-0032

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Sakai-Kato K, Yoshida K, Takechi-Haraya Y, Izutsu KI. Physicochemical characterization of liposomes that mimic the lipid composition of exosomes for effective intracellular trafficking. Langmuir. (2020) 36:12735–44. doi: 10.1021/acs.langmuir.0c02491

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Bartelds R, Nematollahi MH, Pols T, Stuart MC, Pardakhty A, Asadikaram G, et al. Niosomes, an alternative for liposomal delivery. PLoS ONE. (2018) 13:e0194179. doi: 10.1371/journal.pone.0194179

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release. (2021) 329:624–44. doi: 10.1016/j.jconrel.2020.09.055

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Bochicchio S, Sala M, Spensiero A, Scala MC, Gomez-Monterrey IM, Lamberti G, et al. On the design of tailored liposomes for KRX29 peptide delivery. New J Chem. (2017) 41:11280–90. doi: 10.1039/C7NJ03115G

CrossRef Full Text | Google Scholar

28. Zhang T, Su M, Jiang X, Xue Y, Zhang J, Zeng X, et al. Transepithelial transport route and liposome encapsulation of milk-derived ACE-inhibitory peptide Arg-Leu-Ser-Phe-Asn-Pro. J Agr Food Chem. (2019) 67:5544–51. doi: 10.1021/acs.jafc.9b00397

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Nayar R, Schroit AJ, Fidler IJ. Liposome encapsulation of muramyl peptides for activation of macrophage cytotoxic properties. Methods Enzymol. (1986) 132:594–603. doi: 10.1016/S0076-6879(86)32044-5

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Rezaei N, Mehrnejad F, Vaezi Z, Sedghi M, Asghari SM, Naderi-Manesh H, et al. Encapsulation of an endostatin peptide in liposomes: stability, release, and cytotoxicity study. Colloid Surface B. (2020) 185:110552. doi: 10.1016/j.colsurfb.2019.110552

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Heuts J, Varypataki EM, van der Maaden K, Romeijn S, Drijfhout JW, van Scheltinga AT, et al. Cationic liposomes: a flexible vaccine delivery system for physicochemically diverse antigenic peptides. Pharm Res-Dordr. (2018) 35:1–9. doi: 10.1007/s11095-018-2490-6

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Mansourian M, Badiee A, Jalali SA, Shariat S, Yazdani M, Amin M, et al. Effective induction of anti-tumor immunity using p5 HER-2/neu derived peptide encapsulated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunol Lett. (2014) 162:87–93. doi: 10.1016/j.imlet.2014.07.008

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Wang J, Gong J, Wei Z. Strategies for liposome drug delivery systems to improve tumor treatment efficacy. AAPS Pharm Sci Tech. (2022) 23:1–14. doi: 10.1208/s12249-021-02179-4

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Guimarães D, Cavaco-Paulo A, Nogueira E. Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm. (2021) 601:120571. doi: 10.1016/j.ijpharm.2021.120571

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Amin MCIM, Butt AM, Amjad MW, Kesharwani P. Polymeric micelles for drug targeting and delivery. In: Mishra V, Kesharwani P, Mohd Amin MCM, Iyer A, editors. Nanotechnology-based Approaches for Targeting and Delivery of Drugs and Genes. Amsterdam: Elsevier Inc. (2017), p. 167–202. doi: 10.1016/B978-0-12-809717-5.00006-3

CrossRef Full Text | Google Scholar

36. Lu Y, Zhang E, Yang J, Cao Z. Strategies to improve micelle stability for drug delivery. Nano Res. (2018) 11:4985–98. doi: 10.1007/s12274-018-2152-3

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Hanafy NAN, El-Kemary M, Leporatti S. Micelles structure development as a strategy to improve smart cancer therapy. Cancers. (2018) 10:238. doi: 10.3390/cancers10070238

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Smith JD, Cardwell LN, Porciani D, Nolla A, Cornelison BT, Schulte MC, et al. Therapeutic peptide delivery via aptamer-displaying, disulfide-linked peptide amphiphile micelles. Mol Syst Des Eng. (2020) 5:269–83. doi: 10.1039/C9ME00092E

CrossRef Full Text | Google Scholar

39. Smith JD, Cardwell LN, Porciani D, Nguyen JA, Zhang R, Gallazzi F, et al. Aptamer-displaying peptide amphiphile micelles as a cell-targeted delivery vehicle of peptide cargoes. Phys Biol. (2018) 15:065006. doi: 10.1088/1478-3975/aadb68

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Ju P, Hu J, Li F, Cao Y, Li L, Shi D, et al. A biodegradable polyphosphoester-functionalized poly (disulfide) nanocarrier for reduction-triggered intracellular drug delivery. J Mater Chem B. (2018) 6:7263–73. doi: 10.1039/C8TB01566J

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Cheng YJ, Zhang AQ, Hu JJ, He F, Zeng X, Zhang XZ, et al. Multifunctional peptide-amphiphile end-capped mesoporous silica nanoparticles for tumor targeting drug delivery. ACS Appl Mater Inter. (2017) 9:2093–103. doi: 10.1021/acsami.6b12647

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Wu D, Li Y, Shen J, Tong Z, Hu Q, et al. Supramolecular chemotherapeutic drug constructed from pillararene-based supramolecular amphiphile. Chem Commun. (2018) 54:8198–201. doi: 10.1039/C8CC04334E

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Mohammadi A, Jafari SM, Mahoonak AS, Ghorbani M. Liposomal/nanoliposomal encapsulation of food-relevant enzymes and their application in the food industry. Food Bioprocess Tech. (2021) 14:23–38. doi: 10.1007/s11947-020-02513-x

CrossRef Full Text | Google Scholar

44. Moeller EH, Holst B, Nielsen LH, Pedersen PS, Ostergaard J. Stability, liposome interaction, and in vivo pharmacology of ghrelin in liposomal suspensions. Int J Pharmaceut. (2010) 390:13–8. doi: 10.1016/j.ijpharm.2009.05.067

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Ramezanzade L, Hosseini SF, Akbari-Adergani B, Yaghmur A. Cross-linked chitosan-coated liposomes for encapsulation of fish-derived peptide. Lwt. (2021) 150:112057. doi: 10.1016/j.lwt.2021.112057

CrossRef Full Text | Google Scholar

46. Hosseini SF, Soofi M, Rezaei M. Enhanced physicochemical stability of ω-3 PUFAs concentrates-loaded nanoliposomes decorated by chitosan/gelatin blend coatings. Food Chem. (2021) 345:128865. doi: 10.1016/j.foodchem.2020.128865

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Montero P, Mosquera M, Marín-Peñalver D, Alemán A, Martínez-Álvarez Ó, Gómez-Guillén MC, et al. Changes in structural integrity of sodium caseinate films by the addition of nanoliposomes encapsulating an active shrimp peptide fraction. J Food Eng. (2019) 244:47–54. doi: 10.1016/j.jfoodeng.2018.09.024

CrossRef Full Text | Google Scholar

48. da Silva Malheiros P, Daroit DJ, Brandelli A. Food applications of liposome-encapsulated antimicrobial peptides. Trends Food Sci Tech. (2010) 21:284–92. doi: 10.1016/j.tifs.2010.03.003

CrossRef Full Text | Google Scholar

49. Mohan A, Rajendran SR, He QS, Bazinet L, Udenigwe CC. Encapsulation of food protein hydrolysates and peptides: a review. RSC Adv. (2015) 5:79270–8. doi: 10.1039/C5RA13419F

CrossRef Full Text | Google Scholar

50. Maja L, Željko K, Mateja P. Sustainable technologies for liposome preparation. J Supercrit Fluid. (2020) 165:104984. doi: 10.1016/j.supflu.2020.104984

CrossRef Full Text | Google Scholar

51. Antimisiaris SG, Marazioti A, Kannavou M, Natsaridis E, Gkartziou F, Kogkos G, et al. Overcoming barriers by local drug delivery with liposomes. Adv Drug Deliver Rev. (2021) 174:53–86. doi: 10.1016/j.addr.2021.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Kondratowicz A, Weiss M, Juzwa W, Majchrzycki Ł, Lewandowicz G. Characteristics of liposomes derived from egg yolk. Open Chem. (2019) 17:763–78. doi: 10.1515/chem-2019-0070

CrossRef Full Text | Google Scholar

53. Wang T, Xue J, Hu Q, Zhou M, Luo Y. Preparation of lipid nanoparticles with high loading capacity and exceptional gastrointestinal stability for potential oral delivery applications. J Colloid Interf Sci. (2017) 507:119–30. doi: 10.1016/j.jcis.2017.07.090

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Akbari J, Saeedi M, Ahmadi F, Hashemi SMH, Babaei A, et al. Solid lipid nanoparticles and nanostructured lipid carriers: a review of the methods of manufacture and routes of administration. Pharm Dev Technol. (2022) 27:525–44. doi: 10.1080/10837450.2022.2084554

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Westesen K, Bunjes H, Koch MHJ. Physicochemical charac- terization of lipid nanoparticles and evaluation of their drug loading capacity and sustained release potential. J Control Release. (1997) 48:223–36. doi: 10.1016/S0168-3659(97)00046-1

CrossRef Full Text | Google Scholar

56. Dumont C, Bourgeois S, Fessi H, Jannin V. Lipid-based nanosuspensions for oral delivery of peptides, a critical review. Int J Pharmaceut. (2018) 541:117–35. doi: 10.1016/j.ijpharm.2018.02.038

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Christophersen PC, Birch D, Saarinen J, Isomäki A, Nielsen HM, Yang M, et al. Investigation of protein distribution in solid lipid particles and its impact on protein release using coherent anti-Stokes Raman scattering microscopy. J Control Release. (2015) 197:111–20. doi: 10.1016/j.jconrel.2014.10.023

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Su L, Zhou F, Yu M, Ge R, He J, Zhang B, et al. Solid lipid nanoparticles enhance the resistance of oat-derived peptides that inhibit dipeptidyl peptidase IV in simulated gastrointestinal fluids. J Funct Foods. (2020) 65:103773. doi: 10.1016/j.jff.2019.103773

CrossRef Full Text | Google Scholar

59. Dumont C, Bourgeois S, Fessi H, Dugas PY, Jannin V. In-vitro evaluation of solid lipid nanoparticles: ability to encapsulate, release and ensure effective protection of peptides in the gastrointestinal tract. Int J Pharmaceuti. (2019) 565:409–18. doi: 10.1016/j.ijpharm.2019.05.037

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Francis JE, Skakic I, Smooker PM. Design and preparation of solid lipid nanoparticle (SLN)-mediated DNA vaccines. In: Sunil T, editors. Vaccine Design. New York, NY: Humana (2022), p. 355–66. doi: 10.1007/978-1-0716-1892-9_18

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Kadari A, Pooja D, Gora RH, Gudem S, Kolapalli VRM, Kulhari H, et al. Design of multifunctional peptide collaborated and docetaxel loaded lipid nanoparticles for antiglioma therapy. Eur J Pharm Biopharm. (2018) 132:168–79. doi: 10.1016/j.ejpb.2018.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Trapani A, Tripodo G, Mandracchia D, Cioffi N, Ditaranto N, Cerezuela R, et al. Glutathione loaded solid lipid nanoparticles: preparation and in vitro evaluation as delivery systems of the antioxidant peptide to immunocompetent fish cells. J Cellular Biotechnol. (2016) 2:1–14. doi: 10.3233/JCB-15022

CrossRef Full Text | Google Scholar

63. Muntoni E, Marini E, Ahmadi N, Milla P, Ghè C, Bargoni A, et al. Lipid nanoparticles as vehicles for oral delivery of insulin and insulin analogs: preliminary ex vivo and in vivo studies. Acta Diabetol. (2019) 56:1283–92. doi: 10.1007/s00592-019-01403-9

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Xu Y, Zheng Y, Wu L, Zhu X, Zhang Z, Huang Y, et al. Novel solid lipid nanoparticle with endosomal escape function for oral delivery of insulin. ACS Appl Mater Inter. (2018) 10:9315–24. doi: 10.1021/acsami.8b00507

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Hecq J, Amighi K, Goole J. Development and evaluation of insulin-loaded cationic solid lipid nanoparticles for oral delivery. J Drug Deliv Sci Tech. (2016) 36:192–200. doi: 10.1016/j.jddst.2016.10.012

CrossRef Full Text | Google Scholar

66. Gallarate M, Battaglia L, Peira E, Trotta M. Peptide-loaded solid lipid nanoparticles prepared through coacervation technique. Int J Chem Eng. (2011) 2011:1–6. doi: 10.1155/2011/132435

CrossRef Full Text | Google Scholar

67. Souto EB, Baldim I, Oliveira WP, Rao R, Yadav N, Gama FM, et al. SLN and NLC for topical, dermal, and transdermal drug delivery. Expert Opin Drug Deliv. (2020) 17:357–77. doi: 10.1080/17425247.2020.1727883

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Sakellari GI, Zafeiri I, Batchelor H, Spyropoulos F. Formulation design, production and characterisation of solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for the encapsulation of a model hydrophobic active. Food Hydrocoll Health. (2021) 1:100024. doi: 10.1016/j.fhfh.2021.100024

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Gao Y, Wu X, McClements DJ, Cheng C, Xie Y, Liang R, et al. Encapsulation of bitter peptides in water-in-oil high internal phase emulsions reduces their bitterness and improves gastrointestinal stability. Food Chem. (2022) 386:132787. doi: 10.1016/j.foodchem.2022.132787

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Jo Y-J, Karbstein HP, van der Schaaf US. Collagen peptide-loaded W 1 /O single emulsions and W 1 /O/W 2 double emulsions: influence of collagen peptide and salt concentration, dispersed phase fraction and type of hydrophilic emulsifier on droplet stability and encapsulation efficiency. Food Funct. (2019) 10:3312–23. doi: 10.1039/C8FO02467G

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Gharehbeglou P, Jafari SM, Homayouni A, Hamishekar H, Mirzaei H. Fabrication of double W1/O/W2 nano-emulsions loaded with oleuropein in the internal phase (W1) and evaluation of their release rate. Food Hydrocoll. (2019) 89:44–55. doi: 10.1016/j.foodhyd.2018.10.020

CrossRef Full Text | Google Scholar

72. Choi MJ, Choi D, Lee J, Jo YJ. Encapsulation of a bioactive peptide in a formulation of W1/O/W2-type double emulsions: formation and stability. Food Struct. (2020) 25:100145. doi: 10.1016/j.foostr.2020.100145

CrossRef Full Text | Google Scholar

73. Joyce P, Dening TJ, Meola TR, Schultz HB, Holm R, Thomas N, et al. Solidification to improve the biopharmaceutical performance of SEDDS: opportunities and challenges. Adv Drug Deliver Rev. (2019) 142:102–17. doi: 10.1016/j.addr.2018.11.006

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Mahmood A, Bernkop-Schnürch A. SEDDS: a game changing approach for the oral administration of hydrophilic macromolecular drugs. Adv Drug Deliver Rev. (2018) 142:91–101. doi: 10.1016/j.addr.2018.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Hetényi G, Griesser J, Moser M, Demarne F, Jannin V, Bernkop-Schnürch A, et al. Comparison of the protective effect of self-emulsifying peptide drug delivery systems towards intestinal proteases and glutathione. Int J Pharmaceut. (2017) 523:357–65. doi: 10.1016/j.ijpharm.2017.03.027

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Griesser J, Hetényi G, Kadas H, Demarne F, Jannin V, Bernkop-Schnürch A, et al. Self-emulsifying peptide drug delivery systems: how to make them highly mucus permeating. Int J Pharmaceut. (2018) 538:159–66. doi: 10.1016/j.ijpharm.2018.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

77. AboulFotouh K, Allam AA, El-Badry M, El-Sayed AM. Role of self-emulsifying drug delivery systems in optimizing the oral delivery of hydrophilic macromolecules and reducing interindividual variability. Colloids Surface B. (2018) 167:82–92. doi: 10.1016/j.colsurfb.2018.03.034

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Menzel C, Holzeisen T, Laffleur F, Zaichik S, Abdulkarim M, Gumbleton M, et al. In vivo evaluation of an oral self-emulsifying drug delivery system (sedds) for exenatide. J Control Release Soc. (2018) 277:165. doi: 10.1016/j.jconrel.2018.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Zupančič O, Rohrer J, Thanh Lam H, Grießinger JA, Bernkop-Schnürch A. Development and in vitro characterization of self-emulsifying drug delivery system (SEDDS) for oral opioid peptide delivery. Drug Dev Ind Pharm. (2017) 43:1694–702. doi: 10.1080/03639045.2017.1338722

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Kim MR, Feng T, Zhang Q, Chan HYE, Chau Y. Co-encapsulation and co-delivery of peptide drugs via polymeric nanoparticles. Polymers. (2019) 11:288. doi: 10.3390/polym11020288

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Castro KCD, Costa JM, Campos MGN. Drug-loaded polymeric nanoparticles: a review. Int J Polym Mater. (2022) 71:1–13. doi: 10.1080/00914037.2020.1798436

CrossRef Full Text | Google Scholar

82. Evans BC, Fletcher RB, Kilchrist KV, Dailing EA, Mukalel AJ, Colazo JM, et al. An anionic, endosome-escaping polymer to potentiate intracellular delivery of cationic peptides, biomacromolecules, and nanoparticles. Nat Commun. (2019) 10:1–19. doi: 10.1038/s41467-019-12906-y

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Lee C, Choi JS, Kim I, Oh KT, Lee ES, Park ES, et al. Long-acting inhalable chitosan-coated poly (lactic-co-glycolic acid) nanoparticles containing hydrophobically modified exendin-4 for treating type 2 diabetes. Int J Nanomed. (2013) 8:2975. doi: 10.2147/IJN.S48197

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Baghaee PT, Divsalar A, Chamani J, Donya A. Human serum albumin-malathion complex study in the presence of silver nanoparticles at different sizes by multi spectroscopic techniques. J Biomol Struct Dyn. (2019) 37:2254–64. doi: 10.1080/07391102.2018.1491416

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Allahyari M, Mohit E. Peptide/protein vaccine delivery system based on PLGA particles. Hum Vaccines Immunother. (2016) 12:806–28. doi: 10.1080/21645515.2015.1102804

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Castro PM, Baptista P, Madureira AR, Sarmento B, Pintado ME. Combination of PLGA nanoparticles with mucoadhesive guar-gum films for buccal delivery of antihypertensive peptide. Int J Pharmaceut. (2018) 547:593–601. doi: 10.1016/j.ijpharm.2018.05.051

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Menon JU, Ravikumar P, Pise A, Gyawali D, Hsia CC, Nguyen KT, et al. Polymeric nanoparticles for pulmonary protein and DNA delivery. Acta Biomater. (2014) 10:2643–52. doi: 10.1016/j.actbio.2014.01.033

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Zhang X, Sun M, Zheng A, Cao D, Bi Y, Sun J, et al. Preparation and characterization of insulin-loaded bioadhesive PLGA nanoparticles for oral administration. Eur J Pharm Sci. (2012) 45:632–8. doi: 10.1016/j.ejps.2012.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Dyawanapelly S, Koli U, Dharamdasani V, Jain R, Dandekar P. Improved mucoadhesion and cell uptake of chitosan and chitosan oligosaccharide surface-modified polymer nanoparticles for mucosal delivery of proteins. Drug Deliv Transl Res. (2016) 6:365–79. doi: 10.1007/s13346-016-0295-x

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Sheng J, Han L, Qin J, Ru G, Li R, Wu L, et al. N-trimethyl chitosan chloride-coated PLGA nanoparticles overcoming multiple barriers to oral insulin absorption. ACS Appl Mater Inter. (2015) 7:15430–41. doi: 10.1021/acsami.5b03555

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Rastogi R, Anand S, Koul V. Evaluation of pharmacological efficacy of ‘insulin–surfoplex'encapsulated polymer vesicles. Int J Pharmaceut. (2009) 373:107–15. doi: 10.1016/j.ijpharm.2009.01.022

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Kelly III IB, Fletcher RB, McBride JR, Weiss SM, Duvall CL. Tuning composition of polymer and porous silicon composite nanoparticles for early endosome escape of anti-microRNA peptide nucleic acids. ACS Appl Mater Inter. (2020) 12:39602–11. doi: 10.1021/acsami.0c05827

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Souza FR, Fornasier F, Carvalho AS, Silva BM, Lima MC, Pimentel AS, et al. Polymer-coated gold nanoparticles and polymeric nanoparticles as nanocarrier of the BP100 antimicrobial peptide through a lung surfactant model. J Mol Liq. (2020) 314:113661. doi: 10.1016/j.molliq.2020.113661

CrossRef Full Text | Google Scholar

94. Kumar M, Gupta D, Singh G, Sharma S, Bhat M, Prashant CK, et al. Novel polymeric nanoparticles for intracellular delivery of peptide cargos: antitumor efficacy of the BCL-2 conversion peptide NuBCP-9. Delivery of NuBCP in polymeric nanoparticles. Cancer Res. (2014) 74:3271–81. doi: 10.1158/0008-5472.CAN-13-2015

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Hoshino Y, Koide H, Furuya K, Haberaecker III WW, Lee SH, et al. The rational design of a synthetic polymer nanoparticle that neutralizes a toxic peptide in vivo. Proc Natl Acad Sci. (2012) 109:33–8. doi: 10.1073/pnas.1112828109

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Maheri H, Hashemzadeh F, Shakibapour N, Kamelniya E, Malaekeh-Nikouei B, Mokaberi P, et al. Glucokinase activity enhancement by cellulose nanocrystals isolated from jujube seed: a novel perspective for type II diabetes mellitus treatment (in vitro). J Mol Struct. (2022) 1269:133803. doi: 10.1016/j.molstruc.2022.133803

CrossRef Full Text | Google Scholar

97. Luan Q, Zhou W, Zhang H, Bao Y, Zheng M, Shi J, et al. Cellulose-based composite macrogels from cellulose fiber and cellulose nanofiber as intestine delivery vehicles for probiotics. J Agr Food Chem. (2018) 66:339–45. doi: 10.1021/acs.jafc.7b04754

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Silva SS, Mano JF, Reis RL. Ionic liquids in the processing and chemical modification of chitin and chitosan for biomedical applications. Green Chem. (2017) 19:1208–20. doi: 10.1039/C6GC02827F

CrossRef Full Text | Google Scholar

99. Choi D, Min S-G, Jo Y-J. Functionality of porcine skin hydrolysates produced by hydrothermal processing for liposomal delivery system. J Food Biochem. (2018) 42:e12464. doi: 10.1111/jfbc.12464

CrossRef Full Text | Google Scholar

100. Liu H, Wang C, Li C, Qin Y, Wang Z, Yang F, et al. A functional chitosan-based hydrogel as a wound dressing and drug delivery system in the treatment of wound healing. RSC Adv. (2018) 8:7533–49. doi: 10.1039/C7RA13510F

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Genedy HH, Delair T, Montembault A. Chitosan based microRNA nanocarriers. Pharmaceuticals. (2022) 15:1036. doi: 10.3390/ph15091036

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Yu X, Hou J, Shi Y, Su C, Zhao L. Preparation and characterization of novel chitosan-protamine nanoparticles for nucleus-targeted anticancer drug delivery. Int J Nanom. (2016) 11:6035. doi: 10.2147/IJN.S117066

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Gao Z, Li Z, Yan J, Wang P. Irinotecan and 5-fluorouracil-co-loaded, hyaluronic acid-modified layer-by-layer nanoparticles for targeted gastric carcinoma therapy. Drug Des Dev Ther. (2017) 11:2595. doi: 10.2147/DDDT.S140797

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Liu W, Ye A, Liu W, Liu C, Han J, Singh H, et al. Behaviour of liposomes loaded with bovine serum albumin during in vitro digestion. Food Chem. (2015) 175:16–24. doi: 10.1016/j.foodchem.2014.11.108

PubMed Abstract | CrossRef Full Text | Google Scholar

105. O'Neill GJ, Egan T, Jacquier JC, O'Sullivan M, O'Riordan ED. Kinetics of immobilisation and release of tryptophan, riboflavin and peptides from whey protein microbeads. Food Chem. (2015) 180:150–5. doi: 10.1016/j.foodchem.2015.01.131

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Nasri R, Hamdi M, Touir S, Li S, Karra-Chaabouni M, Nasri M, et al. Development of delivery system based on marine chitosan: encapsulationand release kinetic study of antioxidant peptides from chitosan microparticle. Int J Biol Macromol. (2021) 167:1445–51. doi: 10.1016/j.ijbiomac.2020.11.098

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Song X, Chen Y, Zhao G, Sun H, Che H, Leng X, et al. Effect of molecular weight of chitosan and its oligosaccharides on antitumor activities of chitosan-selenium nanoparticles. Carbohyd polym. (2020) 231:115689. doi: 10.1016/j.carbpol.2019.115689

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Xu B, Jiang G, Yu W, Liu D, Liu Y, Kong X, et al. Preparation of poly (lactic-co-glycolic acid) and chitosan composite nanocarriers via electrostatic self assembly for oral delivery of insulin. Mat Sci Eng. (2017) 78:420–8. doi: 10.1016/j.msec.2017.04.113

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Hosseini SF, Soleimani MR, Nikkhah M. Chitosan/sodium tripolyphosphate nanoparticles as efficient vehicles for antioxidant peptidic fraction from common kilka. Int J Biol Macromol. (2018) 111:730–7. doi: 10.1016/j.ijbiomac.2018.01.023

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Safdar R, Thanabalan M. Developments in insulin delivery and potential of chitosan for controlled release application: a review. J Drug Deliv Sci Technol. (2022) 77:103873. doi: 10.1016/j.jddst.2022.103873

CrossRef Full Text | Google Scholar

111. Ariza-Sáenz M, Espina M, Bolaños N, Calpena AC, Gomara MJ, Haro I, et al. Penetration of polymeric nanoparticles loaded with an HIV-1 inhibitor peptide derived from GB virus C in a vaginal mucosa model. Eur J Pharm Biopharm. (2017) 120:98–106. doi: 10.1016/j.ejpb.2017.08.008

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Alles N, Soysa NS, Hussain MA, Tomomatsu N, Saito H, Baron R, et al. Polysaccharide nanogel delivery of a TNF-α and RANKL antagonist peptide allows systemic prevention of bone loss. Eur J Pharm Sci. (2009) 37:83–8. doi: 10.1016/j.ejps.2009.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Santos R, Gomes D, Macedo H, Barros D, Tibério C, Veiga AS, et al. Guar gum as a new antimicrobial peptide delivery system against diabetic foot ulcers Staphylococcus aureus isolates. J Med Microbiol. (2016) 65:1092–9. doi: 10.1099/jmm.0.000329

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Narayanaswamy R, Torchilin VP. Hydrogels and their applications in targeted drug delivery. Molecules. (2019) 24:603. doi: 10.3390/molecules24030603

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Ou Y, Tian M. Advances in multifunctional chitosan-based self-healing hydrogels for biomedical application. J Mater Chem B. (2021).

PubMed Abstract | Google Scholar

116. Yang Z, McClements DJ, Li C, Sang S, Chen L, Long J, et al. Targeted delivery of hydrogels in human gastrointestinal tract: a review. Food Hydrocoll. (2022) 134:108013. doi: 10.1016/j.foodhyd.2022.108013

CrossRef Full Text | Google Scholar

117. Feng JP, Zhu R, Jiang F, Xie J, Gao C, Li M, et al. Melittin-encapsulating peptide hydrogels for enhanced delivery of impermeable anticancer peptides. Biomater Sci. (2020) 8:4559–69. doi: 10.1039/C9BM02080B

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Li HL, Xu L, He XD, Li JX. Exploration on antimicrobial activity of self-assembled peptide (RADA16-I). World Latest Med Inf . (2017) 66:19–21. doi: 10.19613/j.cnki.1671-3141.2017.66.008

CrossRef Full Text | Google Scholar

119. Yang G, Huang T, Wang Y, Wang H, Li Y, Yu K, et al. Sustained release of antimicrobial peptide from self-assembling hydrogel enhanced osteogenesis. J Biomater Sci Polym Ed. (2018) 29:1812–24. doi: 10.1080/09205063.2018.1504191

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Molinos M, Carvalho V, Silva DM, Gama FM. Development of a hybrid dextrin hydrogel encapsulating dextrin nanogel as protein delivery system. Biomacromolecules. (2012) 13:517–27. doi: 10.1021/bm2015834

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Hirakura T, Yasugi K, Nemoto T, Sato M, Shimoboji T, Aso Y, et al. Hybrid hyaluronan hydrogel encapsulating nanogel as a protein nanocarrier: new system for sustained delivery of protein with a chaperone-like function. J Control Release. (2010) 142:483–9. doi: 10.1016/j.jconrel.2009.11.023

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Chang D, Park K, Famili A. Hydrogels for sustained delivery of biologics to the back of the eye. Drug Discov Today. (2019) 24:1470–82. doi: 10.1016/j.drudis.2019.05.037

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Kang JY, Eggert M, Mouli S, Aljuffali I, Fu X, Nie B, et al. Pharmacokinetics, antitumor and cardioprotective effects of liposome-encapsulated phenylaminoethyl selenide in human prostate cancer rodent models. Pharm Res. (2015) 32:852–62. doi: 10.1007/s11095-014-1501-5

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Huang WC, Deng B, Lin C, Carter KA, Geng J, Razi A, et al. A malaria vaccine adjuvant based on recombinant antigen binding to liposomes. Nat Nanotechnol. (2018) 13:1174–81. doi: 10.1038/s41565-018-0271-3

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W, et al. Liposomes: structure, composition, types, and clinical applications. Heliyon. (2022) 8:e09394. doi: 10.1016/j.heliyon.2022.e09394

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Mirchandani Y, Patravale VB, Brijesh S. Solid lipid nanoparticles for hydrophilic drugs. J Control Release. (2021) 335:457–64. doi: 10.1016/j.jconrel.2021.05.032

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Nahum V, Domb AJ. Solid lipid microspheres decorated nanoparticles as drug carriers. Int J Pharm. (2022) 621:121797. doi: 10.1016/j.ijpharm.2022.121797

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Salawi A. Self-emulsifying drug delivery systems: a novel approach to deliver drugs. Drug Deliv. (2022) 29:1811–23. doi: 10.1080/10717544.2022.2083724

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Haddadzadegan S, Dorkoosh F, Bernkop-Schnurch A. Oral delivery of therapeutic peptides and proteins: technology landscape of lipid-based nanocarriers. Adv Drug Deliv Rev. (2022) 182:114097. doi: 10.1016/j.addr.2021.114097

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Tran H. ElSayed ME. Progress and limitations of oral peptide delivery as a potentially transformative therapy. Expert Opin Drug Deliv. (2022) 19:163–78. doi: 10.1080/17425247.2022.2051476

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Qiao R. Functional polymeric nanoparticles for drug delivery. Curr Pharm Design. (2022) 28:339–339. doi: 10.2174/138161282805220111142951

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Dizaj SM, Salatin S, Khezri K, Lee JY, Lotfipour F. Targeting multidrug resistance with antimicrobial peptide-decorated nanoparticles and polymers. Front Microbiol. (2022) 13:831655. doi: 10.3389/fmicb.2022.831655

PubMed Abstract | CrossRef Full Text | Google Scholar

133. de Lima PHC, Tavares AA, de Lima Silva SM, de Moura MR, Aouada FA, Grillo R. Recent advances on nanohybrid systems constituting clay-chitosan with organic molecules-a review. Appl Clay Sci. (2022) 226:106548. doi: 10.1016/j.clay.2022.106548

CrossRef Full Text | Google Scholar

134. Kumar S, Dhiman R, Prudencio CR, da Costa AC, Vibhuti A, Leal E, et al. Chitosan: applications in drug delivery system. Mini Rev Med Chem. (2022) 22. doi: 10.2174/1389557522666220609102010 [Online ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Yu F, Zhu Y, Shang X, Yuan H, Hu F. Rationally-designed chitosan-based polymeric nanomaterials according to intrinsic characteristics for cancer therapy and theranostics: a review. Curr Med Chem. (2022) 30. doi: 10.2174/0929867329666220620164429 [Online ahead of print].

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Yang D. Recent advances in hydrogels. Chem Mater. (2022) 34:1987–9. doi: 10.1021/acs.chemmater.2c00188

CrossRef Full Text | Google Scholar

137. Huang S, Hong X, Zhao M, Liu N, Liu H, Zhao J, et al. Nanocomposite hydrogels for biomedical applications. Bioeng Transl Med. (2022) 7:e10315. doi: 10.1002/btm2.10315

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Abune L, Wang Y. Affinity hydrogels for protein delivery. Trends Pharmacol Sci. (2021) 42:300–12. doi: 10.1016/j.tips.2021.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Limasale YDP, Atallah P, Werner C, Freudenberg U, Zimmermann R. Tuning the local availability of VEGF within glycosaminoglycan-based hydrogels to modulate vascular endothelial cell morphogenesis. Adv Funct Mater. (2020) 30:2000068. doi: 10.1002/adfm.202000068

CrossRef Full Text | Google Scholar

140. Adil MM, Vazin T, Ananthanarayanan B, Rodrigues GM, Rao AT, Kulkarni RU, et al. Engineered hydrogels increase the post-transplantation survival of encapsulated hESC-derived midbrain dopaminergic neurons. Biomaterials. (2017) 136:1–11. doi: 10.1016/j.biomaterials.2017.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: bioactive peptide, delivery system, nanoparticle, peptide drug, bioavailability

Citation: Zhang X, Li X, Zhao Y, Zheng Q, Wu Q and Yu Y (2022) Nanocarrier system: An emerging strategy for bioactive peptide delivery. Front. Nutr. 9:1050647. doi: 10.3389/fnut.2022.1050647

Received: 22 September 2022; Accepted: 08 November 2022;
Published: 05 December 2022.

Edited by:

Mingqian Tan, Dalian Polytechnic University, China

Reviewed by:

Jamshidkhan Chamani, Islamic Azad University of Mashhad, Iran
Sutapa Biswas Majee, NSHM Knowledge Campus, India
Eleonore Fröhlich, Medical University of Graz, Austria

Copyright © 2022 Zhang, Li, Zhao, Zheng, Wu and Yu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Qiang Wu, qiangwusmile@163.com; Yougui Yu, yufly225@163.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.