Skip to main content
Food Science and Biotechnology logoLink to Food Science and Biotechnology
. 2021 Aug 5;30(8):1003–1023. doi: 10.1007/s10068-021-00942-8

Biological activity and processing technologies of edible insects: a review

Jae Hoon Lee 1, Tae-Kyung Kim 1, Chang Hee Jeong 2, Hae In Yong 1, Ji Yoon Cha 1, Bum-Keun Kim 1, Yun-Sang Choi 1,
PMCID: PMC8364587  PMID: 34471556

Abstract

The burgeoning global population growth has raised concerns regarding the expected increase in the demand for food, which could be partially tackled by identifying novel food sources. To this end, edible insects have recently attracted research interest. Several technologies for utilizing edible insect-derived proteins have been introduced; however, research into their functional utilization is insufficient. Herein, we reviewed the relevant literature on the importance of insects as food sources, extraction of edible insects, the nutritional value of insects, biological activities of components, and their applications in food industries. We summarized the studies primarily focused on the functional utilization of edible insects, suggesting that for successful incorporation and growth of edible insects in food and pharmaceutical industries, strategies to improve the extraction methods are required to explore the biological activity of edible insects. Furthermore, the awareness of edible insects with a focus on their allergens warrants consideration.

Keywords: Edible insect, Biological activity, Processing technology, Food resources, Entomophagy

Introduction

By 2050, the global population is expected to exceed 9 billion, and therefore, the demand for food will proportionally increase, requiring the need to identify new food resources (Kim et al., 2019). Proteins are crucial for physical development and maintenance of health, and they require high energy and opportunity costs for large-scale production (Nyangena et al., 2020; Park et al., 2017). Essential amino acids (EAAs) that play major roles in tissue growth, energy production, immune function, and nutrient absorption are not synthesized in the human body; even if they are synthesized, the amount is minimal, and the requirement must be fulfilled by food consumption. Animal proteins are the most effective source of EAAs (del Hierro et al., 2020). Therefore, the issues associated with agricultural land exploitation for livestock breeding and environmental pollution are expected to intensify (Choi et al., 2017; Kim et al., 2021b). However, there is a limit to the increase in protein demand with traditional livestock production methods, and it is necessary to partially address the demand using alternative meat products (da Silva Lucas et al., 2020; Mintah et al., 2020).

Consequently, the world has started to focus on securing plant-based proteins, cultured meat, and edible insect resources to protect food security (Choi et al., 2020; Kim et al., 2021a; Park, 2021). In particular, edible insects are emerging as a substitute for high-protein food that can replace traditional animal proteins (Khampakool et al., 2020). In addition, edible insect proteins have the advantage of the requirement of reduced farmland and greenhouse gas emissions compared to traditional animal proteins and exclude the concerns about infectious diseases associated with livestock (Kim et al., 2019; Kwak et al., 2020). Therefore, edible insects have gained increasing research interest as alternative protein sources. In addition, some studies have explored the scope of using edible insects as alternative lipid sources (da Silva Lucas et al., 2020; Kim et al., 2020). Studies have also shown that steps followed in extraction procedures affect the functional properties (Kim et al., 2020) and digestibility (Lee et al., 2021) of insect proteins. Moreover, the biological activities of edible insects have been explored (di Mattia et al., 2019). Several studies have reported the antioxidant (del Hierro et al., 2020), anti-hypertensive (Hall et al., 2018), anti-cancer (Cho et al., 2019), anti-inflammatory (Baek et al., 2018), anti-obesity (Seo et al., 2017), anti-diabetic (Lacroix et al., 2019), and anti-microbial (Rahnamaeian et al., 2015) activities of edible insect proteins. Thus, edible insects are not only highly nutritious but also possess various biological activities. Accordingly, the possibility of their applications as functional food and pharmaceutical materials is increasing, while edible insect-derived proteins are emerging as an alternative resource that can replace existing proteins and lipids in the food industry.

This review summarizes various studies on edible insects that have been conducted worldwide. In particular, we have reviewed studies investigating the use of edible insects as food sources, nutritional value and biological activity of edible insects, and extraction procedure of proteins from edible insects. Finally, we have described the potential applications of edible insects in the food and pharmaceutical industries.

Edible insects as food resources: limitations and safety

Proteins improve the nutritional, technical, and functional properties of food (FAO/WHO/UNU, 1985; Kinsella and Melachouris, 1976). In particular, from a nutritional perspective, the abundance of EAAs has been used as an important index for choosing an alternative source of protein because EAAs are indispensable; however, as they cannot be synthesized in the human body, the diet supplements are the only alternatives (FAO/WHO/UNU, 1985). Owing to the projected increase in food demand, several studies have explored the viable alternatives, of which edible insects have received the most attention, as edible insect proteins have an excellent amino acid profile. It has been reported that mealworms, which are the most widely consumed insects, have an EAA composition similar to that of soybean and bovine casein (Yi et al., 2013). For example, lysine, a limited EAA in rice, wheat, and maize, is present in edible insects; therefore, edible insects are suggested as a suitable alternative source to complement the quality of plant-based food (Ghosh et al., 2017).

Proteins extracted from edible insects exert antioxidant, anti-diabetic, anti-hypertensive, anti-cancer, anti-microbial, anti-inflammatory, and immunomodulatory effects (Nongonierma and FitzGerald, 2017) compared to other food sources (Mintah et al., 2019; Sila and Bougatef, 2016; Zielińska et al., 2017). The bioactive peptides generated from edible insects through different food processing methods, including fermentation, enzymatic or chemical hydrolysis, and digestion processes (Udenigwe and Aluko, 2012), have been shown to enhance these activities (Cicero et al., 2017). Although the aforementioned activities of edible insects have not been fully explored, we speculate that in-depth studies could reveal the potential of edible insect proteins and their hydrolysates as sustainable novel protein sources for human nutrition.

Despite the multitude of beneficial health attributes, there are some limitations to using edible insects as novel food sources. In particular, food neophobia is a critical problem in incorporating edible insects into the food system. Additionally, insects in various idiomatic phrases, proverbs, and adages reflect a negative image, which might be attributed to the negative perception of edible insects (Meyer-Rochow and Kejonen, 2020). Reportedly, people who avoid unaccustomed food are more likely to be disgusted by insect consumption (la Barbera et al., 2018). Several studies have shown that the attitudes of people toward eating insects change positively (Adámek et al., 2018, Barsics et al., 2017); however, a positive attitude does not mean a preference for entomophagy. With these prejudices, Europeans feel daring, interested, and adventurous regarding the consumption of insects (Tuccillo et al., 2020). Moreover, although South Koreans eat edible insects such as grilled crickets, boiled silkworm pupae, or medicine pellet forms, they are reluctant to visit edible insect restaurants (Hwang and Choe, 2020). However, processed protein powder could be an alternative way to consume edible insects as food sources, and consumers would feel less repulsed when consuming insect protein powders rather than whole-form insects (Orkusz et al., 2020). Therefore, insect-based products could serve as a potential alternative to promote the consumption of edible insects.

Regardless of the acceptance of edible insects as food resources, safety associated with edible insects could be a major factor determining their inclusion in the human diet. The history of entomophagy is longer than that of conventional food resources; however, the emphasis on entomophagy is in progress worldwide, including in the Western society (Tranter, 2013). Several studies have reported food safety hazards and nutritional factors associated with edible insects (Murefu et al., 2019; van Huis et al., 2013). Murefu et al. (2019) reported that processing methods, including boiling, frying, and roasting, could markedly increase the safety of edible insects. In addition, it has been shown that mycotoxins that can cause acute and chronic diseases can be produced in insect guts (Imathiu, 2020; van Huis et al., 2013). The levels of aflatoxin, the most dangerous mycotoxin in edible insects, have been reported to be higher than the regulated limit, which could be due to contamination through open and unhygienic drying conditions (Kachapulula et al., 2018). Furthermore, several studies have reported contamination of various heavy metals in edible insects; however, they have reported no additional hazards compared with conventional animal food sources (Murefu et al., 2019; Poma et al., 2017). In addition, various studies have shown that the amount of antinutrients that imposes several health concerns is not above the regulated limit, and therefore, the potential threat of antinutrients in edible insects is minor (Oibiokpa, 2017; Shantibala et al., 2014). Parasites, pathogenic microorganisms, and pesticide residues in edible insects might be problematic; however, it could be controlled when farming these insects for food but could be a problem when harvesting or hunting wild edible insects (Imathiu, 2020). Moreover, various allergens have been identified in edible insects (de Gier and Verhoeckx, 2018), and allergic reactions mainly involve tropomyosin and arginine kinase (over 70%). However, allergens present in edible insects cannot be controlled well through changes in external factors because they are an endogenous risk factor for health (Murefu et al., 2019). It has also been reported that heating and digestion cannot eliminate the potential allergic reactions of edible insects (Broekman et al., 2015). Furthermore, undeclared allergens could be a threat, even causes of death, to allergen-responsive and sensitive persons (Poms et al., 2004). For instance, people with allergic reactions to crustaceans (arthropods) have shown large potential for cross-reactivity with edible insects (Lopata et al., 2010). Therefore, the edible insect allergens should be identified and declared, and the intake of edible insects as food sources must be carefully considered.

Nutritional value of edible insects

The significance and disadvantages of edible insects and their various nutritional values are listed in Table 1. The primary use of edible insects lies in their protein content, which varies widely from 7 to 91% from fresh matter (Bukkens, 1997; Nakagaki and Defoliart, 1991; van Huis, 2016). These variations might be due to differences in species, breeding, and feeding methods. The feed conversion ratio (FCR), which measures the efficiency of an animal in converting the food provided into the desired output, of edible insects (1.7 kg for 1 kg of cricket) is lower than that of other animals (10, 5, and 2.5 kg for 1 kg of beef, pork, and chicken, respectively). Low FCR is an important factor that renders edible insects an efficient food source with lesser environmental impacts (Govorushko, 2019). Moreover, the amino acid content is also an important factor when choosing a food for protein supply, and most edible insects (> 200 species) contain the recommended level of EAAs. In addition, absorption of digested amino acids is another determining factor for selecting protein sources (Yi et al., 2016). It has been shown that > 50% of insect proteins can be digested by human intestinal conditions in vitro (Nongonierma and FitzGerald, 2017; Yi et al., 2016), indicating that edible insects could serve as an excellent alternative protein source. However, the digestibility values of insect proteins need to be recalculated in some studies because different nitrogen to protein conversion factors (Kp) were used to calculate protein content based on total nitrogen compared to conventional meat sources. The Kp of Tenebrio molitor, Alphitobius diaperinus, and Hermetia illucens larvae is 4.76, that of the proteins extracted from these insects is 5.60, and that of conventional meat sources is 6.25 (Janssen et al., 2017; Kim et al., 2019). The abundant chitin components forming the shell of insects comprise glycoproteins, such as glucosamine, which can be identified using the Kjeldahl method (Janssen et al., 2017).

Table 1.

Significance and disadvantages of edible insects as alternative food and their nutritional value

Nutrienta Content (g/100 g) Significance as an alternative food source Disadvantages References
Protein 7–91

Higher feed conversion ratio than other animals

Satisfy recommended essential amino acid level for humans

High digestibility (> 50%)

Protein nitrogen conversion value is different from conventional animal protein sources because of the high composition of glycoproteins (e.g., chitin) Govorushko (2019), Janssen et al. (2017), Nongonierma and FitzGerald (2017), van Huis (2016), Yi et al. (2016)
Fat 0.1–30 Rich in unsaturated fatty acids High diversity according to sex, environment, stage, and species Bukkens (1997), DeFoliart (1999), Yi et al. (2016)
Carbohydrate 1–10

Significant effect on humoral immunity

Wound-healing

Chitin cannot be digested Long et al. (2007), Xiaoming et al. (2010)
Micronutrient Under 1 High amount of Fe, Cu, Zn, Ca, Na, Mn, P, vitamin B1, B2, B6, D, E, K, and C High diversity according to sex, environment, stage, and species Bukkens (1997), Kim et al. (2019), Xiaoming et al. (2010)

aDry matter was used to determine protein and micronutrient contents, and the fresh matter was used to determine fat and carbohydrate contents

Lipids are major energy and metabolic activity sources, and essential fatty acid deficiency is associated with protein-energy malnutrition in developing countries (Smit et al., 2004). Edible insects contain ~ 30% fat on a fresh weight basis and are rich in unsaturated fatty acids, such as linoleic acid and linolenic acid (DeFoliart, 1999). However, lipid content and fatty acid compositions are affected by the environment, sex, diet, stage of development, and species (Bukkens, 1997). Because of abundant lipid content, edible insects are suggested as an attractive alternative source of essential fatty acids (Yi et al., 2013). However, this aspect has been comparatively less explored as most studies have focused on the protein content of edible insects (Paul et al., 2017). Therefore, further studies are required to investigate the safety, application, and utilization methods of various edible insect lipids.

Carbohydrates are not only the primary sources of energy production but are also major constituents of the animal body (Asp, 1993). Although the polysaccharide content in edible insects is lesser than that of proteins and lipids (1–10%), the polysaccharides in silkworm pupae have been reported to have a substantial effect on humoral immunity (Long et al., 2007; Xiaoming et al., 2010). Chitin, the primary component of exoskeletons of insects, is a long-chain polymer of N-acetyl glucosamine, a derivative of glucose used for wound healing and medical film production (Xiaoming et al., 2010).

Micronutrients constitute a smaller part of the human diet; however, their role is critical in maintaining a healthy life (Mlček et al., 2017). In edible insects, metal ions (e.g., iron, copper, zinc, calcium, sodium, and manganese), phosphorus, and vitamins, such as B1, B2, B6, D, E, K, and C, are abundant, and the levels of these nutrients are similar to those in conventional meat sources (Bukkens, 1997; Kim et al., 2019; Xiaoming et al., 2010). However, these values differ widely depending on the species and origin of the analyzed insects. Overall, it can be concluded that environmental differences are essential in determining the nutritional value of edible insects.

Extraction of nutritional and functional components from edible insects

In Western culture, entomophagy is still not generally established, and consumer acceptability of edible insects is low (Yen, 2009). Thus, extracting the nutritional and functional components from edible insects and using them as food ingredients has been suggested to increase consumer acceptability (Sosa and Fogliano 2017). Therefore, protein, fat, and chitin are primarily extracted from edible insects using various methods (Table 2) to be used as functional ingredients in food and nutraceuticals (Gravel and Doyen, 2020; Okagu et al., 2020; Vercruysse et al., 2005).

Table 2.

Extraction methods for each component and their characteristics

Component Extraction process Characteristics of the extraction method Insect species References
Protein Alkaline and sonication-assisted extraction Improvement in foaming and emulsifying abilities Schistocerca grega, Apis mellifera Mishyna et al. (2019)
Dry fractionation Diversification of chemical composition Tenebrio molitor L Purschke et al. (2018a)
Defatting and sonication Improvement of the yield Tenebrio molitor L., Gryllus bimaculatus, Bombyx mori Choi et al. (2017)
Defatting and alkaline extraction Improvement of water and oil absorption properties Hermetia illucens Mintah et al. (2020)
Enzymatic hydrolysis Enhancement of emulsifying activity, foaming ability, and oil binding capacity Locusta migratoria L Purschke et al. (2018b)
Fat (oil) Water extraction Lower lipid yield but higher ω-3 fatty acid contents than Soxhlet and Folch extractions Tenebrio molitor, Alphitobius diaperinus, Acheta domesticus, Blaptica dubia Tzompa-Sosa et al. (2014)
Folch extraction Higher lipid yield and ω-6 fatty acid contents than Soxhlet and water extractions except for Blaptica dubia Tenebrio molitor, Alphitobius diaperinus, Acheta domesticus, Blaptica dubia Tzompa-Sosa et al. (2014)
Supercritical CO2 extraction Higher monounsaturated fatty acid contents than Soxhlet extraction Acheta domesticus Laroche et al. (2019)
Ultrasound-assisted extraction Lower acid and peroxide levels and higher polyunsaturated fatty acid contents and thermal stability than Soxhlet extraction Clanis bilineata Sun et al. (2018)
Chitin Chemical extraction Simple and inexpensive Pimelia sp. Kaya et al. (2016)
Deep eutectic solvent extraction Low toxicity and biodegradability Hermetia illucens Zhou et al. (2019)

Protein extraction

Insect proteins are considered the primary alternative protein resources to meet the growing demand for animal proteins worldwide because of their high nutritional value (Bruinsma 2003). Therefore, various extraction methods, such as water extraction, dry fractionation, and alkaline extraction, are used to extract high-quality proteins from different insects, and new methods (sonication and ultra-high pressure) are being developed (Melgar-Lalanne et al., 2019). The extraction rate and characteristics of insect proteins may vary depending on the insect species, sex, life stage, diet, and geographical origin (Rumpold and Schlüter, 2013a; 2013b). In particular, extraction methods are an important factor that can influence the technical functionalities of insect proteins (Zhao et al., 2016). Previously, Kim et al. (2020) reported that the proteins extracted from three different edible insects (i.e., T. molitor, Allomyrina dichotoma, and Protaetia brevitarsis) showed improved foam capacity by more than 40% and increased stability by more than 40 min through defatting processes with hexane. Dry fractionation diversified the chemical composition of proteins extracted from mealworm larvae (T. molitor L.) by increasing the fine fraction (< 500 μm), leading to improved nutritional and functional properties as ingredients in the food matrix (Purschke et al., 2018a). Moreover, proteins from grasshopper (Schistocerca gregaria) and honey bees (Apis mellifera) improved the foaming and emulsifying abilities by 10–40% through alkaline and sonication-assisted extractions (Mishyna et al., 2019). In addition, the addition of defatting pretreatment in protein extraction offers the advantage of increasing the protein extraction yield (Choi et al., 2017; Mintah et al., 2020). Overall, it is clear that the changes in the functionalities of insect proteins by the extraction process are closely related to the alteration in surface hydrophobicity, protein charge, and protein composition according to molecular weight.

Lipid extraction

Lipids are the second-largest nutrient component in edible insects after proteins (Xiaoming et al., 2010). In particular, edible insects showed a higher value of essential fatty acids (linoleic acid and α-linolenic acid) than other sources, such as Eucommia ulmodie, Salmo salar, and Camellia sinensis (da Silva Lucas et al., 2020). Therefore, various methods, including water extraction, Folch extraction, Soxhlet method, and supercritical CO2 extraction, have been used to extract these lipid components. However, unlike proteins, the extraction process does not markedly influence the fatty acid composition, but the types of lipids extracted and the extraction yield are influenced by the lipid extraction process (da Silva Lucas et al., 2020). For instance, the lipid yield from four different edible insects (T. molitor, Alphitobius diaperinus, Acheta domesticus, and Blaptica dubiawas) increased from 19 to 60% when obtained by Folch extraction compared with water extraction and Soxhlet extraction, wherein Folch extraction improved the content of ω-6 fatty acids in lipids from T. molitor, A. diaperinus, and A. domesticus (Tzompa-Sosa et al., 2014). Supercritical CO2 extraction has been reported to isolate thermally sensitive components, decrease the oxidation of solutes, produce solvent-free residues, and employ appropriate solvent characteristics (e.g., non-toxic, non-explosive, and chemically inert) (Mariod et al., 2010; Purschke et al., 2017; Roy et al., 2006). According to a previous study, oil from Clanis bilineata obtained via ultrasound-assisted aqueous extraction exhibits lower peroxide, acid, and p-anisidine levels and higher thermal stability and polyunsaturated fatty acid contents compared with oil extracted using the Soxhlet method (Sun et al., 2018). Collectively, these studies indicate that the extraction process can alter the characteristics or types of lipids extracted from edible insects; therefore, it should be carefully selected considering the desired application.

Chitin and chitosan extraction

Currently, the most common sources of chitin are shrimp, shellfish, and crabs. However, extracting chitin from insects can be more advantageous in terms of chemical consumption, extraction method, time, and yield than marine crustaceans (Mohan et al., 2020). Chitin and chitosan, which are non-toxic, biodegradable polymers, are extracted from the exoskeletons of various edible insects. In general, chitin from insects is extracted using a simple and inexpensive chemical extraction process, including demineralization and deproteinization (Mohan et al., 2020). Recently, studies on green extraction methods to overcome the disadvantages of conventional chemical extraction methods—alterations in physicochemical properties, use of expensive chemicals in the purification process, and release of toxic effluent wastewater into the environment—have been emphasized (Dhillon et al., 2013; Huang et al., 2018; Kaur and Dhillon, 2015). For instance, various methods, such as microwave-assisted, phytoextraction, ultrasonic-assisted, and enzymatic methods, have been reported to produce chitin from marine crustaceans (Gartner et al., 2010; Gopal et al., 2019; Hongkulsup et al., 2016; Valdez-Peña et al., 2010). In particular, biological extraction methods by microorganisms, including Bacillus subtilis, Lactobacillus plantarum, and Pseudomonas aeruginosa K-187, have been reported to decrease chitin degradation and impurity levels of extracts (Oh et al., 2000; Rao et al., 2000; Yang et al., 2000). According to Khanafari et al. (2008), the microbial extraction process requires less energy, fewer solvents, less time, and a less complicated procedure than the chemical extraction process. Moreover, the extraction method using deep eutectic solvents (DES) is considered a green alternative because of its low toxicity and cost, ease of synthesis, and biodegradability compared to conventional methods (Paiva et al., 2014; Zhang et al., 2012). Thus, the DES extraction method has been employed to produce chitin from insects (e.g., Hermetia illucens) (Zhou et al., 2019), and acid detergent fiber and acid detergent lignin methods have been used for chitin production from H. illucens (Brigode et al., 2020). Although further research is needed, it is believed that these green extraction methods have the potential to replace chemical methods.

Biological activities of various edible insect components

Antioxidant activity

Various components of edible insects have been reported to have antioxidant activity, from hydrolysates obtained through various enzyme treatments to peptides obtained through purification and identification processes, extracts using water and organic solvents, and chitosan present in the skin of larvae (Table 3). For example, Bombyx mori (silkworm) larvae protein isolate obtained using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) has been reported to have strong radical scavenging activity (IC50 = 57.91 μg/mL) and ferrous ion chelating activity (IC50 = 2.03 mg/mL) (Wu et al., 2011). di Mattia et al. (2019) also reported that water- or lipid-soluble extracts of B. mori have antioxidant activities, as measured using ferric reducing/antioxidant power (FRAP) and 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulfonic acid (ABTS) radical scavenging activity. The extracts showed higher antioxidant activity than orange juice or olive oil, which are functional food known to regulate human antioxidant networks (Foroudi et al., 2014; Zamora-Ros et al., 2013). Adult B. mori (silk moth) hydrolysates obtained using alcalase and alkaline protease also exhibit high radical scavenging activity, as estimated using oxygen radical absorbance capacity (ORAC) and 1,1-diphenyl-2-picrylhydrazyl (DPPH) assays (Liu et al., 2017). This study revealed no change in the DPPH radical scavenging activity of these hydrolysates in the gastrointestinal digestion test.

Table 3.

Functional bioactive components from edible insects

Biological activity Edible insects (stage) Sample Sample preparation Experimental method References
Antioxidant Bombyx mori (Larvae) Hydrolysates (IC50 = 57.91 μg/mL for DPPH; 2.03 mg/mL for ferrous chelating) Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) DPPH assay Wu et al. (2011)
Ferrous ion chelating assay
Reducing power assay
Bombyx mori (Adult) Hydrolysates Hydrolysis using enzyme mixture (alcalase and alkaline protease) DPPH assay Liu et al. (2017)
ORAC assay
Bombyx mori (Not described) Water extracts, lipid extracts Extraction with hexane and then residue extraction with water ABTS assay di Mattia et al. (2019)
FRAP assay
Schistocerca gregaria (Adult) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) DPPH assay Zielińska et al. (2017)
ABTS assay
Ferrous ion chelating assay
Reducing power assay
Musca domestica (Larvae) Water extracts Extraction with water (decoction method) DPPH assay Li et al. (2017)
Musca domestica (Larvae) Hydrolysates Hydrolysis using alcalase or neutral proteinase DPPH assay Zhang et al. (2016)
Superoxide anion radical assay
Hydroxyl radical assay
Ferrous ion chelating assay
Reducing power assay
Musca domestica (Larvae) Chitosan (IC50 = 373 μg/mL for DPPH) Extraction with chemical method (deproteinization, decolorization, decalcification, and deacetylation) DPPH assay Ai et al. (2008)
Reducing power assay
Ferrous ion chelating assay
Gryllodes sigillatus (Adult) Hydrolysates Hydrolysis using alcalase and then further digestion using gastrointestinal enzymes (pepsin, bile salt, pancreatin) DPPH assay Hall et al. (2018)
ABTS assay
FRAP assay
Ferrous ion chelating assay
Gryllodes sigillatus (Adult) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) DPPH assay Zielińska et al. (2017)
ABTS assay
Ferrous ion chelating assay
Reducing power assay
Tenebrio molitor (Larvae) Extracts Ultrasound-assisted extraction or pressurized-liquid extraction using ethanol and ethanol: water (1:1) mixture DPPH assay del Hierro et al. (2020)
Tenebrio molitor (Larvae) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) DPPH assay Zielińska et al. (2017)
ABTS assay
Ferrous ion chelating assay
Reducing power assay
Tenebrio molitor (Not described) Hydrolysates Hydrolysis using Protamex or flavourzyme or alcalase DPPH assay Messina et al. (2019)
Acheta domesticus (Adult) Extracts Ultrasound-assisted extraction or pressurized-liquid extraction using ethanol and ethanol: water (1:1) mixture DPPH assay del Hierro et al. (2020)
Acheta domesticus (Not described) Water extracts, lipid extracts Extraction with hexane and then residue extraction with water ABTS assay di Mattia et al. (2019)
FRAP assay
Acheta domesticus (Not described) Hydrolysates Hydrolysis using Protamex or flavourzyme or alcalase DPPH assay Messina et al. (2019)
Chrysomya megacephala (Larvae) Chitosan (IC50 = 1.2 mg/mL) Extraction with chemical method (deproteinization, decolorization, decalcification, and deacetylation) DPPH assay Song et al. (2013)
Oecophylla smaragdina (Larvae)

Peptide

CTKKHKPNC (IC50 = 48.2 μM for DPPH; 38.4 μM for ABTS)

Digestion using gastrointestinal enzyme (pepsin and trypsin) and purification with ultrafiltration, size-exclusion chromatography, and RP-HPLC DPPH assay Pattarayingsakul et al. (2017)
ABTS assay
Anti-hypertensive Spodoptera littoralis (Larvae)

Peptide

AVF (IC50 = 2,123 μM)

Digestion using gastrointestinal enzymes (pepsin, trypsin, and chymotrypsin) ACE inhibitory activity assay using HHL substrate-spectrophotometric method Vercruysse et al. (2008)
Spodoptera littoralis (Larvae)

Hydrolysates

Sample with gastrointestinal digestion (IC50 = 320 μg/mL), sample with gastrointestinal digestion and mucosal peptidase (IC50 = 211 μg/mL)

Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) and then hydrolysis using mucosal peptidase ACE inhibitory activity assay using HHL substrate-spectrophotometric method Vercruysse et al. (2009a, b)
Bombyx mori (Larvae) Hydrolysates (IC50 = 0.69 mg/mL) Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) ACE inhibitory activity assay using FAPGG substrate-spectrophotometric method Vercruysse et al. (2005)
DTG substrate-HPLC method
Bombyx mori (Larvae) Hydrolysates (IC50 = 8.3 μg/mL) Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) ACE inhibitory activity assay using HHL substrate-spectrophotometric method Wu et al. (2011)
Bombyx mori (Pupae)

Peptide

APPPKK (IC50 = 47 μg/mL)

Hydrolysis using acidic protease and purification with Sephadex gel filtration ACE inhibitory activity assay using HHL substrate-HPLC method Wang et al. (2011)
Bombyx mori (Pupae)

Peptide

ASL (IC50 = 102.15 μM)

Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) and purification with ultrafiltration, gel filtration chromatography, and RP-HPLC ACE inhibitory activity assay using HHL substrate-spectrophotometric method Wu et al. (2015)
Bombus terrestris (Adult) Hydrolysates (IC50 = 4.96 mg/mL) Digestion using gastrointestinal enzymes (pepsin, trypsin, and α-chymotrypsin) ACE inhibitory activity assay using FAPGG substrate-spectrophotometric method Vercruysse et al. (2005)
DTG substrate-HPLC method
Musca domestica (Larvae) Water extracts (IC50 = 430 μg/mL) Extraction with water (decoction method) ACE inhibitory activity assay using the fluorescent substrate-spectrophotometric method Li et al. (2017)
Gryllodes sigillatus (Not described) Hydrolysates Hydrolysis using alcalase and then further digestion using gastrointestinal enzymes (pepsin, bile salt, and pancreatin) ACE inhibitory activity assay using HHL substrate-spectrophotometric method Hall et al. (2018)
Tenebrio molitor (Larvae)

Peptide

YAN (IC50 = 17 μg/mL for ACE inhibition)

Hydrolysis using alcalase and purification with Sephadex gel filtration and RP-HPLC ACE inhibitory activity assay using HHL substrate-HPLC method Dai et al. (2013)
In vivo test using SHR model
Oecophylla smaragdina (Larvae)

Peptide

FFGT (IC50 = 19.5 μM), LSRVP (IC50 = 52.7 μM)

Digestion using gastrointestinal enzymes (pepsin and trypsin) and purification with ultrafiltration, size-exclusion chromatography, and RP-HPLC ACE inhibitory activity assay using the 3HB-GGG substrate-spectrophotometric method Pattarayingsakul et al. (2017)
Anti-cancer Bombyx mori (Larvae) Extracts Fermentation with Aspergillus kawachii and extraction with ethanol The following assay using HepG2 human hepatocellular carcinoma cells (Sulforhodamine B assay, cell cycle analysis, Annexin V staining assay, DNA fragmentation analysis, western blot analysis) Cho et al. (2019)
Calliphora vicina (Larvae)

Peptide

HGVSGHGQHGVHG

Immunization with heat-killed Escherichia coli D31 and Micrococcus luteus A270 as well as isolation peptide from the hemolymph NK activity assay using K562 tumor cells Chernysh et al., (2002)
In vivo interferon induction test using mice
Musca domestica (Larvae) Chitosan Extraction with chemical methods (deproteinization, decolorization, decalcification, and deacetylation) MTT assay using HeLa human cervical carcinoma and mouse sarcoma-180 tumor cells Ai et al. (2008)
Anti-inflammatory Schistocerca gregaria (Adult) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) LOX, COX-2 inhibitory activity assay using spectrophotometric method Zielińska et al. (2017)
Gryllodes sigillatus (Adult) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) LOX, COX-2 inhibitory activity assay using spectrophotometric method Zielińska et al. (2017)
Tenebrio molitor (Larvae) Hydrolysates Digestion using gastrointestinal enzymes (α-amylase, pepsin, pancreatin, and bile extract) LOX, COX-2 inhibitory activity assay using spectrophotometric method Zielińska et al. (2017)
Polyrhachis dives (Not described) 13 nitrogen-containing non-peptide compounds Extraction with ethanol and isolation with various chromatography Anti-proliferation assay using T lymphocytes isolated from mouse-erived spleens Tang et al. (2015)
TNF-α production assay using RAW 264.7 macrophages
COX, Jak3 inhibitory activity assay
Lycorma delicatula (Adult) Extracts Extraction with water IL-13 production assay using ConA-activated splenocytes Baek et al. (2018)
MMP expression assay using LPS-activated RAW 264.7 macrophages
Anti-obesity Tenebrio molitor (Larvae) Extracts Extraction with ethanol The following assays using 3T3-L1 adipocytes were performed: Oil red O staining assay, triglyceride content analysis, PCR, and western blot analysis Seo et al. (2017)
In vivo test using high-fat diet-induced obese mouse
Allomyrina dichotoma (Larvae) Extracts Extraction with ethanol The following assays using 3T3-L1 adipocytes were performed: Oil red O staining assay, triglyceride content analysis, PCR, and western analysis Chung et al. (2014)
Allomyrina dichotoma (Larvae) Powder Freeze-drying and grinding In vivo test using high-fat diet-fed mouse Yoon et al. (2015)
Clanis bilineata (Larvae) Chitooligosaccharides Extraction with chemical method from larvae skin (deproteinization, dehydration, and deacetylation) and then hydrolysis using α-amylase In vivo test using high-fat diet-fed rat Xia et al. (2013)
Anti-diabetic Musca domestica (Larvae) Water extracts (IC50 = 3.52 mg/mL) Extraction with water (decoction method) DPP-IV inhibitory activity assay using Gly-Pro-pNA substrate-spectrophotometric method Li et al. (2017)
Alphitobius diaperinus (Not described) Hydrolysates Digestion using gastrointestinal enzymes (pepsin and pancreatin) and then hydrolysis using thermolysin or alcalase or flavourzyme or papain DPP-IV inhibitory activity assay using Gly-Pro-AMC substrate-spectrophotometric method Lacroix et al. (2019)
Gryllodes sigillatus (Not described) Hydrolysates Hydrolysis using alcalase and then further digestion using gastrointestinal enzymes (pepsin, bile salt, and pancreatin) DPP-IV inhibitory activity assay using Gly-Pro-pNA substrate-spectrophotometric method Hall et al. (2018)
Anti-microbial Bombus terrestris and Bombus pascuorum (Not described) Peptides Immunization with E. coli and isolation peptide from the hemolymph E. coli growth inhibition assay Rahnamaeian et al. (2015)
Galleria mellonella (Larvae) Peptides Immunization with viable E. coli D31 and isolation peptide from the hemolymph Colony counting assay Cytryńska et al. (2007)
Radial diffusion assay
Gut microbiome Tenebrio molitor (Not described) Hydrolysates Digestion using gastrointestinal enzyme (α-amylase, pepsin, pancreatin, and bile solution) and dialysis In vitro gut microbiota simulation model (bacterial composition analyzed by fluorescence in situ hybridization and organic acid production analyzed by GC) de Carvalho et al. (2019)
Gryllodes sigillatus (Not described) Powder Drying and roasting Clinical crossover trial test (twenty healthy adults aged 20–48 participated) Stull et al. (2018)

Furthermore, Musca domestica (housefly) hydrolysates obtained using alcalase and neutral proteinase showed hydroxyl, superoxide anion, DPPH radical scavenging activity, reducing power activity, and metal chelating activity (Zhang et al., 2016). Amino acid analysis of the hydrolysates identified several acidic and basic amino acids, including Glu, Asp, Arg, and Lys, which had been shown to contribute to the antioxidant activity of the hydrolysates. According to Li et al. (2017), aromatic and hydrophobic amino acids present in the water extracts of M. domestica contribute to the DPPH radical scavenging activity of the extract. Furthermore, it has been reported that chitosan from protein-removed M. domestica larvae exerted higher antioxidant activity than ascorbic acid, which was used as a positive control (Ai et al., 2008).

del Hierro et al. (2020) and Messina et al. (2019) confirmed the antioxidant activity of extracts and hydrolysates from T. molitor and A. domesticus. Messina et al. (2019) prepared three different enzyme hydrolysates (Protamex, alcalase, and flavourzyme) using T. molitor and A. domesticus. Comparison of the DPPH radical scavenging activity of each hydrolysate confirmed that the Protamex hydrolysate had the highest antioxidant activity among the T. molitor hydrolysates, whereas the alcalase hydrolysate exerted the highest antioxidant activity among the A. domesticus hydrolysates. Messina et al. (2019) reported that the higher the degree of hydrolysis, the greater the production of low molecular weight peptides, resulting in higher DPPH radical scavenging activity. del Hierro et al. (2020) produced an extract of T. molitor and A. domesticus using water, ethanol, and their mixture. They found that DPPH radical scavenging activity increased as the total phenolic compound content of the two edible insect extracts increased. Furthermore, enzyme hydrolysate from T. molitor (Zielińska et al., 2017) and water and lipid extracts from A. domesticus (di Mattia et al., 2019) have all demonstrated antioxidant activities for scavenging free radicals.

Hall et al. (2018) and Zielińska et al. (2017) have reported the antioxidant activity of the hydrolysates obtained using gastrointestinal enzymes (e.g., pepsin, pancreatin, and α-amylase) of S. gregaria and Gryllodes sigillatus. The antioxidant activity of the hydrolysates was determined using the radical scavenging assay (DPPH and ABTS), metal chelating assay, and FRAP assay. Zielińska et al. (2017) confirmed that a hydrolysate of S. gregaria and G. sigillatus with a low molecular weight of ≤ 3 kDa exhibited excellent antioxidant activity and confirmed an increase in antioxidant activity according to the heat treatment of the hydrolysates. According to Hall et al. (2018), the antioxidant activity of hydrolysates increased when they were treated with gastrointestinal digestion, suggesting that digestion in vivo can help release the bioactive compounds present in the hydrolysates. Pattarayingsakul et al. (2017) identified antioxidant peptides from Oecophylla smaragdina hydrolysates obtained through gastrointestinal digestion followed by their separation and purification using ultrafiltration, size-exclusion chromatography, RP-HPLC, and LC–MS/MS. Finally, they isolated CTKKHKPNC peptide, which showed excellent antioxidant activity (IC50 values: 48.2 μM for DPPH assay and 38.4 μM for ABTS assay).

Song et al. (2013) studied chitosan isolated from Chrysomya megacephala (blowfly) larvae using chemical methods, including deproteinization, decolorization, decalcification, and deacetylation. Using the DPPH assay, C. megacephala chitosan showed excellent antioxidant activity compared to commercial chitosan, with an IC50 value of 1.2 mg/mL. This study confirmed that the content of active hydroxyl and amino groups in the chitosan polymer chains is related to the antioxidant activity. Additionally, it was reported that the ease of breaking the inter- and intra-hydrogen bonds due to low molecular weight would have contributed to the high antioxidant activity.

Anti-hypertensive activity

High blood pressure is a well-known major risk factor for cardiovascular diseases (Pattarayingsakul et al., 2017). Angiotensin-converting enzyme (ACE) plays a crucial role in the renin-angiotensin system. ACE converts angiotensin I into angiotensin II, inducing vasoconstriction and increasing blood pressure (Vercruysse et al., 2009a). Therefore, ACE inhibitors have been studied as anti-hypertensive therapeutic agents, and several studies on ACE inhibitory activity have been conducted to investigate the anti-hypertensive activities of edible insects (Table 3).

Studies on hydrolysates and peptides with anti-hypertensive activity have been reported in larvae and pupae of B. mori (Vercruysse et al., 2005; Wang et al., 2011; Wu et al., 2011; Wu et al., 2015). Vercruysse et al. (2005) hydrolyzed B. mori larvae with pepsin, trypsin, and α-chymotrypsin (gastrointestinal digestion) to obtain enzymatic hydrolysates. In this study, two methods (FAPGG substrate-spectrophotometric method and DTG substrate-HPLC method) were used to determine ACE inhibitory activity. After gastrointestinal digestion, the ACE inhibitory activity was greatly increased. The IC50 value decreased from 72.47 (non-hydrolyzed sample) to 0.69 mg/mL (gastrointestinal digestion) via DTG substrate-HPLC method. Similarly, Wu et al. (2011) estimated the ACE inhibitory activities of the hydrolysates obtained via gastrointestinal enzyme digestion using the HHL substrate-spectrophotometric method. The IC50 value of the hydrolysate was 8.3 μg/mL, which was close to that of captopril (5.3 μg/mL) used as a positive control. This was expected because of the increased C-terminal aromatic amino acid residue levels of the hydrolysate. It has been shown that α-chymotrypsin (classified as endo-peptidase) used for hydrolysis is known to cleave the C-terminal aromatic amino acid residue. Furthermore, Wang et al. (2011) and Wu et al. (2015) performed enzymatic hydrolysis in B. mori pupae. They then purified the hydrolysate following several steps (e.g., ultrafiltration, gel filtration, and RP-HPLC) to obtain specific peptides APPPKK (IC50 value: 47 μg/mL) and ASL (IC50 value: 102.15 μM) with ACE inhibitory activity. In these studies, a molecular docking analysis was conducted, and as a result, it was confirmed that the peptides were bound to the active site pocket of ACE. Furthermore, Wu et al. (2015), using the Lineweaver-Bruk plot analysis, reported that ASL inhibited ACE in a competitive inhibition pattern.

Furthermore, Vercruysse et al. (2008) and Vercruysse et al. (2009b) produced hydrolysates and peptides from Spodoptera littoralis (cotton leafworm) larvae and evaluated ACE inhibitory activity using the HHL substrate-spectrophotometric method. Vercruysse et al. (2008) hydrolyzed the non-water-soluble fraction of S. littoralis using gastrointestinal enzymes and identified a tri-peptide, AVF (IC50 value: 2,213 μM) with excellent ACE inhibitory activity through sequential separation and purification steps. Additionally, Vercruysse et al. (2009a) reported that when mucosal peptidase was treated after gastrointestinal digestion, the IC50 value decreased from 320 to 211 μg/mL, increasing ACE inhibitory activity.

The anti-hypertensive activity of peptides from T. molitor larvae was reported by Dai et al. (2013). In their study, the HHL substrate-HPLC method was used to determine the ACE inhibitory activity of the T. molitor alcalase hydrolysate. In addition, tri-peptide (YAN, IC50 value: 17 μg/mL) with excellent ACE inhibitory activity was isolated via separation and purification. Using this tri-peptide-containing fraction, an in vivo test was performed on a spontaneously hypertensive rat model, and it was confirmed that systolic blood pressure was reduced in a dose-dependent manner.

In addition, extracts of M. domestica (Li et al., 2017) and hydrolysates of Bombus terrestris (Vercruysse et al., 2005), G. sigillatus (Hall et al., 2018), and O. smaragdina (Pattarayingsakul et al., 2017) have been reported to have anti-hypertensive activity.

Anti-cancer and anti-inflammatory activity

Abnormal growth and proliferation of cells in the body are typical characteristics of cancer (Lee and Paik, 2019). Therefore, studies on substances that effectively inhibit the abnormal proliferation of such cancer cells are actively conducted, and several studies using food-derived proteins and peptides have been reported (Chalamaiah et al., 2018; Karami et al., 2019). In addition, inflammation has long been known to be associated with cancer pathophysiology, and it is known that the immune system, consisting of a network of cells, tissues, and organs, can effectively prevent cancer cell growth (Chalamaiah et al., 2018). Cancer immunotherapy has been reported as an important aspect of cancer research and treatment (Lee and Paik, 2019). Therefore, studies are being actively conducted to identify substances with anti-cancer and anti-inflammatory activities in edible insects (Table 3).

Several studies have reported that various components of edible insects have anti-cancer activity. Cho et al. (2019) obtained an ethanol extract from fermented B. mori. Briefly, the authors fermented B. mori for 3 days using Aspergillus kawachii before proceeding with extraction, followed by ethanol extraction to obtain the fermented ethanol extract. The fermented ethanol extract of B. mori showed higher inhibition activity of HepG2 hepatocellular carcinoma growth than the unfermented extract. The fermented extract showed increased DNA fragmentation, nucleic condensation, and an apoptotic cell population (Cho et al., 2019). In particular, the fermented extract increased the expression of pro-apoptotic proteins (Bax, and caspase-3, 8, and 9) while inhibiting the expression of the anti-apoptotic protein (Bcl-2), inducing cell apoptosis.

Chernysh et al. (2002) obtained the specific peptide alloferon 1 (HGVSGHGQHGVHG) from Calliphora vicina by immunization with a heat-killed bacterial mixture (Escherichia coli D31 and Micrococcus luteus A270). Alloferon 1 exhibited strong anti-cancer activity by stimulating the natural killer (NK) activity of lymphocytes in mouse spleen or human peripheral blood. It was confirmed that these simulated lymphocytes inhibited the growth of K562 tumor cells. In addition, the authors reported that in mice intranasally administered alloferon 1, interferon synthesis was effectively induced. Furthermore, it has been shown that chitosan isolated from M. domestica larvae has anti-cancer activity (Ai et al., 2008). Briefly, chitosan (1 mg/mL) effectively inhibited the growth of HeLa (50.8%) and S-180 (52.9%) tumor cells in an in vitro MTT assay (Ai et al., 2008).

Edible insects that have anti-inflammatory activity include Polyrhachis dives (Tang et al., 2015), Lycorma delicatula (Baek et al., 2018), G. sigillatus, T. molitor, and S. gregaria (Zielińska et al., 2017). Tang et al. (2015), using ethanol extraction and chromatography separation, obtained 13 nitrogen-containing non-peptide compounds that showed anti-inflammatory activity. The authors showed that some of these compounds effectively inhibited the proliferation of ConA-stimulated T lymphocytes isolated from mouse spleens. In addition, they claimed that the identified compound not only inhibited the production of TNF-α in RAW 264.7 macrophages induced by lipopolysaccharide (LPS) but also inhibited cyclooxygenase (COX)-1 and Jak3 kinase activities, which are associated with inflammatory responses.

Furthermore, the aqueous fraction of L. delicatula has been reported to show anti-inflammatory activity in LPS-induced RAW 264.7 macrophages and ConA-induced splenocytes (Baek et al., 2018). Briefly, the secretion of the pro-inflammatory cytokine, interleukin (IL)-13, was inhibited by the aqueous fraction of L. delicatula in splenocytes in a dose-dependent manner. In addition, the increased expression of matrix metalloproteinases (MMPs) in LPS-stimulated RAW 264.7 macrophages was decreased by treatment with L. delicatula aqueous fraction. These MMPs are known to induce the elevation of inflammatory mediators (growth factors, inflammatory cytokines, and adhesion molecules) and exacerbate the inflammatory response (Hong et al., 2015).

Zielińska et al. (2017) reported that hydrolysates obtained from G. sigillatus, T. molitor, and S. gregaria showed inhibitory activity against lipoxygenase (LOX) and CoX-2. LOX and COX-2 are associated with the production of leukotrienes and prostaglandins, respectively, and overproduction of these substances can lead to dysregulated inflammatory responses (Shrivastava et al., 2017). According to Zielińska et al. (2017), the inhibitory activity of edible insect hydrolysates after the in vitro absorption process was higher than before.

Anti-obesity and anti-diabetic activity

Obesity and diabetes are metabolic disorders that cause various health problems and are the leading causes of death worldwide (Bais and Patel, 2020). In addition, obesity is a major risk factor for type 2 diabetes (Chung et al., 2014). Accordingly, research has been actively conducted to identify substances with anti-obesity and anti-diabetic activities in edible insects (Table 3).

Chung et al. (2014) and Yoon et al. (2015) reported that A. dichotoma (Korean horn beetle) has anti-obesity activity. Chung et al. (2014) obtained ethanol extracts of A. dichotoma and measured the anti-obesity activity through lipid accumulation and adipocyte-specific gene expression analysis using 3T3-L1 adipocytes. They showed that the treatment with extracts substantially inhibited intracellular lipid accumulation and reduced triglyceride content. Furthermore, extracts of A. dichotoma inhibited the expression of CCAAT/enhancer-binding protein (C/EBP)-α, peroxisome proliferator-activated receptor (PPAR), fatty acid synthase, adipocyte fatty acid-binding protein (aP2), lipoprotein lipase (LPL), and stearoyl-coenzyme desaturase-1, which are related to adipogenesis and lipogenesis. Yoon et al. (2015) studied the anti-obesity effect of A. dichotoma in vivo using a high-fat diet (HFD) model. Like in vitro studies, administration of A. dichotoma was shown to inhibit the expression of PPAR-γ, C/EBP-α, and LPL in HFD-fed mice. In addition, serum analysis showed reduced contents of triglycerides and leptin, which are positively correlated with adiposity (Yoon et al., 2015).

T. molitor larvae have been reported to exert anti-obesity activity by inhibiting adipogenesis through the AMP-activated protein kinase and mitogen-activated protein kinase signaling pathways in 3T3-L1 adipocytes and attenuating the expression of adipocyte-specific marker genes and the body weight gain in an HFD mouse model (Seo et al., 2017).

Additionally, Xia et al. (2013) reported that chitooligosaccharides isolated from C. bilineata larvae have hypolipidemic activity. In this study, chitin was extracted from the skin of C. bilineata larvae and then hydrolyzed by α-amylase to produce chitooligosaccharides. These chitooligosaccharides reduced weight gain in HFD-fed rats and increased fecal fat and cholesterol contents, indicating that chitooligosaccharides have a strong binding capacity for fat and cholesterol. In addition, via plasma lipid content analysis, it was confirmed that the content of triacylglycerol, total cholesterol, and low-density lipoprotein cholesterol was lowered, whereas high-density lipoprotein cholesterol content was increased. Thus, it was confirmed that chitooligosaccharides derived from C. bilineata have an excellent effect on improving blood lipid levels in HFD mice.

Edible insects that have anti-diabetic activity include M. domestica (Li et al., 2017), A. diaperinus (Lacroix et al., 2019), and G. sigillatus (Hall et al., 2018). These three edible insects were shown to inhibit dipeptidyl peptidase (DPP)-IV enzyme activity, a serine protease that hydrolyzes incretin hormones. DPP-IV inhibitors are effective against type 2 diabetes, as they inhibit the breakdown of the incretin hormone, which plays a major role in regulating blood glucose levels (Seshadri and Kirubha, 2009).

Lacroix et al. (2019) obtained hydrolysates of A. diaperinus by gastrointestinal digestion followed by hydrolysis using four enzymes (thermolysin, alcalase, flavourzyme, and papain). Among them, it was confirmed that thermolysin hydrolysate showed strong DPP-IV inhibitory activity. In contrast, Hall et al. (2018) first hydrolyzed G. sigillatus with alcalase, followed by a gastrointestinal digestion test to obtain the hydrolysate. In this work, the authors confirmed that gastrointestinal digestion conditions enhanced the DPP-IV inhibitory activity of the hydrolysate. Furthermore, Li et al. (2017) conducted water extraction of M. domestica, separated the fractions through ultrafiltration, and compared the DPP-IV inhibitory activity according to the molecular weight. It has been reported that the high molecular weight fraction of water extract has a high DPP-IV inhibitory activity. Amino acid analysis revealed that the high molecular weight fraction contained high levels of Ile, Leu, Val, Pro, Gly, Ala, and Phe amino acids. These amino acids identified in other studies reporting DPP-IV inhibitory activity (Xia et al., 2017) are speculated to contribute to the DPP-IV inhibitory activity of M. domestica.

Anti-microbial activity and effects on the gut microbiome

The gut microbiota is composed of a wide variety of microbes that interact with the immune system and play an important role in human health (Zong et al., 2020). Changes in the gut microbiota composition are associated with several diseases (Mahnic et al., 2020). Reportedly, anti-microbial peptide (AMP), a defense system found in all organisms, plays a major role in host-microbe interactions (Zong et al., 2020). Since the isolation of cecropin from Hyalophora cecropia in 1980 (Hultmark et al., 1980), multiple AMPs have been isolated from various edible insects (Table 3).

AMPs derived from edible insects were produced through immunization with E. coli (Cytryńska et al., 2007; Rahnamaeian et al., 2015). Cytryńska et al. (2007) isolated and purified eight peptides from G mellonella larvae and determined their anti-microbial activity against Gram-positive and Gram-negative bacteria. The authors reported that seven of the eight purified peptides showed anti-microbial activity against at least one or more Gram-positive bacteria, whereas, against Gram-negative bacteria, only one purified peptide (Gm cecropin D-like peptide) had anti-microbial activity against E. coli D31. In addition, peptides from Galleria mellonella inhibited the growth of several yeast and filamentous fungi. Rahnamaeian et al. (2015) studied two AMPs, abaecin and hymenoptaecin, isolated from B. pascuorum and B. terrestris. The AMPs, when applied individually to E. coli D31, did not exert distinct anti-microbial activity; however, the growth of E. coli was effectively inhibited when both AMPs were treated simultaneously. These results suggested that anti-microbial activity exerted by the AMPs could be interdependent, where one AMP damaged the bacterial envelope, allowing the other AMP to pass through the membrane and access the intracellular target. In addition, it has been reported that AMPs isolated from Spodoptera littoralis (Seufi et al., 2011), Anopheles gambiae (Vizioli et al., 2000), and Papilio xuthus (Kim et al., 2010) have anti-microbial activity against various Gram-positive and Gram-negative bacteria.

To the best of our knowledge, no previous studies have directly confirmed the effects of AMP-derived edible insects on the gut microbiota. However, a few studies have reported the effects of edible insect ingestion on the gut microbiota. For instance, de Carvalho et al. (2019) studied the effect of T. molitor flour on the human gut microbiota using an in vitro digestion model. In this study, the in vitro digestion of T. molitor was performed by implementing the digestion process using α-amylase, pepsin, pancreatin, and bile solution and absorption into the small intestine through the dialysis step. The effect of the obtained sample on the gut microbiota was confirmed by measuring the growth of fecal bacteria and the production of organic acids. As a result, it was confirmed that T. molitor flour has a positive effect on the growth of Bacteroidaceae and Prevotellaceae, which can produce propionate, a beneficial organic salt. Additionally, the organic acid analyses performed in this study confirmed that T. molitor has a positive effect on the production of short-chain fatty acids (SCFAs), especially acetate and propionate (de Carvalho et al., 2019). These SCFAs help regulate intermediate and peripheral metabolism by acting as signaling molecules between the microbiota and the human host.

Stull et al. (2018), using a crossover trial study, reported that the consumption of G. sigillatus increased Bifidobacterium animalis growth, a well-known beneficial probiotic that improves gastrointestinal function, prevents diarrhea, and reduces the side effects of antibiotic use. In addition, G. sigillatus consumption has been reported to markedly reduce the amount of plasma TNF-α, suggesting that it may help reduce systemic inflammation.

Food application of edible insect

Various species of edible insects are consumed as food in more than 100 countries worldwide. In addition, the species of insects consumed vary from country to country (Kim et al., 2019). Edible insects have historically been cultivated or harvested in the wild and consumed in Asia, South America, Central America, and Africa, where they are considered a part of the traditional diet (Dobermann et al., 2017). However, interest in edible insects has increased in Europe and North America over the past decade (Melgar-Lalanne et al., 2019). Approximately 130 brands and companies have been identified that commercialize edible insect-based food, and several types of processed food products using edible insects, including pasta, chips, snacks, biscuits, chocolate, candy, energy bar, protein bar, protein powder, beer, bread, meatballs, burgers, and sausages, have been reported (Kim et al., 2019; Melgar-Lalanne et al., 2019). As mentioned above, edible insects have a high level of protein and a well-balanced amino acid profile. In addition, it has been confirmed that studies using proteins occupy a large part of research on the biological activities of edible insects. Therefore, it has been reported that protein occupies a large part in applying edible insects to food.

Reportedly, edible insect proteins positively affect the quality and nutritional aspects of meat products by being applied as an additive to various meat products. Kim et al. (2016) and Kim et al. (2017) manufactured emulsified meat products containing edible insect proteins. The authors reported that sausages containing edible insect protein could be manufactured without a substantial difference in quality characteristics compared to existing products. In addition, Scholliers et al. (2020) reported that cooked sausages made by partially replacing meat with insect proteins had positive effects on cooking loss. Park et al. (2017) reported that meat batter formulated with B. mori powder had improved nutritional value and reduced cooking loss. However, the addition of transglutaminase further improved the texture and physicochemical properties of meat batter, suggesting the combination of silkworm pupae and transglutaminase as a new nutritional and functional source.

Preparation of processed food products using edible insect powder as an ingredient is a widely used method to avoid the reluctance of consuming whole insects (Ordoñez-Araque and Egas-Montenegro, 2021). In addition, because of its high-protein content, low-fat content, rich dietary fiber, and rich minerals such as calcium, phosphorus, and iron, many studies have confirmed their potential as additives or substitutes for grain-based food (Osimani et al., 2018). For instance, Haber et al. (2019) produced bread with added S. gregaria powder, which increased the nutritional value and reported no marked difference in color, texture, and taste parameters compared to conventional bread. Similarly, Biró et al. (2019) reported that buckwheat pasta prepared from flour enriched with B. mori powder showed higher protein content and masked the disliked flavor imparted by buckwheat. Furthermore, Azzollini et al. (2018) prepared a snack with added T. molitor powder, which showed improved nutritional properties and increased digestibility of the snack. Collectively, these studies demonstrated that the addition of edible insects increased the nutritional value of the product and had compensatory advantages for existing problems. These studies (Azzollini et al., 2018; Biró et al., 2019; Haber et al., 2019) suggest that determining the appropriate amount of edible insects to add is of paramount importance to prepare a product with fortified nutrition without compromising its physicochemical properties.

In conclusion, edible insects are a promising novel protein source globally, as they have excellent nutritional composition, especially amino acids. As described in this review, when edible insects are appropriately processed, including protein, lipid, chitin, and chitosan extraction processes, they can be used in various fields as an excellent food resource. Recent studies have also reported various biological activities of edible insects, including antioxidant, anti-hypertensive, anti-cancer, anti-inflammatory, anti-obesity, anti-diabetic, and anti-microbial activities. According to the functional properties of these edible insects, they could also serve as potential novel medicinal resources.

However, despite the considerable value of edible insects, further research is needed to use edible insects in the food and pharmaceutical industries. First, the consumer safety of insect proteins must be considered. Safety problems, such as allergies to edible insects, have been reported, but in-depth studies to decipher the specific causes and control methods are still needed. Second, functional components of edible insects, such as bioactive peptides, need to be identified, and their biological activity should be validated by performing adequate in vitro and in vivo experiments prior to determining their biological activity in humans. Finally, it is necessary to establish a system to standardize the use of edible insects in the food and pharmaceutical industries. If the above problems are solved, in the future, the edible insect industry will expand.

Acknowledgements

This research was funded by the Main Research Program (E0211100-01) of the Korea Food Research Institute.

Declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Footnotes

Publisher's Note

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

Contributor Information

Jae Hoon Lee, Email: Leejaehoon@kfri.re.kr.

Tae-Kyung Kim, Email: privacykin@naver.comr.

Chang Hee Jeong, Email: jeongch@wikim.re.kr.

Hae In Yong, Email: awsm_y@kfri.re.kr.

Ji Yoon Cha, Email: Chajiyoon@kfri.re.kr.

Bum-Keun Kim, Email: bkkim@kfri.re.kr.

Yun-Sang Choi, Email: kcys0517@kfri.re.kr.

References

  1. Adámek M, Adámková A, Mlček J, Borkovcová M, Bednářová M. Acceptability and sensory evaluation of energy bars and protein bars enriched with edible insect. Potravinarstvo Slovak Journal of Food Sciences. 2018;12:431–437. [Google Scholar]
  2. Ai H, Wang F, Yang Q, Zhu F, Lei C. Preparation and biological activities of chitosan from the larvae of housefly, Musca Domestica. Carbohydrate Polymers. 2008;72:419–423. doi: 10.1016/j.carbpol.2007.09.010. [DOI] [Google Scholar]
  3. Asp NG. Nutritional importance and classification of food carbohydrates. In: Meuser F, Manners DJ, Seibel W, editors. Plant Polymeric Carbohydrates. Cambridge: Woodhead Publishing; 1993. pp. 121–126. [Google Scholar]
  4. Azzollini D, Derossi A, Fogliano V, Lakemond C, Severini C. Effects of formulation and process conditions on microstructure, texture and digestibility of extruded insect-riched snacks. Innovative Food Science and Emerging Technologies. 2018;45:344–353. doi: 10.1016/j.ifset.2017.11.017. [DOI] [Google Scholar]
  5. Baek SH, Joung O, Lee HY, Shin JC, Choi WS, Lee TH, Hwang JS, Nam SH, Son HU, Lee SH. Anti-oxidative fraction of Lycorma delicatula alleviates inflammatory indicators. Natural Product Communications. 2018;13:431–434. doi: 10.1177/1934578X1801300413. [DOI] [Google Scholar]
  6. Bais S, Patel NJ. In vitro anti diabetic and anti obesity effect of J. communis extract on 3T3L1 mouse adipocytes: A possible role of MAPK/ERK activation. Obesity Medicine. 2020;18:100219. doi: 10.1016/j.obmed.2020.100219. [DOI] [Google Scholar]
  7. Barsics F, Megido RC, Brostaux Y, Barsics C, Blecker C, Haubruge E, Francis F. Could new information influence attitudes to foods supplemented with edible insects? British Food Journal. 2017;119:2027–2039. doi: 10.1108/BFJ-11-2016-0541. [DOI] [Google Scholar]
  8. Biró B, Fodor R, Szedljak I, Pásztor-Huszár K, Gere A. Buckwheat-pasta enriched with silkworm powder: Technological analysis and sensory evaluation. LWT-Food Science and Technology. 2019;116:108542. doi: 10.1016/j.lwt.2019.108542. [DOI] [Google Scholar]
  9. Brigode C, Hobbi P, Jafari H, Verwilghen F, Baeten E, Shavandi A. Isolation and physicochemical properties of chitin polymer from insect farm side stream as a new source of renewable biopolymer. Journal of Cleaner Production. 2020;275:122924. doi: 10.1016/j.jclepro.2020.122924. [DOI] [Google Scholar]
  10. Broekman H, Knulst A, den Hartog Jager S, Monteleone F, Gaspari M, de Jong G, Houben G, Verhoeckx K. Effect of thermal processing on mealworm allergenicity. Molecular Nutrition and Food Research. 2015;59:1855–1864. doi: 10.1002/mnfr.201500138. [DOI] [PubMed] [Google Scholar]
  11. Bruinsma J. World agriculture: towards 2015/2030: an FAO perspective. Earthscan (2003)
  12. Bukkens SG. The nutritional value of edible insects. Ecology of Food and Nutrition. 1997;36:287–319. doi: 10.1080/03670244.1997.9991521. [DOI] [Google Scholar]
  13. Chalamaiah M, Yu W, Wu J. Immunomodulatory and anticancer protein hydrolysates (peptides) from food proteins: A review. Food Chemistry. 2018;245:205–222. doi: 10.1016/j.foodchem.2017.10.087. [DOI] [PubMed] [Google Scholar]
  14. Chernysh S, Kim S, Bekker G, Pleskach V, Filatova N, Anikin V, Platonov V, Bulet P. Antiviral and antitumor peptides from insects. Proceedings of the National Academy of Sciences. 2002;99:12628–12632. doi: 10.1073/pnas.192301899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cho HD, Min HJ, Won YS, Ahn HY, Cho YS, Seo KI. Solid state fermentation process with Aspergillus kawachii enhances the cancer-suppressive potential of silkworm larva in hepatocellular carcinoma cells. BMC Complementary and Alternative Medicine. 2019;19:241. doi: 10.1186/s12906-019-2649-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Choi BD, Wong NA, Auh JH. Defatting and sonication enhances protein extraction from edible insects. Korean Journal for Food Science of Animal Resources. 2017;37:955–961. doi: 10.5851/kosfa.2017.37.6.955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Choi KH, Yoon JW, Kim M, Jeong J, Ryu M, Park S, Jo C, Lee CK. Optimization of culture conditions for maintaining pig muscle stem cells in vitro. Food Science of Animal Resources. 2020;40:659–667. doi: 10.5851/kosfa.2020.e39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chung MY, Yoon YI, Hwang JS, Goo TW, Yun EY. Anti-obesity effect of Allomyrina dichotoma (Arthropoda: Insecta) larvae ethanol extract on 3T3-L1 adipocyte differentiation. Entomological Research. 2014;44:9–16. doi: 10.1111/1748-5967.12044. [DOI] [Google Scholar]
  19. Cicero AFG, Fogacci F, Colletti A. Potential role of bioactive peptides in prevention and treatment of chronic diseases: A narrative review. British Journal of Pharmacology. 2017;174:1378–1394. doi: 10.1111/bph.13608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cytryńska M, Mak P, Zdybicka-Barabas A, Suder P, Jakubowicz T. Purification and characterization of eight peptides from Galleria mellonella immune hemolymph. Peptides. 2007;28:533–546. doi: 10.1016/j.peptides.2006.11.010. [DOI] [PubMed] [Google Scholar]
  21. da Silva Lucas AJ, de Oliveira LM, da Rocha M, Prentice C. Edible insects: An alternative of nutritional, functional and bioactive compounds. Food Chemistry. 2020;311:126022. doi: 10.1016/j.foodchem.2019.126022. [DOI] [PubMed] [Google Scholar]
  22. Dai CH, Ma HL, Luo L, Yin XL. Angiotensin I-converting enzyme (ACE) inhibitory peptide derived from Tenebrio molitor (L.) larva protein hydrolysate. European Food Research and Technology. 2013;236:681–689. doi: 10.1007/s00217-013-1923-z. [DOI] [Google Scholar]
  23. de Gier S, Verhoeckx K. Insect (food) allergy and allergens. Molecular Immunology. 2018;100:82–106. doi: 10.1016/j.molimm.2018.03.015. [DOI] [PubMed] [Google Scholar]
  24. de Carvalho NM, Walton G, Poveda C, Silva S, Amorim M, Madureira A, Pintado M, Gibson G, Jauregi P. Study of in vitro digestion of Tenebrio molitor flour for evaluation of its impact on the human gut microbiota. Journal of Functional Foods. 2019;59:101–109. doi: 10.1016/j.jff.2019.05.024. [DOI] [Google Scholar]
  25. DeFoliart GR. Insects as food: why the western attitude is important. Annual Review of Entomology. 1999;44:21–50. doi: 10.1146/annurev.ento.44.1.21. [DOI] [PubMed] [Google Scholar]
  26. del Hierro JN, Gutiérrez-Docio A, Otero P, Reglero G, Martin D. Characterization, antioxidant activity, and inhibitory effect on pancreatic lipase of extracts from the edible insects Acheta domesticus and Tenebrio molitor. Food Chemistry. 2020;309:125742. doi: 10.1016/j.foodchem.2019.125742. [DOI] [PubMed] [Google Scholar]
  27. Dhillon GS, Kaur S, Brar SK, Verma M. Green synthesis approach: Extraction of chitosan from fungus mycelia. Critical Reviews in Biotechnology. 2013;33:379–403. doi: 10.3109/07388551.2012.717217. [DOI] [PubMed] [Google Scholar]
  28. di Mattia C, Battista N, Sacchetti G, Serafini M. Antioxidant activities in vitro of water and liposoluble extracts obtained by different species of edible insects and invertebrates. Frontiers in Nutrition. 2019;6:106. doi: 10.3389/fnut.2019.00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dobermann D, Swift JA, Field LM. Opportunities and hurdles of edible insects for food and feed. Nutrition Bulletin. 2017;42:293–308. doi: 10.1111/nbu.12291. [DOI] [Google Scholar]
  30. FAO/WHO/UNU . Energy and protein requirements: report of a joint FAO/WHO/UNU expert consultation. Geneva: Food and Agriculture Organization/World Health Organization/United Nations University; 1985. p. 206. [Google Scholar]
  31. Foroudi S, Potter AS, Stamatikos A, Patil BS, Deyhim F. Drinking orange juice increases total antioxidant status and decreases lipid peroxidation in adults. Journal of Medicinal Food. 2014;17:612–617. doi: 10.1089/jmf.2013.0034. [DOI] [PubMed] [Google Scholar]
  32. Gartner C, Peláez CA, López BL. Characterization of chitin and chitosan extracted from shrimp shells by two methods. e-Polymers. 2010;10:69. doi: 10.1515/epoly.2010.10.1.748. [DOI] [Google Scholar]
  33. Ghosh S, Lee SM, Jung C, Meyer-Rochow VB. Nutritional composition of five commercial edible insects in South Korea. Journal of Asia-Pacific Entomology. 2017;20:686–694. doi: 10.1016/j.aspen.2017.04.003. [DOI] [Google Scholar]
  34. Gopal J, Muthu M, Dhakshanamurthy T, Kim KJ, Hasan N, Kwon SJ, Chun S. Sustainable ecofriendly phytoextract mediated one pot green recovery of chitosan. Scientific Reports. 2019;9:13832. doi: 10.1038/s41598-019-50133-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Govorushko S. Global status of insects as food and feed source: A review. Trends in Food Science and Technology. 2019;91:436–445. doi: 10.1016/j.tifs.2019.07.032. [DOI] [Google Scholar]
  36. Gravel A, Doyen A. The use of edible insect proteins in food: Challenges and issues related to their functional properties. Innovative Food Science and Emerging Technologies. 2020;59:102272. doi: 10.1016/j.ifset.2019.102272. [DOI] [Google Scholar]
  37. Haber M, Mishyna M, Martinez JI, Benjamin O. The influence of grasshopper (Schistocerca gregaria) powder enrichment on bread nutritional and sensorial properties. LWT-Food Science and Tehcnology. 2019;115:108395. doi: 10.1016/j.lwt.2019.108395. [DOI] [Google Scholar]
  38. Hall F, Johnson PE, Liceaga A. Effect of enzymatic hydrolysis on bioactive properties and allergenicity of cricket (Gryllodes sigillatus) protein. Food Chemistry. 2018;262:39–47. doi: 10.1016/j.foodchem.2018.04.058. [DOI] [PubMed] [Google Scholar]
  39. Hong JM, Kwon OK, Shin IS, Song HH, Shin NR, Jeon CM, Oh SR, Han SB, Ahn KS. Anti-inflammatory activities of Physalis alkekengi var. franchetii extract through the inhibition of MMP-9 and AP-1 activation. Immunobiology. 2015;220:1–9. doi: 10.1016/j.imbio.2014.10.004. [DOI] [PubMed] [Google Scholar]
  40. Hongkulsup C, Khutoryanskiy VV, Niranjan K. Enzyme assisted extraction of chitin from shrimp shells (Litopenaeus vannamei) Journal of Chemical Technology and Biotechnology. 2016;91:1250–1256. doi: 10.1002/jctb.4714. [DOI] [Google Scholar]
  41. Huang WC, Zhao D, Guo N, Xue C, Mao X. Green and facile production of chitin from crustacean shells using a natural deep eutectic solvent. Journal of Agricultural and Food Chemistry. 2018;66:11897–11901. doi: 10.1021/acs.jafc.8b03847. [DOI] [PubMed] [Google Scholar]
  42. Hultmark D, Steiner H, Rasmuson T, Boman HG. Insect immunity. Purification and properties of three inducible bactericidal proteins from hemolymph of immunized pupae of Hyalophora cecropia. European Journal of Biochemistry. 1980;106:7–16. doi: 10.1111/j.1432-1033.1980.tb05991.x. [DOI] [PubMed] [Google Scholar]
  43. Hwang J, Choe JY. How to enhance the image of edible insect restaurants: Focusing on perceived risk theory. International Journal of Hospitality Management. 2020;87:102464. doi: 10.1016/j.ijhm.2020.102464. [DOI] [Google Scholar]
  44. Imathiu S. Benefits and food safety concerns associated with consumption of edible insects. NFS Journal. 2020;18:1–11. doi: 10.1016/j.nfs.2019.11.002. [DOI] [Google Scholar]
  45. Janssen RH, Vincken JP, van den Broek LA, Fogliano V, Lakemond CM. Nitrogen-to-protein conversion factors for three rdible insects: Tenebrio molitor, Alphitobius diaperinus, and Hermetia illucens. Journal of Agricultural and Food Chemistry. 2017;65:2275–2278. doi: 10.1021/acs.jafc.7b00471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kachapulula PW, Akello J, Bandyopadhyay R, Cotty PJ. Aflatoxin contamination of dried insects and fish in Zambia. Journal of Food Protection. 2018;81:1508–1518. doi: 10.4315/0362-028X.JFP-17-527. [DOI] [PubMed] [Google Scholar]
  47. Karami Z, Peighambardoust SH, Hesari J, Akbari-Adergani B, Andreu D. Antioxidant, anticancer and ACE-inhibitory activities of bioactive peptides from wheat germ protein hydrolysates. Food Bioscience. 2019;32:100450. doi: 10.1016/j.fbio.2019.100450. [DOI] [Google Scholar]
  48. Kaur S, Dhillon GS. Recent trends in biological extraction of chitin from marine shell wastes: A review. Critical Reviews in Biotechnology. 2015;35:44–61. doi: 10.3109/07388551.2013.798256. [DOI] [PubMed] [Google Scholar]
  49. Kaya M, Sargin I, Erdonmez D. Microbial biofilm activity and physicochemical characterization of biodegradable and edible cups obtained from abdominal exoskeleton of an insect. Innovative Food Science and Emerging Technologies. 2016;36:68–74. doi: 10.1016/j.ifset.2016.05.018. [DOI] [Google Scholar]
  50. Khampakool A, Soisungwan S, You S, Park SH. Infrared assisted freeze-drying (IRAFD) to produce shelf-stable insect food from Protaetia brevitarsis (white-spotted flower chafer) larva. Food Science of Animal Resources. 2020;40:813–830. doi: 10.5851/kosfa.2020.e60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Khanafari A, Marandi R, Sanati S. Recovery of chitin and chitosan from shrimp waste by chemical and microbial methods. Iranian Journal of Health and Environment. 2008;5:19–24. [Google Scholar]
  52. Kim SR, Hong MY, Park SW, Choi KH, Yun EY, Goo TW, Kang SW, Suh HJ, Kim I, Hwang JS. Characterization and cDNA cloning of a cecropin-like antimicrobial peptide, papiliocin, from the swallowtail butterfly, Papilio Xuthus. Molecules and Cells. 2010;29:419–423. doi: 10.1007/s10059-010-0050-y. [DOI] [PubMed] [Google Scholar]
  53. Kim HW, Setyabrata D, Lee YJ, Jones OG, Kim YHB. Pre-treated mealworm larvae and silkworm pupae as a novel protein ingredient in emulsion sausages. Innovative Food Science and Emerging Technologies. 2016;38:116–123. doi: 10.1016/j.ifset.2016.09.023. [DOI] [Google Scholar]
  54. Kim HW, Setyabrata D, Lee Y, Jones OG, Kim YHB. Effect of house cricket (Acheta domesticus) flour addition on physicochemical and textural properties of meat emulsion under various formulations. Journal of Food Science. 2017;82:2787–2793. doi: 10.1111/1750-3841.13960. [DOI] [PubMed] [Google Scholar]
  55. Kim TK, Yong HI, Kim YB, Kim HW, Choi YS. Edible insects as a protein source: A review of public perception, processing technology, and research trends. Food Science of Animal Resources. 2019;39:521–540. doi: 10.5851/kosfa.2019.e53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kim TK, Yong HI, Chun HH, Lee MA, Kim YB, Choi YS. Changes of amino acid composition and protein technical functionality of edible insects by extracting steps. Journal of Asia-Pacific Entomology. 2020;23:298–305. doi: 10.1016/j.aspen.2019.12.017. [DOI] [Google Scholar]
  57. Kim TK, Yong HI, Kang MC, Jung S, Jang HW, Choi YS. Effects of high hydrostatic pressure on technical functional properties of edible insect protein. Food Science of Animal Resources. 2021;41:185–195. doi: 10.5851/kosfa.2020.e85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kim TK, Yong HI, Kim YB, Jung S, Kim HW, Choi YS. Effects of organic solvent on functional properties of defatted proteins extracted from Protaetia brevitarsis larvae. Food Chemistry. 2021;336:127679. doi: 10.1016/j.foodchem.2020.127679. [DOI] [PubMed] [Google Scholar]
  59. Kinsella JE, Melachouris N. Functional properties of proteins in foods: A survey. Critical Reviews in Food Science and Nutrition. 1976;7:219–280. doi: 10.1080/10408397609527208. [DOI] [Google Scholar]
  60. Kwak KW, Kim SY, An KS, Kim YS, Park K, Kim E, Hwang JS, Kim MA, Ryu HY, Yoon HJ. Subacute oral toxicity evaluation of freeze-dried powder of Locusta migratoria. Food Science of Animal Resources. 2020;40:795–812. doi: 10.5851/kosfa.2020.e55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. la Barbera F, Verneau F, Amato M, Grunert K. Understanding Westerners’ disgust for the eating of insects: The role of food neophobia and implicit associations. Food Quality and Preference. 2018;64:120–125. doi: 10.1016/j.foodqual.2017.10.002. [DOI] [Google Scholar]
  62. Lacroix IME, Dávalos Terán ID, Fogliano V, Wichers HJ. Investigation into the potential of commercially available lesser mealworm (A. diaperinus) protein to serve as sources of peptides with DPP-IV inhibitory activity. International Journal of Food Science and Technology. 2019;54:696–704. doi: 10.1111/ijfs.13982. [DOI] [Google Scholar]
  63. Laroche M, Perreault V, Marciniak A, Gravel A, Chamberland J, Doyen A. Comparison of conventional and sustainable lipid extraction methods for the production of oil and protein isolate from edible insect meal. Foods. 2019;8:572. doi: 10.3390/foods8110572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Lee JH, Paik HD. Anticancer and immunomodulatory activity of egg proteins and peptides: A review. Poultry Science. 2019;98:6505–6516. doi: 10.3382/ps/pez381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Lee S, Jo K, Yong HI, Choi YS, Jung S. Comparison of the in vitro protein digestibility of Protaetia brevitarsis larvae and beef loin before and after defatting. Food Chemistry. 2021;338:128073. doi: 10.1016/j.foodchem.2020.128073. [DOI] [PubMed] [Google Scholar]
  66. Li H, Inoue A, Taniguchi S, Yukutake T, Suyama K, Nose T, Maeda I. Multifunctional biological activities of water extract of housefly larvae (Musca domestica) PharmaNutrition. 2017;5:119–126. doi: 10.1016/j.phanu.2017.09.001. [DOI] [Google Scholar]
  67. Liu Y, Wan S, Liu J, Zou Y, Liao S. Antioxidant activity and stability study of peptides from enzymatically hydrolyzed male silkmoth. Journal of Food Processing and Preservation. 2017;41:e13081. doi: 10.1111/jfpp.13081. [DOI] [Google Scholar]
  68. Long S, Ying F, Zhao H, Tao M, Xin Z. Studies on alkaline solution extraction of polysaccharide from silkworm pupa and its immuno-modulating activities. Forest Research. 2007;20:782–786. [Google Scholar]
  69. Lopata AL, O'Hehir RE, Lehrer SB. Shellfish allergy. Clinical and Experimental Allergy. 2010;40:850–858. doi: 10.1111/j.1365-2222.2010.03513.x. [DOI] [PubMed] [Google Scholar]
  70. Mahnic A, Breskvar M, Dzeroski S, Skok P, Pintar S, Rupnik M. Distinct types of gut microbiota dysbiosis in hospitalized gastroenterological patients are disease non-related and characterized with the predominance of either Enterobacteriaceae or Enterococcus. Frontiers in Microbiology. 2020;11:120. doi: 10.3389/fmicb.2020.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Mariod AA, Abdelwahab SI, Gedi MA, Solati Z. Supercritical carbon dioxide extraction of sorghum bug (Agonoscelis pubescens) oil using response surface methodology. Journal of the American Oil Chemists' Society. 2010;87:849–856. doi: 10.1007/s11746-010-1565-2. [DOI] [Google Scholar]
  72. Melgar-Lalanne G, Hernández-Álvarez AJ, Salinas-Castro A. Edible insects processing: Traditional and innovative technologies. Comprehensive Reviews in Food Science and Food Safety. 2019;18:1166–1191. doi: 10.1111/1541-4337.12463. [DOI] [PubMed] [Google Scholar]
  73. Messina CM, Gaglio R, Morghese M, Tolone M, Arena R, Moschetti G, Santulli A, Francesca N, Settanni L. Microbiological profile and bioactive properties of insect powders used in food and feed formulations. Foods. 2019;8:400. doi: 10.3390/foods8090400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Meyer-Rochow VB, Kejonen A. Could Western attitudes towards edible insects possibly be influenced by idioms containing unfavourable references to insects, spiders and other invertebrates? Foods. 2020;9:172. doi: 10.3390/foods9020172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Mintah BK, He R, Dabbour M, Xiang J, Agyekum AA, Ma H. Techno-functional attribute and antioxidative capacity of edible insect protein preparations and hydrolysates thereof: Effect of multiple mode sonochemical action. Ultrasonics Sonochemistry. 2019;58:104676. doi: 10.1016/j.ultsonch.2019.104676. [DOI] [PubMed] [Google Scholar]
  76. Mintah BK, He R, Agyekum AA, Dabbour M, Golly MK, Ma H. Edible insect protein for food applications: Extraction, composition, and functional properties. Journal of Food Process Engineering. 2020;43:e13362. doi: 10.1111/jfpe.13362. [DOI] [Google Scholar]
  77. Mishyna M, Martinez JI, Chen J, Benjamin O. Extraction, characterization and functional properties of soluble proteins from edible grasshopper (Schistocerca gregaria) and honey bee (Apis mellifera) Food Research International. 2019;116:697–706. doi: 10.1016/j.foodres.2018.08.098. [DOI] [PubMed] [Google Scholar]
  78. Mlček J, Adamek M, Adámková A, Borkovcová M, Bednářová M, Skácel J. Detection of selected heavy metals and micronutrients in edible insect and their dependency on the feed using XRF spectrometry. Potravinarstvo Slovak Journal of Food Sciences. 2017;11:725–730. [Google Scholar]
  79. Mohan K, Ganesan AR, Muralisankar T, Jayakumar R, Sathishkumar P, Uthayakumar V, Chandirasekar R, Revathi N. Recent insights into the extraction, characterization, and bioactivities of chitin and chitosan from insects. Trends in Food Science and Technology. 2020;105:17–42. doi: 10.1016/j.tifs.2020.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Murefu TR, Macheka L, Musundire R, Manditsera FA. Safety of wild harvested and reared edible insects: A review. Food Control. 2019;101:209–224. doi: 10.1016/j.foodcont.2019.03.003. [DOI] [Google Scholar]
  81. Nakagaki BJ, Defoliart GR. Comparison of diets for mass-rearing Acheta domesticus (Orthoptera: Gryllidae) as a novelty food, and comparison of food conversion efficiency with values reported for livestock. Journal of Economic Entomology. 1991;84:891–896. doi: 10.1093/jee/84.3.891. [DOI] [Google Scholar]
  82. Nongonierma AB, FitzGerald RJ. Unlocking the biological potential of proteins from edible insects through enzymatic hydrolysis: A review. Innovative Food Science and Emerging Technologies. 2017;43:239–252. doi: 10.1016/j.ifset.2017.08.014. [DOI] [Google Scholar]
  83. Nyangena DN, Mutungi C, Imathiu S, Kinyuru J, Affognon H, Ekesi S, Nakimbugwe D, Fiaboe KK. Effects of traditional processing techniques on the nutritional and microbiological quality of four edible insect species used for food and feed in East Africa. Foods. 2020;9:574. doi: 10.3390/foods9050574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Oh YS, Shih L, Tzeng YM, Wang SL. Protease produced by Pseudomonas aeruginosa K-187 and its application in the deproteinization of shrimp and crab shell wastes. Enzyme and Microbial Technology. 2000;27:3–10. doi: 10.1016/S0141-0229(99)00172-6. [DOI] [PubMed] [Google Scholar]
  85. Oibiokpa FI. Nutrient and antinutrient compositions of some edible insect species in Northern Nigeria. Fountain Journal of Natural and Applied Sciences. 2017;6:9–24. doi: 10.53704/fujnas.v6i1.159. [DOI] [Google Scholar]
  86. Okagu OD, Verma O, McClements DJ, Udenigwe CC. Utilization of insect proteins to formulate nutraceutical delivery systems: Encapsulation and release of curcumin using mealworm protein-chitosan nano-complexes. International Journal of Biological Macromolecules. 2020;151:333–343. doi: 10.1016/j.ijbiomac.2020.02.198. [DOI] [PubMed] [Google Scholar]
  87. Ordoñez-Araque R, Egas-Montenegro E. Edible insects: A food alternative for the sustainable development of the planet. International Journal of Gastronomy and Food Science. 2021;23:100304. doi: 10.1016/j.ijgfs.2021.100304. [DOI] [Google Scholar]
  88. Orkusz A, Wolanska W, Harasym J, Piwowar A, Kapelko M. Consumers' attitudes facing rntomophagy: Polish case perspectives. International Journal of Environmental Research and Public Health. 2020;17:2427. doi: 10.3390/ijerph17072427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Osimani A, Milanović V, Cardinali F, Roncolini A, Garofalo C, Clementi F, Pasquini M, Mozzon M, Foligni R, Raffaelli N. Bread enriched with cricket powder (Acheta domesticus): A technological, microbiological and nutritional evaluation. Innovative Food Science and Emerging Technologies. 2018;48:150–163. doi: 10.1016/j.ifset.2018.06.007. [DOI] [Google Scholar]
  90. Paiva A, Craveiro R, Aroso I, Martins M, Reis RL, Duarte ARC. Natural deep eutectic solvents–solvents for the 21st century. ACS Sustainable Chemistry and Engineering. 2014;2:1063–1071. doi: 10.1021/sc500096j. [DOI] [Google Scholar]
  91. Park YW. The impact of plant-based non-dairy alternative milk on the dairy industry. Food Science of Animal Resources. 2021;41:8–15. doi: 10.5851/kosfa.2020.e82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Park YS, Choi YS, Hwang KE, Kim TK, Lee CW, Shin DM, Han SG. Physicochemical properties of meat batter added with edible silkworm pupae (Bombyx mori) and transglutaminase. Korean Journal for Food Science of Animal Resources. 2017;37:351–359. doi: 10.5851/kosfa.2017.37.3.351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Pattarayingsakul W, Nilavongse A, Reamtong O, Chittavanich P, Mungsantisuk I, Mathong Y, Prasitwuttisak W, Panbangred W. Angiotensin-converting enzyme inhibitory and antioxidant peptides from digestion of larvae and pupae of Asian weaver ant, Oecophylla smaragdina, Fabricius. Journal of the Science of Food and Agriculture. 2017;97:3133–3140. doi: 10.1002/jsfa.8155. [DOI] [PubMed] [Google Scholar]
  94. Paul A, Frederich M, Megido RC, Alabi T, Malik P, Uyttenbroeck R, Francis F, Blecker C, Haubruge E, Lognay G, Danthine S. Insect fatty acids: A comparison of lipids from three orthopterans and Tenebrio molitor L. larvae. Journal of Asia-Pacific Entomology. 2017;20:337–340. doi: 10.1016/j.aspen.2017.02.001. [DOI] [Google Scholar]
  95. Poma G, Cuykx M, Amato E, Calaprice C, Focant JF, Covaci A. Evaluation of hazardous chemicals in edible insects and insect-based food intended for human consumption. Food and Chemical Toxicology. 2017;100:70–79. doi: 10.1016/j.fct.2016.12.006. [DOI] [PubMed] [Google Scholar]
  96. Poms RE, Klein CL, Anklam E. Methods for allergen analysis in food: A review. Food Additives and Contaminants. 2004;21:1–31. doi: 10.1080/02652030310001620423. [DOI] [PubMed] [Google Scholar]
  97. Purschke B, Stegmann T, Schreiner M, Jäger H. Pilot-scale supercritical CO2 extraction of edible insect oil from Tenebrio molitor L. larvae–Influence of extraction conditions on kinetics, defatting performance and compositional properties. European Journal of Lipid Science and Technology. 2017;119:1600134. doi: 10.1002/ejlt.201600134. [DOI] [Google Scholar]
  98. Purschke B, Brüggen H, Scheibelberger R, Jäger H. Effect of pre-treatment and drying method on physico-chemical properties and dry fractionation behaviour of mealworm larvae (Tenebrio molitor L.) European Food Research and Technology. 2018;244:269–280. doi: 10.1007/s00217-017-2953-8. [DOI] [Google Scholar]
  99. Purschke B, Tanzmeister H, Meinlschmidt P, Baumgartner S, Lauter K, Jäger H. Recovery of soluble proteins from migratory locust (Locusta migratoria) and characterisation of their compositional and techno-functional properties. Food Research International. 2018;106:271–279. doi: 10.1016/j.foodres.2017.12.067. [DOI] [PubMed] [Google Scholar]
  100. Rahnamaeian M, Cytryńska M, Zdybicka-Barabas A, Dobslaff K, Wiesner J, Twyman RM, Zuchner T, Sadd BM, Regoes RR, Schmid-Hempel P. Insect antimicrobial peptides show potentiating functional interactions against Gram-negative bacteria. Proceedings of the Royal Society B: Biological Sciences. 2015;282:20150293. doi: 10.1098/rspb.2015.0293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Rao M, Munoz J, Stevens W. Critical factors in chitin production by fermentation of shrimp biowaste. Applied Microbiology and Biotechnology. 2000;54:808–813. doi: 10.1007/s002530000449. [DOI] [PubMed] [Google Scholar]
  102. Roy BC, Sasaki M, Goto M. Effect of temperature and pressure on the extraction yield of oil from sunflower seed with supercritical carbon dioxide. Journal of Applied Sciences. 2006;6:71–75. doi: 10.3923/jas.2006.71.75. [DOI] [Google Scholar]
  103. Rumpold BA, Schlüter OK. Nutritional composition and safety aspects of edible insects. Molecular Nutrition and Food Research. 2013;57:802–823. doi: 10.1002/mnfr.201200735. [DOI] [PubMed] [Google Scholar]
  104. Rumpold BA, Schlüter OK. Potential and challenges of insects as an innovative source for food and feed production. Innovative Food Science and Emerging Technologies. 2013;17:1–11. doi: 10.1016/j.ifset.2012.11.005. [DOI] [Google Scholar]
  105. Scholliers J, Steen L, Fraeye I. Partial replacement of meat by superworm (Zophobas morio larvae) in cooked sausages: Effect of heating temperature and insect: Meat ratio on structure and physical stability. Innovative Food Science and Emerging Technologies. 2020;66:102535. doi: 10.1016/j.ifset.2020.102535. [DOI] [Google Scholar]
  106. Seo M, Goo TW, Chung MY, Baek M, Hwang JS, Kim M, Yun EY. Tenebrio molitor larvae inhibit adipogenesis through AMPK and MAPKs signaling in 3T3-L1 adipocytes and obesity in high-fat diet-induced obese mice. International Journal of Molecular Sciences. 2017;18:518. doi: 10.3390/ijms18030518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Seshadri K, Kirubha M. Gliptins: A new class of oral antidiabetic agents. Indian Journal of Pharmaceutical Sciences. 2009;71:608. doi: 10.4103/0250-474X.59541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Seufi AM, Hafez EE, Galal FH. Identification, phylogenetic analysis and expression profile of an anionic insect defensin gene, with antibacterial activity, from bacterial-challenged cotton leafworm, Spodoptera Littoralis. BMC Molecular Biology. 2011;12:1–14. doi: 10.1186/1471-2199-12-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Shantibala T, Lokeshwari RK, Debaraj H. Nutritional and antinutritional composition of the five species of aquatic edible insects consumed in Manipur, India. Journal of Insect Science. 2014;14:14. doi: 10.1093/jis/14.1.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Shrivastava SK, Srivastava P, Bandresh R, Tripathi PN, Tripathi A. Design, synthesis, and biological evaluation of some novel indolizine derivatives as dual cyclooxygenase and lipoxygenase inhibitor for anti-inflammatory activity. Bioorganic and Medicinal Chemistry. 2017;25:4424–4432. doi: 10.1016/j.bmc.2017.06.027. [DOI] [PubMed] [Google Scholar]
  111. Sila A, Bougatef A. Antioxidant peptides from marine by-products: Isolation, identification and application in food systems. A review. Journal of Functional Foods. 2016;21:10–26. doi: 10.1016/j.jff.2015.11.007. [DOI] [Google Scholar]
  112. Smit EN, Muskiet FA, Boersma ER. The possible role of essential fatty acids in the pathophysiology of malnutrition: A review. Prostaglandins, Leukotrienes and Essential Fatty Acids. 2004;71:241–250. doi: 10.1016/j.plefa.2004.03.019. [DOI] [PubMed] [Google Scholar]
  113. Song C, Yu H, Zhang M, Yang Y, Zhang G. Physicochemical properties and antioxidant activity of chitosan from the blowfly Chrysomya megacephala larvae. International Journal of Biological Macromolecules. 2013;60:347–354. doi: 10.1016/j.ijbiomac.2013.05.039. [DOI] [PubMed] [Google Scholar]
  114. Sosa DAT, Fogliano V. Potential of insect-derived ingredients for food applications. Chap. 9. In: Shields DC, editor. Insect Physiology and Ecology. London: IntechOpen; 2017. pp. 215–231. [Google Scholar]
  115. Stull VJ, Finer E, Bergmans RS, Febvre HP, Longhurst C, Manter DK, Patz JA, Weir TL. Impact of edible cricket consumption on gut microbiota in healthy adults, a double-blind, randomized crossover trial. Scientific Reports. 2018;8:10762. doi: 10.1038/s41598-018-29032-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Sun M, Xu X, Zhang Q, Rui X, Wu J, Dong M. Ultrasonic-assisted aqueous extraction and physicochemical characterization of oil from Clanis bilineata. Journal of Oleo Science. 2018;67:151–165. doi: 10.5650/jos.ess17108. [DOI] [PubMed] [Google Scholar]
  117. Tang JJ, Fang P, Xia HL, Tu ZC, Hou BY, Yan YM, Di L, Zhang L, Cheng YX. Constituents from the edible Chinese black ants (Polyrhachis dives) showing protective effect on rat mesangial cells and anti-inflammatory activity. Food Research International. 2015;67:163–168. doi: 10.1016/j.foodres.2014.11.022. [DOI] [Google Scholar]
  118. Tranter H. Insects creeping into english diets: Introducing entomophagy to school children in a provincial town. MS thesis, University of East Anglia, Norwich, UK (2013)
  119. Tuccillo F, Marino MG, Torri L. Italian consumers’ attitudes towards entomophagy: Influence of human factors and properties of insects and insect-based food. Food Research International. 2020;137:109619. doi: 10.1016/j.foodres.2020.109619. [DOI] [PubMed] [Google Scholar]
  120. Tzompa-Sosa DA, Yi L, van Valenberg HJ, van Boekel MA, Lakemond CM. Insect lipid profile: Aqueous versus organic solvent-based extraction methods. Food Research International. 2014;62:1087–1094. doi: 10.1016/j.foodres.2014.05.052. [DOI] [Google Scholar]
  121. Udenigwe CC, Aluko RE. Food protein-derived bioactive peptides: Production, processing, and potential health benefits. Journal of Food Science. 2012;77:R11–24. doi: 10.1111/j.1750-3841.2011.02455.x. [DOI] [PubMed] [Google Scholar]
  122. Valdez-Peña AU, Espinoza-Perez JD, Sandoval-Fabian GC, Balagurusamy N, Hernandez-Rivera A, De-la-Garza-Rodriguez IM, Contreras-Esquivel JC. Screening of industrial enzymes for deproteinization of shrimp head for chitin recovery. Food Science and Biotechnology. 2010;19:553–557. doi: 10.1007/s10068-010-0077-z. [DOI] [Google Scholar]
  123. van Huis A. Edible insects are the future? Proceedings of the Nutrition Society. 2016;75:294–305. doi: 10.1017/S0029665116000069. [DOI] [PubMed] [Google Scholar]
  124. van Huis A, van Itterbeeck J, Klunder H, Mertens E, Halloran A, Muir G, Vantomme P. Edible insects: Future prospects for food and feed security. Rome: Food and Agriculture Organization of the United Nations; 2013. [Google Scholar]
  125. Vercruysse L, van Camp J, Smagghe G. ACE inhibitory peptides derived from enzymatic hydrolysates of animal muscle protein: A review. Journal of Agricultural and Food Chemistry. 2005;53:8106–8115. doi: 10.1021/jf0508908. [DOI] [PubMed] [Google Scholar]
  126. Vercruysse L, Smagghe G, Matsui T, Van Camp J. Purification and identification of an angiotensin I converting enzyme (ACE) inhibitory peptide from the gastrointestinal hydrolysate of the cotton leafworm, Spodoptera Littoralis. Process Biochemistry. 2008;43:900–904. doi: 10.1016/j.procbio.2008.04.014. [DOI] [Google Scholar]
  127. Vercruysse L, Smagghe G, Beckers T, Van Camp J. Antioxidative and ACE inhibitory activities in enzymatic hydrolysates of the cotton leafworm, Spodoptera Littoralis. Food Chemistry. 2009;114:38–43. doi: 10.1016/j.foodchem.2008.09.011. [DOI] [Google Scholar]
  128. Vercruysse L, Smagghe G, van der Bent A, van Amerongen A, Ongenaert M, van Camp J. Critical evaluation of the use of bioinformatics as a theoretical tool to find high-potential sources of ACE inhibitory peptides. Peptides. 2009;30:575–582. doi: 10.1016/j.peptides.2008.06.027. [DOI] [PubMed] [Google Scholar]
  129. Vizioli J, Bulet P, Charlet M, Lowenberger C, Blass C, Müller HM, Dimopoulos G, Hoffmann J, Kafatos F, Richman A. Cloning and analysis of a cecropin gene from the malaria vector mosquito, Anopheles Gambiae. Insect Molecular Biology. 2000;9:75–84. doi: 10.1046/j.1365-2583.2000.00164.x. [DOI] [PubMed] [Google Scholar]
  130. Wang W, Wang N, Zhou Y, Zhang Y, Xu L, Xu J, Feng F, He G. Isolation of a novel peptide from silkworm pupae protein components and interaction characteristics to angiotensin I-converting enzyme. European Food Research and Technology. 2011;232:29–38. doi: 10.1007/s00217-010-1358-8. [DOI] [Google Scholar]
  131. Wu QY, Jia JQ, Tan GX, Xu JL, Gui ZZ. Physicochemical properties of silkworm larvae protein isolate and gastrointestinal hydrolysate bioactivities. African Journal of Biotechnology. 2011;10:6145–6153. [Google Scholar]
  132. Wu Q, Jia J, Yan H, Du J, Gui Z. A novel angiotensin-I converting enzyme (ACE) inhibitory peptide from gastrointestinal protease hydrolysate of silkworm pupa (Bombyx mori) protein: biochemical characterization and molecular docking study. Peptides. 2015;68:17–24. doi: 10.1016/j.peptides.2014.07.026. [DOI] [PubMed] [Google Scholar]
  133. Xia Z, Chen J, Wu S. Hypolipidemic activity of the chitooligosaccharides from Clanis bilineata (Lepidoptera), an edible insect. International Journal of Biological Macromolecules. 2013;59:96–98. doi: 10.1016/j.ijbiomac.2013.04.017. [DOI] [PubMed] [Google Scholar]
  134. Xia EQ, Zhu SS, He MJ, Luo F, Fu CZ, Zou TB. Marine peptides as potential agents for the management of type 2 diabetes mellitus-a prospect. Marine Drugs. 2017;15:88. doi: 10.3390/md15040088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Xiaoming C, Ying F, Hong Z. Review of the nutritive value of edible insects. In: Durst PB, Johnson DV, Leslie RN, Shono K, editors. Forest insects as food: Humans bite back. Bangkok: RAP Publication; 2010. pp. 85–92. [Google Scholar]
  136. Yang JK, Shih L, Tzeng YM, Wang SL. Production and purification of protease from a Bacillus subtilis that can deproteinize crustacean wastes. Enzyme and Microbial Technology. 2000;26:406–413. doi: 10.1016/S0141-0229(99)00164-7. [DOI] [PubMed] [Google Scholar]
  137. Yen AL. Edible insects: Traditional knowledge or western phobia? Entomological Research. 2009;39:289–298. doi: 10.1111/j.1748-5967.2009.00239.x. [DOI] [Google Scholar]
  138. Yi L, Lakemond CM, Sagis LM, Eisner-Schadler V, van Huis A, van Boekel MA. Extraction and characterisation of protein fractions from five insect species. Food Chemistry. 2013;141:3341–3348. doi: 10.1016/j.foodchem.2013.05.115. [DOI] [PubMed] [Google Scholar]
  139. Yi LY, van Boekel MAJS, Boeren S, Lakemond CMM. Protein identification and in vitro digestion of fractions from Tenebrio molitor. European Food Research and Technology. 2016;242:1285–1297. doi: 10.1007/s00217-015-2632-6. [DOI] [Google Scholar]
  140. Yoon YI, Chung MY, Hwang JS, Han MS, Goo TW, Yun EC. Allomyrina dichotoma (Arthropoda: Insecta) larvae confer resistance to obesity in mice fed a high-fat diet. Nutrients. 2015;7:1978–1991. doi: 10.3390/nu7031978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Zamora-Ros R, Serafini M, Estruch R, Lamuela-Raventós RM, Martínez-González MA, Salas-Salvadó J, Fiol M, Lapetra J, Arós F, Covas MI, Andres-Lacueva C, PREDIMED Study Investigators Mediterranean diet and non enzymatic antioxidant capacity in the PREDIMED study: Evidence for a mechanism of antioxidant tuning. Nutrition, Metabolism and Cardiovascular Diseases. 2013;23:1167–1174. doi: 10.1016/j.numecd.2012.12.008. [DOI] [PubMed] [Google Scholar]
  142. Zhang Q, Vigier KDO, Royer S, Jerome F. Deep eutectic solvents: Syntheses, properties and applications. Chemical Society Reviews. 2012;41:7108–7146. doi: 10.1039/c2cs35178a. [DOI] [PubMed] [Google Scholar]
  143. Zhang H, Wang P, Zhang AJ, Li X, Zhang JH, Qin QL, Wu YJ. Antioxidant activities of protein hydrolysates obtained from the housefly larvae. Acta Biologica Hungarica. 2016;67:236–246. doi: 10.1556/018.67.2016.3.2. [DOI] [PubMed] [Google Scholar]
  144. Zhao X, Vázquez-Gutiérrez JL, Johansson DP, Landberg R, Langton M. Yellow mealworm protein for food purposes-extraction and functional properties. PLoS ONE. 2016;11:e0147791. doi: 10.1371/journal.pone.0147791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Zhou P, Li J, Yan T, Wang X, Huang J, Kuang Z, Ye M, Pan M. Selectivity of deproteinization and demineralization using natural deep eutectic solvents for production of insect chitin (Hermetia illucens) Carbohydrate Polymers. 2019;225:115255. doi: 10.1016/j.carbpol.2019.115255. [DOI] [PubMed] [Google Scholar]
  146. Zielińska E, Baraniak B, Karaś M. Antioxidant and anti-inflammatory activities of hydrolysates and peptide fractions obtained by enzymatic hydrolysis of selected heat-treated edible insects. Nutrients. 2017;9:970. doi: 10.3390/nu9090970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Zong X, Fu J, Xu B, Wang Y, Jin M. Interplay between gut microbiota and antimicrobial peptides. Animal Nutrition. 2020;6:389–396. doi: 10.1016/j.aninu.2020.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Food Science and Biotechnology are provided here courtesy of Springer

RESOURCES