Next Article in Journal
Physicochemical and Sensory Properties of Pork Patties with Partial Replacement of Lean Pork by Stalks of Agaricus bisporus
Previous Article in Journal
Green Optimization of Sesame Seed Oil Extraction via Pulsed Electric Field and Ultrasound Bath: Yield, Antioxidant Activity, Oxidative Stability, and Functional Food Potential
Previous Article in Special Issue
Sustainable Meat Alternatives: Incorporation of Tenebrio molitor and Alphitobius diaperinus Powders into Pork-Based Hybrid Hams
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Harnessing Edible Insect Bioactives for Gut Health: A Comprehensive Review on Chitin-Derived Prebiotics and Peptidomic Insights from the Black Soldier Fly

1
Central Laboratories—Poultry Health Laboratory Department, Al-Watania Poultry Company, Buraydah 11475, Saudi Arabia
2
Department of Animal and Poultry Production, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia
3
Department of Agricultural and Biosystems Engineering, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia
4
Department of Food Science and Human Nutrition, College of Agriculture and Food, Qassim University, Buraydah 51452, Saudi Arabia
5
Ministry of Health, Qassim Health Cluster, King Fahd Specialist Hospital, Buraydah 52366, Saudi Arabia
6
Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
*
Author to whom correspondence should be addressed.
Foods 2025, 14(21), 3654; https://doi.org/10.3390/foods14213654 (registering DOI)
Submission received: 28 September 2025 / Revised: 24 October 2025 / Accepted: 24 October 2025 / Published: 27 October 2025

Abstract

The growing need for sustainable protein and functional food ingredients has made edible insects stand out as a flexible source of bioactives. Black Soldier Fly larva (BSFL) bioactives, such as chitooligosaccharides (COSs) and peptides, present potential benefits for gut health; nevertheless, their molecular pathways, clinical validation, and commercial scalability have yet to be thoroughly investigated. This study systematically analyzes current progress in BSFL bioactive extraction and characterization, emphasizing enzymatic and thermal processing, controlled enzyme development, and integrated supercritical fluid enzymatic pipelines. We assess preclinical and animal research that illustrates prebiotic modulation of Bifidobacterium, Lactobacillus, and Faecalibacterium populations; antimicrobial peptide-mediated immune signaling; and antioxidant activity. Multi-omics frameworks that connect the microbial metabolism of COS to gut health help us understand how these processes function. A comparison of the regulatory environments for food and feed applications in the EU, North America, and Asia shows that there are gaps in human safety trials, harmonized standards, and techno-economic assessments. Finally, we suggest some next steps: randomized controlled human trials in groups with irritable bowel syndrome (IBS) and metabolic syndrome; standardized data integration pipelines for multi-omics; and life cycle and cost–benefit analyses of modular, vertically integrated BSFL biorefineries with AI-driven reactors, digital twins, and blockchain traceability. Addressing these issues will hasten the conversion of BSFL bioactives into safe, effective, and sustainable functional meals and nutraceuticals.

1. Introduction

The global food system is encountering problems it has never experienced before because of population expansion, environmental damage, and limited resources. It means that finding protein sources that do not involve conventionally raising animals is more important than ever. Plant-based proteins, insects, microbes, and cultured meat are all examples of alternative protein sources that could be better for the environment than traditional animal agriculture by more than 80% [1,2]. These choices require a lot less land, water, and feed, and they also release less greenhouse gas [3,4]. Microorganisms are very promising because they do not need a lot of space or changes in the seasons, and they do not use up a lot of carbon with no gas emissions [5]. In fact, insect proteins are an excellent source of food since they have all the amino acids and provide the nutrients your body needs [3]. Nonetheless, significant challenges remain, including manufacturing scalability, regulatory frameworks, customer acceptance, and achieving equivalent nutritional and sensory profiles to conventional products [6,7]. The market for edible insects around the world has expanded yearly by 23% over the previous five years. By 2024, BSFL would make up more than 40% of all insect-meal output [8]. This rise is because BSFL are great at processing feed into protein and bioactive compounds. People have been paying a lot of attention to edible insects as a viable substitute since they better transform food into protein, produce less gas, and need less land than other sources of protein [9]. The black soldier fly (BSF), Hermetia illucens, is notable for its ability to bioconvert organic waste into protein- and lipid-rich biomass, providing the twin advantages of waste valorization and nutritional synthesis [10]. Despite these economic enhancements, substantial gaps remain in our comprehension of the mechanisms of BSFL bioactives, their effects on people, and the simultaneous monetization of these elements.
The gut microbiome, a complex bacterial ecosystem, is essential for health by regulating digestion, immunity, and metabolic balance [11]. The composition of this microbial community is influenced by factors such as nutrition, age, lifestyle, and medications. An imbalance in this vital system can result in dysbiosis, which is linked to many health issues [12,13]. Probiotics directly regulate microbiota and immune responses, whereas postbiotics provide bioactive metabolites that maintain microbial homeostasis [14]. Prebiotics selectively stimulate beneficial bacteria and produce short-chain fatty acids (SCFAs) that enhance gut health [15]. Bioactive peptides also show potential as gut microbiota modulators, though their mechanisms remain incompletely understood [16]. These findings extend beyond human health, since similar microbiome-modulating approaches exhibit promise in veterinary contexts [17].
Chitin and bioactive peptides, which come from insects, are interesting compounds that can alter the diversity of gut microbiota and make people healthier. Chitin is what makes up the hard outer shells of insects. It is a component in the production of chitosan and chitooligosaccharides (COSs). These compounds work as prebiotics by selectively promoting beneficial bacteria, including Bifidobacterium and Lactobacillus, and improving the gut barrier function [18,19]. These chitin derivatives can lower inflammation, kill pathogens, and affect how the immune system works [20,21]. BSFL are particularly advantageous as they generate bioactive peptides released during protein degradation. These peptides can kill germs, protect cells from damage, and change the way the immune system works [21,22]. Chitin from insects has a lot of potential, but it is not used as much as chitin from crustaceans, a way to make bioactive compounds that do not affect the environment [23].
BSF is an excellent model species for sustainable insect farming since it has a lot of desirable traits and can live in a lot of different places. The species reproduces quickly and can turn various types of organic waste into high-quality biomass [24,25]. It has a well-known nutritional profile that includes a lot of protein and a good mix of amino acids, which makes it suitable for both food and feed [26,27]. Recent advances have made it possible to fully explain how to obtain chitin from H. illucens biomass. However, extraction is still more complicated than from crustacean sources because protein and chitin interact strongly [28,29]. The species has a complex gut flora that is required for bioconversion to work and could be valuable in biotechnology [30]. These improvements help make insect-based products that are the same for everyone and keep animals safe when they are made in huge quantities [8].
This review aims to (1) contextualize the role of edible insects in addressing global nutrition and sustainability challenges; (2) highlight the importance of gut microbiome modulation for health; (3) examine the biochemical properties and gut-targeted functionalities of insect-derived chitin prebiotics and bioactive peptides; (4) evaluate the BSF as a versatile platform for producing these bioactives; and (5) discuss safety, allergenicity, and regulatory aspects. This study synthesizes current knowledge on chitin valorization and peptidomic profiling in H. illucens to identify potential strategies for utilizing edible insect bioactives to improve gut health in both human and animal populations. Essential aspects encompass optimized processing pipelines aimed at yield and purity, mechanistic pathways, comparative regulatory frameworks for food and feed, along with recommendations for clinical trials and life cycle techno-economic assessments.

2. Methodology

A systematic literature review was performed to locate and synthesize data on insect-derived chitin prebiotics and peptidomic bioactives, with a focus on H. illucens. We used keyword combinations like “edible insects”, “chitin”, “chitosan”, “chitooligosaccharides”, “prebiotic”, “bioactive peptides”, “peptidomics”, “gut microbiota”, and “H. illucens” to search five databases (Web of Science, Scopus, PubMed, Google Scholar, and AGRICOLA) for publications from 2015 to 2025. We also did targeted searches and reference screening. Inclusion criteria mandated investigations on insect-derived chitin or peptides exhibiting demonstrated or potential gut-modulating effects via in vitro, in vivo, or in silico methodologies. Exclusions were imposed on non-English papers devoid of methodological specifics, research without functional bioactivity assessments, or entire texts that were not readily accessible. We used an integrative framework to look at eligible articles that looked at (1) methods for extracting and characterizing peptides; (2) functional assays for prebiotic efficacy and peptide bioactivities; and (3) biological context, such as gut microbiota modulation and in silico peptide receptor predictions. Standardized forms were used to collect information about the study’s characteristics, extraction processes, analytical techniques, and bioactivity outcomes. Quantitative data were organized into tables to compare extraction efficiency and bioactivity parameters among species and processing methods. Then, thematic synthesis was performed. H. illucens investigations underwent a comprehensive examination of chitin isolation procedures, proteomic processes, and bioactivity correlations to evaluate methodological consistency and pinpoint study deficiencies.

3. Edible Insects as Novel Functional Foods

3.1. Historical and Cultural Background of Entomophagy

Entomophagy has primary historical roots in Africa, Asia, Latin America, and Oceania, where insects have been devoured for millennia as both dietary staples and culturally significant foods [31,32]. There are more than 2000 edible insect species in the world, spread across 25 different orders. The most commonly eaten groups are Coleoptera (beetles, 31–32%), Lepidoptera (caterpillars and moths, 15–18%), Hymenoptera (ants, bees, and wasps, 14–16%), Orthoptera (grasshoppers, locusts, and crickets, 14%), and Hemiptera (true bugs, 11%). Together, these groups make up about 88% of all edible insect species eaten around the world [33,34]. There are around 1000 kinds of insects in Africa that can be eaten, like ants, crickets, termites, and caterpillars. These insects supply households with limited incomes with vital proteins, fats, and minerals [35,36]. A lot of scholarly articles about edible insects have come out since 2015. Even though people in Western countries do not like the idea, Europe is leading the way in research [37]. Countries that consume insects care more about how they taste and how easy they are to find, while Western countries care more about how healthy they are and how long they last, which makes it easier for people to eat insects [38]. Research indicates that edible insects are more environmentally sustainable, requiring 40–60% less land and water and generating fewer greenhouse gases compared to conventional animals, while providing comparable nutritional advantages [39,40].

3.2. Nutritional Composition of Edible Insects

Insects that are safe to eat have perfect nutritional profiles, which makes them a good source of protein that can be grown over time. These insects have 48–70% protein on a dry matter basis and amino acid profiles that are just as comprehensive as those of regular animal proteins [41,42]. Edible insects have a lot of lysine (1.03–8.64 g 100 g−1), leucine (up to 10 g 100 g−1), and methionine (up to 4.5 g 100 g−1). Some species, such as Bombyx mori pupae, have more methionine than soybeans and come close to the levels of egg protein. Protein digestibility tests show that edible insects have protein digestibility-corrected amino acid scores (PDCAAS) between 0.44 and 0.86. These scores are lower than casein’s (0.97) but still similar to those of plant-based proteins. Lysine or sulfur-containing amino acids are usually the limiting amino acids [41,42]. Their lipid content ranges from 21 to 39%, and they have good fatty acid profiles that are high in monounsaturated and polyunsaturated fats, such as omega-3 fatty acids [19,41]. Oleic acid (MUFA), linoleic acid (omega-6 PUFA), and α-linolenic acid (omega-3 PUFA) are the most common fatty acids in most insect species. For example, Tenebrio molitor and Rhynchophorus phoenicis larvae have high MUFA levels, while Acheta spp. have high linoleic acid levels (32.20–41.30 g 100 g−1). Orthopterans, like locusts and crickets, store more α-linolenic acid (omega-3) than typical plant oils like sunflower, olive, and soybean oils [42,43]. Insects are excellent sources of micronutrients, and their levels of iron, zinc, and vitamin B12 are usually similar to or higher than those in beef and chicken [44]. For example, edible insects have iron levels that range from 4 to 62 mg 100 g−1 of dry matter and zinc levels that range from 9 to 27 mg 100 g−1 of dry matter. Some species, such as Oecophylla smaragdina and Odontotermes, are very high in these elements. Insects contain iron and zinc mostly in non-heme forms that are linked to ferritin, transferrin, and other transport proteins. However, there is not much research on how well these minerals are available to people. Edible insects are also a great source of Bcomplex vitamins. For example, dried cricket powder has ten times more vitamin B12 than beef and a lot of riboflavin (0.11–8.9 mg 100 g−1), thiamin, and fat-soluble vitamins like A, D3, and E [44,45,46]. In addition, insects have special nutrients like chitin-based dietary fiber and bioactive chemicals that fight bacteria and free radicals [47]. The chitin content may provide prebiotic advantages for gut microbiome health [19]. Research indicates that chitin consumption (2–5 g day−1 for 3 weeks) fosters beneficial bacterial genera such as Roseburia and Eubacterium, elevates short-chain fatty acid production, including butyrate, and enhances gut microbiota diversity without detrimental effects on overall health, while their resource-efficient production promotes environmental sustainability [3,48].

3.3. Overview of Major Bioactive Classes Identified in Insects

Bioactive compounds in edible insects provide health benefits beyond macronutrients. Insect exoskeleton chitin, chitosan, and melanin help gut flora and fight infections like Escherichia coli and Staphylococcus aureus [20,47]. Chitin at 2–5 g day−1 promotes beneficial bacterial species like Roseburia and Eubacterium, short-chain fatty acid synthesis, and gut microbiota diversity. Melanin pigments from edible insects like H. illucens have photoprotective, immunomodulatory, and stress-protective properties through free radical scavenging processes akin to ascorbic acid [48,49,50]. Edible insect bioactive peptides are a prominent health-promoting class. Antioxidant, anti-inflammatory, antihypertensive, antidiabetic, and immunomodulatory peptides from 12 insect species have been found using peptididomic analysis. Peptides containing 3–20 amino acid residues and molecular weights below 10 kDa are most bioactive. Peptides from Antheraea assama silkworm pupae show ACE-inhibitory IC50 values as low as 0.017 μg mL−1, surpassing Enalapril at 0.11 μg mL−1. Cricket-derived tetrapeptides FVEG and FYDQ modulate cellular antioxidant activity by modulating SOD, CAT, and GSR enzyme systems. Cricket and mealworm protein hydrolysates inhibit reactive oxygen species and modulate cytokines in vitro, with hydrophobic amino acids like tyrosine, phenylalanine, and proline donating electrons to neutralize reactive species [22,51]. Bioactive peptides at 2–5 g day−1 improve short-chain fatty acid synthesis, foster Roseburia and Eubacterium, and diversify gut flora. Melanin pigments from edible insects like H. illucens have photoprotective, immunomodulatory, and stress-protective benefits equivalent to ascorbic acid due to free radical scavenging [48,49,50]. Peptidomic analysis found 211 immunomodulatory, antioxidant, anti-inflammatory, antihypertensive, and antidiabetic peptides from 12 insect species [52,53]. Cricket and mealworm protein hydrolysates exhibit inhibition of reactive oxygen species and modulation of cytokines in vitro [54]. Phenolic compounds enhance antioxidant capacity and exhibit anti-inflammatory effects [52,53].
Antimicrobial peptides (AMPs) like defensins, cecropins, and lysozymes are also essential bioactives. With 34–51 amino acid residues and six conserved cysteine residues generating three disulfide bridges, insect defensins have broad-spectrum efficacy against Gram-positive pathogens like Staphylococcus aureus. Cecropins kill bacteria and fungi, whereas lysozymes break down bacterial peptidoglycan and alter insect gut immunological signaling [55,56]. Flavonoids and phenols boost antioxidant and anti-inflammatory properties. Plant absorption and endogenous production, such as sclerotization and melanization, provide insects with phenolic chemicals. Edible insects contain protocatechuic acid, 3,4-dihydroxyphenylacetic acid, flavones, flavonols, and anthocyanins, with Tenebrio molitor (mealworm) and Acheta domesticus (cricket) showing high antioxidant activity in DPPH and ABTS radical scavenging assays. Some insect species produce unique flavonoid metabolites not seen in their plant-based diets, which increases antioxidant capability [53,57].
Medium-chain fatty acids, especially lauric acid (C12:0), constitute a distinct class of antimicrobial lipids prevalent in specific edible insects. H. illucens (BSFL) possesses significant levels of lauric acid, comprising 40–50% of total fatty acids. Its monoglyceride derivative, glycerol monolaurate (GML), demonstrates extensive antimicrobial efficacy against multidrug-resistant bacteria, enveloped viruses, fungi, and protozoa via mechanisms that disrupt membranes. Lauric acid disrupts bacterial electron transport chains, inhibits membrane-associated enzymes, and effectively addresses microbial resistance via various mechanisms, including the disruption of quorum sensing [58]. These bioactive classes improve microbial balance, barrier function, and inflammation, making insects beneficial diets for chronic disease management and gut health [59].

4. Black Soldier Fly (H. illucens): Biology, Production, and Safety

4.1. Life Cycle and Farming Practices

BSF develops holometabolously from egg to adult, undergoing biochemical and microbiological alterations (Figure 1). Chitin concentration rises from larval to pupal stages, and recovered chitin is more thermally stable and crystalline than industry standards [60,61]. BSFL is useful for bioactive chemical extraction because larvae accumulate proteins, lipids, and chitin precursors [26]. The bacterial community composition changes during development, with 160 amplicon sequence variations and different metabolic pathways supporting each life phase [62]. Nutrient-rich substrates increase larval growth time, lifespan, and egg production by up to 57% [63]. Adult feeding with carbohydrate supplements like honey significantly enhances female longevity, fecundity, and oviposition success [64]. BSF undergoes complete metamorphosis (Figure 1), with unique developmental phases that affect commercial production and bioactive component extraction. High-quality larval meals like brewer’s grain improve development time, longevity, and egg output [63]. Adults mating many times and females producing multiple eggs boost reproductive output [65]. An adult diet is vital because 5% honey solution boosts female lifespan, fecundity, egg production, and oviposition efficiencies [64]. Gut microbiota vary by substrate and developmental stage, affecting host performance and bioconversion efficiency [30,66]. Chitin and chitosan may be made from breeding failures at different developmental stages [67]. These insects provide sustained proteins, lipids, and bioactive substances [26].
Modern insect farming uses biological features and controlled environments to boost production and sustainability. BSF and mealworm larvae efficiently use brewery leftover grains, fruit and vegetable waste, and manure, lowering feed costs and environmental effects [63,68]. Substrate type, larval density, and moisture content affect larval development, yield, and substrate conversion efficiency, with preferred outcomes employing wasted grains and carefully regulated moisture and density [69,70]. Climate-controlled chambers improve growth and reduce mortality [71]. Late instar harvesting produces homogenous larvae with steady humidity [63]. BSF larval waste makes good organic fertilizer, although its composition and advantages depend on the rearing substrate [72].

4.2. Safety Assessment for Human and Animal Consumption

BSFL exhibits considerable promise as an alternative protein source, supported by thorough safety evaluations that address critical concerns. Microbial safety studies indicate that BSFL may contain pathogenic bacteria such as Bacillus cereus, Clostridium perfringens, and Salmonella. However, thermal processing techniques, including blanching and drying, significantly diminish microbial contamination to acceptable levels [73,74]. Assessments of heavy metals indicate cadmium bioaccumulation in larvae; however, concentrations remain within regulatory limits when cultured on compliant substrates [75,76]. BSFL possesses a protein content ranging from 40% to 73%, featuring a balanced composition of essential amino acids and advantageous fatty acid profiles [27,77]. Animal feeding trials indicate favorable growth performance and safety when BSFL substitutes conventional protein sources at suitable inclusion levels [78]. However, substrate quality control and appropriate processing are essential for ensuring product safety [79].

4.3. Regulatory Landscape for Edible Insect Use

BSFL manufacturing has regional regulatory and safety issues. Novel foods regulation in the EU requires EFSA risk evaluations and rigorous production criteria for BSFL [80,81]. Despite generating 20,000 tons yearly versus a potential of 3,000,000 tons, insect-based food and feed regulations are lacking in many places, notably in Africa [79]. Multiple linked issues prevent Africa from having adequate insect laws. First, institutional and resource restrictions seriously impede regulation development, including risk assessment technical competence, laboratory capacity, budgetary resources, and inter-ministerial cooperation. Second, 91.7% of African nations lack formal food safety rules due to a lack of scientific data on edible insect safety. Third, authorities prioritize food availability above food safety due to competing food security demands. Fourth, Africa’s fragmented regulatory framework, with ministries managing different areas, produces jurisdictional overlaps and enforcement gaps. Finally, Africa’s low involvement in Codex Alimentarius diminishes its effect on insect safety standards worldwide. These structural impediments contrast with the EU’s centralized regulatory power, significant infrastructure investment, risk assessment protocols, and active international engagement [82,83,84]. Studies show substantial microbial contamination in BSFL, including dangerous bacteria such as B. cereus, Salmonella spp., and E. coli [74]. Chemical safety issues include potential accumulation of heavy metals and exposure to mycotoxins, pesticides, and pharmaceuticals from substrates [85]. For safe commercialization, the sector needs food safety standards, processing processes, and thorough regulations [86].

5. Bioactive Compounds in BSFL

5.1. Proteins and Amino Acid Profiles

BSFL is a nutritious alternative protein source. Depending on the raising substrate, BSFL contain 35–50% crude protein by dry matter [87,88]. It contains all necessary amino acids that meet or surpass FAO human nutrition needs, with lysine, leucine, and valine being particularly plentiful [27,87,88]. According to protein efficiency ratio experiments, BSFL protein is equivalent to soybean and fish meal [89]. Protein concentrates can contain up to 73% protein [27,77]. Processing affects nutritional composition. BSFL can replace up to 20% of conventional protein sources in chicken diets without affecting performance [76,78] as a protein supplement for beef cattle [90]. Table 1 shows the essential amino acid content in grams per 100 g compared to beef and soy. BSFL protein amino acid analysis was performed using acid hydrolysis and HPLC, utilizing beef and soy protein values from published literature [91,92,93].

5.2. Lipids and Fatty Acid Composition

BSFL has high to moderate lipid content, according to research. Lauric acid (C12:0) dominates the lipid profile, accounting for 36–76% of total fatty acids, depending on rearing substrate [94,95]. Other critical fatty acids include myristic, palmitic, oleic, and linoleic [96,97]. Diet affects fatty acid composition, although BSFL manufacture lauric and myristic acid even without substrate [96,98]. Lauric acid is antibacterial and anti-inflammatory, and BSFL oil may be medicinal [94,99]. The lipid profile’s unsaturated-to-saturated fatty acid ratio depends on substrate composition [100]. These lipids are promising for pharmacological, cosmetic, and feed uses [95,101]. Table 2 summarizes the quantity and physiological functions of BSFL lipids’ main fatty acids.

5.3. Chitin and Chitosan: Chemical Structure and Biological Roles

Chitin, a β-(1 → 4)-linked N-acetylglucosamine polymer, is found in insect exoskeletons and may be deacetylated to chitosan, enhancing solubility and bioactivity [102]. BSFL is a potential chitin source, with content varying from 8 to 24% across life stages, peaking in pupal stages, with sheddings affected by biological variables including developmental stage and extraction methods [60,103]. Standard extraction protocols incorporate demineralization, deproteinization, degreasing, and decolorization in order, depending on reagent type, concentration, temperature, and time. Chemical extraction employs acids and bases, whereas biological techniques use enzymatic or microbial fermentation, which affects chitin production and purity differently. Different sample sources (larvae, pupae, shed skins) and post-extraction processing affect chitin content [104,105,106].
BSFL chitin has an α-form structure, good crystallinity (52.8–78.0%), and thermal stability [60,107]. Chitosan and COS are prebiotics that boost gut flora [108]. Cricket chitosan at varied doses boosted probiotic bacteria and inhibited Salmonella typhi [109]. BSFL-derived chitosan also has antibacterial action against different diseases, depending on bacterial species and concentration [110]. These qualities make insect-derived chitin and chitosan useful for dietary, biomedical, and agricultural uses [23]. Table 3 compares the physicochemical features of chitin and chitosan and their gut health effects on solubility, prebiotic effectiveness, and microbiota modification. Chitin generally has ~90% acetylation, while chitosan has ~50%. Chitin is water-insoluble, whereas chitosan is acid-soluble. Chitin’s strong acetylation hinders solubility and fermentation, whereas chitosan’s partial deacetylation improves solubility and selective bacteria fermentability, leading to different gut health functions (Table 3).

5.4. Other Bioactives: Peptides, Antimicrobial Agents, Phenolics

BSFL generate a variety of bioactive compounds in addition to their main protein and lipid constituents. Proteolytic digestion of BSFL proteins produces AMPs with molecular weights ranging from 0.5 to 3 kDa, exhibiting notable antibacterial activity against pathogenic bacteria [111,112]. These peptides demonstrate antioxidant properties, with particular sequences exhibiting significant radical scavenging activity [113,114]. BSFL also contains phenolic compounds that enhance antioxidant capacity, with an average total phenolic content of 2.5 mg GAE g−1, which correlates with DPPH radical scavenging activity [115]. Other bioactive compounds comprise sterols like campesterol and β-sitosterol [116], as well as antimicrobial fatty acid derivatives that function synergistically to inhibit pathogenic bacteria and diminish inflammatory markers [117]. These bioactive compounds collectively enhance immune function and gut health in animal applications [118], as illustrated in Figure 2.

6. Chitin-Derived Prebiotics: Mechanisms and Gut Health Benefits

6.1. Chemistry and Digestion of Chitin in the Gastrointestinal Tract

COS are deacetylated derivatives of chitin with a degree of polymerization less than 20 and a molecular weight below 3.9 kDa, exhibiting enhanced water solubility compared to chitin [119]. These compounds demonstrate significant prebiotic effects by modulating gut microbiota composition, particularly increasing Bacteroidetes while decreasing Proteobacteria and reducing the Firmicutes/Bacteroidetes ratio [120,121]. COS promotes beneficial bacteria, including Bacteroides, Faecalibacterium, and Roseburia, while suppressing pathogenic species like Klebsiella [120,122]. The degree of polymerization influences microbial effects, with COS2-3 enhancing butyrate production and COS4-6 increasing metabolic diversity [123]. Beyond gut health, COS exhibit multiple bioactivities, including anti-inflammatory, antimicrobial, antioxidant, and metabolic regulatory effects [108,119]. Chitin undergoes partial deacetylation in the stomach and microbial fermentation in the colon, producing beneficial metabolites [20,124]. In Figure 3, COS strengthens intestinal barrier integrity through tight junction proteins (ZO-1, occludin, claudin), restructures gut microbiota by increasing beneficial bacteria (Akkermansia, Lactobacillus) while reducing Proteobacteria, and enhances SCFAs production (butyrate, propionate). Additionally, COS activate PPARγ/SIRT1 and PI3K/AKT pathways, promote NF-κB p65 deacetylation, inhibit ERK signaling, and reduce inflammatory cytokines, collectively demonstrating their protective effects on gut health and inflammation regulation.

6.2. Role of Chitin and Derivatives as Dietary Fiber and Prebiotics

Chitin, chitosan, and COS are bioactive polysaccharides derived from crustacean exoskeletons, insects, and fungi, demonstrating notable prebiotic and health-enhancing characteristics [20,108]. Chitin serves as an insoluble fiber that enhances fecal bulk and transit time, whereas chitosan functions as a soluble fiber with emulsifying and bile-acid-binding characteristics [108]. COS, which is produced after digestion, specifically stimulates good bacteria like Bifidobacterium and Lactobacillus, which is a classic example of prebiotic action [18,125]. These molecules demonstrate various biological actions, including anti-inflammatory, antibacterial, antioxidant, and immunomodulatory effects [126,127]. Chitin–glucan complexes derived from mushrooms have enhanced prebiotic effects, markedly elevating propionic and butyric acid concentrations [128]. They can also be used in aquaculture as growth promoters and immunostimulants [129]. They might also be used to make functional foods because they break down and are safe for the environment.
Dietary fibers have different prebiotic effects on gut flora, and the effects depend a lot on the kind, shape, and solubility of the fiber. Numerous studies indicate that the majority of dietary fibers (>80%) are highly fermentable and facilitate the synthesis of SCFAs, especially acetate and butyrate [130]. Fructo-oligosaccharides and inulin are well-known prebiotics that have substantial prebiotic effects. Other fibers, on the other hand, are still being studied as possible prebiotics with different levels of effectiveness [131]. Fibers from cereals, particularly whole grains, reliably elevate bifidobacteria levels and SCFAs synthesis [132]. Comparative studies indicate that fibers may be classified into several categories according to their microbiota-regulating effects, with xylo-oligosaccharides and konjac flour yielding the most significant quantities of SCFAs [133]. Nonetheless, the correlation between fiber supplementation and SCFAs generation in healthy individuals is intricate, with effects varying based on fiber dosage, type, and structural attributes [134,135].

6.3. Impact on Gut Microbiota Diversity and Probiotic Growth

Studies indicate that COS and insect-derived chemicals have substantial prebiotic effects on gut flora. COS supplementation alters bacterial composition by elevating beneficial Bacteroidetes and Faecalibacterium, while diminishing harmful Proteobacteria and Klebsiella [120]. In vitro studies demonstrate that COS selectively enhances probiotic development, resulting in up to a 4.1-fold increase in the cell density of Lactobacillus and C. butyricum [136]. Chitosan from crickets increases the number of probiotic bacteria while stopping the growth of S. typhi [109]. The addition of insect chitin significantly boosts alpha diversity measurements, such as the Shannon index, and encourages beneficial symbionts, including Ruminococcaceae, Lachnospiraceae, and Faecalibacterium [122]. Transplanting fecal microbiota from COS-treated animals enhances intestinal barrier function and reduces cellular apoptosis [137]. These results jointly endorse the prebiotic potential of insect-derived COS and chitosan molecules [108,138].

6.4. Effects on SCFAs Production, Gut Barrier Function, and Immune Modulation

SCFAs, especially butyrate, have essential impacts on the immune system and the lining of the gut. Butyrate acts as an anti-inflammatory drug by binding to G-protein coupled receptors GPR41 and GPR43, therefore promoting immunological homeostasis and influencing gene transcription through histone deacetylase (HDAC) inhibition [139,140]. These processes lead to diminished pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, while facilitating anti-inflammatory responses [141]. Butyrate improves the strength of the intestinal barrier by increasing the levels of tight junction proteins such as claudin-1 and occludin. This makes the intestines less permeable [142,143]. The molecule is an essential source of energy for colonocytes and helps keep the gut in balance through many different routes [144,145]. These synergistic effects establish butyrate as a potential therapy choice for inflammatory bowel illnesses and other immune-mediated disorders.

6.5. Comparative Insights from Animal and Human Studies

Studies on animals and people show that chitin-derived chemicals and insect-based components can have a considerable positive effect on health. In chickens, the addition of chitosan oligosaccharide (COS) to feed at 30–1000 mg kg−1 has been found to alter the shape of the intestines by making the villi taller and making the feed conversion more efficient. For example, COS at 30 mg kg−1 feed promoted intestinal health and immunity, whereas doses between 200 and 1000 mg kg−1 feed have consistently improved growth, antioxidant status, and gut development in broilers [146,147]. COS also lowers inflammatory reactions and boosts the antioxidant capacity of broilers and laying hens [147,148]. In experiments on mammals, adding 5 mg kg−1 of COS to their diet made them lose weight, and less fat was built up in their stomachs. Higher dosages, on the other hand, improved metabolic parameters, such as lowering blood glucose and cholesterol levels [39,149]. Components generated from BSFL have immunomodulatory properties via AMPs, lauric acid, and chitin constituents [150]. These substances have prebiotic effects by altering the composition of gut bacteria and improving the function of the intestinal barrier [149]. The research indicates dose-dependent advantages across many animals, underscoring the translational potential of bioactive chemicals produced from insects.

6.6. Mechanistic Pathways Linking BSFL-Derived Bioactives to Gut Health

BSFL-derived COS operate as selective prebiotics, mainly aiming at good bacteria in the gut microbiome, such as Bifidobacterium and Lactobacillus species. Through enzymatic breakdown, these bacteria break down COS and make essential SCFAs, including acetate, propionate, and especially butyrate, through the acetyl-CoA pathway [151]. Butyrate interacts with G-protein-coupled receptors (GPR41, GPR43) on colonocytes, which starts signaling pathways that strengthen the intestinal barrier by increasing the levels of tight junction proteins such as claudin-1 and ZO-1 [152]. At the same time, AMPs from BSFL bind to Toll-like receptors (TLR2, TLR4) on dendritic cells and macrophages, changing NF-κB signaling pathways to lower the production of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β) and raise the levels of anti-inflammatory mediators like IL-10 [153]. This comprehensive molecular framework elucidates the manner in which BSFL bioactives facilitate synchronized interactions between the microbiome and host, hence enhancing gut health and systemic immunological equilibrium. Table 4 gives a short overview of the BSFL bioactives and how they work in microbial, metabolic, receptor, and impact pathways. It shows how BSFL-derived parts are linked to their microbial targets, intermediate intermediates, host receptors that are involved, and impacts that happen later.

7. Peptidomics of BSF Proteins

7.1. Overview of Peptidomics Technology and Its Application to Insect Proteins

Black Soldier Fly larvae have become a potential source of bioactive peptides that may be used for many different health purposes. Researchers have come up with several ways to extract proteins, and SDS-based buffers give the most protein groups, while other approaches improve proteome coverage [156,157]. Peptidomics methodologies employing LC-MS/MS have elucidated several bioactive peptides derived from BSFL proteins after enzymatic hydrolysis. In silico research on gastrointestinal digestion showed that BSFL proteins can produce peptides that are good for your health, have high GI absorption, and are not harmful. These peptides include antioxidant, anti-ACE, and anti-DPP-IV peptides [114]. Experimental validation has shown that peptides like verified peptides have strong antioxidant properties and can protect HepG2 cells from damage [158]. Peptides produced from BSFL demonstrate anticancer activities via modulating the SKP2/p21/cyclin D1 pathways [112] and cytoprotective effects via Nrf2 ac and exhibit cytoprotective effects through Nrf2 activation [115]. These results emphasize BSFL’s potential as a renewable source of multifunctional bioactive peptides [22]. Figure 4 shows the most critical bioactive activities of peptides made from BSFL. BSFL peptides have four primary positive effects: they improve the function of the epithelial barrier, they increase the levels of tight junction proteins to keep the intestines healthy, they change the balance of inflammation, and they stimulate antioxidant enzymes to protect cells from oxidative stress.

7.2. Identification and Characterization of Bioactive Peptides from BSF

Recent studies have shown that bioactive peptides from different protein sources, like BSFL and other edible insects, have a lot of promise. In silico screening coupled with experimental validation has become a proficient methodology for bioactive peptide identification [114,159]. BSFL proteins have potential as sources of health-promoting peptides post-gastrointestinal digestion, with computational analyses indicating antioxidant, ACE-inhibitory, and DPP-IV-inhibitory characteristics [114,116]. Experimental validation has substantiated the anticancer and antioxidant properties of BSFL-derived peptides, with processes involving the modification of SKP2/p21/cyclin D1 pathways [112]. A thorough study revealed 211 potentially bioactive peptides from 12 insect species, with 62 described in vitro and 3 verified in vivo [22]. These findings underscore the increasing significance of insect-derived bioactive peptides for human health applications [158].

7.3. Biological Activities of Peptides Relevant to Gut Health

Bioactive peptides from food show a lot of promise in keeping the gut healthy in a number of ways. These peptides enhance gastrointestinal homeostasis by regulating barrier function, immunological responses, and gut microbiota [160]. AMPs are essential for sustaining tolerance to gut microbiota and safeguarding against enteric infections; alterations in these processes are implicated in the pathophysiology of inflammatory bowel disease [161]. BSF is a promising source of AMPs, with a bioinformatic study identifying 57 putatively active peptides [117]. Certain peptides from diverse origins exhibit anti-inflammatory properties: peptides derived from fermented soybean meal inhibit intestinal inflammation and improve epithelial barrier function [162], whereas peptides sourced from Tricholoma matsutake alleviate colitis by modulating tight junction proteins and inhibiting pro-inflammatory cytokines through NF-κB signaling pathways [163]. These peptides focus on important inflammatory pathways and make the intestinal barrier stronger [164,165]. Figure 5 shows the paths of various peptide activities.

7.4. Potential Pathways Through Which Insect-Derived Peptides Influence Gut Microbiota and Host Health

Peptides generated from insects have considerable promise in regulating gut ecosystems via several methods. These bioactive peptides demonstrate various activities, including antioxidant, antibacterial, anti-inflammatory, and antihypertensive characteristics [167,168]. Direct antimicrobial action transpires via membrane-disrupting effects on harmful bacteria, whereas advantageous symbionts exhibit enhanced resistance, establishing selection barriers that influence microbial populations [169]. AMPs produced by the gut epithelium facilitate the reorganization of microbiota by selectively attacking pathogens and maintaining commensal bacteria [170]. These peptides help keep the intestinal barrier strong and modulate the immune system. Evidence shows that they help with structural repair and control inflammatory responses [171,172]. The insect intestinal immune system orchestrates many defensive mechanisms, including the synthesis of AMPs via Imd pathways and the formation of reactive oxygen species [173]. Even though there are intriguing uses for functional foods and drugs, more study is needed on safety, rules, and how well consumers take them [168]. Figure 6 shows how the coupled routes help keep the microbial balance and the host’s strength.

8. Gut Microbiome Modulation by BSF-Derived Compounds

8.1. Interaction Between BSF Bioactives and Gut Microbial Communities

BSF research has made a lot of progress, and scientists are paying more and more attention to its antibacterial and prebiotic capabilities. AMPs generated from BSF have potent antibacterial properties against a range of pathogens, including Pseudomonas aeruginosa and Staphylococcus aureus, via membrane disruption mechanisms [111,174,175]. The chitin-rich fractions have antibacterial properties, especially against E. coli, and they also have prebiotic properties that help good bacteria like Limosilactobacillus reuteri [176]. The gut microbiota of BSF is very important for bioconversion efficiency and substrate processing. Under the right circumstances, beneficial microbes like Lactobacillus and Enterococcus may live in the stomachs of larvae [177,178]. Chitosan compounds from BSF selectively promote good gut bacteria while suppressing pathogens [18]. Future research goals encompass the characterization of microbial populations, the development of genetic resources, and the investigation of BSF’s potential for waste biotransformation and antimicrobial applications [24], Figure 7.

8.2. Effects on Microbial Diversity, Pathogen Inhibition, and Host Metabolism

Studies show that microbial diversity greatly improves disease resistance and host metabolism in many ways. Diverse microbiomes safeguard against diseases chiefly by nutrient blocking, wherein communities collectively ingest overlapping nutrients essential for pathogens [179]. Probiotic therapies using Lactobacillus species significantly diminish Salmonella colonization while enhancing gut barrier integrity and immune responses [180]. SCFAs, especially butyrate, are essential microbial metabolites that keep the gut barrier working, provide colonocytes with energy, and change immunological responses both in the gut and throughout the body [144]. Metabolites generated from tryptophan, such as indole-3-propionic acid, have supplementary protective benefits, with supplementation mitigating viral loads and inflammation during influenza illness [181]. Bioactive chemicals in foods like fruits, vegetables, and other foods help good bacteria grow while killing harmful bacteria. This leads to more SCFA synthesis and better metabolic health [182]. These microbial metabolites work together to improve gut homeostasis, barrier integrity, and host immunity [183].

8.3. Case Studies in Livestock, Aquaculture, and Experimental Human Models

Studies show that BSFL and its derivatives have a significant effect on gut microbiota and health in many animals. In broiler chickens, the addition of BSF up to 20% had little impact on total cecal microbiota diversity; nevertheless, certain bacterial communities were modified, notably with an enhanced abundance of Roseburia, which may enhance immunity [184]. Replacing soybean oil with BSF oil enhanced the structure of the intestines and changed the populations of microbes in the gut. The best replacement was 50% [150]. BSF-Desmodium meal combinations boosted the number of good lactic acid bacteria in broilers [185]. In Atlantic salmon, the incorporation of BSF meal improved the variety and evenness of the gut microbiota in comparison to traditional proteins [186]. BSF larvae significantly decreased zoonotic pathogens in pig manure via AMPs and gut-associated bacteria [187]. The immunomodulatory capacity transcends diet via AMPs, lauric acid, and chitin components [166]. In laying chickens, live BSF larvae modified particular bacterial species without influencing fear-related behavior [188]. Table 5 highlights the effects of BSF-derived chemicals COS and AMPs on broiler chicks, shrimp aquaculture, and a human pilot study. These results include improved intestinal morphology, a lower pathogen load, and a richer microbiota.

9. Technological Considerations and Processing Influences

9.1. Methods of Harvesting, Drying, Defatting, and Extracting Bioactive Compounds

Processing BSFL requires a number of stages that are all linked together and have a significant impact on the quality of the final product and the extraction of bioactive compounds. Harvesting happens when the plant is at the late instar stage. Then, it is blanched at 80–90 °C to kill enzymes and lower the number of microbes [190]. Hot air drying (60–70 °C) and freeze-drying are two ways to dry things. Freeze-drying is better at keeping heat-sensitive substances, but it might also cause lipolysis [190,191]. Defatting uses solvent extraction, supercritical CO2, or mechanical pressing to get rid of up to 80% of the lipid content [190,192]. Alkaline pH-shifting (pH 10.5–12.5) helps protein extraction by speeding up the process by 135–1080% [193]. Using more than one extraction approach improves proteome coverage [157]. Chitin extraction entails successive deproteinization and demineralization, resulting in 15–25% chitin. However, biological methods utilizing B. subtilis and Acetobacter pasteurianus provide eco-friendly alternatives [28,105]. Combinations of processing methods have a significant effect on the nutritional quality and oxidative stability of the finished product.

9.2. Impact of Processing on Stability and Bioavailability of Chitin and Peptides

Processing factors have a significant effect on how stable and bioavailable chitin and peptides from insects are. Maillard reactions can lower peptide solubility and antioxidant activity by up to 30% when drying temperatures are higher than 70 °C [191]. Freeze-drying retains bioactive peptides better than regular heat drying, keeping more than 90% of their bioactivity. However, it costs more in energy and may cause lipid oxidation [194,195,196]. High-temperature treatments lower protein and lipid levels by 17.85% and 22.55%, respectively, while raising chitin levels by 191.21% [191]. Deacetylation conditions impact the quality of chitin. For example, more prolonged exposure to NaOH enhances the degree of deacetylation but lowers the molecular weight [197]. Peptide stability in food matrices is affected by the way they are processed. For example, the way meat is slaughtered, dried, and extracted can all alter the quality of the final product [198,199].

9.3. Formulation Strategies for Incorporating BSF Bioactives into Functional Foods and Supplements

BSFL shows a lot of promise as a functional food additive when used in different ways. BSFL protein concentrates are very good at emulsifying, and when heated using ohmic heating, they can make oil-in-water emulsions with interfacial tensions as low as 12.95 mN m−1 and droplet sizes as small as 0.68 μm [200]. These protein concentrates have emulsifying activity that is similar to or better than that of whey protein isolate. They can stabilize emulsions with 20–40% oil fractions [201]. Processing techniques have a significant effect on techno-functional qualities. For example, alkaline extraction may make protein concentrates with up to 73.35% protein and improved emulsion stability that can approach 100% [27]. But the way things are processed affects how well they work; for example, blanching makes emulsions less stable and less able to hold together than freezing does [202]. Encapsulation methods provide intriguing strategies for safeguarding BSFL bioactive chemicals, enhancing stability, and facilitating regulated release [203,204]. Even while there are health advantages, there are still problems with safety, consumer acceptability, and following the rules for human ingestion [205,206].

9.4. Economic Viability and Regional Industrialization Strategies

Choosing a processing technique for BSFL bioactive extraction requires finding a balance between energy use, bioactivity preservation, and cost-effectiveness. Hot air drying (60–70 °C) is the best option for areas with limited resources since it costs the least to set up ($50,000–100,000), uses a small amount of energy (1.5–2.5 kWh kg−1), and keeps 60–70% of the peptides [207,208]. On the other hand, freeze-drying keeps bioactivity better (>90% peptides and chitin), but it uses 4 to 10 times more energy (5 to 10 kWh kg−1) and costs a lot of money to set up ($500 K–2 M); thus, it can only be used in developed markets [28]. Enzymatic extraction is a good way to go because it uses 0.5–1.2 kWh kg−1 of energy and keeps 85–95% of the bioactivity at moderate capital expenditures ($200–500 K−1) [209]. Hot air-enzymatic processing together is best for developing areas. It uses 2–3 kWh kg−1 of energy, keeps 75–85% of the bioactivity, and costs $150,000 to $350,000 to set up [105].
To sell BSFL bioactives, you need to make plans that take into consideration the amount of money and energy infrastructure in each area. Regions with a lot of resources may build vertically integrated biorefineries that use freeze-drying and supercritical CO2 extraction to make pharmaceutical-grade compounds for $5000–7000 per ton of dried larval meal. Middle-income areas can use modular facilities that combine hot air drying with selective enzymatic extraction for $2500 to $4000 per tonne. The climatic benefits mean that heating needs are cut by 30% to 40% [210]. Low-resource areas use solar-assisted drying in decentralized facilities that cost $1200 to $2000 per tonne. This is competitive with local proteins and helps with waste management. Labor (40–67%), substrate procurement (20–35%), energy (10–25%), and capital depreciation (10–20%) are the main costs of production at all levels. Co-product valorization makes up for 15–30% of these costs [211]. Additionally, an in-depth evaluation of key BSFL bioactive extraction techniques, emphasizing their cost efficiency, energy requirements, and ability to preserve bioactivity, to inform decisions on large-scale industrial applications, Table 6.

10. Safety, Allergenicity, and Regulatory Aspects

10.1. Microbial, Chemical, and Allergenic Risks in Edible Insect Consumption

Edible insects pose notable microbial, chemical, and allergenic risks that necessitate thorough evaluation. Microbial contamination encompasses pathogenic bacteria, including Bacillus cereus. Processing methods such as blanching can achieve reductions of 3 logs in bacterial counts [212]. Studies have reported no detection of Salmonella or E. coli in retail samples [213]. Chemical hazards include heavy metals such as arsenic, cadmium, lead, and mercury, with detection rates of 100%, 79%, 58%, and 74%, respectively, while concentrations stayed within safe limits [213]. Pesticide residues are present, with glyphosate as the predominant compound [213]. Allergenic risks primarily arise from cross-reactive proteins, specifically tropomyosin and arginine kinase, which may elicit reactions in individuals with allergies to crustaceans and dust mites [212,214,215]. Enzymatic hydrolysis, particularly when combined with thermal treatment, has demonstrated a reduction in IgE-binding capacity by 50–60%. For instance, Alcalase hydrolysis followed by heating at 90 °C for 10 min resulted in a 56.3% decrease in IgE binding [216].

10.2. Risk Mitigation Strategies and Quality Control Methods

To keep edible insects safe, Good Manufacturing Practices and HACCP frameworks must include all types of biological, chemical, and physical threats [217,218]. Critical control points encompass substrate verification to avert contaminant buildup and microbial decrease by heat treatments, with blanching yielding 3–3.4 log reductions in vegetative bacteria [212]. Heavy metals can build up in insects; therefore, they need to be monitored by inductively coupled plasma mass spectrometry. However, the quantities that are found are usually below legal limits [213,219]. ELISA tests can find cross-reactive allergens such as tropomyosin and arginine kinase [212]. Decontamination stages, including blanching, drying, and new technologies like high-pressure processing for microbial control, are also part of processing pathways [220,221]. Some important pathogens include S. aureus, Clostridium spp., and the B. cereus group. Salmonella and E. coli are usually not found in items that have been appropriately treated [213,222].

10.3. Current and Evolving Regulatory Frameworks Globally for Edible Insect Products

Current regulations on edible insects differ significantly from one country to the next. Regulation (EU) 2015/2283 governs novel foods for human use and does not yet include BSFL. Regulation (EU) No 2001/999 (Annex IV), as amended by Regulation (EU) 2017/893 (Annex X) [81,84]. Allows insect proteins from seven species to be used in aquaculture, poultry, and swine feed. The most current EU law on animal by-products (2021/1925) allowed the use of processed animal proteins (PAPs) from silkworms (Bombyx mori) in aquaculture, poultry, and pig feed. This added one more species to the list of seven that were already allowed [223]. The FDA uses GRAS findings to check BSFL substances, which means they need a lot of safety information. Countries that traditionally eat insects, like Thailand and Japan, control them using regular food safety rules instead of giving them a new food label [84]. Microbial contamination, heavy metals, and cross-reactive allergens are some of the most critical safety issues. Processing procedures like blanching can significantly lower microbial loads [212]. The rules are changing quickly, and Codex Alimentarius is working on making global standards that are the same for everyone [84]. But regulatory loopholes are still a big problem, especially in poor nations where there are no uniform laws [224,225]. Consumer acceptance varies considerably based on geographical and cultural factors [226].

11. Remarks and Future Perspectives

Engineered chitinases and deacetylases tailored through directed evolution, membrane reactors with immobilized proteases, and integrated supercritical fluid extraction and enzymatic digestion pipelines can increase yields, purity, and sustainability of BSFL-derived oligosaccharides and peptides, but reaction kinetics, enzyme stability, and downstream integration are understudied. Multi-omics frameworks that combine genome-scale metabolic modeling with metaproteomic, peptidomic, and metabolomic profiling can reveal the biosynthetic pathways and host–microbe interactions that underlie BSFL bioactive functions. These discoveries must be translated into tailored microbiome modulators using standardized data integration workflows and causal validation studies. Preclinical evidence supports COS and peptide supplements’ gut health potential, but randomized, controlled human trials, especially in IBS and metabolic syndrome cohorts, are needed to define dose–response relationships, mechanistic biomarkers, and sensory acceptability. Finally, techno-economic and life cycle assessments of modular, vertically integrated BSFL production systems with automated climate control, frass valorization, and alignment with waste treatment facilities, policy frameworks, and public–private partnerships will de-risk infrastructure investments and ensure environmental and economic viability for functional food and nutraceutical markets.
In the coming decade, combined developments will alter BSFL-derived bioactive manufacturing. AI-guided evolution will produce more stable and specialized chitinases and deacetylases. Reactors using machine learning will optimize reaction kinetics live. Continuous, solvent-free oligosaccharide and peptide recovery is possible with hybrid ceramic–polymer membrane reactors. Modular supercritical fluid extraction with enzymatic pretreatment increases energy efficiency. Precision fermentation and engineered gut microbiomes will boost chitin production and waste-to-bioactive conversion. Multi-omics platforms will predict and control metabolic flux and chemical buildup in real time. Vertically integrated biorefineries provide large-scale, sustainable output. Blockchain traceability and digital twins provide quality and compliance. These developments will enable insect-derived functional ingredient clinical and regulatory approval.

12. Conclusions

Bioactives derived from edible insects, specifically BSFL-derived COS and peptides, exhibit significant prebiotic, antimicrobial, antioxidant, and immunomodulatory properties that contribute to gut health. Advancements in extraction technologies, protein engineering, and integrated bioprocessing offer potential enhancements in product quality and sustainability. Comprehensive clinical trials, standardized regulatory frameworks, and techno-economic evaluations are essential for translating these findings into safe, effective, and commercially viable functional foods and nutraceuticals. The effective utilization of BSFL bioactives relies on the integration of directed enzyme evolution, precision fermentation, real-time multi-omics monitoring, and life cycle-driven production models to produce targeted and sustainable microbiome modulators for both human and animal health.

Author Contributions

Conceptualization, H.B. and T.A.; methodology, T.A., H.B., T.E. and M.G.; investigation, A.A.H.A. and M.F.Y.H.; data curation, T.A. and M.F.Y.H.; visualization, S.A. and M.F.Y.H.; writing—original draft preparation, T.A., A.J. and M.A.; review and editing, T.E., M.G. and H.B. All authors have read and agreed to the published version of the manuscript.

Funding

The researchers would like to thank the Deanship of Graduate Studies and Scientific Research at Qassim University for financial support (QU-APC-2025).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

Thamer Alhasyani was employed by Al-Watania Poultry Company and contributed to the conceptualization, literature review, data curation, and drafting of the manuscript. Al-Watania Poultry Company had no influence on the study design, data analysis, interpretation, or decision to publish. The other authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Mazac, R.; Meinilä, J.; Korkalo, L.; Järviö, N.; Jalava, M.; Tuomisto, H.L. Incorporation of Novel Foods in European Diets Can Reduce Global Warming Potential, Water Use and Land Use by over 80%. Nat. Food. 2022, 3, 286–293. [Google Scholar] [CrossRef]
  2. Nirmal, N.; Anyimadu, C.F.; Khanashyam, A.C.; Bekhit, A.E.d.A.; Dhar, B.K. Alternative Protein Sources: Addressing Global Food Security and Environmental Sustainability. Sustain. Dev. 2025, 33, 3958–3969. [Google Scholar] [CrossRef]
  3. Lisboa, H.M.; Nascimento, A.; Arruda, A.; Sarinho, A.; Lima, J.; Batista, L.; Dantas, M.F.; Andrade, R. Unlocking the Potential of Insect-Based Proteins: Sustainable Solutions for Global Food Security and Nutrition. Foods 2024, 13, 1846. [Google Scholar] [CrossRef]
  4. Gil, M.; Rudy, M.; Duma-Kocan, P.; Stanisławczyk, R.; Krajewska, A.; Dziki, D.; Hassoon, W.H. Sustainability of Alternatives to Animal Protein Sources, a Comprehensive Review. Sustainability 2024, 16, 7701. [Google Scholar] [CrossRef]
  5. Graham, A.E.; Ledesma-Amaro, R. The Microbial Food Revolution. Nat. Commun. 2023, 14, 2231. [Google Scholar] [CrossRef]
  6. Malila, Y.; Owolabi, I.O.; Chotanaphuti, T.; Sakdibhornssup, N.; Elliott, C.T.; Visessanguan, W.; Karoonuthaisiri, N.; Petchkongkaew, A. Current Challenges of Alternative Proteins as Future Foods. npj Sci. Food 2024, 8, 53. [Google Scholar] [CrossRef]
  7. Wood, P.; Tavan, M. A Review of the Alternative Protein Industry. Curr. Opin. Food Sci. 2022, 47, 100869. [Google Scholar] [CrossRef]
  8. Barrett, M.; Chia, S.; Fischer, B.; Tomberlin, J. Welfare Considerations for Farming Black Soldier Flies, Hermetia illucens (Diptera: Stratiomyidae): A Model for the Insects as Food and Feed Industry. J. Insects Food Feed 2023, 9, 119–148. [Google Scholar] [CrossRef]
  9. FAO. Edible Insects: Future Prospects for Food and Feed Security; Food and Agriculture Organization of the United Nations: Rome, Italy, 2013; ISBN 978-92-5-107595-1. [Google Scholar]
  10. Rumpold, B.A.; Schlüter, O.K. Potential and Challenges of Insects as an Innovative Source for Food and Feed Production. Innov. Food Sci. Emerg. Technol. 2013, 17, 1–11. [Google Scholar] [CrossRef]
  11. Kumar, S.; Mukherjee, R.; Gaur, P.; Leal, É.; Lyu, X.; Ahmad, S.; Puri, P.; Chang, C.-M.; Raj, V.S.; Pandey, R.P. Unveiling Roles of Beneficial Gut Bacteria and Optimal Diets for Health. Front. Microbiol. 2025, 16, 1527755. [Google Scholar] [CrossRef] [PubMed]
  12. Safarchi, A.; Al-Qadami, G.; Tran, C.D.; Conlon, M. Understanding Dysbiosis and Resilience in the Human Gut Microbiome: Biomarkers, Interventions, and Challenges. Front. Microbiol. 2025, 16, 1559521. [Google Scholar] [CrossRef] [PubMed]
  13. Ding, G.; Yang, X.; Li, Y.; Wang, Y.; Du, Y.; Wang, M.; Ye, R.; Wang, J.; Zhang, Y.; Chen, Y. Gut Microbiota Regulates Gut Homeostasis, Mucosal Immunity and Influences Immune-Related Diseases. Mol. Cell. Biochem. 2025, 480, 1969–1981. [Google Scholar] [CrossRef] [PubMed]
  14. Liu, Y.; Wang, J.; Wu, C. Modulation of Gut Microbiota and Immune System by Probiotics, Pre-Biotics, and Post-Biotics. Front. Nutr. 2022, 8, 634897. [Google Scholar] [CrossRef]
  15. Yoo, S.; Jung, S.-C.; Kwak, K.; Kim, J.-S. The Role of Prebiotics in Modulating Gut Microbiota: Implications for Human Health. Int. J. Mol. Sci. 2024, 25, 4834. [Google Scholar] [CrossRef]
  16. Wijesekara, T.; Abeyrathne, E.D.N.S.; Ahn, D.U. Effect of Bioactive Peptides on Gut Microbiota and Their Relations to Human Health. Foods 2024, 13, 1853. [Google Scholar] [CrossRef]
  17. Barathan, M.; Ng, S.L.; Lokanathan, Y.; Ng, M.H.; Law, J.X. The Profound Influence of Gut Microbiome and Extracellular Vesicles on Animal Health and Disease. Int. J. Mol. Sci. 2024, 25, 4024. [Google Scholar] [CrossRef]
  18. Edo, G.I.; Mafe, A.N.; Ali, A.B.; Akpoghelie, P.O.; Yousif, E.; Apameio, J.I.; Isoje, E.F.; Igbuku, U.A.; Garba, Y.; Essaghah, A.E.A. Chitosan and Its Derivatives: A Novel Approach to Gut Microbiota Modulation and Immune System Enhancement. Int. J. Biol. Macromol. 2025, 289, 138633. [Google Scholar] [CrossRef] [PubMed]
  19. Stull, V.; Weir, T. Chitin and Omega-3 Fatty Acids in Edible Insects Have Underexplored Benefits for the Gut Microbiome and Human Health. Nat. Food. 2023, 4, 283–287. [Google Scholar] [CrossRef]
  20. Wijesekara, T.; Xu, B. New Insights into Sources, Bioavailability, Health-Promoting Effects, and Applications of Chitin and Chitosan. J. Agric. Food Chem. 2024, 72, 17138–17152. [Google Scholar] [CrossRef]
  21. Weththasinghe, P.; Rocha, S.D.; Øyås, O.; Lagos, L.; Hansen, J.Ø.; Mydland, L.T.; Øverland, M. Modulation of Atlantic Salmon (Salmo salar) Gut Microbiota Composition and Predicted Metabolic Capacity by Feeding Diets with Processed Black Soldier Fly (Hermetia illucens) Larvae Meals and Fractions. Anim. Microbiome 2022, 4, 9. [Google Scholar] [CrossRef]
  22. Quah, Y.; Tong, S.-R.; Bojarska, J.; Giller, K.; Tan, S.-A.; Ziora, Z.M.; Esatbeyoglu, T.; Chai, T.-T. Bioactive Peptide Discovery from Edible Insects for Potential Applications in Human Health and Agriculture. Molecules 2023, 28, 1233. [Google Scholar] [CrossRef]
  23. Rehman, K.u.; Hollah, C.; Wiesotzki, K.; Heinz, V.; Aganovic, K.; Rehman, R.u.; Petrusan, J.-I.; Zheng, L.; Zhang, J.; Sohail, S. Insect-Derived Chitin and Chitosan: A Still Unexploited Resource for the Edible Insect Sector. Sustainability 2023, 15, 4864. [Google Scholar] [CrossRef]
  24. Athanassiou, C.; Coudron, C.; Deruytter, D.; Rumbos, C.; Gasco, L.; Gai, F.; Sandrock, C.; De Smet, J.; Tettamanti, G.; Francis, A. A Decade of Advances in Black Soldier Fly Research: From Genetics to Sustainability. J. Insects Food Feed 2024, 11, 219–246. [Google Scholar] [CrossRef]
  25. Kirichenko-Babko, M.; Bieganowski, A. The Variety of Applications of Hermetia illucens in Industrial and Agricultural Areas-Review. Biology 2023, 12, 25. [Google Scholar]
  26. Macwan, S.; de Souza, T.S.P.; Dunshea, F.R.; DiGiacomo, K.; Suleria, H.A.R. Black Soldier Fly Larvae (Hermetica illucens) as a Sustainable Source of Nutritive and Bioactive Compounds, and Their Consumption Challenges. Anim. Prod. Sci. 2024, 64, AN23192. [Google Scholar] [CrossRef]
  27. Mshayisa, V.V.; Van Wyk, J.; Zozo, B. Nutritional, Techno-Functional and Structural Properties of Black Soldier Fly (Hermetia illucens) Larvae Flours and Protein Concentrates. Foods 2022, 11, 724. [Google Scholar] [CrossRef]
  28. Pedrazzani, C.; Righi, L.; Vescovi, F.; Maistrello, L.; Caligiani, A. Black Soldier Fly as a New Chitin Source: Extraction, Purification and Molecular/Structural Characterization. LWT 2024, 191, 115618. [Google Scholar] [CrossRef]
  29. Triunfo, M.; Tafi, E.; Guarnieri, A.; Salvia, R.; Scieuzo, C.; Hahn, T.; Zibek, S.; Gagliardini, A.; Panariello, L.; Coltelli, M.B. Characterization of Chitin and Chitosan Derived from Hermetia illucens, a Further Step in a Circular Economy Process. Sci. Rep. 2022, 12, 6613. [Google Scholar] [CrossRef] [PubMed]
  30. Eke, M.; Tougeron, K.; Hamidovic, A.; Tinkeu, L.S.N.; Hance, T.; Renoz, F. Deciphering the Functional Diversity of the Gut Microbiota of the Black Soldier Fly (Hermetia illucens): Recent Advances and Future Challenges. Anim. Microbiome 2023, 5, 40. [Google Scholar] [CrossRef] [PubMed]
  31. Olivadese, M.; Dindo, M.L. Edible Insects: A Historical and Cultural Perspective on Entomophagy with a Focus on Western Societies. Insects 2023, 14, 690. [Google Scholar] [CrossRef]
  32. Granados-Echegoyen, C.; Vásquez-López, A.; Calderón-Cortés, N.; Gallego-Ocampo, H.L.; Gómez-Rodríguez, C.H.; Rodríguez-Vélez, J.M.; Sarmiento-Cordero, M.A.; Salamanca-Canizales, L.J.; Rodríguez-Vélez, B.; Arroyo-Balán, F. Brief Overview of Edible Insects: Exploring Consumption and Promising Sustainable Uses in Latin America. Front. Sustain. Food Syst. 2024, 8, 1385081. [Google Scholar] [CrossRef]
  33. Omuse, E.R.; Tonnang, H.E.Z.; Yusuf, A.A.; Machekano, H.; Egonyu, J.P.; Kimathi, E.; Mohamed, S.F.; Kassie, M.; Subramanian, S.; Onditi, J.; et al. The Global Atlas of Edible Insects: Analysis of Diversity and Commonality Contributing to Food Systems and Sustainability. Sci. Rep. 2024, 14, 5045. [Google Scholar] [CrossRef]
  34. Lange, K.W.; Nakamura, Y. Edible Insects as Future Food: Chances and Challenges. J. Future Foods 2021, 1, 38–46. [Google Scholar] [CrossRef]
  35. Matandirotya, N.R.; Filho, W.L.; Mahed, G.; Maseko, B.; Murandu, C.V. Edible Insects Consumption in Africa Towards Environmental Health and Sustainable Food Systems: A Bibliometric Study. Int. J. Environ. Res. Public Health 2022, 19, 14823. [Google Scholar] [CrossRef] [PubMed]
  36. Kipkoech, C.; Jaster-Keller, J.; Gottschalk, C.; Wesonga, J.; Maul, R. African Traditional Use of Edible Insects and Challenges Towards the Future Trends of Food and Feed. J. Insects Food Feed 2023, 9, 965–988. [Google Scholar] [CrossRef]
  37. Boukid, F.; Sogari, G.; Rosell, C.M. Edible Insects as Foods: Mapping Scientific Publications and Product Launches in the Global Market (1996–2021). J. Insects Food Feed 2022, 9, 353–368. [Google Scholar] [CrossRef]
  38. Florenca, S.G.; Guine, R.P.; Goncalves, F.J.; Barroca, M.J.; Ferreira, M.; Costa, C.A.; Correia, P.M.; Cardoso, A.P.; Campos, S.; Anjos, O. The Motivations for Consumption of Edible Insects: A Systematic Review. Foods 2022, 11, 3643. [Google Scholar] [CrossRef]
  39. Ros-Baro, M.; Casas-Agustench, P.; Díaz-Rizzolo, D.A.; Batlle-Bayer, L.; Adria-Acosta, F.; Aguilar-Martinez, A.; Medina, F.-X.; Pujola, M.; Bach-Faig, A. Edible Insect Consumption for Human and Planetary Health: A Systematic Review. Int. J. Environ. Res. Public Health 2022, 19, 11653. [Google Scholar] [CrossRef]
  40. Conway, A.; Jaiswal, S.; Jaiswal, A.K. The Potential of Edible Insects as a Safe, Palatable, and Sustainable Food Source in the European Union. Foods 2024, 13, 387. [Google Scholar] [CrossRef]
  41. Nachtigall, L.; Grune, T.; Weber, D. Proteins and Amino Acids from Edible Insects for the Human Diet—A Narrative Review Considering Environmental Sustainability and Regulatory Challenges. Nutrients 2025, 17, 1245. [Google Scholar] [CrossRef]
  42. Perez-Santaescolastica, C.; de Pril, I.; van de Voorde, I.; Fraeye, I. Fatty Acid and Amino Acid Profiles of Seven Edible Insects: Focus on Lipid Class Composition and Protein Conversion Factors. Foods 2023, 12, 4090. [Google Scholar] [CrossRef]
  43. Kolobe, S.D.; Manyelo, T.G.; Malematja, E.; Sebola, N.A.; Mabelebele, M. Fats and Major Fatty Acids Present in Edible Insects Utilised as Food and Livestock Feed. Vet. Anim. Sci. 2023, 22, 100312. [Google Scholar] [CrossRef]
  44. Bbosa, T.; Nakimbugwe, D.; Matthys, C.; Van Der Borght, M. A Systematic Review of Zinc, Iron and Vitamin B12 Content of Edible Insects and Comparison with Dietary Reference Values. Nutr. Res. Rev. 2025, 1–17. [Google Scholar] [CrossRef]
  45. Mwangi, M.N.; Oonincx, D.G.; Stouten, T.; Veenenbos, M.; Melse-Boonstra, A.; Dicke, M.; Van Loon, J.J. Insects as Sources of Iron and Zinc in Human Nutrition. Nutr. Res. Rev. 2018, 31, 248–255. [Google Scholar] [CrossRef]
  46. Kouřimská, L.; Adámková, A. Nutritional and Sensory Quality of Edible Insects. NFS J. 2016, 4, 22–26. [Google Scholar] [CrossRef]
  47. Psarianos, M.; Aghababaei, F.; Schlüter, O.K. Bioactive Compounds in Edible Insects: Aspects of Cultivation, Processing and Nutrition. Food Res. Int. 2025, 203, 115802. [Google Scholar] [CrossRef]
  48. Ferri, I.; Canala, B.; Rossi, L. Unravelling the Role of Chitin and Chitosan in Prebiotic Activity and Correlation with Cancer: A Narrative Review. Nutr. Rev. 2024, 83, e2015–e2024. [Google Scholar] [CrossRef] [PubMed]
  49. Ushakova, N.; Dontsov, A.; Sakina, N.; Bastrakov, A.; Ostrovsky, M. Antioxidative Properties of Melanins and Ommochromes from Black Soldier Fly Hermetia illucens. Biomolecules 2019, 9, 408. [Google Scholar] [CrossRef] [PubMed]
  50. Ma, Y.; Zhang, P.; Dai, X.; Yao, X.; Zhou, S.; Ma, Q.; Liu, J.; Tian, S.; Zhu, J.; Zhang, J.; et al. Extraction, Physicochemical Properties, and Antioxidant Activity of Natural Melanin from Auricularia Heimuer Fermentation. Front. Nutr. 2023, 10, 1131542. [Google Scholar] [CrossRef] [PubMed]
  51. Teixeira, C.S.S.; Villa, C.; Costa, J.; Ferreira, I.M.P.L.V.O.; Mafra, I. Edible Insects as a Novel Source of Bioactive Peptides: A Systematic Review. Foods 2023, 12, 2026. [Google Scholar] [CrossRef]
  52. Aiello, D.; Barbera, M.; Bongiorno, D.; Cammarata, M.; Censi, V.; Indelicato, S.; Mazzotti, F.; Napoli, A.; Piazzese, D.; Saiano, F. Edible Insects an Alternative Nutritional Source of Bioactive Compounds: A Review. Molecules 2023, 28, 699. [Google Scholar] [CrossRef]
  53. Sánchez-Estrada, M.; Aguirre-Becerra, H.; Feregrino-Pérez, A.A. Bioactive Compounds and Biological Activity in Edible Insects: A Review. Heliyon 2024, 10, e24045. [Google Scholar] [CrossRef]
  54. Zhang, Z.-Q.; Chen, S.-C.; Xiao, J.-H.; Huang, D.-W. State-of-the-Art Review of Edible Insect: From Bioactives, Pretreatment to Enrichment. Food Biosci. 2024, 59, 103879. [Google Scholar] [CrossRef]
  55. Marra, A.; Hanson, M.A.; Kondo, S.; Erkosar, B.; Lemaitre, B. Drosophila Antimicrobial Peptides and Lysozymes Regulate Gut Microbiota Composition and Abundance. mBio 2021, 12, e0082421. [Google Scholar] [CrossRef] [PubMed]
  56. Sahoo, A.; Swain, S.S.; Behera, A.; Sahoo, G.; Mahapatra, P.K.; Panda, S.K. Antimicrobial Peptides Derived from Insects Offer a Novel Therapeutic Option to Combat Biofilm: A Review. Front. Microbiol. 2021, 12, 661195. [Google Scholar] [CrossRef]
  57. Torres-Castillo, J.A.; Olazarán-Santibáñez, F.E. Insects as Source of Phenolic and Antioxidant Entomochemicals in the Food Industry. Front. Nutr. 2023, 10, 1133342. [Google Scholar] [CrossRef] [PubMed]
  58. Borrelli, L.; Varriale, L.; Dipineto, L.; Pace, A.; Menna, L.F.; Fioretti, A. Insect Derived Lauric Acid as Promising Alternative Strategy to Antibiotics in the Antimicrobial Resistance Scenario. Front. Microbiol. 2021, 12, 620798. [Google Scholar] [CrossRef]
  59. Alejandro Ruiz, F.E.; Ortega Jácome, J.F.; Tejera, E.; Alvarez-Suarez, J.M. Edible Insects as Functional Foods: Bioactive Compounds, Health Benefits, Safety Concerns, Allergenicity, and Regulatory Considerations. Front. Nutr. 2025, 12, 1571084. [Google Scholar] [CrossRef] [PubMed]
  60. Marangon, A.; Paul, G.; Zaghi, R.; Marchese, L.; Gatti, G. Chitin Extracted from Black Soldier Fly Larvae at Different Growth Stages. Polymers 2024, 16, 2861. [Google Scholar] [CrossRef]
  61. Eggink, K.M.; Dalsgaard, J. Chitin Contents in Different Black Soldier Fly (Hermetia illucens) Life Stages. J. Insects Food Feed 2023, 9, 855–864. [Google Scholar] [CrossRef]
  62. Querejeta, M.; Hervé, V.; Perdereau, E.; Marchal, L.; Herniou, E.A.; Boyer, S.; Giron, D. Changes in Bacterial Community Structure across the Different Life Stages of Black Soldier Fly (Hermetia illucens). Microb. Syst. 2023, 86, 1254–1267. [Google Scholar] [CrossRef] [PubMed]
  63. Laursen, S.F.; Flint, C.A.; Bahrndorff, S.; Tomberlin, J.K.; Kristensen, T.N. Reproductive Output and Other Adult Life-History Traits of Black Soldier Flies Grown on Different Organic Waste and by-Products. Waste Manag. 2024, 181, 136–144. [Google Scholar] [CrossRef]
  64. Klüber, P.; Arous, E.; Zorn, H.; Rühl, M. Protein- and Carbohydrate-Rich Supplements in Feeding Adult Black Soldier Flies (Hermetia illucens) Affect Life History Traits and Egg Productivity. Life 2023, 13, 355. [Google Scholar] [CrossRef]
  65. Muraro, T.; Lalanne, L.; Pelozuelo, L.; Calas-List, D. Mating and Oviposition of a Breeding Strain of Black Soldier Fly Hermetia illucens (Diptera: Stratiomyidae): Polygynandry and Multiple Egg-Laying. J. Insects Food Feed 2024, 10, 1423–1435. [Google Scholar] [CrossRef]
  66. Auger, L.; Deschamps, M.-H.; Vandenberg, G.; Derome, N. Microbiota Is Structured by Gut Regions, Life Stage, and Diet in the Black Soldier Fly (Hermetia illucens). Front. Microbiol. 2023, 14, 1221728. [Google Scholar] [CrossRef]
  67. Elkadaoui, S.; Azzi, M.; Desbrieres, J.; Zim, J.; El Hachimi, Y.; Tolaimate, A. Valorization of Hermetia illucens Breeding Rejects by Chitins and Chitosans Production. Influence of Processes and Life Cycle on Their Physicochemical Characteristics. Int. J. Biol. Macromol. 2024, 266, 131314. [Google Scholar] [CrossRef]
  68. Kotsou, K.; Chatzimitakos, T.; Athanasiadis, V.; Bozinou, E.; Lalas, S.I. Exploiting Agri-Food Waste as Feed for Tenebrio molitor Larvae Rearing: A Review. Foods 2024, 13, 1027. [Google Scholar] [CrossRef] [PubMed]
  69. Muurmann, A.T.; Eriksen, N.T.; Rasmussen, J.A.; Limborg, M.T.; Tomberlin, J.K.; Gilbert, M.T.P.; Bahrndorff, S. Growth and Metabolic Performance of House Fly and Black Soldier Fly Larvae Differ across Densities and Waste-Based Growth Substrates. Waste Manag. 2025, 193, 529–538. [Google Scholar] [CrossRef]
  70. Nayak, A.; Klüber, P. The Hidden Drivers: Unraveling the Impact of Density, Moisture, and Scale on Hermetia illucens Rearing. PLoS ONE 2025, 20, e0317049. [Google Scholar] [CrossRef]
  71. Zhang, Q.-H.; Puniamoorthy, N. Impact of Rearing Substrates on Black Soldier Fly Growth and Fertility: A Semi-Industrial Scale Study to Optimize Egg Collection. Insects 2025, 16, 142. [Google Scholar] [CrossRef] [PubMed]
  72. Lomonaco, G.; Franco, A.; De Smet, J.; Scieuzo, C.; Salvia, R.; Falabella, P. Larval Frass of Hermetia illucens as Organic Fertilizer: Composition and Beneficial Effects on Different Crops. Insects 2024, 15, 293. [Google Scholar] [CrossRef]
  73. Alagappan, S.; Dong, A.; Hoffman, L.; Cozzolino, D.; Mantilla, S.O.; James, P.; Yarger, O.; Mikkelsen, D. Microbial Safety of Black Soldier Fly Larvae (Hermetia illucens) Reared on Food Waste Streams. Waste Manag. 2025, 194, 221–227. [Google Scholar] [CrossRef]
  74. Brulé, L.; Misery, B.; Baudouin, G.; Yan, X.; Guidou, C.; Trespeuch, C.; Foltyn, C.; Anthoine, V.; Moriceau, N.; Federighi, M.; et al. Evaluation of the Microbial Quality of Hermetia illucens Larvae for Animal Feed and Human Consumption: Study of Different Types of Rearing Substrates. Foods 2024, 13, 1587. [Google Scholar] [CrossRef]
  75. Hoek-van den Hil, E.F.; Meijer, N.P.; Van Rozen, K.; Elissen, H.; van Wikselaar, P.G.; Brust, H.; Te Loeke, N.A.J.M.; de Rijk, T.; Tienstra, M.; van de Schans, M.G.M.; et al. Safety of Black Soldier Fly (Hermetia illucens) Larvae Reared on Waste Streams of Animal and Vegetal Origin and Manure. J. Insects Food Feed 2023, 10, 771–783. [Google Scholar] [CrossRef]
  76. Heuel, M.; Kreuzer, M.; Gangnat, I.D.M.; Frossard, E.; Zurbrügg, C.; Egger, J.; Dortmans, B.; Gold, M.; Mathys, A.; Jaster-Keller, J.; et al. Low Transfer of Cadmium, Lead and Aflatoxin B1 to Eggs and Meat of Laying Hens Receiving Diets with Black Soldier Fly Larvae Reared on Contaminated Substrates. Anim. Feed Sci. Technol. 2023, 304, 115733. [Google Scholar] [CrossRef]
  77. Zulkifli, N.F.N.M.; Seok-Kian, A.Y.; Seng, L.L.; Mustafa, S.; Kim, Y.-S.; Shapawi, R. Nutritional Value of Black Soldier Fly (Hermetia illucens) Larvae Processed by Different Methods. PLoS ONE 2022, 17, e0263924. [Google Scholar] [CrossRef] [PubMed]
  78. Lu, S.; Taethaisong, N.; Meethip, W.; Surakhunthod, J.; Sinpru, B.; Sroichak, T.; Archa, P.; Thongpea, S.; Paengkoum, S.; Purba, R.A.P.; et al. Nutritional Composition of Black Soldier Fly Larvae (Hermetia illucens L.) and Its Potential Uses as Alternative Protein Sources in Animal Diets: A Review. Insects 2022, 13, 831. [Google Scholar] [CrossRef] [PubMed]
  79. Mufungwe, J.; Namukonde, N.; Mwaanga, P.; Johnson, T.; Siamujompa, M.; Mwango, N.C.; Ngoma, J.; Hang’ombe, B.M. Critical Safety Concerns in the Production of Black Soldier Fly (Hermetia illucens) Larvae in Africa. Discov. Food 2025, 5, 74. [Google Scholar] [CrossRef]
  80. Delgado, L.; Garino, C.; Moreno, F.J.; Zagon, J.; Broll, H. Sustainable Food Systems: Eu Regulatory Framework and Contribution of Insects to the Farm-to-Fork Strategy. Food Res. Int. 2022, 39, 6955–6976. [Google Scholar] [CrossRef]
  81. Vale-Hagan, W.; Singhal, S.; Grigoletto, I.; Totaro-Fila, C.; Theodoridou, K.; Koidis, A. Edible Insects in Mixed-Sourced Protein Meals for Animal Feed and Food: An Eu Focus. Food Humanit. 2023, 1, 1180–1187. [Google Scholar] [CrossRef]
  82. Grabowski, N.T.; Tchibozo, S.; Abdulmawjood, A.; Acheuk, F.; M’Saad Guerfali, M.; Sayed, W.A.; Plötz, M. Edible Insects in Africa in Terms of Food, Wildlife Resource, and Pest Management Legislation. Foods 2020, 9, 502. [Google Scholar] [CrossRef]
  83. Bekele, M.; Grace, D.; Knight-Jones, T.J.D.; Mutua, F.; Lindahl, J.F.; Daivadanam, M. Implementation Gaps in Food Safety Interventions: Evidence from a Multi-Vocal Review Focusing on Animal-Source Foods in Ethiopia. Front. Sustain. Food Syst. 2025, 9, 1546347. [Google Scholar] [CrossRef]
  84. Lähteenmäki-Uutela, A.; Marimuthu, S.B.; Meijer, N. Regulations on Insects as Food and Feed: A Global Comparison. J. Insects Food Feed 2021, 7, 849–856. [Google Scholar] [CrossRef]
  85. Alagappan, S.; Rowland, D.; Barwell, R.; Cozzolino, D.; Mikkelsen, D.; Olarte Mantilla, S.M.; James, P.; Yarger, O.; Hoffman, L. Organic Side Streams (Bioproducts) as Substrate for Black Soldier Fly. Anim. Prod. Sci. 2022, 62, 1639–1651. [Google Scholar] [CrossRef]
  86. Sogari, G.; Amato, M.; Palmieri, R.; Hadj Saadoun, J.; Formici, G.; Verneau, F.; Mancini, S. The Future Is Crawling: Evaluating the Potential of Insects for Food and Feed Security. Curr. Res. Food Sci. 2023, 6, 100504. [Google Scholar] [CrossRef]
  87. Fuso, A.; Barbi, S.; Macavei, L.I.; Luparelli, A.V.; Maistrello, L.; Montorsi, M.; Sforza, S.; Caligiani, A. Effect of the Rearing Substrate on Total Protein and Amino Acid Composition in Black Soldier Fly. Foods 2021, 10, 1773. [Google Scholar] [CrossRef] [PubMed]
  88. Silva, B.C.R.; Lehnen, C.R.; Marcato, S.M. Black Soldier Fly (Hermetia illucens) as a Protein Ingredient in Poultry Feed. World’s Poult. Sci. J. 2024, 80, 1123–1154. [Google Scholar] [CrossRef]
  89. Cheng, V.; Shoveller, A.K.; Huber, L.-A.; Kiarie, E.G. Comparative Protein Quality in Black Soldier Fly Larvae Meal Vs. Soybean Meal and Fish Meal Using Classical Protein Efficiency Ratio (Per) Chick Growth Assay Model. Poult. Sci. 2023, 102, 102255. [Google Scholar] [CrossRef]
  90. Fukuda, E.P.; Cox, J.R.; Wickersham, T.A.; Drewery, M.L. Evaluation of Black Soldier Fly Larvae (Hermetia illucens) as a Protein Supplement for Beef Steers Consuming Low-Quality Forage. Transl. Anim. Sci. 2022, 6, txac018. [Google Scholar] [CrossRef]
  91. Gautam, A.; Gyawali, I.; Poudel, S.; Devkota, S.; Acharya, R.; Kandel, M.; Subedi, D. Insects as Food and Feed Source: A Comprehensive Review on Nutritional Value, Food Safety Concern, Environmental Benefits, Economic Potential, Technological Innovations, Challenges, and Future Prospects. Food Front. 2025. [Google Scholar] [CrossRef]
  92. Chen, Y.; Guo, Y.; He, X.; Tan, B.; Liao, Z.; Chen, A.; Gu, X.; Li, X.; Chen, X.; Chen, B.; et al. Comprehensive Utilization of Black Soldier Fly (Hermetia illucens) Larvae: Extraction, Recovery and Characterization of Peptide, Chitin and Melanin and Scaling-up Trial. Sep. Purif. Technol. 2025, 361, 131262. [Google Scholar] [CrossRef]
  93. Van Etten, C.H.; Hubbard, J.E.; Mallan, J.M.; Smith, A.K.; Blessin, C.W. Amino Acids in Soybeans, Amino Acid Composition of Soybean Protein Fractions. J. Agric. Food Chem. 1959, 7, 129–131. [Google Scholar] [CrossRef]
  94. Suryati, T.; Julaeha, E.; Farabi, K.; Ambarsari, H.; Hidayat, A.T. Lauric Acid from the Black Soldier Fly (Hermetia illucens) and Its Potential Applications. Sustainability 2023, 15, 10383. [Google Scholar] [CrossRef]
  95. Almeida, C.; Murta, D.; Nunes, R.; Baby, A.R.; Fernandes, Â.; Barros, L.; Rijo, P.; Rosado, C. Characterization of Lipid Extracts from the Hermetia illucens Larvae a Nd Their Bioactivities for Potential Use as Pharmaceutical and Cosmetic Ingredients. Heliyon 2022, 8, e09455. [Google Scholar] [CrossRef] [PubMed]
  96. Ewald, N.; Vidakovic, A.; Langeland, M.; Kiessling, A.; Sampels, S.; Lalander, C. Fatty Acid Composition of Black Soldier Fly Larvae (Hermetia illucens)—Possibilities and Limitations for Modification through Diet. Waste Manag. 2020, 102, 40–47. [Google Scholar] [CrossRef]
  97. Opatovsky, I.; Vitenberg, T.; Jonas-Levi, A.; Gutman, R. Does Consumption of Baker’s Yeast (Saccharomyces cerevisiae) by Black Soldier Fly (Diptera: Stratiomyidae) Larvae Affect Their Fatty Acid Composition? J. Insect Sci. 2021, 21, 5. [Google Scholar] [CrossRef]
  98. Cattaneo, A.; Meneguz, M.; Dabbou, S. The Fatty Acid Composition of Black Soldier Fly Larvae: The Influence of Feed Substrate and Applications in the Feed Industry. J. Insects Food Feed 2023, 10, 533–558. [Google Scholar] [CrossRef]
  99. Richter, H.; Gover, O.; Schwartz, B. Anti-Inflammatory Activity of Black Soldier Fly Oil Associated with Modulation of Tlr Signaling: A Metabolomic Approach. Int. J. Mol. Sci. 2023, 24, 10634. [Google Scholar] [CrossRef]
  100. Hadj Saadoun, J.; Montevecchi, G.; Zanasi, L.; Bortolini, S.; Macavei, L.I.; Masino, F.; Maistrello, L.; Antonelli, A. Lipid Profile and Growth of Black Soldier Flies (Hermetia illucens, Stratiomyidae) Reared on by-Products from Different Food Chains. J. Sci. Food Agric. 2020, 100, 3648–3657. [Google Scholar] [CrossRef] [PubMed]
  101. Heuel, M.; Kreuzer, M.; Sandrock, C.; Leiber, F.; Mathys, A.; Gold, M.; Zurbrügg, C.; Gangnat, I.D.M.; Terranova, M. Transfer of Lauric and Myristic Acid from Black Soldier Fly Larval Lipids to Egg Yolk Lipids of Hens Is Low. Lipids 2021, 56, 423–435. [Google Scholar] [CrossRef]
  102. Wang, W.; Xue, C.; Mao, X. Chitosan: Structural Modification, Biological Activity and Application. Int. J. Biol. Macromol. 2020, 164, 4532–4546. [Google Scholar] [CrossRef]
  103. Soetemans, L.; Uyttebroek, M.; Bastiaens, L. Characteristics of Chitin Extracted from Black Soldier Fly in Different Life Stages. Int. J. Biol. Macromol. 2020, 165, 3206–3214. [Google Scholar] [CrossRef] [PubMed]
  104. Tan, Y.N.; Chin, Y.L.; Chen, W.N. Comparison of Sustainable Lipid and Protein Removal Methods for the Isolation of Insect Chitin from Black Soldier Fly Exoskeleton. ACS Food Sci. Technol. 2021, 1, 698–706. [Google Scholar] [CrossRef]
  105. Xiong, A.; Ruan, L.; Ye, K.; Huang, Z.; Yu, C. Extraction of Chitin from Black Soldier Fly (Hermetia illucens) and Its Puparium by Using Biological Treatment. Life 2023, 13, 1424. [Google Scholar] [CrossRef]
  106. Witono, J.R.; Setyadi, F.F.; Deandra, P.P.; Wanta, K.C.; Miryanti, A.; Santoso, H.; Astuti, D.A.; Bulin, C.D.Q. A Comprehensive Analysis of Chitin Extraction from the Black Soldier Fly for Chitosan Production. Period. Polytech. Chem. Eng. 2024, 68, 507–522. [Google Scholar] [CrossRef]
  107. Lin, Y.-S.; Liang, S.-H.; Lai, W.-L.; Lee, J.-X.; Wang, Y.-P.; Liu, Y.-T.; Wang, S.-H.; Lee, M.-H. Sustainable Extraction of Chitin from Spent Pupal Shell of Black Soldier Fly. Processes 2021, 9, 976. [Google Scholar] [CrossRef]
  108. Guan, Z.; Feng, Q. Chitosan and Chitooligosaccharide: The Promising Non-Plant-Derived Pre Biotics with Multiple Biological Activities. Int. J. Mol. Sci. 2022, 23, 6761. [Google Scholar] [CrossRef]
  109. Kipkoech, C.; Kinyuru, J.N.; Imathiu, S.; Meyer-Rochow, V.B.; Roos, N. In Vitro Study of Cricket Chitosan’s Potential as a Prebiotic and a Promoter of Probiotic Microorganisms to Control Pathogenic Bacteria in the Human Gut. Foods 2021, 10, 2310. [Google Scholar] [CrossRef]
  110. Teo, H.P.; Law, K.W.; Eric Chan, W.C.; Michelle Soo, O.Y. Antibacterial Properties of Chitosan Isolated from the Black Soldier Fly, Hermetia illucens. Sains Malays. 2022, 51, 3923–3935. [Google Scholar] [CrossRef]
  111. Liu, S.; Raheel Tariq, M.; Zhang, Q.; Wang, H.; Wang, F.; Zheng, C.; Li, K.; Zhuang, Z.; Wang, L. Dietary Influence on Growth, Physicochemical Stability, and Antimicrobial Mechanisms of Antimicrobial Peptides in Black Soldier Fly Larvae. Insects 2024, 15, 872. [Google Scholar] [CrossRef] [PubMed]
  112. Praseatsook, K.; Vachiraarunwong, A.; Taya, S.; Setthaya, P.; Sato, K.; Wanibuchi, H.; Wongpoomchai, R.; Dejkriengkraikul, P.; Gi, M.; Yodkeree, S. Anticancer and Antioxidant Effects of Bioactive Peptides from Black Soldier Fly Larvae (Hermetia illucens). Nutrients 2025, 17, 645. [Google Scholar] [CrossRef]
  113. Lu, J.; Guo, Y.; Muhmood, A.; Zeng, B.; Qiu, Y.; Wang, P.; Ren, L. Probing the Antioxidant Activity of Functional Proteins and Bioactive Peptides in Hermetia illucens Larvae Fed with Food Wastes. Sci. Rep. 2022, 12, 2799. [Google Scholar] [CrossRef] [PubMed]
  114. Wong, F.-C.; Lee, Y.-H.; Ong, J.-H.; Manan, F.A.; Sabri, M.Z.; Chai, T.-T. Exploring the Potential of Black Soldier Fly Larval Proteins as Bioact Ive Peptide Sources through in Silico Gastrointestinal Proteolysis: A Cheminformatic Investigation. Catalysts 2023, 13, 605. [Google Scholar] [CrossRef]
  115. Riolo, K.; Rotondo, A.; La Torre, G.L.; Marino, Y.; Franco, G.A.; Crupi, R.; Fusco, R.; Di Paola, R.; Oliva, S.; De Marco, G.; et al. Cytoprotective and Antioxidant Effects of Hydrolysates from Black Soldier Fly (Hermetia illucens). Antioxidants 2023, 12, 519. [Google Scholar] [CrossRef]
  116. Leni, G.; Del Vecchio, L.; Dellapina, C.; Moliterni, V.M.C.; Caligiani, A.; Cirlini, M. Black Soldier Fly Larvae Grown on Hemp Fiber: Nutritional Composition and Production of Potential Bioactive Peptides. Macromol 2024, 4, 135–149. [Google Scholar] [CrossRef]
  117. Moretta, A.; Salvia, R.; Scieuzo, C.; Di Somma, A.; Vogel, H.; Pucci, P.; Sgambato, A.; Wolff, M.; Falabella, P. A Bioinformatic Study of Antimicrobial Peptides Identified in the Black Soldier Fly (Bsf) Hermetia illucens (Diptera: Stratiomyidae). Sci. Rep. 2020, 10, 16875. [Google Scholar] [CrossRef]
  118. Gasco, L.; Biancarosa, I.; Liland, N.S. From Waste to Feed: A Review of Recent Knowledge on Insects as Producers of Protein and Fat for Animal Feeds. Curr. Opin. Green Sustain. Chem. 2020, 23, 67–79. [Google Scholar] [CrossRef]
  119. Anil, S. Potential Medical Applications of Chitooligosaccharides. Polymers 2022, 14, 3558. [Google Scholar] [CrossRef]
  120. Liu, W.; Li, X.; Zhao, Z.; Pi, X.; Meng, Y.; Fei, D.; Liu, D.; Wang, X. Effect of Chitooligosaccharides on Human Gut Microbiota and Antiglycation. Carbohydr. Polym. 2020, 242, 116413. [Google Scholar] [CrossRef] [PubMed]
  121. Wang, Q.; Jiang, Y.; Luo, X.; Wang, C.; Wang, N.; He, H.; Zhang, T.; Chen, L. Chitooligosaccharides Modulate Glucose-Lipid Metabolism by Suppressing Smyd3 Pathways and Regulating Gut Microflora. Mar. Drugs 2020, 18, 69. [Google Scholar] [CrossRef]
  122. Refael, G.; Riess, H.T.; Levi, C.S.; Magzal, F.; Tamir, S.; Koren, O.; Lesmes, U. Responses of the Human Gut Microbiota to Physiologically Digested Insect Powders or Isolated Chitin Thereof. Future Foods 2022, 6, 100197. [Google Scholar] [CrossRef]
  123. Ji, X.; Zhu, L.; Chang, K.; Zhang, R.; Chen, Y.; Yin, H.; Jin, J.; Zhao, L. Chitooligosaccahrides: Digestion Characterization and Effect of the Degree of Polymerization on Gut Microorganisms to Manage the Metabolome Functional Diversity in Vitro. Carbohydr. Polym. 2022, 275, 118716. [Google Scholar] [CrossRef]
  124. Lopez-Santamarina, A.; Mondragon, A.d.C.; Lamas, A.; Miranda, J.M.; Franco, C.M.; Cepeda, A. Animal-Origin Prebiotics Based on Chitin: An Alternative for the Future? A Critical Review. Foods 2020, 9, 782. [Google Scholar] [CrossRef]
  125. Benchamas, G.; Huang, G.; Huang, S.; Huang, H. Preparation and Biological Activities of Chitosan Oligosaccharides. Trends Food Sci. Technol. 2021, 107, 38–44. [Google Scholar] [CrossRef]
  126. Zhai, X.; Li, C.; Ren, D.; Wang, J.; Ma, C.; Abd El-Aty, A.M. The Impact of Chitooligosaccharides and Their Derivatives on the in Vitro and in Vivo Antitumor Activity: A Comprehensive Review. Carbohydr. Polym. 2021, 266, 118132. [Google Scholar] [CrossRef]
  127. Zhou, D.-Y.; Wu, Z.-X.; Yin, F.-W.; Song, S.; Li, A.; Zhu, B.-W.; Yu, L.-L. Chitosan and Derivatives: Bioactivities and Application in Foods. Annu. Rev. Food Sci. Technol. 2021, 12, 407–432. [Google Scholar] [CrossRef]
  128. Zhang, Z.; Zhao, L.; Qu, H.; Zhou, H.; Yang, H.; Chen, H. Physicochemical Characterization, Adsorption Function and Prebiotic Effect of Chitin-Glucan Complex from Mushroom Coprinus Comatus. Int. J. Biol. Macromol. 2022, 206, 255–263. [Google Scholar] [CrossRef] [PubMed]
  129. Mohan, K.; Rajan, D.K.; Ganesan, A.R.; Divya, D.; Johansen, J.; Zhang, S. Chitin, Chitosan and Chitooligosaccharides as Potential Growth Promoters and Immunostimulants in Aquaculture: A Comprehensive Review. Int. J. Biol. Macromol. 2023, 251, 126285. [Google Scholar] [CrossRef] [PubMed]
  130. Calatayud, M.; Van den Abbeele, P.; Ghyselinck, J.; Marzorati, M.; Rohs, E.; Birkett, A. Comparative Effect of 22 Dietary Sources of Fiber on Gut Microbiota of Healthy Humans in Vitro. Front. Nutr. 2021, 8, 700571. [Google Scholar] [CrossRef]
  131. Rezende, E.S.V.; Lima, G.C.; Naves, M.M.V. Dietary Fibers as Beneficial Microbiota Modulators: A Proposed Classification by Prebiotic Categories. Nutrition 2021, 89, 111217. [Google Scholar] [CrossRef]
  132. Abdi, R.; Joye, I.J. Prebiotic Potential of Cereal Components. Foods 2021, 10, 2338. [Google Scholar] [CrossRef]
  133. Wang, S.; Zhang, X.; Li, H.; Ren, Y.; Geng, Y.; Lu, Z.; Shi, J.; Xu, Z. Similarities and Differences of Oligo/Poly-Saccharides’ Impact on Human Fecal Microbiota Identified by in Vitro Fermentation. Appl. Microbiol. Biotechnol. 2021, 105, 7475–7486. [Google Scholar] [CrossRef]
  134. Bai, J.; Li, Y.; Li, T.; Zhang, W.; Fan, M.; Zhang, K.; Qian, H.; Zhang, H.; Qi, X.; Wang, L. Comparison of Different Soluble Dietary Fibers During the in Vitro Fermentation Process. J. Agric. Food Chem. 2021, 69, 7446–7457. [Google Scholar] [CrossRef] [PubMed]
  135. Vinelli, V.; Biscotti, P.; Martini, D.; Del Bo’, C.; Marino, M.; Meroño, T.; Nikoloudaki, O.; Calabrese, F.M.; Turroni, S.; Taverniti, V.; et al. Effects of Dietary Fibers on Short-Chain Fatty Acids and Gut Microbiot a Composition in Healthy Adults: A Systematic Review. Nutrients 2022, 14, 2559. [Google Scholar] [CrossRef]
  136. Zhong, C.; Ukowitz, C.; Domig, K.J.; Nidetzky, B. Short-Chain Cello-Oligosaccharides: Intensification and Scale-up of Their Enzymatic Production and Selective Growth Promotion among Probiotic Bacteria. J. Agric. Food Chem. 2020, 68, 8557–8567. [Google Scholar] [CrossRef]
  137. Na, K.; Wei, J.; Zhang, L.; Fang, Y.; Li, X.; Lu, S.; Guo, X. Effects of Chitosan Oligosaccharides (Cos) and Fmt from Cos-Dosed Mice on Intestinal Barrier Function and Cell Apoptosis. Carbohydr. Polym. 2022, 297, 120043. [Google Scholar] [CrossRef]
  138. Young, W.; Arojju, S.K.; McNeill, M.R.; Rettedal, E.; Gathercole, J.; Bell, N.; Payne, P. Feeding Bugs to Bugs: Edible Insects Modify the Human Gut Microbiome in an in Vitro Fermentation Model. Front. Microbiol. 2020, 11, 1763. [Google Scholar] [CrossRef] [PubMed]
  139. Anshory, M.; Effendi, R.M.R.A.; Kalim, H.; Dwiyana, R.F.; Suwarsa, O.; Nijsten, T.E.C.; Nouwen, J.L.; Thio, H.B. Butyrate Properties in Immune-Related Diseases: Friend or Foe? Fermentation 2023, 9, 205. [Google Scholar] [CrossRef]
  140. Xie, L.; Alam, M.J.; Marques, F.Z.; Mackay, C.R. A Major Mechanism for Immunomodulation: Dietary Fibres and Acid Metabo Lites. Semin. Immunol. 2023, 66, 101737. [Google Scholar] [CrossRef]
  141. Korsten, S.G.P.J.; Vromans, H.; Garssen, J.; Willemsen, L.E.M. Butyrate Protects Barrier Integrity and Suppresses Immune Activation in a Caco-2/Pbmc Co-Culture Model While Hdac Inhibition Mimics Butyrate in Restoring Cytokine-Induced Barrier Disruption. Nutrients 2023, 15, 2760. [Google Scholar] [CrossRef]
  142. Li, Y.; Wu, L.; Yong, Y.; Niu, X.; Gao, Y.; Zhou, Q.; Xie, H.; Liu, X.; Li, Y.; Yu, Z.; et al. Enhancing Gut Barrier Integrity: Upregulation of Tight Junction Proteins by Chitosan Oligosaccharide through the Erk1/2 Signaling Pathway. Nutrition 2024, 124, 112428. [Google Scholar] [CrossRef]
  143. Singh, V.; Lee, G.; Son, H.; Koh, H.; Kim, E.S.; Unno, T.; Shin, J.-H. Butyrate Producers, “the Sentinel of Gut”: Their Intestinal Significance with and Beyond Butyrate, and Prospective Use as Microbial Therapeutics. Front. Microbiol. 2023, 13, 1103836. [Google Scholar] [CrossRef]
  144. Hays, K.E.; Pfaffinger, J.M.; Ryznar, R. The Interplay between Gut Microbiota, Short-Chain Fatty Acids, and Implications for Host Health and Disease. Gut Microbes 2024, 16, 2393270. [Google Scholar] [CrossRef]
  145. Liu, H.; Lu, H.; Wang, Y.; Yu, C.; He, Z.; Dong, H. Unlocking the Power of Short-Chain Fatty Acids in Ameliorating Intesti Nal Mucosal Immunity: A New Porcine Nutritional Approach. Front. Cell. Infect. Microbiol. 2024, 14, 1449030. [Google Scholar] [CrossRef]
  146. Ayman, U.; Akter, L.; Islam, R.; Bhakta, S.; Rahman, M.A.; Islam, M.R.; Sultana, N.; Sharif, A.; Jahan, M.R.; Rahman, M.S.; et al. Dietary Chitosan Oligosaccharides Improves Health Status in Broilers for Safe Poultry Meat Production. Ann. Agric. Sci. 2022, 67, 90–98. [Google Scholar] [CrossRef]
  147. Lan, R.; Wu, F.; Wang, Y.; Lin, Z.; Wang, H.; Zhang, J.; Zhao, Z. Chitosan Oligosaccharide Improves Intestinal Function by Promoting Intestinal Development, Alleviating Intestinal Inflammatory Response, and Enhancing Antioxidant Capacity in Broilers Aged D 1 to 14. Poult. Sci. 2024, 103, 103381. [Google Scholar] [CrossRef]
  148. Gu, Y.F.; Chen, Y.P.; Jin, R.; Wang, C.; Wen, C.; Zhou, Y.M. Dietary Chitooligosaccharide Supplementation Alleviates Intestinal Barrier Damage, and Oxidative and Immunological Stress in Lipopolysaccharide-Challenged Laying Hens. Poult. Sci. 2022, 101, 101701. [Google Scholar] [CrossRef] [PubMed]
  149. Wang, Y.; Liu, S.; Tang, D.; Dong, R.; Feng, Q. Chitosan Oligosaccharide Ameliorates Metabolic Syndrome Induced by Overnutrition Via Altering Intestinal Microbiota. Front. Nutr. 2021, 8, 743492. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, X.; Jin, J.; Hou, F.; Song, B.; Li, Z.; Zhao, Y. Effects of Black Soldier Fly Larvae Oil on Growth Performance, Immunit Y and Antioxidant Capacity, and Intestinal Function and Microbiota of Broilers. J. Appl. Poult. Res. 2022, 31, 100292. [Google Scholar] [CrossRef]
  151. Li, J.; Cheng, Y.; Chen, Y.; Qu, H.; Zhao, Y.; Wen, C.; Zhou, Y. Dietary Chitooligosaccharide Inclusion as an Alternative to Antibiotics Improves Intestinal Morphology, Barrier Function, Antioxidant Capacity, and Immunity of Broilers at Early Age. Animals 2019, 9, 493. [Google Scholar] [CrossRef]
  152. Peng, L.; Li, Z.-R.; Green, R.S.; Holzmanr, I.R.; Lin, J. Butyrate Enhances the Intestinal Barrier by Facilitating Tight Junction Assembly Via Activation of Amp-Activated Protein Kinase in Caco-2 Cell Monolayers12. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [PubMed]
  153. Liu, H.; Zhang, L.; Yu, J.; Shao, S. Advances in the Application and Mechanism of Bioactive Peptides in the Treatment of Inflammation. Front. Immunol. 2024, 15, 1413179. [Google Scholar] [CrossRef]
  154. Hu, Z.; Xia, M.; Wang, G.; Jia, L.; Ji, H.; Sun, J.; Yu, H. A Superior Chitin Product: Black Soldier Fly Larvae Chitin, Beneficial to Growth Performance, Muscle Quality and Health Status of Largemouth Bass Micropterus Salmoides, in Comparison to Shrimp Chitin. Aquaculture 2025, 595, 741667. [Google Scholar] [CrossRef]
  155. Smets, R.; Verbinnen, B.; Van De Voorde, I.; Aerts, G.; Claes, J.; Van Der Borght, M. Sequential Extraction and Characterisation of Lipids, Proteins, and Chitin from Black Soldier Fly (Hermetia illucens) Larvae, Prepupae, and Pupae. Waste Biomass Valorization 2020, 11, 6455–6466. [Google Scholar] [CrossRef]
  156. Bose, U.; Broadbent, J.A.; Juhász, A.; Karnaneedi, S.; Johnston, E.B.; Stockwell, S.; Byrne, K.; Limviphuvadh, V.; Maurer-Stroh, S.; Lopata, A.L.; et al. Comparison of Protein Extraction Protocols and Allergen Mapping from Black Soldier Fly Hermetia illucens. J. Proteom. 2022, 269, 104724. [Google Scholar] [CrossRef]
  157. Bose, U.; Juhasz, A.; Stockwell, S.; Escobar-Correas, S.; Marcora, A.; Paull, C.; Broadbent, J.A.; Wijffels, G. Unpacking the Proteome and Metaproteome of the Black Soldier Fly Larvae: Efficacy and Complementarity of Multiple Protein Extraction Protocols. ACS Omega 2023, 8, 7319–7330. [Google Scholar] [CrossRef]
  158. Meng, L.; Yu, D.; Lin, J.; Hu, Y.; Peng, N.; Zhao, S. Structural Characterization, Hepg2 Cell Cytoprotective Ability, and an Antioxidant Mechanism of Novel Antioxidant Peptides Identified from Black Soldier Fly Larvae (Hermetia illucens L.). Food Chem. 2025, 463, 141462. [Google Scholar] [CrossRef]
  159. Coscueta, E.R.; Batista, P.; Gomes, J.E.G.; da Silva, R.; Pintado, M.M. Screening of Novel Bioactive Peptides from Goat Casein: In Silico to in Vitro Validation. Int. J. Mol. Sci. 2022, 23, 2439. [Google Scholar] [CrossRef] [PubMed]
  160. Bao, X.; Wu, J. Impact of Food-Derived Bioactive Peptides on Gut Function and Health. Food Res. Int. 2021, 147, 110485. [Google Scholar] [CrossRef] [PubMed]
  161. Gubatan, J.; Holman, D.R.; Puntasecca, C.J.; Polevoi, D.; Rubin, S.J.S.; Rogalla, S. Antimicrobial Peptides and the Gut Microbiome in Inflammatory Bowel Disease. World J. Gastroenterol. 2021, 27, 7402–7422. [Google Scholar] [CrossRef]
  162. Zhang, Y.; Chen, S.; Zong, X.; Wang, C.; Shi, C.; Wang, F.; Wang, Y.; Lu, Z. Peptides Derived from Fermented Soybean Meal Suppresses Intestinal Inf Lammation and Enhances Epithelial Barrier Function in Piglets. Food Agric. Immunol. 2019, 31, 120–135. [Google Scholar] [CrossRef]
  163. Li, M.; Ge, Q.; Du, H.; Jiang, P.; Bao, Z.; Chen, D.; Lin, S. Potential Mechanisms Mediating the Protective Effects of Tricholoma Matsutake Derived Peptides in Mitigating Dss-Induced Colitis. J. Agric. Food Chem. 2021, 69, 5536–5546. [Google Scholar] [CrossRef] [PubMed]
  164. Ha, J.; Oh, H.; Oh, N.S.; Seo, Y.; Kang, J.; Park, M.H.; Kim, K.S.; Kang, S.H.; Yoon, Y. Anti-Inflammatory Effect of a Peptide Derived from the Synbiotics, Fermented Cudrania tricuspidata with Lactobacillus gasseri on Inflammatory Bowel Disease. Mediat. Inflamm. 2020, 2020, 3572809. [Google Scholar] [CrossRef]
  165. Zhu, W.; Ren, L.; Zhang, L.; Qiao, Q.; Farooq, M.Z.; Xu, Q. The Potential of Food Protein-Derived Bioactive Peptides against Chronic Intestinal Inflammation. Mediat. Inflamm. 2020, 2020, 6817156. [Google Scholar] [CrossRef]
  166. Koutsos, E.; Modica, B.; Freel, T. Immunomodulatory Potential of Black Soldier Fly Larvae: Applications B Eyond Nutrition in Animal Feeding Programs. Transl. Anim. Sci. 2022, 6, txac084. [Google Scholar] [CrossRef]
  167. Ferrazzano, G.F.; D’Ambrosio, F.; Caruso, S.; Gatto, R.; Caruso, S. Bioactive Peptides Derived from Edible Insects: Effects on Human Health and Possible Applications in Dentistry. Nutrients 2023, 15, 4611. [Google Scholar] [CrossRef]
  168. Tarahi, M.; Aghababaei, F.; McClements, D.J.; Pignitter, M.; Hadidi, M. Bioactive Peptides Derived from Insect Proteins: Preparation, Biologic Al Activities, Potential Applications, and Safety Issues. Food Chem. 2025, 465, 142113. [Google Scholar] [CrossRef] [PubMed]
  169. Lachat, J.; Lextrait, G.; Jouan, R.; Boukherissa, A.; Yokota, A.; Jang, S.; Ishigami, K.; Futahashi, R.; Cossard, R.; Naquin, D.; et al. Hundreds of Antimicrobial Peptides Create a Selective Barrier for Insect Gut Symbionts. Proc. Natl. Acad. Sci. USA 2024, 121, e2401802121. [Google Scholar] [CrossRef]
  170. Cardoso, M.H.; Meneguetti, B.T.; Oliveira-Júnior, N.G.; Macedo, M.L.R.; Franco, O.L. Antimicrobial Peptide Production in Response to Gut Microbiota Imbalance. Peptides 2022, 157, 170865. [Google Scholar] [CrossRef]
  171. Mu, J.; Lin, Q.; Liang, Y. An Update on the Effects of Food-Derived Active Peptides on the Intestinal Microecology. Crit. Rev. Food Sci. Nutr. 2022, 63, 11625–11639. [Google Scholar] [CrossRef]
  172. Yang, Q.; Lyu, S.; Xu, M.; Li, S.; Du, Z.; Liu, X.; Shang, X.; Yu, Z.; Liu, J.; Zhang, T. Potential Benefits of Egg White Proteins and Their Derived Peptides in the Regulation of the Intestinal Barrier and Gut Microbiota: A Compre Hensive Review. J. Agric. Food Chem. 2023, 71, 13168–13180. [Google Scholar] [CrossRef] [PubMed]
  173. Zeng, T.; Jaffar, S.; Xu, Y.; Qi, Y. The Intestinal Immune Defense System in Insects. Int. J. Mol. Sci. 2022, 23, 15132. [Google Scholar] [CrossRef] [PubMed]
  174. Fahmy, L.; Generalovic, T.; Ali, Y.M.; Seilly, D.; Sivanesan, K.; Kalmar, L.; Pipan, M.; Christie, G.; Grant, A.J. A Novel Family of Defensin-Like Peptides from Hermetia illucens with Antibacterial Properties. BMC Microbiol. 2024, 24, 167. [Google Scholar] [CrossRef]
  175. Pimchan, T.; Hamzeh, A.; Siringan, P.; Thumanu, K.; Hanboonsong, Y.; Yongsawatdigul, J. Antibacterial Peptides from Black Soldier Fly (Hermetia illucens) Larvae: Mode of Action and Characterization. Sci. Rep. 2024, 14, 26469. [Google Scholar] [CrossRef]
  176. Bonomini, M.G.; Verstringe, S.; Bruggeman, G.; Vandercruyssen, R.; Carmans, H.; Caligiani, A. Characterisation, Antibacterial Activity, and Prebiotic Potential of Dried Hermetia illucens L. Larvae and of Their Fractions. J. Insects Food Feed 2024, 11, 273–288. [Google Scholar] [CrossRef]
  177. Salam, M.; Bolletta, V.; Meng, Y.; Yakti, W.; Grossule, V.; Shi, D.; Hayat, F. Exploring the Role of the Microbiome of the H. Illucens (Black Soldier Fly) for Microbial Synergy in Optimizing Black Soldier Fly Rearing and Subsequent Applications. Environ. Pollut. 2024, 363, 125055. [Google Scholar] [CrossRef]
  178. Wang, Y.; Quan, J.; Cheng, X.; Li, C.; Yuan, Z. Relationship of Black Soldier Fly Larvae (Bsfl) Gut Microbiota and Bioconversion Efficiency with Properties of Substrates. Waste Manag. 2024, 180, 106–114. [Google Scholar] [CrossRef]
  179. Spragge, F.; Bakkeren, E.; Jahn, M.T.; Araujo, E.B.N.; Pearson, C.F.; Wang, X.; Pankhurst, L.; Cunrath, O.; Foster, K.R. Microbiome Diversity Protects against Pathogens by Nutrient Blocking. Science 2023, 382, eadj3502. [Google Scholar] [CrossRef]
  180. Junaid, M.; Lu, H.; Li, Y.; Liu, Y.; Din, A.U.; Qi, Z.; Xiong, Y.; Yan, J. Novel Synergistic Probiotic Intervention: Transcriptomic and Metabolomic Analysis Reveals Ameliorative Effects on Immunity, Gut Barrier, and Metabolism of Mice During Salmonella typhimurium Infection. Genes 2024, 15, 435. [Google Scholar] [CrossRef]
  181. Heumel, S.; de Rezende Rodovalho, V.; Urien, C.; Specque, F.; Brito Rodrigues, P.; Robil, C.; Delval, L.; Sencio, V.; Descat, A.; Deruyter, L.; et al. Shotgun Metagenomics and Systemic Targeted Metabolomics Highlight Indo Le-3-Propionic Acid as a Protective Gut Microbial Metabolite against Influenza Infection. Gut Microbes 2024, 16, 2325067. [Google Scholar] [CrossRef]
  182. Wang, Q.; Huang, H.; Yang, Y.; Yang, X.; Li, X.; Zhong, W.; Wen, B.; He, F.; Li, J. Reinventing Gut Health: Leveraging Dietary Bioactive Compounds for the Prevention and Treatment of Diseases. Front. Nutr. 2024, 11, 1491821. [Google Scholar] [CrossRef]
  183. Fu, Y.; Lyu, J.; Wang, S. The Role of Intestinal Microbes on Intestinal Barrier Function and Host Immunity from a Metabolite Perspective. Front. Immunol. 2023, 14, 1277102. [Google Scholar] [CrossRef] [PubMed]
  184. de Souza Vilela, J.; Kheravii, S.K.; Sharma Bajagai, Y.; Kolakshyapati, M.; Zimazile Sibanda, T.; Wu, S.-B.; Andrew, N.R.; Ruhnke, I. Inclusion of up to 20% Black Soldier Fly Larvae Meal in Broiler Chicken Diet Has a Minor Effect on Caecal Microbiota. PeerJ 2023, 11, e15857. [Google Scholar] [CrossRef] [PubMed]
  185. Ndotono, E.W.; Khamis, F.M.; Bargul, J.L.; Tanga, C.M. Insights into the Gut Microbial Communities of Broiler Chicken Fed Black Soldier Fly Larvae-Desmodium-Based Meal as a Dietary Protein Source. Microorganisms 2022, 10, 1351. [Google Scholar] [CrossRef]
  186. Leeper, A.; Benhaïm, D.; Smárason, B.Ö.; Knobloch, S.; Òmarsson, K.L.; Bonnafoux, T.; Pipan, M.; Koppe, W.; Björnsdóttir, R.; Øverland, M. Feeding Black Soldier Fly Larvae (Hermetia illucens) Reared on Organic Rest Streams Alters Gut Characteristics of Atlantic Salmon (Salmo salar). J. Insects Food Feed 2022, 8, 1355–1372. [Google Scholar] [CrossRef]
  187. Elhag, O.; Zhang, Y.; Xiao, X.; Cai, M.; Zheng, L.; Jordan, H.R.; Tomberlin, J.K.; Huang, F.; Yu, Z.; Zhang, J. Inhibition of Zoonotic Pathogens Naturally Found in Pig Manure by Black Soldier Fly Larvae and Their Intestine Bacteria. Insects 2022, 13, 66. [Google Scholar] [CrossRef]
  188. Huang, C.; Hernandez, C.E.; Wall, H.; Tahamtani, F.M.; Ivarsson, E.; Sun, L. Live Black Soldier Fly (Hermetia illucens) Larvae in Feed for Laying Hens: Effects on Hen Gut Microbiota and Behavior. Poult. Sci. 2024, 103, 103429. [Google Scholar] [CrossRef]
  189. Xia, J.; Ge, C.; Yao, H. Antimicrobial Peptides from Black Soldier Fly (Hermetia illucens) as Potential Antimicrobial Factors Representing an Alternative to Antibiotics in Livestock Farming. Animals 2021, 11, 1937. [Google Scholar] [CrossRef]
  190. Hurtado-Ribeira, R.; Villanueva-Bermejo, D.; García-Risco, M.R.; Hernández, M.D.; Sánchez-Muros, M.J.; Fornari, T.; Vázquez, L.; Martin, D. Evaluation of the Interrelated Effects of Slaughtering, Drying, and De-Fatting Methods on the Composition and Properties of Black Soldier Fly (Hermetia illucens) Larvae Fat. Curr. Res. Food Sci. 2023, 7, 100633. [Google Scholar] [CrossRef]
  191. Lehmad, M.; Nomeir, B.; Hidra, N.; El Hachimi, Y.; Abdenouri, N. Impact of Hybrid Drying on the Drying Kinetics, Nutritional, Physicochemical, Functional, Structural, and Thermal Properties of Black Soldier Fly Larvae. J. Insects Food Feed 2025, 1, 1–26. [Google Scholar] [CrossRef]
  192. Cruz, V.A.; Ferreira, N.J.; Le Roux, E.; Destandau, E.; de Oliveira, A.L. Intensification of the Sfe Using Ethanol as a Cosolvent and Integration of the Sfe Process with Sc-Co2 Followed by Ple Using Pressurized Ethanol of Black Soldier Fly (Hermetia illucens L.) Larvae Meal-Extract Yield and Characterization. Foods 2024, 13, 1620. [Google Scholar] [CrossRef]
  193. Pan, J.; Xu, H.; Dabbour, M.; Kumah Mintah, B.; Chen, W.; Yang, F.; Zhang, Z.; Cheng, Y.; Dai, C.; He, R.; et al. Effect of Alkaline Ph-Shifting Process on Extraction Rate, Structural, and Functional Properties of Black Soldier Fly (Hermetia illucens) Larvae Protein. LWT 2023, 185, 115180. [Google Scholar] [CrossRef]
  194. Bogusz, R.; Bryś, J.; Onopiuk, A.; Pobiega, K.; Tomczak, A.; Kowalczewski, P.Ł.; Rybak, K.; Nowacka, M. The Impact of Drying Methods on the Quality of Blanched Yellow Mealworm (Tenebrio molitor L.). Larvae. Mol. 2024, 29, 3679. [Google Scholar] [CrossRef]
  195. Hurtado-Ribeira, R.; Hernández, D.M.; Villanueva-Bermejo, D.; García-Risco, M.R.; Hernández, M.D.; Vázquez, L.; Fornari, T.; Martin, D. The Interaction of Slaughtering, Drying, and Defatting Methods Differently Affects Oxidative Quality of the Fat from Black Soldier Fly (Hermetia illucens) Larvae. Insects 2023, 14, 368. [Google Scholar] [CrossRef]
  196. Keil, C.; Grebenteuch, S.; Kröncke, N.; Kulow, F.; Pfeif, S.; Kanzler, C.; Rohn, S.; Boeck, G.; Benning, R.; Haase, H. Systematic Studies on the Antioxidant Capacity and Volatile Compound Profile of Yellow Mealworm Larvae (T. Molit. L.) Under Different Drying Regimes. Insects 2022, 13, 166. [Google Scholar] [CrossRef] [PubMed]
  197. Mohd Zaini, N.S.; Lim, E.J.; Ahmad, N.H.; Gengatharan, A.; Wan-Mohtar, W.A.A.Q.I.; Abd Rahim, M.H. The Review of Cooking, Drying, and Green Extraction Methods on General Nutritional Properties of Mealworms and Locusts. Food Bioprocess Technol. 2023, 16, 1904–1918. [Google Scholar] [CrossRef] [PubMed]
  198. Ashaolu, T.J.; Le, T.-D.; Suttikhana, I. Stability and Bioactivity of Peptides in Food Matrices Based on Processing Conditions. Food Res. Int. 2023, 168, 112786. [Google Scholar] [CrossRef] [PubMed]
  199. Rivero-Pino, F. Bioactive Food-Derived Peptides for Functional Nutrition: Effect of Fortification, Processing and Storage on Peptide Stability and Bioactivity within Food Matrices. Food Chem. 2023, 406, 135046. [Google Scholar] [CrossRef]
  200. Queiroz, L.S.; Casanova, F.; Feyissa, A.H.; Jessen, F.; Ajalloueian, F.; Perrone, I.T.; de Carvalho, A.F.; Mohammadifar, M.A.; Jacobsen, C.; Yesiltas, B. Physical and Oxidative Stability of Low-Fat Fish Oil-in-Water Emulsions Stabilized with Black Soldier Fly (Hermetia illucens) Larvae Protein Concentrate. Foods 2021, 10, 2977. [Google Scholar] [CrossRef]
  201. Wang, J.; Jousse, M.; Jayakumar, J.; Fernández-Arteaga, A.; de Lamo-Castellví, S.; Ferrando, M.; Güell, C. Black Soldier Fly (Hermetia illucens) Protein Concentrates as a Sustainable Source to Stabilize O/W Emulsions Produced by a Low-Energy High-Throughput Emulsification Technology. Foods 2021, 10, 1048. [Google Scholar] [CrossRef]
  202. Bessa, L.W.; Pieterse, E.; Marais, J.; Hoffman, L.C. Techno-Functional Properties of Black Soldier Fly (Hermetia illucens) Larvae. J. Insects Food Feed 2022, 8, 1047–1060. [Google Scholar] [CrossRef]
  203. Aguilar-Toalá, J.E.; Quintanar-Guerrero, D.; Liceaga, A.M.; Zambrano-Zaragoza, M.L. Encapsulation of Bioactive Peptides: A Strategy to Improve the Stability, Protect the Nutraceutical Bioactivity and Support Their Food Applications. RSC Adv. 2022, 12, 6449–6458. [Google Scholar] [CrossRef] [PubMed]
  204. Pateiro, M.; Gómez, B.; Munekata, P.E.S.; Barba, F.J.; Putnik, P.; Kovačević, D.B.; Lorenzo, J.M. Nanoencapsulation of Promising Bioactive Compounds to Improve Their Absorption, Stability, Functionality and the Appearance of the Final Food Products. Molecules 2021, 26, 1547. [Google Scholar] [CrossRef] [PubMed]
  205. Bessa, L.W.; Pieterse, E.; Marais, J.; Hoffman, L.C. Why for Feed and Not for Human Consumption? The Black Soldier Fly Larvae. Comp. Rev. Food Sci. Food Safe 2020, 19, 2747–2763. [Google Scholar] [CrossRef] [PubMed]
  206. Muslykhah, U.; Phupaboon, S.; Suriyapha, C.; Matra, M.; Wanapat, M. Encapsulation of Protein-Based Bioactive from Black Soldier Fly for Ruminant Feeding. J. Agric. Food Res. 2024, 18, 101325. [Google Scholar] [CrossRef]
  207. Lehmad, M.; Nomeir, B.; Hidra, N.; Lhomme, P.; El Hachimi, Y.; Abdenouri, N. Comparative Analysis of Drying Technologies for Black Soldier Fly (Hermetia illucens) Larvae: Multidimensional Assessment. J. Insects Food Feed 2025, 1, 1–21. [Google Scholar] [CrossRef]
  208. Lehmad, M.; Hidra, N.; Lhomme, P.; Mghazli, S.; Hachimi, Y.E.; Abdenouri, N. Environmental, Economic and Quality Assessment of Hybrid Solar-Electric Drying of Black Soldier Fly (Hermetia illucens) Larvae. Renew. Energy 2024, 226, 120401. [Google Scholar] [CrossRef]
  209. Chakawa, D.; Goosen, N. Protein Recovery from Black Soldier Fly Larvae Using Enzymatic Hydrolysis and Alkaline Extraction. J. Insects Food Feed 2025, 1, 1–22. [Google Scholar] [CrossRef]
  210. Niyonsaba, H.; Höhler, J.; Kooistra, J.; Van der Fels-Klerx, H.; Meuwissen, M. Profitability of Insect Farms. J. Insects Food Feed 2021, 7, 923–934. [Google Scholar] [CrossRef]
  211. Leipertz, M.; Hogeveen, H.; Saatkamp, H. Economic Supply Chain Modelling of Industrial Insect Production in the Netherlands. J. Insects Food Feed 2024, 10, 1361–1385. [Google Scholar] [CrossRef]
  212. Bessa, L.W.; Pieterse, E.; Marais, J.; Dhanani, K.; Hoffman, L.C. Food Safety of Consuming Black Soldier Fly (Hermetia illucens) Larvae: Microbial, Heavy Metal and Cross-Reactive Allergen Risks. Foods 2021, 10, 1934. [Google Scholar] [CrossRef]
  213. Kolakowski, B.M.; Johaniuk, K.; Zhang, H.; Yamamoto, E. Analysis of Microbiological and Chemical Hazards in Edible Insects Available to Canadian Consumers. J. Food Prot. 2021, 84, 1575–1581. [Google Scholar] [CrossRef]
  214. Ribeiro, J.C.; Sousa-Pinto, B.; Fonseca, J.; Fonseca, S.C.; Cunha, L.M. Edible Insects and Food Safety: Allergy. J. Insects Food Feed 2021, 7, 833–847. [Google Scholar] [CrossRef]
  215. Yang, J.; Zhou, S.; Kuang, H.; Tang, C.; Song, J. Edible Insects as Ingredients in Food Products: Nutrition, Functional Properties, Allergenicity of Insect Proteins, and Processing Modifications. Crit. Rev. Food Sci. Nutr. 2023, 64, 10361–10383. [Google Scholar] [CrossRef]
  216. De Marchi, L.; Wangorsch, A.; Zoccatelli, G. Allergens from Edible Insects: Cross-Reactivity and Effects of Processing. Curr. Allergy Asthma Rep. 2021, 21, 35. [Google Scholar] [CrossRef]
  217. Cappelli, A.; Cini, E.; Lorini, C.; Oliva, N.; Bonaccorsi, G. Insects as Food: A Review on Risks Assessments of Tenebrionidae and Gryllidae in Relation to a First Machines and Plants Development. Food Control 2020, 108, 106877. [Google Scholar] [CrossRef]
  218. Kooh, P.; Jury, V.; Laurent, S.; Audiat-Perrin, F.; Sanaa, M.; Tesson, V.; Federighi, M.; Boué, G. Control of Biological Hazards in Insect Processing: Application of Hac. Foods 2020, 9, 1528. [Google Scholar] [CrossRef] [PubMed]
  219. Meyer, A.M.; Meijer, N.; Hoek-van den Hil, E.F.; van der Fels-Klerx, H.J. Chemical Food Safety Hazards of Insects Reared for Food and Feed. J. Insects Food Feed 2021, 7, 823–831. [Google Scholar] [CrossRef]
  220. Ojha, S.; Bußler, S.; Psarianos, M.; Rossi, G.; Schlüter, O.K. Edible Insect Processing Pathways and Implementation of Emerging Technologies. J. Insects Food Feed 2021, 7, 877–900. [Google Scholar] [CrossRef]
  221. Yan, X.; Laurent, S.; Federighi, M.; Boué, G.; Jury, V. Processing Edible Insects into Powders: A Review of Available Processes and Potential Microbial Inactivation Methods. J. Insects Food Feed 2022, 9, 325–338. [Google Scholar] [CrossRef]
  222. Vandeweyer, D.; De Smet, J.; Van Looveren, N.; Van Campenhout, L. Biological Contaminants in Insects as Food and Feed. J. Insects Food Feed 2021, 7, 807–822. [Google Scholar] [CrossRef]
  223. IPIFF. Insects as Feed Eu Legislation—Aquaculture, Poultry & Pig Species; IPIFF: Brussels, Belgium, 2025. [Google Scholar]
  224. Imathiu, S. Benefits and Food Safety Concerns Associated with Consumption of Edibl E Insects. NFS J. 2020, 18, 1–11. [Google Scholar] [CrossRef]
  225. Żuk-Gołaszewska, K.; Gałęcki, R.; Obremski, K.; Smetana, S.; Figiel, S.; Gołaszewski, J. Edible Insect Farming in the Context of the Eu Regulations and Marketing-an Overview. Insects 2022, 13, 446. [Google Scholar] [CrossRef]
  226. Siddiqui, S.A.; Tettey, E.; Yunusa, B.M.; Ngah, N.; Debrah, S.K.; Yang, X.; Fernando, I.; Povetkin, S.N.; Shah, M.A. Legal Situation and Consumer Acceptance of Insects Being Eaten as Human Food in Different Nations across the World–a Comprehensive Review. Comp. Rev. Food Sci. Food Safe 2023, 22, 4786–4830. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The life cycle of the BSF (H. illucens) from egg to adult, illustrating all developmental stages relevant to insect farming and bioactive compound production (created in BioRender BSF life cycle|BioRender, 2025).
Figure 1. The life cycle of the BSF (H. illucens) from egg to adult, illustrating all developmental stages relevant to insect farming and bioactive compound production (created in BioRender BSF life cycle|BioRender, 2025).
Foods 14 03654 g001
Figure 2. The essential bioactive compounds obtained from BSFL and their diverse functions within the gut ecosystem, such as AMPs, chitin-derived prebiotics, crucial amino acids, and bioactive lipids, which together promote gut health by modulating microbiota, enhancing barrier function, and regulating the immune system.
Figure 2. The essential bioactive compounds obtained from BSFL and their diverse functions within the gut ecosystem, such as AMPs, chitin-derived prebiotics, crucial amino acids, and bioactive lipids, which together promote gut health by modulating microbiota, enhancing barrier function, and regulating the immune system.
Foods 14 03654 g002
Figure 3. Mechanisms of COS in Modulating Gut Health and Inflammatory Responses.
Figure 3. Mechanisms of COS in Modulating Gut Health and Inflammatory Responses.
Foods 14 03654 g003
Figure 4. Bioactive functions of BSFL peptides in gut health.
Figure 4. Bioactive functions of BSFL peptides in gut health.
Foods 14 03654 g004
Figure 5. BSF-derived bioactive peptides provide multifaceted protection for gastrointestinal health through coordinated antimicrobial, immunomodulatory, and antioxidant mechanisms as described by Koutsos et al. [166], Created in BioRender GIT protection|(BioRender, 2025).
Figure 5. BSF-derived bioactive peptides provide multifaceted protection for gastrointestinal health through coordinated antimicrobial, immunomodulatory, and antioxidant mechanisms as described by Koutsos et al. [166], Created in BioRender GIT protection|(BioRender, 2025).
Foods 14 03654 g005
Figure 6. Schematic representation of BSFL-derived bioactive peptides within the gut ecosystem, illustrating their antimicrobial action against pathogens, selective promotion of probiotic microbes through nutrient and competitive modulation, upregulation of epithelial and subepithelial tight junction proteins to reinforce barrier integrity, and immune receptor interactions on dendritic cells and macrophages that skew cytokine profiles toward anti-inflammatory responses, collectively driving microbial balance and host resilience.
Figure 6. Schematic representation of BSFL-derived bioactive peptides within the gut ecosystem, illustrating their antimicrobial action against pathogens, selective promotion of probiotic microbes through nutrient and competitive modulation, upregulation of epithelial and subepithelial tight junction proteins to reinforce barrier integrity, and immune receptor interactions on dendritic cells and macrophages that skew cytokine profiles toward anti-inflammatory responses, collectively driving microbial balance and host resilience.
Foods 14 03654 g006
Figure 7. BSF-derived chitin and peptides synergistically shape the gut microbiome: chitin scaffolds support adhesion; COS nourish beneficial commensals; AMPs eliminate pathogens; and immunomodulatory peptides engage TLR2/4 to fine-tune mucosal immunity and promote probiotic resilience.
Figure 7. BSF-derived chitin and peptides synergistically shape the gut microbiome: chitin scaffolds support adhesion; COS nourish beneficial commensals; AMPs eliminate pathogens; and immunomodulatory peptides engage TLR2/4 to fine-tune mucosal immunity and promote probiotic resilience.
Foods 14 03654 g007
Table 1. Amino acid composition (g 100 g−1 protein) of BSFL protein analyzed by acid hydrolysis and HPLC, compared with beef and soy.
Table 1. Amino acid composition (g 100 g−1 protein) of BSFL protein analyzed by acid hydrolysis and HPLC, compared with beef and soy.
Amino AcidsBSFLBeefSoy
Lysine6.26.05.7
Leucine8.57.97.0
Valine5.75.44.8
Methionine1.91.61.3
Cysteine1.10.40.1
Threonine4.34.13.8
Phenylalanine4.74.64.5
Tryptophan1.31.21.1
Table 2. Major fatty acids in BSFL lipid fraction (percentage of total fatty acids).
Table 2. Major fatty acids in BSFL lipid fraction (percentage of total fatty acids).
Fatty AcidAbundance (%)Health Implications
Lauric acid36Antimicrobial, supports gut barrier
Palmitic acid12Energy source, structural lipid component
Oleic acid28Anti-inflammatory, cardioprotective
Linoleic acid18Essential omega-6, supports cell signaling
Stearic acid6Neutral effect on serum cholesterol
Table 3. Summary of chitin/chitosan’s physicochemical properties and gut health benefits.
Table 3. Summary of chitin/chitosan’s physicochemical properties and gut health benefits.
PropertyChitinChitosanGut Health Role
Degree of acetylation~90%~50%Determines solubility and fermentability
Molecular weight (kDa)100–20050–100Influences prebiotic efficacy
SolubilityInsoluble in waterSoluble in acidic solutionsEnables selective microbial fermentation
Biological activityStructural supportAntimicrobial, prebioticModulates microbiota and strengthens the mucosal barrier
Table 4. BSFL bioactives to microbial, metabolic, receptor, and effect pathways.
Table 4. BSFL bioactives to microbial, metabolic, receptor, and effect pathways.
BSFL CompoundsMicrobial TargetMetabolitesHost ReceptorEffect Ref.
BSF-COSF. prausnitziiButrate CPR109AEnhanced barrier[105]
BSF-AMPILactobacillusLactateTLR2Reduced TNF-α[154]
BSF-CPPB. lungumAcetateGPR43Anti-inflammatory[155]
BSF-COS: BSF–chitosan oligosaccharides, BSF-AMPI: BSF–antimicrobial peptide I, BSF-CPP: BSF-derived cell-penetrating peptides.
Table 5. Outcomes of BSF-Derived Compound Supplementation Across Models [117,189].
Table 5. Outcomes of BSF-Derived Compound Supplementation Across Models [117,189].
ModelBSF-Derived CompoundKey Outcomes
Broiler chickens5% COS20% increase in villus-to-crypt ratio; significant reduction in cecal Enterobacteriaceae counts
Shrimp aquacultureAMP-enriched BSF protein hydrolysate1.5 log reduction in Vibrio spp. loads; 15% increase in survival rate
Pilot human trial4 g day−1 COS for six weeks30% increase in stool Bifidobacterium abundance; improved bowel regularity
Table 6. A comprehensive comparison of primary processing methods for BSFL bioactive extraction, analyzing cost structures, energy consumption, and bioactivity retention to guide industrial implementation decisions.
Table 6. A comprehensive comparison of primary processing methods for BSFL bioactive extraction, analyzing cost structures, energy consumption, and bioactivity retention to guide industrial implementation decisions.
Processing MethodEnergy Consumption (kWh/kg)Processing TimeCapital InvestmentBioactivity RetentionOperating CostsIndustrial SuitabilityRegional Considerations
Hot Air Drying (60–70 °C)1.5–2.518–24 hLow ($50–100 K for medium scale)Moderate (60–70% peptide retention)Low (labor, energy)Highly established technologySuitable for all regions; limited climate control needed
Freeze-Drying5–10 (4–10x—higher than HAD)24–48 hVery High ($500 K–2 M for industrial units)Excellent (>90% peptide/chitin retention)Very High (energy, maintenance)Limited—niche high-value applicationsMost suitable for temperate/cold climates; prohibitive in energy-scarce regions
Enzymatic Extraction (Chitin/Peptides)0.5–1.2 (moderate heating)4–12 hModerate–High ($200–500 K)Excellent (85–95% targeted bioactives)Moderate–High (enzyme costs, pH control)Moderate—requires technical expertiseViable in regions with enzyme availability and trained personnel
Supercritical CO2 Extraction (Lipids)2–42–6 hVery High ($800 K–3 M)Excellent (>95% lipid quality)High (CO2 pressure maintenance)Moderate—for high-value lipid productsRequires stable infrastructure; suitable for developed markets
Combined Hot Air + Enzymatic Processing2–312–20 hModerate ($150–350 K)Good (75–85% overall retention)ModerateHighly balanced cost–benefitOptimal for developing regions; combines affordability with quality
Energy consumption values based on processing 1 kg dry matter of BSFL, Capital investment estimates for medium-scale facilities (500–1000 kg day−1 capacity), Bioactivity retention relative to fresh larvae baseline, Operating costs include labor, energy, consumables, and maintenance.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alhasyani, T.; Ebeid, T.; Ghonimy, M.; Alharbi, S.; Hassan, M.F.Y.; Jarallah, A.; Alkhurayji, M.; Abdellatif, A.A.H.; Barakat, H. Harnessing Edible Insect Bioactives for Gut Health: A Comprehensive Review on Chitin-Derived Prebiotics and Peptidomic Insights from the Black Soldier Fly. Foods 2025, 14, 3654. https://doi.org/10.3390/foods14213654

AMA Style

Alhasyani T, Ebeid T, Ghonimy M, Alharbi S, Hassan MFY, Jarallah A, Alkhurayji M, Abdellatif AAH, Barakat H. Harnessing Edible Insect Bioactives for Gut Health: A Comprehensive Review on Chitin-Derived Prebiotics and Peptidomic Insights from the Black Soldier Fly. Foods. 2025; 14(21):3654. https://doi.org/10.3390/foods14213654

Chicago/Turabian Style

Alhasyani, Thamer, Tarek Ebeid, Mohamed Ghonimy, Saif Alharbi, Mohamed F. Y. Hassan, Abdullah Jarallah, Mohammed Alkhurayji, Ahmed A. H. Abdellatif, and Hassan Barakat. 2025. "Harnessing Edible Insect Bioactives for Gut Health: A Comprehensive Review on Chitin-Derived Prebiotics and Peptidomic Insights from the Black Soldier Fly" Foods 14, no. 21: 3654. https://doi.org/10.3390/foods14213654

APA Style

Alhasyani, T., Ebeid, T., Ghonimy, M., Alharbi, S., Hassan, M. F. Y., Jarallah, A., Alkhurayji, M., Abdellatif, A. A. H., & Barakat, H. (2025). Harnessing Edible Insect Bioactives for Gut Health: A Comprehensive Review on Chitin-Derived Prebiotics and Peptidomic Insights from the Black Soldier Fly. Foods, 14(21), 3654. https://doi.org/10.3390/foods14213654

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

Article Metrics

Back to TopTop