Next Article in Journal
Effective and Stable Senomorphic Apigenin Delivery System Obtained by Supercritical Carbon Dioxide Processing
Previous Article in Journal
Sexually Transmitted Infections: Usefulness of Molecular Methods for Microorganism Detection in Stored Sexual Assault Samples
Previous Article in Special Issue
The Probiotic Strain Clostridium butyricum TO-A Produces Butyrate by Utilizing Lactate and Acetate
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release

1
Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Ministry of Education), School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
2
Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(17), 8119; https://doi.org/10.3390/ijms26178119
Submission received: 14 July 2025 / Revised: 3 August 2025 / Accepted: 20 August 2025 / Published: 22 August 2025

Abstract

Bioactive peptides encrypted in bovine β-casein display diverse physiological functions, including antihypertensive, antioxidative, antimicrobial, and immunomodulatory activities. These peptides are normally released during gastrointestinal digestion or microbial fermentation, especially by proteolytic systems of lactic acid bacteria (LAB). However, peptide yields vary widely among LAB strains, reflecting strain-specific protease repertoires. To overcome these limitations, the scientific goal of this study is to provide a comprehensive synthesis of how synthetic biology, molecular biotechnology, and systems-level approaches can be leveraged to enhance the targeted discovery and production of β-casein-derived bioactive peptides. Genome engineering tools such as clustered regularly interspaced short palindromic repeats associated system (CRISPR/Cas) systems have been applied to modulate gene expression and metabolic flux in LAB, while inducible expression platforms allow on-demand peptide production. Additionally, cell-free systems based on LAB lysates further provide rapid prototyping for high-throughput screening. Finally, multi-omics approaches, including genomics, transcriptomics, proteomics, and metabolomics, further help pinpoint regulatory bottlenecks and facilitate rational strain optimization. This review provides a comprehensive overview of bioactive peptides derived from bovine β-casein and highlights recent progress in LAB-based strategies—both natural and engineered—for their efficient release. These advances pave the way for developing next-generation functional fermented foods enriched with targeted bioactivities.

Graphical Abstract

1. Introduction

Fermented dairy products are produced through the fermentation of milk. Buttermilk, cream, butter, yogurt, and cheese are among the most well-known examples [1]. Milk remains one of the most widely consumed foods globally, as reported in OECD–FAO Agricultural Outlook 2024–2033, world milk production—around 81% from cows, 15% from buffaloes, and 4% from goats, sheep, and camels—is forecast to grow at 1.6% per year over the next decade, reaching 1085 million tons by 2033 [2]. Dairy products exhibit remarkable diversity due to the complex composition of milk and the wide range of microorganisms that can thrive in it. Dairy products are a valuable source of essential nutrients, including high-quality proteins, omega-3 fatty acids, oleic acid, conjugated linoleic acid, vitamins (e.g., A, D, B12), minerals (e.g., calcium, phosphorus, zinc), and bioactive peptides [3]. In addition to their nutritional benefits, products such as yogurt and cheese offer appealing organoleptic qualities—namely, desirable texture and flavor—that enhance the human daily diet [4] (Figure 1a).
The proteins in milk are broadly classified into two main groups, caseins and whey proteins. Approximately 80% of the total protein content in bovine milk is composed of caseins, with the rest being whey proteins. According to their primary amino acid sequence homology, bovine caseins can be divided into four subtypes: αs1-casein, αs2-casein, β-casein, and κ-casein (Figure 1b). The whey protein fraction mainly consists of α-lactalbumin, β-lactoglobulin, lactoferrin, immunoglobulins, serum albumin, and the secretory component [5]. Casein can be utilized in its native form or processed into caseinates and peptides. In particular, hydrolysates of β-Casein have drawn increasing attention for their potential applications in functional foods, owing to the bioactive peptides they release [6]. Typically, these peptides consist of 2 to 20 amino acids (AAs) [7]. Within the precursor protein sequence, they are inactive until liberated by (i) digestion in the gastrointestinal tract [7], (ii) microbial fermentation involving proteolytic microorganisms [8], or (iii) enzymatic hydrolysis using plants or microorganisms [9]. Once released, these peptides may exert various beneficial effects on the cardiovascular, immune, nervous, and/or human digestive system [10] (Figure 1c).
Among the four casein subtypes, β-casein stands out as a particularly effective precursor for generating bioactive peptides due to several favorable structural characteristics. Its primary sequence is particularly rich in proline, glutamine, and phosphorylated serine residues, which are commonly found in bioactive motifs associated with antioxidant, antihypertensive, immunomodulatory, or mineral-binding properties [11]. Moreover, β-casein exhibits a relatively open and flexible tertiary structure due to its high proportion of hydrophilic residues, making it more accessible to proteolytic cleavage compared to other milk proteins. These features facilitate the targeted release of functional peptides, both in vivo and under controlled fermentation processes [12,13].
The enzymatic accessibility of β-casein makes it an ideal substrate for controlled proteolysis during dairy fermentation. In this context, lactic acid bacteria (LAB) play a central role in the proteolytic breakdown of caseins during dairy fermentation. LAB possess a sophisticated proteolytic system composed of cell-envelope proteinases, intracellular peptidases, and peptide transporters, which allow LAB to hydrolyze caseins into small peptides and free amino acids [14,15]. This natural capacity not only contributes to the development of flavor and texture in fermented dairy products but also provides a biological route for releasing health-promoting peptides from milk proteins. However, spontaneous peptide production by wild-type LAB is often inefficient, strain-dependent, and rarely targeted toward sequences with specific bioactivities.
To overcome these limitations, a suite of molecular strategies—including genetic engineering, synthetic biology, and omics-guided strain optimization—has been increasingly employed to enhance the proteolytic capacity and peptide specificity of LAB [16]. These strategies enable the precise modification of key enzymes, transport systems, and regulatory elements involved in casein hydrolysis. As a result, engineered LAB strains are now emerging as powerful biocatalysts for the tailored production of bioactive peptides from β-casein in a controlled, food-grade manner. The integration of such molecular tools into LAB-based systems represents a promising avenue for developing next-generation functional dairy ingredients and fermented nutraceuticals.
Figure 1. (a) Representative milk-derived (dairy) products. Clockwise, starting at noon: cheese, sour cream, soured or buttermilk, ice cream, butter, yogurt. In the center: the raw starting material, milk. (b) Pie chart showing the milk protein composition and percentage ratios; the stacked bar chart displays the casein composition and percentage distribution [17]. (c) Bovine β-casein-derived peptide bioactivities benefiting human health.
Figure 1. (a) Representative milk-derived (dairy) products. Clockwise, starting at noon: cheese, sour cream, soured or buttermilk, ice cream, butter, yogurt. In the center: the raw starting material, milk. (b) Pie chart showing the milk protein composition and percentage ratios; the stacked bar chart displays the casein composition and percentage distribution [17]. (c) Bovine β-casein-derived peptide bioactivities benefiting human health.
Ijms 26 08119 g001

2. Functional Diversity of Bioactive Peptides from Bovine β-Casein

Bovine β-casein is a rich source of encrypted peptides with diverse biological activities, which are released during digestion or microbial fermentation. These peptides can target major physiological systems, including the gastrointestinal, cardiovascular, nervous, and immune systems (Figure 1c) [6,11]. The functional diversity of β-casein-derived peptides reflects differences in their amino acid composition, structure, and susceptibility to proteolytic cleavage. This section summarizes the major classes of bioactive peptides derived from bovine β-casein and their corresponding physiological roles.

2.1. Caseinophosphopeptides (CPPs): Mineral-Binding and Immunomodulatory Peptides

Among the different functional peptides, caseinophosphopeptides (CPPs) are well-characterized for their mineral-binding capabilities. Derived from casein, CPPs are phosphorylated peptides capable of binding and solubilizing minerals, including zinc and calcium [18]. Phosphate groups are attached to serine residues through monoester bonds, with bovine β-casein containing five phosphorylatable serines. Two major CPPs identified from β-casein are fragment 16–40 (f(16–40)) and fragment 16–43 (f(16–43)) [19,20,21,22]. In rats ingesting β-casein, the peptide f(16–40) has been detected in the small intestine, where it elevates soluble calcium concentrations in the distal ileum and facilitates passive calcium absorption [20]. In addition, f(16–40) promotes the proliferation of mouse spleen cells and rabbit Peyer’s patch cells [22]. Similarly, the peptide f(16–43) demonstrates immune-stimulatory properties, including stimulation of immunoglobulin production and cytokine secretion in human immune cells, as well as increased proliferation and interleukin-6 (IL-6) expression in B lymphocytes [19,20].

2.2. β-Casomorphins (β-CMs): Opioid-like Peptides

Another major category of β-casein-derived peptides is β-casomorphins (β-CMs), which possess opioid-like activity. These peptides act primarily as μ-opioid receptor agonists and have been linked to various biological functions [23,24]. The heptapeptide β-CM7, the first exogenous opioid peptide discovered [25], not only exhibits opioid activity but also enhances mucin production in the gastrointestinal tract through the µ-opioid pathway [26]. It also shows hypoglycemic effects in diabetic rats by increasing plasma insulin levels and shows increased angiotensin-converting enzyme (ACE) inhibitory activity [27,28]. The release of β-CM7 varies among milk types, with A1 milk generating more than A2, which may explain differential effects on gut and neurological health [29]. Recent studies have further highlighted the role of β-CM7 in gut–brain axis modulation by affecting gut permeability, inflammation, and central nervous system signaling [30]. Another peptide, β-CM5, is noted as the strongest opioid agonist among β-CMs and exhibits high resistance to proteolytic digestion. It has been shown to alter gut motility in rats [31,32] and stimulate neurite outgrowth in neuronal cell lines [33], as well as improve learning and memory disturbances related to cholinergic dysfunction [34].

2.3. DPP-IV Inhibitory Peptides: Metabolic Regulation

In addition to opioid activity, certain peptides derived from β-casein have shown potential in metabolic regulation [35]. Of particular interest are peptides that inhibit dipeptidyl peptidase-4 (DPP-IV), an enzyme that degrades incretin hormones like glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), which are essential for insulin secretion [36]. To date, 21 DPP-IV inhibitory peptides have been identified from bovine β-casein, ranging from 3 to 9 amino acids with IC50 values between 46 and 1300 µM. About half of these peptides originate from fragment f(75–95), and many possess a characteristic proline at the second position from the N-terminus, suggesting a potential structural motif underlying DPP-IV-inhibitory activity. Tripeptides conforming to the X-Pro-X motif function as competitive inhibitors of DPP-IV, for instance, LPL and LPQ display IC50 values of 241.4 µM and 82 µM, respectively. Nonetheless, detecting such small peptides using mass spectrometry (MS) remains challenging; advances in Liquid Chromatography–Mass Spectrometry (LC-MS) techniques are facilitating the discovery of additional short DPP-IV inhibitors.

2.4. Antimicrobial Peptides (AMPs)

Beyond metabolic regulation, β-casein-derived peptides also exhibit antimicrobial activity [37]. Antimicrobial peptides (AMPs) are generally short, positively charged peptides capable of disrupting microbial membranes. Such peptides from food sources are of interest due to their safety and natural origin [38]. Several antimicrobial peptides have been identified from bovine caseins following enzymatic hydrolysis. For example, two peptides, casecidin-15 and casecidin-17, derived from the C-terminal region of β-casein, showed inhibitory effects against Escherichia coli and exhibited amphiphilic structures that support their antimicrobial function [39]. Furthermore, plasmin digestion of β-casein revealed eight antibacterial peptides, notably βC8(VKEAMAPK), βC12(EAMAPKHK), and βC14(VLPVPQKAVPYPQR), which were highly active against Gram-positive bacteria and characterized by their hydrophobicity and C-terminal basic residues [40]. These findings highlight β-casein as a promising source of natural antimicrobial agents, with potential applications in enhancing the safety and functionality of dairy-based products.

2.5. Immunomodulatory Peptides

Several β-casein peptides function as immunomodulators, modulating immune responses without the side effects of synthetic drugs like cyclosporine and glucocorticoids [41], which provides a promising strategy for dietary modulation of the immune response [42,43]. These peptides can enhance lymphocyte proliferation, regulate cytokine expression, and attenuate inflammatory responses. For instance, β-casein-derived peptides such as QEPVL have been demonstrated to alleviate inflammation and restore gut microbiota balance in DSS-induced colitis models, likely through the inhibition of the NLRP3/NF-κB signaling pathway [44].

2.6. ACE-Inhibitory Peptides: Antihypertensive Potential

Among the most studied functionalities of β-casein-derived peptides is their ability to inhibit ACE, a key enzyme in blood pressure regulation [45,46,47]. Therefore, ACE inhibitors are helpful agents in the therapeutic treatment of hypertension. Synthetic ACE inhibitors such as captopril and enalapril are effective but often have adverse side effects, prompting interest in natural alternatives [48]. ACE-inhibitory activity is exhibited by more than half of the bioactive peptides derived from bovine β-casein (Figure 2a). About 56% of these peptides display IC50 values below 100 μM, whereas fewer than 10% have IC50 values in the range from 500 μM to 1000 μM (Figure 2b–d). Notably, the most studied ACE-I tripeptides, VPP and IPP, were identified through the fermentation of skim milk [49] and have been commercialized in Calpis sour milk (Calpis Co., Sumida City, Japan) and Evolus® (Valio, Finland) [50]. The IC50 values of VPP and IPP are reported to be 9 μM and 5 μM, respectively, in an in vitro ACE inhibition assay. Identified as the most potent peptide to date, Met-Ala-Pro (MAP) exhibits an IC50 of 0.8 μM in an in vitro ACE inhibition assay and has also demonstrated pronounced blood pressure-lowering effects in an in vivo hypertensive rat model (3 mg/kg dose, p < 0.01 at 8 h post-administration) [51].

2.7. Other Bioactivities

Beyond the aforementioned functions, peptides derived from bovine β-casein also exhibit diverse biological activities, including antioxidant effects [52], prolyl endopeptidase inhibition [53], and blood coagulation inhibition [54]. Recent work further highlights their value in lipid and glucose homeostasis. Jiang et al. [55] isolated β-casein peptides VLPVPQ and LQPE, which lowered micellar cholesterol solubility and suppressed the cholesterol transporter NPC1L1 in Caco-2 cells while upregulating efflux transporters ABCA1 and ABCG8. Similarly, Gong et al. [56] reported that the goat-milk peptide QEPVLGPVRGPFP from β-casein attenuated high-glucose-induced insulin resistance in HepG2 cells by enhancing glucose uptake and glycogen synthesis, while downregulating gluconeogenic genes. Synthetic peptides such as SQSKVLPVPQK also promoted trans-intestinal cholesterol excretion via ABCG5 activation and downregulated hepatic bile acid synthesis through the FGF15/19–LXRα axis [57]. Collectively, these findings underline the hypolipidemic and insulin-sensitizing potential of β-casein–derived peptides, supporting their inclusion in functional foods targeting metabolic disorders.
To catalogue these multifunctional peptides in a consistent and traceable manner, we compiled bovine β-casein-derived bioactive peptides from three widely used databases of milk-derived peptides: BIOPEP [58], MBPDB [59], and EROP-Moscow [60], retrieving a total of 176 entries. To ensure data quality and relevance, we applied four exclusion criteria: (i) no supporting reference; (ii) only hypothetical or predicted activity; (iii) discrepancies between database sequences and original literature; and (iv) bitter taste reported without any functional bioactivity. For instance, peptides such as PGP, PHQ, FPPQS, VLP, and GPV were present only in BIOPEP without literature support, while MBPDB recorded LVYPFPGPIP as an ACE inhibitor, although the cited publication reported activity for LVYPFPGPIH. After applying these quality control steps, 140 unique β-casein-derived bioactive peptides were retained and are listed in Table 1. A comprehensive diagram summarizing the theoretical curation process and exclusion steps is presented in Figure 3.

3. Proteolytic Systems of Lactic Acid Bacteria and Natural Peptide Liberation from β-Casein

Having established the diverse bioactivities of β-casein–derived peptides and their promising roles in metabolic and cardiovascular health, it is essential to understand how these peptides are generated at the molecular level. In dairy fermentations, lactic acid bacteria (LAB) are the primary microbial agents responsible for protein breakdown and peptide release. These Gram-positive, low GC-content bacteria are widely used in the fermentation of milk, meat, fish, and vegetables, and are classified as “generally recognized as safe (GRAS)” by the U.S. Food and Drug Administration (FDA) [14,164]. Since their first industrial application in cheese and sour milk in the 1890s, LAB have gained increasing economic and scientific interest [165,166,167]. Beyond acidification and flavor development, LAB have also been used as biocatalysts for the release of bioactive peptides, such as antihypertensive peptides, antimicrobial peptides, and immunomodulatory peptides [168]. One of the most relevant features of LAB in dairy fermentation is their ability to release bioactive peptides from β-casein via a sophisticated proteolytic system. Their proteolytic machinery not only serves nutritional needs for bacterial growth but also drives the liberation of bioactive sequences embedded in food proteins. This section outlines the molecular mechanisms involved and highlights the diversity of peptides generated.

3.1. Extracellular Proteolysis of β-Casein

Casein is degraded by dairy LAB through a proteolytic system to release amino acids necessary for cell growth (Figure 4). Simultaneously, the freed amino acids and small peptides play a role in the sensory characteristics of the dairy product. Among LAB, Lactococcus lactis is the most studied model for proteolysis. Its proteolytic system initiates casein degradation through a cell-envelope proteinase, which hydrolyses the extracellular protein (β-casein in this case) into the oligopeptides of varying length [14,15]. Specifically, the PrtP enzyme, which encodes the cell-envelope proteinase PrtP, is preceded by the divergently transcribed prtM gene, which encodes a lipoprotein, PrtM, which is anchored in the cell membrane and is required for the maturation and activation of PrtP [169]. Interestingly, while both prtP and prtM genes are present in Lacticaseibacillus paracasei [170], only prtP is found in Lactobacillus helveticus, Lb delbrueckii subsp. bulgaricus, and Lc. thermophilus [171,172], indicating species-specific differences in the proteolytic system.

3.2. Peptide Transport and Intracellular Degradation

Following initial cleavage of extracellular β-casein into oligopeptides, transport systems such as Opp, DtpT, and Dpp mediate peptide uptake: Opp handles peptides of 5–35 amino acids, whereas DtpT and Dpp are specialized for di- and tripeptides [173]. Once internalized, peptides will be further degraded in the cytoplasm by a suite of 15 intracellular peptidases with defined cleavage specificities [15]. Residual amino acids and peptides not utilized by the cell are released into the environment, where some of the peptides have potent biological functions [17].

3.3. Release of Functional Peptides

Certain LAB strains have demonstrated the ability to release bioactive peptides from food proteins, especially from caseins. The extent of hydrolysis and the resulting bioactivity are influenced both by the proteolytic enzymes expressed by the strains and the amino acid sequences of various casein types. As an example, an antimicrobial peptide released from human β-casein by Lb. helveticus PR4 exhibits activity against a broad spectrum of pathogens, such as Enterococcus faecium, Salmonella spp., St. aureus, E. coli, Bacillus megaterium, Yersinia enterocolitica, and L. innocua [78]. Moreover, milk fermented with Bifidobacterium bifidum MF 20/5, which is isolated from the Japanese probiotic product Bion-3®, was found to contain the peptide VLPVPQK with antioxidant activity [82]. In addition, LC analysis of Gouda-style cheese identified four antioxidative peptides characterized by an N-terminal X-Pro motif, all displaying IC50 values below 200 μM. Among these, LPQNIPPL exhibited the strongest antioxidative activity [104].
Beyond antimicrobial and antioxidant activities, LAB can also liberate immunomodulatory peptides. For example, milk fermented by Lb. helveticus LH-2 was shown to stimulate murine macrophage activity; four novel immunomodulatory peptides were subsequently identified [174]. Other strains, including Enterococcus faecalis CECT5727 and BCS27, Lb. delbrueckii, Lb. helveticus, Sa. cerevisiae, Lb. delbrueckii subsp. bulgaricus SS1, Lb. delbrueckii subsp. lactis, and Lc. lactis subsp. cremoris FT4 can generate ACE-inhibitory peptides from casein, demonstrating the role of LAB as biocatalysts for natural antihypertensive compounds (Table 1) [50,69].
Taken together, these findings illustrate the complexity and efficiency of LAB proteolytic systems in releasing functional peptides from β-casein. Understanding these microbial mechanisms provides a foundation for selecting or engineering strains optimized for peptide production in functional dairy and nutraceutical applications. However, when redirected toward the intentional production of specific bioactive peptides, native LAB systems reveal notable limitations. These include strain-dependent variability in protease activity, low or inconsistent yields of target peptides, and limited specificity for cleaving desired sequences. In addition, uncontrolled proteolysis can result in over-hydrolysis, degrading functional peptides and reducing their bioactivity. This unpredictability hampers both mechanistic studies and industrial applications, where reproducibility, efficacy, and scalability are essential. These challenges underscore the need for rationally designed LAB strains with greater control over proteolytic precision. In the following section, we highlight emerging molecular strategies—such as genome editing, synthetic biology, and omics-driven optimization—designed to overcome these limitations and fully harness LAB’s biosynthetic potential for targeted bioactive peptide production.

4. Molecular Strategies for Enhancing LAB-Mediated Liberation of Bioactive Peptides

The preceding section illustrated how wild-type LAB release bioactive fragments from β-casein via a sequential route of extracellular proteolysis, peptide transport, and intracellular trimming. However, the native process often under- or over-hydrolyses target peptides. The challenge is, therefore, not only to liberate the desired peptide but also to protect it from further degradation. This section summarizes the molecular toolkit now available to rebalance LAB proteolysis at three levels: promoter-driven control of key enzymes, chromosome-level editing, and high-throughput discovery platforms (Figure 5).

4.1. Inducible Systems for Targeted Regulation of Proteolysis

Because Lc. lactis is the best-characterized LAB; its proteolytic genes provide a model blueprint for targeted manipulation. Over recent decades, a range of genome engineering technologies for lactic acid bacteria (LAB) has emerged [175]. In Lc. lactis, gene expression can be precisely controlled by employing the Nisin-Controlled Expression (NICE) platform, which is widely regarded as the leading inducible expression system for this species. This platform functions through a quorum-sensing two-component regulatory circuit consisting of NisR and NisK proteins. Activation occurs upon the detection of nisin, an antimicrobial peptide naturally synthesized by specific Lc. lactis strains [176]. This system offers tight control over gene expression and is widely applied in Lc. lactis as a food-grade expression platform, particularly for the production of nisin, an important antimicrobial peptide used in the food industry.
In addition to the NICE system, alternative inducible platforms have been introduced to diversify regulatory options and avoid potential cross-reactivity. For instance, Zinc has also been employed as an inducer in LAB expression systems. Mu et al. [177] developed a zinc-responsive system named Zirex, which includes the pneumococcal repressor SczA and the PczcD promoter. This system enables gene expression under elevated Zn2+ concentrations. Another notable system is the xylose-inducible expression system (XIES), in which the PxylT promoter is activated by xylose but repressed by more common sugars such as glucose, fructose, and mannose. This sugar-switchable system is particularly attractive in industrial applications due to the low cost and non-toxic nature of xylose as an inducer [178]. More recently, an agmatine-inducible expression system has been introduced, composed of the agmatine-sensing transcriptional activator AguR and the PaguB promoter. This system offers tight control and dose-dependent gene expression, making it a promising complement or alternative to the traditional NICE system [179].
Together, these inducible promoter systems enable precise temporal and quantitative control over genes encoding proteases, transporters, or peptidases, allowing researchers to upregulate or downregulate specific steps in the proteolytic cascade to favor the accumulation of targeted bioactive peptides from β-casein.

4.2. Genome Editing Tools for Stable Manipulation of Proteolytic Pathways

Beyond transient expression control, stable genome editing provides a powerful avenue to reprogram LAB strains for optimized peptide release. Gene knockout can be accomplished in Lc. lactis by the double crossover method using the non-replicative pORI integration vector [180] or plasmid pCS1966 [181]. Although precise, these traditional methods are time-consuming and labor-intensive, often requiring several weeks to obtain a single marker-free mutant.
To accelerate strain development, novel genome engineering technologies have also been established in LAB. These include bacteriophage-derived recombinase enzymes for single-stranded DNA recombineering in Lb. reuteri and Lc. lactis [182], which facilitates single-stranded DNA recombineering and enables site-specific nucleotide substitutions with higher efficiency. The clustered regularly interspaced short palindromic repeats associated system (CRISPR/Cas) genome editing technology, which combines clustered regularly interspaced short palindromic repeats with Cas proteins, has emerged as one of the most powerful and widely adopted gene editing tools, particularly for eukaryotic organisms where alternative targeted approaches are limited [183]. Successful applications of the CRISPR/Cas9 system have also been reported in various bacterial genera, including Escherichia, Bacillus, Streptococcus, and Clostridium [184,185,186,187]. Within LAB, a CRISPR/Cas9-assisted single-stranded DNA recombineering system in Lb. reuteri employed Cas9-induced double-strand breaks (DSBs) as a counter-selection mechanism [188]. Furthermore, an inducible dual-promoter plasmid was constructed in Lc lactis, building on the previously described NICE system. This plasmid harbors the dCas9 and sgRNA genes under the control of the nisin-inducible promoter PnisA and was successfully utilized to repress transcription of the target gene htrA [189].
However, conventional genome editing in Lc. lactis typically relies on the RecA-mediated double-crossover recombination using non-replicative or conditionally replicative plasmids [181,190]. Gene knockout or insertion in Lc. lactis typically follows a two-step protocol—vector integration followed by co-integrate resolution—which generally requires approximately three weeks to generate a single mutant. The resulting marker-free strain can then serve as the starting point for subsequent rounds, allowing the introduction of multiple mutations into the same genetic background. However, this approach is limited by the considerable time needed to obtain a strain carrying multiple mutations. Leveraging the powerful CRISPR/Cas-based tools to engineer the proteolytic system of Lc. lactis could shorten the time required and lessen the workload. Moreover, utilizing CRISPR/dCas9, rather than CRISPR/Cas9, is more appropriate for Lc. lactis, as this organism lacks a non-homologous end joining (NHEJ) pathway and therefore cannot repair double-strand DNA breaks.
To overcome this, researchers have developed several alternative strategies. One widely adopted approach employs the Streptococcus pyogenes Cas9D10A nickase (SpCas9D10A), which introduces single-strand breaks (SSBs) instead of DSBs, thereby reducing cytotoxicity. In Lb. casei LC2W, a system combining SpCas9D10A with target-specific sgRNA and repair templates successfully circumvented DSB lethality [191]. Similarly, a single-plasmid SpCas9D10A-based editing system was developed for the probiotic Companilactobacillus crustorum, incorporating an SpCas9D10A expression cassette, the repD/repE replicon, ColE1 origin, sgRNA scaffold, and antibiotic resistance markers [192]. A comparable single-plasmid platform was also established in Lb. acidophilus NCFM, featuring a Cas9D10A gene under a constitutive promoter (P6), along with sgRNA and donor templates, enabling gene deletion and insertion [193]. These findings suggest that the exogenous CRISPR-Cas9D10A system can function in LAB even when native CRISPR-Cas machinery is present, highlighting its potential portability to other health-promoting and industrially relevant Lactobacillus species.

4.3. Cell-Free Protein Synthesis for Rapid Discovery and Screening

While gene regulation and genome editing empower rational strain design, cell-free systems offer complementary platforms for discovering and applying bioactive peptides from LAB. For example, cell-free culture supernatants (CFCS) of LAB, which contain bacteriocins and other metabolites, have been shown to inhibit Listeria monocytogenes in milk, meat, and broth via class IIa bacteriocins like leucocin, pediocin, and sakacin, though effectiveness varies with food matrix and storage conditions [194]. Similarly, the treatment of ribbonfish fillets with LAB-CFCS improved physicochemical quality, extended shelf life, and reduced spoilage at both refrigeration and room temperature (Das et al., 2021) [195]. LAB-CFCS also inhibited Staphylococcus aureus growth and biofilm formation by disrupting adhesion, matrix production, and metabolic pathways [196].
Beyond harvesting natural products, cell-free protein synthesis (CFPS) systems allow for in vitro engineering and screening of synthetic pathways. For instance, a CFPS platform was developed based on the nisin biosynthetic pathway to overcome the toxicity and complexity associated with producing lanthipeptides in living cells. Using this platform, researchers could mine the NCBI database for nisin analogs and, within a single day, identify four novel lanthipeptides with antibacterial activity, demonstrating its high-throughput capacity. Moreover, the CFPS was coupled with a functional assay against gram-negative bacteria, leading to the discovery of a potent nisin mutant (M5) with enhanced activity. Importantly, the CFPS-guided approach not only accelerated discovery but also improved the in vivo titers of nisin and its analogs by informing strain engineering. Given the conserved biosynthetic principles of lanthipeptides, this platform provides a broadly applicable and versatile method for discovering and overproducing a wide range of bioactive peptides beyond nisin [197]. These complementary strategies demonstrate the power of cell-free approaches to discover, optimize, and apply LAB-derived bioactive peptides, especially those challenging to express in living hosts.

4.4. Multi-Omics Strategies for System-Level Optimization

In addition to targeted molecular engineering, multi-omics technologies offer a powerful systems-level approach to further optimize the release of bioactive peptides from β-casein by LAB. While traditional methods often rely on single-strain cultures and gene-targeted analysis, multi-omics enables the resolution of dynamic molecular interactions that underlie peptide formation from bovine β-casein in complex environments [198]. Multi-omics technologies—spanning genomics, transcriptomics, proteomics, and metabolomics—provide a powerful systems-level approach to understanding and optimizing the release of bioactive peptides by LAB.
Recent multi-omics studies have demonstrated that mixed LAB fermentation (e.g., Lactobacillus plantarum and L. acidophilus) can break down structurally complex substrates like bee pollen and activate amino acid metabolic pathways [199]. In broader fermented food systems, multi-omics has been used to map microbial contributions to metabolite production and to reveal strain-specific proteolytic capacities that influence functional peptide profiles [200]. Moreover, in broader fermented food systems, multi-omics has been utilized to map microbial contributions to metabolite production and to reveal strain-specific proteolytic capacities that influence functional peptide profiles. For instance, metabolomic and peptidomic analyses have been used to identify bioactive peptides released during fermentation with novel probiotics like Lacticaseibacillus rhamnosus and Lactiplantibacillus plantarum, which revealed significant upregulation of ACE-I peptides. In a study of fresh cheese fermented with three novel probiotics, 112 bioactive peptides were significantly upregulated, highlighting the potential of metabolomics and peptidomics for understanding peptide biosynthesis [201]. Additionally, an integrated approach using metabolomics and peptidomics was employed to delineate the characteristic metabolites and peptides in milk fermented with Lpb. plantarum L3. This study revealed that Lpb. plantarum L3 secreted a complex array of peptidases that cleaved bovine caseins at specific sites to release bioactive peptides, including ACE-I peptides, antioxidant peptides, and antimicrobial peptides. The enzyme systems secreted by Lpb. plantarum L3 preferentially cleaved lysine-based peptide bonds, and the released peptides exhibited various bioactivities [202].
These findings underscore the importance of multi-omics approaches in optimizing fermentation conditions to enhance the functional properties of fermented dairy products. By combining the metabolomic and peptidomic data, this integrated multi-omics approach provides valuable insights into the complex enzymatic processes that regulate the release of bioactive peptides during fermentation. Such knowledge can be applied to optimize LAB strains for specific peptide production, leading to improved functional foods with targeted health benefits.
The convergence of inducible promoters, CRISPR toolkits, CFPS prototyping, and multi-omics analytics now enables precision fermentation: LAB strains engineered to release specific β-casein peptides at an industrial scale while minimizing unwanted side-products. These molecular strategies not only enhance functional-food development but also expand LAB’s potential as GRAS biotherapeutic platforms.

5. Challenges, Regulatory Considerations, and Future Perspectives

While recent advances in molecular engineering, high-throughput screening, and multi-omics have significantly expanded our ability to discover and produce bioactive peptides from bovine β-casein using LAB, the successful industrial translation of these peptides into food-grade ingredients or supplements remains limited. One of the primary technical challenges lies in peptide stability [203]. Bioactive peptides are often highly susceptible to degradation by gastrointestinal digestion or during food processing procedures such as heating, pH fluctuation, or enzymatic cross-reactions with other food matrix components. This instability may significantly compromise their bioavailability and health-promoting effects in vivo.
Moreover, large-scale production of bioactive peptides using LAB fermentation involves multifaceted optimization. These include ensuring strain stability, maintaining robust proteolytic activity across batches, and avoiding the accumulation of bitter peptides or off-flavors during extended fermentation [204]. The dynamic nature of microbial proteolysis, driven by environmental factors such as substrate concentration, fermentation pH, oxygen levels, and nutrient availability, further complicates standardized production [172]. Strain-specific differences in proteolytic specificity further complicate process standardization and require careful strain selection or genetic tailoring. Moreover, the downstream steps—such as peptide concentration, purification, and formulation for delivery—are also resource-intensive and limit scalability. The use of genetically modified lactic acid bacteria (GM-LAB) offers a promising approach to addressing many of these issues. Through synthetic biology tools such as CRISPR/Cas9 or integrative plasmid systems, LAB can be engineered to enhance their proteolytic systems, selectively release target peptides, or eliminate bitter peptide byproducts. Furthermore, GM-LAB can be designed to selectively produce desired bioactive peptides, facilitating easier downstream purification.
However, the use of genetically modified LAB (GM-LAB) introduces regulatory and consumer perception challenges. Regulatory frameworks vary significantly across regions: in the United States, GRAS designation provides a relatively flexible route for GM strain approval, contingent on evidence of safety. In contrast, the European Union operates the more stringent Qualified Presumption of Safety (QPS), which requires detailed evaluations of each genetically modified strain, including potential toxicity, genetic stability, and absence of transferable antibiotic resistance markers. Additionally, public skepticism toward genetically modified organisms (GMOs), especially in the food and health domains, adds a further layer of complexity. Hence, even for strains with robust safety data, product developers may prefer non-GMO LAB strains to avoid labeling controversies and market resistance, especially in export-oriented and infant products.
Looking forward, the convergence of synthetic biology, artificial intelligence (AI), and precision fermentation is expected to define the next frontier in LAB-based bioactive peptide development. Synthetic biology offers the possibility of designing “next-generation” LAB chassis tailored for controlled proteolysis and defined peptide profiles. To fully realize this potential, interdisciplinary collaboration between food engineers, computational biologists, and fermentation experts will be crucial. Food engineers can bring expertise in optimizing fermentation conditions, scaling up production, and ensuring the consistency of peptide output, while computational biologists will focus on creating predictive models, integrating omics data, and using AI tools to optimize peptide sequence design and bioactivity predictions. This collaboration will ensure that both the fermentation processes and the bioinformatics approaches are aligned to maximize the efficiency and specificity of peptide production. Coupled with multi-omics and high-throughput screening platforms, the rational design of LAB protease repertoires could achieve both diversity and specificity in peptide output. Moreover, food engineers will play a pivotal role in translating computational predictions into practical applications by optimizing fermentation conditions such as nutrient availability, temperature, pH, and oxygen levels, which are crucial for the desired expression of bioactive peptides. Meanwhile, computational biologists, aided by machine learning algorithms, can continually improve their models by analyzing the resulting data from fermentation trials. This dynamic feedback loop will allow for a more iterative and fine-tuned approach to peptide production. AI-driven peptide discovery tools—trained on large datasets of sequence–function relationships—are being increasingly applied to predict peptide bioactivities and optimize sequences with enhanced stability, bioavailability, or receptor binding affinity. The synergy between computational biology and fermentation science will allow for the development of peptides with more predictable and controllable characteristics, ultimately reducing the time and resources required for optimization.
Furthermore, precision fermentation platforms promise reproducible, scalable, and cost-effective peptide production with minimal batch variability. To maximize the impact of these platforms, both computational biologists and food engineers will need to work together to fine-tune the optimization parameters and integrate real-time monitoring technologies, ensuring that the fermentation process remains robust and adaptable to various production scales. In the era of personalized nutrition and microbiome-aware interventions, collaborative efforts between these interdisciplinary teams will also facilitate the development of modular LAB consortia that can be designed to generate peptide profiles specifically tailored to individual health needs or microbiota compositions. This flexibility will allow for personalized approaches to peptide delivery, enhancing the efficacy of bioactive peptides in promoting human health and well-being.
However, despite its scientific advantages, the current implementation of synthetic biology-assisted peptide production using LAB may involve higher upfront costs compared to conventional strain isolation and screening approaches. These expenses primarily arise from two domains: the development of engineered strains, including genetic modification and fermentation process optimization, and the subsequent investment in regulatory compliance, which is essential for product approval and market access. These stages require significant resource input and specialized expertise, posing a barrier to small- and medium-sized enterprises. Nevertheless, this strategy offers a much higher ceiling in terms of efficiency and innovation potential. Once a robust chassis strain is established, it can be rapidly reprogrammed or modularly adjusted to produce a variety of bioactive peptides with different functions, significantly reducing time and cost for each new target. Moreover, the integration of machine learning and AI into strain design and peptide prediction workflows holds the promise of accelerating discovery, optimizing peptide properties, and expanding the functional diversity of the final products. In this era of digital biology, AI-powered peptide biomanufacturing systems could unlock unprecedented levels of precision, scalability, and customization in LAB-driven production platforms.

6. Conclusions

Lactic acid bacteria (LAB) represent a natural and versatile microbial platform for the targeted release of bioactive peptides from bovine β-casein, offering an effective strategy to valorize dairy proteins into functional ingredients. The sophisticated proteolytic system of LAB—particularly in strains like Lactococcus lactis and Lactobacillus helveticus—enables precise cleavage of casein sequences to liberate peptides with documented antihypertensive, antioxidative, antimicrobial, immunomodulatory, and metabolic regulatory functions.
These peptides contribute not only to the sensory and nutritional value of fermented dairy products but also hold promise as therapeutic agents in the context of functional foods and nutraceuticals. Despite substantial progress in peptide identification and characterization, challenges remain in optimizing peptide yield, specificity, and stability during industrial production. Recent advances in molecular tools such as CRISPR-based genome modulation (CRISPRi/a), inducible expression systems (e.g., NICE, Zirex, XIES, and agmatine-regulated promoters), and cell-free platforms have opened new avenues to engineer LAB for enhanced peptide liberation. Furthermore, multi-omics approaches allow for a systems-level understanding of LAB proteolysis and metabolic networks, offering data-driven insights into strain selection, process tuning, and pathway optimization.
With the convergence of synthetic biology, fermentation engineering, and computational biology, LAB-based biomanufacturing is poised to enter a new era of precision and efficiency. Synthetic biology, by enabling the rational design of LAB chassis, allows for the precise tailoring of microbial systems that can be optimized for specific proteolytic tasks. LAB strains can now be engineered to possess enhanced, targeted proteases that selectively cleave casein peptides with higher specificity and yield. This engineering opens the door to a range of bioactive peptides with improved bioavailability and potency. Enzymatic engineering is crucial in optimizing the processes by which these peptides are liberated, focusing on modulating the activity of endogenous proteases or introducing novel ones, thus enhancing peptide yield while preventing the production of unwanted byproducts such as bitter peptides. Furthermore, the integration of computational biology, coupled with AI-driven platforms, accelerates the design and optimization process. By using predictive models based on large datasets of fermentation variables, machine learning can help predict optimal fermentation conditions in real-time, improving scalability and consistency across production batches. These advances will allow for unprecedented control over peptide production, providing a clear path to scalability in industrial applications. As computational tools integrate multi-omics data, the feedback loop between real-time fermentation monitoring and process adjustments will enable continuous improvements to both strain design and fermentation parameters, leading to more efficient, cost-effective, and reproducible peptide production. Moreover, with the increasing ability to predict and tailor the bioactivity of peptides, these technologies hold the potential to support the development of highly specialized, personalized bioactive peptides for a variety of health-related applications, from chronic disease prevention to targeted gut microbiome modulation.
However, as we progress toward this new era, regulatory considerations remain a critical component in ensuring the safe and ethical deployment of GM-LAB in the commercial production of bioactive peptides. Regulatory frameworks across different regions present unique challenges, which often require extensive testing and detailed safety documentation, particularly for novel GM strains in food applications, including assessments of potential toxicity, genetic stability, and the absence of transferable antibiotic resistance markers. This creates a significant regulatory burden for companies seeking to commercialize GM-LAB-derived products, which can delay product introduction and increase costs. Furthermore, public perception of GMOs, especially in the food and health sectors, remains a significant challenge. Even with robust safety data, consumer skepticism can hinder market acceptance, particularly for sensitive product categories like infant nutrition. To mitigate these challenges, the convergence of synthetic biology, enzymatic engineering, computational biology, and regulatory policy will be essential. It is important to continue engaging with regulatory authorities to ensure clear, standardized guidelines for GM organisms in food production. Furthermore, public awareness campaigns and stakeholder engagement will play a vital role in building trust and acceptance of GM-LAB-derived products. Collaborations between industry stakeholders, regulatory bodies, and consumers will help streamline approval processes, reduce market entry barriers, and ensure that bioactive peptides are delivered safely, ethically, and efficiently to consumers.
As consumer interest grows in gut health, personalized nutrition, and sustainable protein use, LAB-derived bioactive peptides stand at the forefront of functional food innovation. Realizing their full potential will require continued interdisciplinary collaboration—bridging microbiology, food technology, biotechnology, and regulatory policy—to accelerate the transition from bench to product and, ultimately, deliver scientifically validated, safe, and effective peptide-based interventions to the global market.

Author Contributions

Conceptualization, C.H. and L.C.; methodology, C.H.; validation, C.H.; formal analysis, C.H.; investigation, C.H.; resources, C.H.; data curation, L.C.; writing—original draft preparation, C.H.; writing—review and editing, C.H. and L.C.; visualization, C.H.; supervision, L.C.; project administration, C.H. and L.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank Jan Kok from the University of Groningen for his valuable comments and suggestions on the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
LABlactic acid bacteria
AAamino acid
CPPscaseinophosphopeptides
β-CMsβ-casomorphins
ACEangiotensin-converting enzyme
DPP-IVdipeptidyl peptidase-4
GIPinsulinotropic polypeptide
GLP-1glucagon-like peptide-1
MSmass spectrometry
LC-MSLiquid Chromatography–Mass Spectrometry
AMPsantimicrobial peptides
GRASgenerally recognized as safe
FDAU.S. Food and Drug Administration
NICENIsin-Controlled Expression
XIESxylose-inducible expression system
CRISP/Casclustered regularly interspaced short palindromic repeats associated system
NHEJnon-homologous end joining
SpCas9D10AS. pyogenes Cas9D10A nickase
SSBssingle-strand breaks
DSBsdouble-strand breaks
CFPScell-free protein synthesis
CFCScell-free culture supernatants
GM-LABgenetically modified lactic acid bacteria
QPSQualified Presumption of Safety
GMOsgenetically modified organisms
AIartificial intelligence

References

  1. Turner-Ravana, N. Milk and Dairy Foods: Their Functionality in Human Health and Disease. J. Nutr. Educ. Behav. 2021, 53, 546. [Google Scholar] [CrossRef]
  2. OECD Publishing. OECD-FAO Agricultural Outlook 2024–2033; Organisation for Economic Co-operation and Development OECD: Paris, France, 2024; ISBN 978-92-64-59211-7. [Google Scholar]
  3. Saxelin, M.; Korpela, R.; Mäyrä-Mäkinen, A. Introduction: Classifying Functional Dairy Products; Woodhead Publishing Ltd.: Cambridge, UK, 2003. [Google Scholar]
  4. Giromini, C.; Fekete, Á.A.; Pinotti, L.; Baldi, A. Milk Proteins: Their Role in Cardiovascular Health. Milk Dairy Foods 2020, 145–172. [Google Scholar] [CrossRef]
  5. Eigel, W.; Butler, J.; Ernstrom, C.; Farrell, H., Jr.; Harwalkar, V.; Jenness, R.; Whitney, R.M. Nomenclature of Proteins of Cow’s Milk: Fifth Revision. J. Dairy Sci. 1984, 67, 1599–1631. [Google Scholar] [CrossRef]
  6. Atamer, Z.; Post, A.E.; Schubert, T.; Holder, A.; Boom, R.M.; Hinrichs, J. Bovine β-Casein: Isolation, Properties and Functionality. A Review. Int. Dairy J. 2017, 66, 115–125. [Google Scholar] [CrossRef]
  7. Korhonen, H.; Pihlanto, A. Bioactive Peptides: Production and Functionality. Int. Dairy J. 2006, 16, 945–960. [Google Scholar] [CrossRef]
  8. Gobbetti, M.; Minervini, F.; Rizzello, C.G. Bioactive Peptides in Dairy Products. Handb. Food Prod. Manuf. 2007, 489–517. [Google Scholar] [CrossRef]
  9. Kilara, A.; Panyam, D. Peptides from Milk Proteins and Their Properties. Crit. Rev. Food Sci. Nutr. 2003, 43, 607–633. [Google Scholar] [CrossRef]
  10. Korhonen, H. Milk-Derived Bioactive Peptides: From Science to Applications. J. Funct. Foods 2009, 1, 177–187. [Google Scholar] [CrossRef]
  11. Chen, X.; Fan, R.; Wang, Y.; Munir, M.; Li, C.; Wang, C.; Hou, Z.; Zhang, G.; Liu, L.; He, J. Bovine Milk β-Casein: Structure, Properties, Isolation, and Targeted Application of Isolated Products. Compr. Rev. Food Sci. Food Saf. 2024, 23, e13311. [Google Scholar] [CrossRef] [PubMed]
  12. Chaudhari, H.; Hati, S. Production and Biofunctionality of Milk-Derived Bioactive Peptides. In Microbiome-Gut-Brain Axis: Implications on Health; Sayyed, R.Z., Khan, M., Eds.; Springer Nature: Singapore, 2022; pp. 297–316. ISBN 978-981-16-1626-6. [Google Scholar]
  13. Auestad, N.; Layman, D.K. Dairy Bioactive Proteins and Peptides: A Narrative Review. Nutr. Rev. 2021, 79, 36–47. [Google Scholar] [CrossRef]
  14. Kok, J. Genetics of the Proteolytic System of Lactic Acid Bacteria. FEMS Microbiol. Rev. 1990, 7, 15–41. [Google Scholar] [CrossRef]
  15. Savijoki, K.; Ingmer, H.; Varmanen, P. Proteolytic Systems of Lactic Acid Bacteria. Appl. Microbiol. Biotechnol. 2006, 71, 394–406. [Google Scholar] [CrossRef]
  16. Huang, C.; Kok, J. Editing of the Proteolytic System of Lactococcus lactis Increases Its Bioactive Potential. Appl. Environ. Microbiol. 2020, 86, e01319-20. [Google Scholar] [CrossRef]
  17. Caballero, B.; Finglas, P.; Toldrá, F. Encyclopedia of Food and Health; Academic Press: Cambridge, MA, USA, 2015; ISBN 0-12-384953-5. [Google Scholar]
  18. FitzGerald, R. Potential Uses of Caseinophosphopeptides. Int. Dairy J. 1998, 8, 451–457. [Google Scholar] [CrossRef]
  19. Kawahara, T.; Katayama, D.; Otani, H. Effect of β-Casein (1-28) on Proliferative Responses and Secretory Functions of Human Immunocompetent Cell Lines. Biosci. Biotechnol. Biochem. 2004, 68, 2091–2095. [Google Scholar] [CrossRef] [PubMed]
  20. Sato, R.; Shindo, M.; Gunshin, H.; Noguchi, T.; Naito, H. Characterization of Phosphopeptide Derived from Bovine β-Casein: An Inhibitor to Intra-Intestinal Precipitation of Calcium Phosphate. Biochim. Biophys. Acta (BBA)-Protein Struct. Mol. Enzymol. 1991, 1077, 413–415. [Google Scholar] [CrossRef]
  21. Tobita, K.; Kawahara, T.; Otani, H. Bovine β-Casein (1−28), a Casein Phosphopeptide, Enhances Proliferation and IL-6 Expression of Mouse CD19+ Cells via Toll-like Receptor 4. J. Agric. Food Chem. 2006, 54, 8013–8017. [Google Scholar] [CrossRef] [PubMed]
  22. Hata, I.; Higashiyama, S.; Otani, H. Identification of a Phosphopeptide in Bovine As1-Casein Digest as a Factor Influencing Proliferation and Immunoglobulin Production in Lymphocyte Cultures. J. Dairy Res. 1998, 65, 569–578. [Google Scholar] [CrossRef] [PubMed]
  23. Contet, C.; Kieffer, B.L.; Befort, K. Mu Opioid Receptor: A Gateway to Drug Addiction. Curr. Opin. Neurobiol. 2004, 14, 370–378. [Google Scholar] [CrossRef]
  24. Mansour, A.; Watson, S. Anatomical Distribution of Opioid Receptors in Mammalians: An Overview. Opioids 1993, 79–105. [Google Scholar] [CrossRef]
  25. Brantl, V.; Teschemacher, H.; Henschen, A.; Lottspeich, F. Novel Opioid Peptides Derived from Casein (β-Casomorphins). I. Isolation from Bovine Casein Peptone. Hoppe Seyler’s Z. Physiol. Chem. 1979, 360, 1211–1216. [Google Scholar] [CrossRef] [PubMed]
  26. Zoghbi, S.; Trompette, A.; Claustre, J.; Homsi, M.E.; Garzón, J.; Jourdan, G.; Scoazec, J.-Y.; Plaisancié, P. β-Casomorphin-7 Regulates the Secretion and Expression of Gastrointestinal Mucins through a μ-Opioid Pathway. Am. J. Physiol.-Gastrointest. Liver Physiol. 2006, 290, G1105–G1113. [Google Scholar] [CrossRef] [PubMed]
  27. Hernández-Ledesma, B.; Amigo, L.; Ramos, M.; Recio, I. Release of Angiotensin Converting Enzyme-Inhibitory Peptides by Simulated Gastrointestinal Digestion of Infant Formulas. Int. Dairy J. 2004, 14, 889–898. [Google Scholar] [CrossRef]
  28. Yin, H.; Miao, J.; Ma, C.; Sun, G.; Zhang, Y. β-Casomorphin-7 Cause Decreasing in Oxidative Stress and Inhibiting NF-κB-iNOS-NO Signal Pathway in Pancreas of Diabetes Rats. J. Food Sci. 2012, 77, C278–C282. [Google Scholar] [CrossRef]
  29. Escobar-Charry, M.A.; Quintanilla-Carvajal, M.X. An Overview of Bovine Beta-Casomorphin-7 (b-BCM7) and Its Potential Impact on Microbiota Regulation and Digestive Health. J. Funct. Foods 2025, 128, 106812. [Google Scholar] [CrossRef]
  30. Robinson, S.R.; Greenway, F.L.; Deth, R.C.; Fayet-Moore, F. Effects of Different Cow-Milk Beta-Caseins on the Gut–Brain Axis: A Narrative Review of Preclinical, Animal, and Human Studies. Nutr. Rev. 2025, 83, e1259–e1269. [Google Scholar] [CrossRef]
  31. Dalziel, J.; Spencer, N.; Dunstan, K.; Lynch, A.; Haggarty, N.; Gopal, P.; Roy, N. An In Vitro Rat Model of Colonic Motility to Determine the Effect of β-Casomorphin-5 on Propagating Contractions. Food Funct. 2014, 5, 2768–2774. [Google Scholar] [CrossRef]
  32. Daniel, H.; Vohwinkel, M.; Rehner, G. Effect of Casein and β-Casomorphins on Gastrointestinal Motility in Rats. J. Nutr. 1990, 120, 252–257. [Google Scholar] [CrossRef]
  33. Sakaguchi, M.; Murayama, K.; Jinsmaa, Y.; Yoshikawa, M.; Matsumura, E. Neurite Outgrowth-Stimulating Activities of β-Casomorphins in Neuro-2a Mouse Neuroblastoma Cells. Biosci. Biotechnol. Biochem. 2003, 67, 2541–2547. [Google Scholar] [CrossRef]
  34. Sakaguchi, M.; Koseki, M.; Wakamatsu, M.; Matsumura, E. Effects of Systemic Administration of β-Casomorphin-5 on Learning and Memory in Mice. Eur. J. Pharmacol. 2006, 530, 81–87. [Google Scholar] [CrossRef]
  35. Tulipano, G. Role of Bioactive Peptide Sequences in the Potential Impact of Dairy Protein Intake on Metabolic Health. Int. J. Mol. Sci. 2020, 21, 8881. [Google Scholar] [CrossRef]
  36. Nongonierma, A.B.; FitzGerald, R.J. Prospects for the Management of Type 2 Diabetes Using Food Protein-Derived Peptides with Dipeptidyl Peptidase IV (DPP-IV) Inhibitory Activity. Curr. Opin. Food Sci. 2016, 8, 19–24. [Google Scholar] [CrossRef]
  37. Rossolini, G.M.; Arena, F.; Pecile, P.; Pollini, S. Update on the Antibiotic Resistance Crisis. Curr. Opin. Pharmacol. 2014, 18, 56–60. [Google Scholar] [CrossRef] [PubMed]
  38. Mahlapuu, M.; Håkansson, J.; Ringstad, L.; Björn, C. Antimicrobial Peptides: An Emerging Category of Therapeutic Agents. Front. Cell. Infect. Microbiol. 2016, 6, 194. [Google Scholar] [CrossRef]
  39. Birkemo, G.; O’sullivan, O.; Ross, R.; Hill, C. Antimicrobial Activity of Two Peptides Casecidin 15 and 17, Found Naturally in Bovine Colostrum. J. Appl. Microbiol. 2009, 106, 233–240. [Google Scholar] [CrossRef]
  40. Sedaghati, M.; Ezzatpanah, H.; Boojar, M.M.A.; Ebrahimi, M.T.; Kobarfard, F. Isolation and Identification of Some Antibacterial Peptides in the Plasmin-Digest of β-Casein. LWT-Food Sci. Technol. 2016, 68, 217–225. [Google Scholar] [CrossRef]
  41. Gertsch, J.; Viveros-Paredes, J.M.; Taylor, P. Plant Immunostimulants—Scientific Paradigm or Myth? J. Ethnopharmacol. 2011, 136, 385–391. [Google Scholar] [CrossRef]
  42. Chalamaiah, M.; Yu, W.; Wu, J. Immunomodulatory and Anticancer Protein Hydrolysates (Peptides) from Food Proteins: A Review. Food Chem. 2018, 245, 205–222. [Google Scholar] [CrossRef] [PubMed]
  43. Reyes-Díaz, A.; González-Córdova, A.F.; Hernández-Mendoza, A.; Reyes-Díaz, R.; Vallejo-Cordoba, B. Immunomodulation by Hydrolysates and Peptides Derived from Milk Proteins. Int. J. Dairy Technol. 2018, 71, 1–9. [Google Scholar] [CrossRef]
  44. Liu, T.; Feng, J.; Han, H.; Huang, J.; Zhang, L.; Hettinga, K.; Zhou, P. Anti-Inflammatory Effects of Dietary β-Casein Peptides and Its Peptide QEPVL in a DSS-Induced Inflammatory Bowel Disease Mouse Model. Food Biosci. 2023, 56, 103375. [Google Scholar] [CrossRef]
  45. Bernstein, K.E.; Khan, Z.; Giani, J.F.; Cao, D.-Y.; Bernstein, E.A.; Shen, X.Z. Angiotensin-Converting Enzyme in Innate and Adaptive Immunity. Nat. Rev. Nephrol. 2018, 14, 325–336. [Google Scholar] [CrossRef] [PubMed]
  46. Sheih, I.-C.; Fang, T.J.; Wu, T.-K. Isolation and Characterisation of a Novel Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptide from the Algae Protein Waste. Food Chem. 2009, 115, 279–284. [Google Scholar] [CrossRef]
  47. Messerli, F.H.; Williams, B.; Ritz, E. Essential Hypertension. Lancet 2007, 370, 591–603. [Google Scholar] [CrossRef]
  48. Irvin, J.D.; Viau, J.M. Safety Profiles of the Angiotensin Converting Enzyme Inhibitors Captopril and Enalapril. Am. J. Med. 1986, 81, 46–50. [Google Scholar] [CrossRef] [PubMed]
  49. Nakamura, Y.; Yamamoto, N.; Sakai, K.; Okubo, A.; Yamazaki, S.; Takano, T. Purification and Characterization of Angiotensin I-Converting Enzyme Inhibitors from Sour Milk. J. Dairy Sci. 1995, 78, 777–783. [Google Scholar] [CrossRef]
  50. Rai, A.K.; Sanjukta, S.; Jeyaram, K. Production of Angiotensin I Converting Enzyme Inhibitory (ACE-I) Peptides during Milk Fermentation and Their Role in Reducing Hypertension. Crit. Rev. Food Sci. Nutr. 2017, 57, 2789–2800. [Google Scholar] [CrossRef]
  51. Tonouchi, H.; Suzuki, M.; Uchida, M.; Oda, M. Antihypertensive Effect of an Angiotensin Converting Enzyme Inhibitory Peptide from Enzyme Modified Cheese. J. Dairy Res. 2008, 75, 284–290. [Google Scholar] [CrossRef]
  52. Farvin, K.S.; Baron, C.P.; Nielsen, N.S.; Otte, J.; Jacobsen, C. Antioxidant Activity of Yoghurt Peptides: Part 2–Characterisation of Peptide Fractions. Food Chem. 2010, 123, 1090–1097. [Google Scholar] [CrossRef]
  53. Smacchi, E.; Gobbetti, M. Peptides from Several Italian Cheeses Inhibitory to Proteolytic Enzymes of Lactic Acid Bacteria, Pseudomonas fluorescens ATCC 948 and to the Angiotensin I-Converting Enzyme. Enzym. Microb. Technol. 1998, 22, 687–694. [Google Scholar] [CrossRef]
  54. Liu, H.; Tu, M.; Cheng, S.; Chen, H.; Wang, Z.; Du, M. An Anticoagulant Peptide from Beta-Casein: Identification, Structure and Molecular Mechanism. Food Funct. 2019, 10, 886–892. [Google Scholar] [CrossRef]
  55. Jiang, X.; Pan, D.; Zhang, T.; Liu, C.; Zhang, J.; Su, M.; Wu, Z.; Zeng, X.; Sun, Y.; Guo, Y. Novel Milk Casein–Derived Peptides Decrease Cholesterol Micellar Solubility and Cholesterol Intestinal Absorption in Caco-2 Cells. J. Dairy Sci. 2020, 103, 3924–3936. [Google Scholar] [CrossRef]
  56. Gong, H.; Gao, J.; Wang, Y.; Luo, Q.W.; Guo, K.R.; Ren, F.Z.; Mao, X.Y. Identification of Novel Peptides from Goat Milk Casein That Ameliorate High-Glucose-Induced Insulin Resistance in HepG2 Cells. J. Dairy Sci. 2020, 103, 4907–4918. [Google Scholar] [CrossRef]
  57. Lee, S.; Youn, B. Hypolipidemic Roles of Casein-Derived Peptides by Regulation of Trans-Intestinal Cholesterol Excretion and Bile Acid Synthesis. Nutrients 2020, 12, 3058. [Google Scholar] [CrossRef]
  58. Minkiewicz, P.; Dziuba, J.; Iwaniak, A.; Dziuba, M.; Darewicz, M. BIOPEP Database and Other Programs for Processing Bioactive Peptide Sequences. J. AOAC Int. 2008, 91, 965–980. [Google Scholar] [CrossRef]
  59. Nielsen, S.D.; Beverly, R.L.; Qu, Y.; Dallas, D.C. Milk Bioactive Peptide Database: A Comprehensive Database of Milk Protein-Derived Bioactive Peptides and Novel Visualization. Food Chem. 2017, 232, 673–682. [Google Scholar] [CrossRef] [PubMed]
  60. Zamyatnin, A.A.; Borchikov, A.S.; Vladimirov, M.G.; Voronina, O.L. The EROP-Moscow Oligopeptide Database. Nucleic Acids Res. 2006, 34, D261–D266. [Google Scholar] [CrossRef]
  61. Wang, Z.; Zhang, S.; Wang, W.; Feng, F.; Shan, W. A Novel Angiotensin I Converting Enzyme Inhibitory Peptide from the Milk Casein: Virtual Screening and Docking Studies. Agric. Sci. China 2011, 10, 463–467. [Google Scholar] [CrossRef]
  62. Otani, H.; Nakano, K.; Kawahara, T. Stimulatory Effect of a Dietary Casein Phosphopeptide Preparation on the Mucosal IgA Response of Mice to Orally Ingested Lipopolysaccharide from Salmonella typhimurium. Biosci. Biotechnol. Biochem. 2003, 67, 729–735. [Google Scholar] [CrossRef]
  63. Kawahara, T.; Otani, H. Stimulatory Effects of Casein Phosphopeptide (CPP-III) on mRNA Expression of Cytokines in Caco-2 Cells. Biosci. Biotechnol. Biochem. 2004, 68, 1779–1781. [Google Scholar] [CrossRef] [PubMed]
  64. Otani, H.; Watanabe, T.; Tashiro, Y. Effects of Bovine β-Casein (1-28) and Its Chemically Synthesized Partial Fragments on Proliferative Responses and Immunoglobulin Production in Mouse Spleen Cell Cultures. Biosci. Biotechnol. Biochem. 2001, 65, 2489–2495. [Google Scholar] [CrossRef] [PubMed]
  65. Silanikove, N.; Shamay, A.; Shinder, D.; Moran, A. Stress down Regulates Milk Yield in Cows by Plasmin Induced β-Casein Product That Blocks K+ Channels on the Apical Membranes. Life Sci. 2000, 67, 2201–2212. [Google Scholar] [CrossRef]
  66. Pizzano, R.; Nicolai, M.A.; Padovano, P.; Ferranti, P.; Barone, F.; Addeo, F. Immunochemical Evaluation of Bovine β-Casein and Its 1−28 Phosphopeptide in Cheese during Ripening. J. Agric. Food Chem. 2000, 48, 4555–4560. [Google Scholar] [CrossRef]
  67. Gagnaire, V.; Carpino, S.; Pediliggieri, C.; Jardin, J.; Lortal, S.; Licitra, G. Uncommonly Thorough Hydrolysis of Peptides during Ripening of Ragusano Cheese Revealed by Tandem Mass Spectrometry. J. Agric. Food Chem. 2011, 59, 12443–12452. [Google Scholar] [CrossRef] [PubMed]
  68. Pinto, G.; Caira, S.; Cuollo, M.; Lilla, S.; Chianese, L.; Addeo, F. Bioactive Casein Phosphopeptides in Dairy Products as Nutraceuticals for Functional Foods. In Milk Protein; IntechOpen: London, UK, 2012; ISBN 953-51-0743-7. [Google Scholar]
  69. Gobbetti, M.; Ferranti, P.; Smacchi, E.; Goffredi, F.; Addeo, F. Production of Angiotensin-I-Converting-Enzyme-Inhibitory Peptides in Fermented Milks Started by Lactobacillus delbrueckii subsp. Bulgaricus SS1 and Lactococcus lactis subsp. Cremoris FT4. Appl. Environ. Microbiol. 2000, 66, 3898–3904. [Google Scholar] [CrossRef]
  70. Nongonierma, A.B.; FitzGerald, R.J. Structure Activity Relationship Modelling of Milk Protein-Derived Peptides with Dipeptidyl Peptidase IV (DPP-IV) Inhibitory Activity. Peptides 2016, 79, 1–7. [Google Scholar] [CrossRef] [PubMed]
  71. Lopez-Exposito, I.; Minervini, F.; Amigo, L.; Recio, I. Identification of Antibacterial Peptides from Bovine κ-Casein. J. Food Prot. 2006, 69, 2992–2997. [Google Scholar] [CrossRef]
  72. Donkor, O.; Henriksson, A.; Singh, T.; Vasiljevic, T.; Shah, N.P. ACE-Inhibitory Activity of Probiotic Yoghurt. Int. Dairy J. 2007, 17, 1321–1331. [Google Scholar] [CrossRef]
  73. Azevedo, R.A.; Ferreira, A.K.; Auada, A.V.V.; Pasqualoto, K.F.M.; Marques-Porto, R.; Maria, D.A.; Lebrun, I. Antitumor Effect of Cationic INKKI Peptide from Bovine β-Casein on Melanoma B16F10. J. Cancer Ther. 2012, 3. [Google Scholar] [CrossRef]
  74. Lebrun, I.; Cavallaro, V.; Juliano, L.; Juliano, M.A.; de Sousa e Silva, M.C.C. Effects Of′ Casoparan′, a Peptide Isolated from Casein Hydrolysates with Mastoparan-like Properties. Mediat. Inflamm. 2004, 13, 263–268. [Google Scholar] [CrossRef]
  75. Yamamoto, N.; Akino, A.; Takano, T. Antihypertensive Effect of the Peptides Derived from Casein by an Extracellular Proteinase from Lactobacillus helveticus CP790. J. Dairy Sci. 1994, 77, 917–922. [Google Scholar] [CrossRef]
  76. Kohmura, M.; Nio, N.; Ariyoshi, Y. Inhibition of Angiotensin-Converting Enzyme by Synthetic Peptide Fragments of Various β-Caseins. Agric. Biol. Chem. 1990, 54, 1101–1102. [Google Scholar] [CrossRef] [PubMed]
  77. Asano, M.; Nio, N.; Ariyoshi, Y. Inhibition of Prolyl Endopeptidase by Synthetic β-Casein Peptides and Their Derivatives with a C-Terminal Prolinol or Prolinal. Biosci. Biotechnol. Biochem. 1992, 56, 976–977. [Google Scholar] [CrossRef]
  78. Minervini, F.; Algaron, F.; Rizzello, C.G.; Fox, P.; Monnet, V.; Gobbetti, M. Angiotensin I-Converting-Enzyme-Inhibitory and Antibacterial Peptides from Lactobacillus helveticus PR4 Proteinase-Hydrolyzed Caseins of Milk from Six Species. Appl. Environ. Microbiol. 2003, 69, 5297–5305. [Google Scholar] [CrossRef]
  79. Otte, J.; Shalaby, S.M.; Zakora, M.; Nielsen, M.S. Fractionation and Identification of ACE-Inhibitory Peptides from α-Lactalbumin and β-Casein Produced by Thermolysin-Catalysed Hydrolysis. Int. Dairy J. 2007, 17, 1460–1472. [Google Scholar] [CrossRef]
  80. Quirós, A.; Ramos, M.; Muguerza, B.; Delgado, M.A.; Miguel, M.; Aleixandre, A.; Recio, I. Identification of Novel Antihypertensive Peptides in Milk Fermented with Enterococcus faecalis. Int. Dairy J. 2007, 17, 33–41. [Google Scholar] [CrossRef]
  81. Pihlanto, A.; Virtanen, T.; Korhonen, H. Angiotensin I Converting Enzyme (ACE) Inhibitory Activity and Antihypertensive Effect of Fermented Milk. Int. Dairy J. 2010, 20, 3–10. [Google Scholar] [CrossRef]
  82. Gonzalez-Gonzalez, C.; Gibson, T.; Jauregi, P. Novel Probiotic-Fermented Milk with Angiotensin I-Converting Enzyme Inhibitory Peptides Produced by Bifidobacterium bifidum MF 20/5. Int. J. Food Microbiol. 2013, 167, 131–137. [Google Scholar] [CrossRef]
  83. Nakano, D.; Ogura, K.; Miyakoshi, M.; Ishii, F.; Kawanishi, H.; Kurumazuka, D.; Kwak, C.; Ikemura, K.; Takaoka, M.; Moriguchi, S. Antihypertensive Effect of Angiotensin I-Converting Enzyme Inhibitory Peptides from a Sesame Protein Hydrolysate in Spontaneously Hypertensive Rats. Biosci. Biotechnol. Biochem. 2006, 70, 1118–1126. [Google Scholar] [CrossRef]
  84. Eisele, T.; Stressler, T.; Kranz, B.; Fischer, L. Bioactive Peptides Generated in an Enzyme Membrane Reactor Using Bacillus lentus Alkaline Peptidase. Eur. Food Res. Technol. 2013, 236, 483–490. [Google Scholar] [CrossRef]
  85. Abubakar, A.; Saito, T.; Kitazawa, H.; Kawai, Y.; Itoh, T. Structural Analysis of New Antihypertensive Peptides Derived from Cheese Whey Protein by Proteinase K Digestion. J. Dairy Sci. 1998, 81, 3131–3138. [Google Scholar] [CrossRef]
  86. Chang, K.-J.; Su, Y.F.; Brent, D.A.; Chang, J. Isolation of a Specific Mu-Opiate Receptor Peptide, Morphiceptin, from an Enzymatic Digest of Milk Proteins. J. Biol. Chem. 1985, 260, 9706–9712. [Google Scholar] [CrossRef] [PubMed]
  87. FitzGerald, R.; Meisel, H. Milk Protein Hydrolysates and Bioactive Peptides. In Advanced Dairy Chemistry—1 Proteins: Part A/Part B; Springer: Berlin/Heidelberg, Germany, 2003; pp. 675–698. [Google Scholar]
  88. Meisel, H. Chemical Characterization and Opioid Activity of an Exorphin Isolated from In Vivo Digests of Casein. FEBS Lett. 1986, 196, 223–227. [Google Scholar] [CrossRef] [PubMed]
  89. Garg, S.; Apostolopoulos, V.; Nurgali, K.; Mishra, V.K. Evaluation of in Silico Approach for Prediction of Presence of Opioid Peptides in Wheat. J. Funct. Foods 2018, 41, 34–40. [Google Scholar] [CrossRef]
  90. Meisel, H.; Frister, H.; Schlimme, E. Biologically Active Peptides in Milk Proteins. Z. Für Ernährungswiss. 1989, 28, 267–278. [Google Scholar] [CrossRef]
  91. Sienkiewicz-Szłapka, E.; Jarmołowska, B.; Krawczuk, S.; Kostyra, E.; Kostyra, H.; Iwan, M. Contents of Agonistic and Antagonistic Opioid Peptides in Different Cheese Varieties. Int. Dairy J. 2009, 19, 258–263. [Google Scholar] [CrossRef]
  92. Haq, M.R.U.; Kapila, R.; Saliganti, V. Consumption of β-Casomorphins-7/5 Induce Inflammatory Immune Response in Mice Gut Through Th2 Pathway. J. Funct. Foods 2014, 8, 150–160. [Google Scholar] [CrossRef]
  93. Claustre, J.; Toumi, F.; Trompette, A.; Jourdan, G.; Guignard, H.; Chayvialle, J.A.; Plaisancié, P. Effects of Peptides Derived from Dietary Proteins on Mucus Secretion in Rat Jejunum. Am. J. Physiol.-Gastrointest. Liver Physiol. 2002, 283, G521–G528. [Google Scholar] [CrossRef] [PubMed]
  94. Osborne, S.; Chen, W.; Addepalli, R.; Colgrave, M.; Singh, T.; Tran, C.; Day, L. In Vitro Transport and Satiety of a Beta-Lactoglobulin Dipeptide and Beta-Casomorphin-7 and Its Metabolites. Food Funct. 2014, 5, 2706–2718. [Google Scholar] [CrossRef]
  95. Dubynin, V.; Asmakova, L.; Sokhanenkova, N.Y.; Bespalova, Z.D.; Nezavibat’ko, V.; Kamenskii, A. Comparative Analysis of Neurotropic Activity of Exorphines, Derivatives of Dietary Proteins. Bull. Exp. Biol. Med. 1998, 125, 131–134. [Google Scholar] [CrossRef]
  96. Martínez-Maqueda, D.; Miralles, B.; De Pascual-Teresa, S.; Reverón, I.; Muñoz, R.; Recio, I. Food-Derived Peptides Stimulate Mucin Secretion and Gene Expression in Intestinal Cells. J. Agric. Food Chem. 2012, 60, 8600–8605. [Google Scholar] [CrossRef]
  97. Trompette, A.; Claustre, J.; Caillon, F.; Jourdan, G.; Chayvialle, J.A.; Plaisancie, P. Milk Bioactive Peptides and β-Casomorphins Induce Mucus Release in Rat Jejunum. J. Nutr. 2003, 133, 3499–3503. [Google Scholar] [CrossRef]
  98. Kayser, H.; Meisel, H. Stimulation of Human Peripheral Blood Lymphocytes by Bioactive Peptides Derived from Bovine Milk Proteins. FEBS Lett. 1996, 383, 18–20. [Google Scholar] [CrossRef]
  99. Hatzoglou, A.; Bakogeorgou, E.; Hatzoglou, C.; Martin, P.-M.; Castanas, E. Antiproliferative and Receptor Binding Properties of α-and β-Casomorphins in the T47D Human Breast Cancer Cell Line. Eur. J. Pharmacol. 1996, 310, 217–223. [Google Scholar] [CrossRef]
  100. Brantl, V.; Teschemacher, H.; Bläsig, J.; Henschen, A.; Lottspeich, F. Opioid Activities of β-Casomorphins. Life Sci. 1981, 28, 1903–1909. [Google Scholar] [CrossRef]
  101. Gach, K.; Do-Rego, J.C.; Fichna, J.; Storr, M.; Delbro, D.; Toth, G.; Janecka, A. Synthesis and Biological Evaluation of Novel Peripherally Active Morphiceptin Analogs. Peptides 2010, 31, 1617–1624. [Google Scholar] [CrossRef]
  102. Jinsmaa, Y.; Yoshikawa, M. Enzymatic Release of Neocasomorphin and β-Casomorphin from Bovine β-Casein. Peptides 1999, 20, 957–962. [Google Scholar] [CrossRef]
  103. Saito, T.; Nakamura, T.; Kitazawa, H.; Kawai, Y.; Itoh, T. Isolation and Structural Analysis of Antihypertensive Peptides That Exist Naturally in Gouda Cheese. J. Dairy Sci. 2000, 83, 1434–1440. [Google Scholar] [CrossRef] [PubMed]
  104. Uenishi, H.; Kabuki, T.; Seto, Y.; Serizawa, A.; Nakajima, H. Isolation and Identification of Casein-Derived Dipeptidyl-Peptidase 4 (DPP-4)-Inhibitory Peptide LPQNIPPL from Gouda-Type Cheese and Its Effect on Plasma Glucose in Rats. Int. Dairy J. 2012, 22, 24–30. [Google Scholar] [CrossRef]
  105. Wang, W.; Gu, F.; Wei, C.; Tang, Y.; Zheng, X.; Ren, M.; Qin, Y. PGPIPN, a Therapeutic Hexapeptide, Suppressed Human Ovarian Cancer Growth by Targeting BCL2. PLoS ONE 2013, 8, e60701. [Google Scholar] [CrossRef]
  106. Migliore-Samour, D.; Floc’h, F.; Jolles, P. Biologically Active Casein Peptides Implicated in Immunomodulation. J. Dairy Res. 1989, 56, 357–362. [Google Scholar] [CrossRef] [PubMed]
  107. Migliore-Samour, D.; Jolles, P. Casein, a Prohormone with an Immunomodulating Role for the Newborn? Experientia 1988, 44, 188–193. [Google Scholar] [CrossRef]
  108. Zhao, M.; Wei, C.; Yang, X.; Zhou, J.; Wang, J.; Gu, F.; Lei, T.; Qin, Y. The Milk-Derived Hexapeptide PGPIPN Inhibits the Invasion and Migration of Human Ovarian Cancer Cells by Regulating the Expression of MTA1 and NM23H1 Genes. Int. J. Oncol. 2016, 48, 1721–1729. [Google Scholar] [CrossRef]
  109. Qi, N.; Liu, C.; Yang, H.; Shi, W.; Wang, S.; Zhou, Y.; Wei, C.; Gu, F.; Qin, Y. Therapeutic Hexapeptide (PGPIPN) Prevents and Cures Alcoholic Fatty Liver Disease by Affecting the Expressions of Genes Related with Lipid Metabolism and Oxidative Stress. Oncotarget 2017, 8, 88079. [Google Scholar] [CrossRef]
  110. Gu, F.; Xi, H.; Ruan, X.; Xu, Q.; Wang, S.; Qin, Y.-D. Protective Effect of Immunomodulating Peptide (PGPIPN) Derived from Beta-Casomorphin in Bovine Milk on Acute Alcohol-Induced Liver Injury. Chin. Pharmacol. Bull. 2018, 712–716. [Google Scholar] [CrossRef]
  111. Norris, R.; Poyarkov, A.; O’Keeffe, M.B.; FitzGerald, R.J. Characterisation of the Hydrolytic Specificity of Aspergillus niger Derived Prolyl Endoproteinase on Bovine β-Casein and Determination of ACE Inhibitory Activity. Food Chem. 2014, 156, 29–36. [Google Scholar] [CrossRef]
  112. Quirós, A.; del Mar Contreras, M.; Ramos, M.; Amigo, L.; Recio, I. Stability to Gastrointestinal Enzymes and Structure–Activity Relationship of β-Casein-Peptides with Antihypertensive Properties. Peptides 2009, 30, 1848–1853. [Google Scholar] [CrossRef] [PubMed]
  113. Narva, M.; Rissanen, J.; Halleen, J.; Vapaatalo, H.; Väänänen, K.; Korpela, R. Effects of Bioactive Peptide, Valyl-Prolyl-Proline (VPP), and Lactobacillus helveticus Fermented Milk Containing VPP on Bone Loss in Ovariectomized Rats. Ann. Nutr. Metab. 2007, 51, 65–74. [Google Scholar] [CrossRef] [PubMed]
  114. Cicero, A.F.; Rosticci, M.; Veronesi, M.; Bacchelli, S.; Strocchi, E.; Melegari, C.; Grandi, E.; Borghi, C. Hemodynamic Effects of Lactotripeptides from Casein Hydrolysate in Mediterranean Normotensive Subjects and Patients with High-Normal Blood Pressure: A Randomized, Double-Blind, Crossover Clinical Trial. J. Med. Food 2010, 13, 1363–1368. [Google Scholar] [CrossRef]
  115. Jäkälä, P.; Turpeinen, A.M.; Rajakari, K.; Korpela, R.; Vapaatalo, H. Biological Effects of Casein-Derived Tripeptide Powders Are Not Affected by Fermentation Process. Int. Dairy J. 2010, 20, 366–370. [Google Scholar] [CrossRef]
  116. Yamada, A.; Sakurai, T.; Ochi, D.; Mitsuyama, E.; Yamauchi, K.; Abe, F. Antihypertensive Effect of the Bovine Casein-Derived Peptide Met-Lys-Pro. Food Chem. 2015, 172, 441–446. [Google Scholar] [CrossRef]
  117. Jing, P.; Qian, B.; He, Y.; Zhao, X.; Zhang, J.; Zhao, D.; Lv, Y.; Deng, Y. Screening Milk-Derived Antihypertensive Peptides Using Quantitative Structure Activity Relationship (QSAR) Modelling and In Vitro/In Vivo Studies on Their Bioactivity. Int. Dairy J. 2014, 35, 95–101. [Google Scholar] [CrossRef]
  118. Chakrabarti, S.; Jahandideh, F.; Davidge, S.T.; Wu, J. Milk-Derived Tripeptides IPP (Ile-Pro-Pro) and VPP (Val-Pro-Pro) Enhance Insulin Sensitivity and Prevent Insulin Resistance in 3T3-F442A Preadipocytes. J. Agric. Food Chem. 2018, 66, 10179–10187. [Google Scholar] [CrossRef]
  119. Chakrabarti, S.; Liao, W.; Davidge, S.T.; Wu, J. Milk-Derived Tripeptides IPP (Ile-Pro-Pro) and VPP (Val-Pro-Pro) Differentially Modulate Angiotensin II Effects on Vascular Smooth Muscle Cells. J. Funct. Foods 2017, 30, 151–158. [Google Scholar] [CrossRef]
  120. Aihara, K.; Osaka, M.; Yoshida, M. Oral Administration of the Milk Casein-Derived Tripeptide Val-Pro-Pro Attenuates High-Fat Diet-Induced Adipose Tissue Inflammation in Mice. Br. J. Nutr. 2014, 112, 513–519. [Google Scholar] [CrossRef]
  121. Chakrabarti, S.; Wu, J. Milk-Derived Tripeptides IPP (Ile-Pro-Pro) and VPP (Val-Pro-Pro) Promote Adipocyte Differentiation and Inhibit Inflammation in 3T3-F442A Cells. PLoS ONE 2015, 10, e0117492. [Google Scholar] [CrossRef]
  122. Nongonierma, A.B.; FitzGerald, R.J. Inhibition of Dipeptidyl Peptidase IV (DPP-IV) by Proline Containing Casein-Derived Peptides. J. Funct. Foods 2013, 5, 1909–1917. [Google Scholar] [CrossRef]
  123. Bruno, J.; Nicolas, A.; Pesenti, S.; Schwarz, J.; Simon, J.-L.; Léonil, J.; Plaisancié, P. Variants of β-Casofensin, a Bioactive Milk Peptide, Differently Modulate the Intestinal Barrier: In Vivo and Ex Vivo Studies in Rats. J. Dairy Sci. 2017, 100, 3360–3372, Erratum in J. Dairy Sci. 2018, 101, 5667. [Google Scholar] [CrossRef]
  124. Plaisancié, P.; Boutrou, R.; Estienne, M.; Henry, G.; Jardin, J.; Paquet, A.; Léonil, J. β-Casein (94-123)-Derived Peptides Differently Modulate Production of Mucins in Intestinal Goblet Cells. J. Dairy Res. 2015, 82, 36–46. [Google Scholar] [CrossRef]
  125. Plaisancié, P.; Claustre, J.; Estienne, M.; Henry, G.; Boutrou, R.; Paquet, A.; Léonil, J. A Novel Bioactive Peptide from Yoghurts Modulates Expression of the Gel-Forming MUC2 Mucin as Well as Population of Goblet Cells and Paneth Cells along the Small Intestine. J. Nutr. Biochem. 2013, 24, 213–221. [Google Scholar] [CrossRef]
  126. Bessette, C.; Benoit, B.; Sekkal, S.; Bruno, J.; Estienne, M.; Léonil, J.; Ferrier, L.; Théodorou, V.; Plaisancié, P. Protective Effects of Β-casofensin, a Bioactive Peptide from Bovine Β-casein, against Indomethacin-induced Intestinal Lesions in Rats. Mol. Nutr. Food Res. 2016, 60, 823–833. [Google Scholar] [CrossRef]
  127. Rival, S.G.; Boeriu, C.G.; Wichers, H.J. Caseins and Casein Hydrolysates. 2. Antioxidative Properties and Relevance to Lipoxygenase Inhibition. J. Agric. Food Chem. 2001, 49, 295–302. [Google Scholar] [CrossRef]
  128. Pan, M.; Huo, Y.; Wang, C.; Zhang, Y.; Dai, Z.; Li, B. Positively Charged Peptides from Casein Hydrolysate Show Strong Inhibitory Effects on LDL Oxidation and Cellular Lipid Accumulation in Raw264.7 Cells. Int. Dairy J. 2019, 91, 119–128. [Google Scholar] [CrossRef]
  129. Miao, J.; Liao, W.; Pan, Z.; Wang, Q.; Duan, S.; Xiao, S.; Yang, Z.; Cao, Y. Isolation and Identification of Iron-Chelating Peptides from Casein Hydrolysates. Food Funct. 2019, 10, 2372–2381. [Google Scholar] [CrossRef]
  130. Pihlanto-Leppälä, A.; Rokka, T.; Korhonen, H. Angiotensin I Converting Enzyme Inhibitory Peptides Derived from Bovine Milk Proteins. Int. Dairy J. 1998, 8, 325–331. [Google Scholar] [CrossRef]
  131. Perpetuo, E.A.; Juliano, L.; Lebrun, I. Biochemical and Pharmacological Aspects of Two Bradykinin-Potentiating Peptides Obtained from Tryptic Hydrolysis of Casein. J. Protein Chem. 2003, 22, 601–606. [Google Scholar] [CrossRef]
  132. Hayes, M.; Stanton, C.; Slattery, H.; O’sullivan, O.; Hill, C.; Fitzgerald, G.; Ross, R. Casein Fermentate of Lactobacillus Animalis DPC6134 Contains a Range of Novel Propeptide Angiotensin-Converting Enzyme Inhibitors. Appl. Environ. Microbiol. 2007, 73, 4658–4667. [Google Scholar] [CrossRef]
  133. Nongonierma, A.B.; Paolella, S.; Mudgil, P.; Maqsood, S.; FitzGerald, R.J. Identification of Novel Dipeptidyl Peptidase IV (DPP-IV) Inhibitory Peptides in Camel Milk Protein Hydrolysates. Food Chem. 2018, 244, 340–348. [Google Scholar] [CrossRef]
  134. Sanchez-Rivera, L.; Ares, I.; Miralles, B.; Gómez-Ruiz, J.Á.; Recio, I.; Martínez-Larrañaga, M.R.; Anadon, A.; Martínez, M.A. Bioavailability and Kinetics of the Antihypertensive Casein-Derived Peptide HLPLP in Rats. J. Agric. Food Chem. 2014, 62, 11869–11875. [Google Scholar] [CrossRef]
  135. Sánchez-Rivera, L.; Santos, P.F.; Miralles, B.; Carrón, R.; Montero, M.J.; Recio, I. Peptide Fragments from β-Casein f (134–138), HLPLP, Generated by the Action of Rat Blood Plasma Peptidases Show Potent Antihypertensive Activity. Food Res. Int. 2016, 88, 348–353. [Google Scholar] [CrossRef]
  136. Robert, M.-C.; Razaname, A.; Mutter, M.; Juillerat, M.A. Identification of Angiotensin-I-Converting Enzyme Inhibitory Peptides Derived from Sodium Caseinate Hydrolysates Produced by Lactobacillus helveticus NCC 2765. J. Agric. Food Chem. 2004, 52, 6923–6931. [Google Scholar] [CrossRef]
  137. Nongonierma, A.B.; FitzGerald, R.J. Susceptibility of Milk Protein-Derived Peptides to Dipeptidyl Peptidase IV (DPP-IV) Hydrolysis. Food Chem. 2014, 145, 845–852. [Google Scholar] [CrossRef]
  138. Maeno, M.; Yamamoto, N.; Takano, T. Identification of an Antihypertensive Peptide from Casein Hydrolysate Produced by a Proteinase from Lactobacillus helveticus CP790. J. Dairy Sci. 1996, 79, 1316–1321. [Google Scholar] [CrossRef]
  139. Phelan, M.; Khaldi, N.; Shields, D.C.; Kerins, D.M. Angiotensin Converting Enzyme and Nitric Oxide Inhibitory Activities of Novel Milk Derived Peptides. Int. Dairy J. 2014, 35, 38–42. [Google Scholar] [CrossRef]
  140. Liu, D.; Sun, H.; Zhang, L.; Li, S.; Qin, Z. High-Level Expression of Milk-Derived Antihypertensive Peptide in Escherichia coli and Its Bioactivity. J. Agric. Food Chem. 2007, 55, 5109–5112, Erratum in J. Agric. Food Chem. 2008, 56, 1524. [Google Scholar] [CrossRef]
  141. Kumar, N.; Devi, S.; Mada, S.B.; Reddi, S.; Kapila, R.; Kapila, S. Anti-Apoptotic Effect of Buffalo Milk Casein Derived Bioactive Peptide by Directing Nrf2 Regulation in Starving Fibroblasts. Food Biosci. 2020, 35, 100566. [Google Scholar] [CrossRef]
  142. Maruyama, S.; Nakagomi, K.; Tomizuka, N.; Suzuki, H. Angiotensin I-Converting Enzyme Inhibitor Derived from an Enzymatic Hydrolysate of Casein. II. Isolation and Bradykinin-Potentiating Activity on the Uterus and the Ileum of Rats. Agric. Biol. Chem. 1985, 49, 1405–1409. [Google Scholar]
  143. Maruyama, S.; Mitachi, H.; Tanaka, H.; Tomizuka, N.; Suzuki, H. Studies on the Active Site and Antihypertensive Activity of Angiotensin I-Converting Enzyme Inhibitors Derived from Casein. Agric. Biol. Chem. 1987, 51, 1581–1586. [Google Scholar] [CrossRef]
  144. Tsopmo, A.; Romanowski, A.; Banda, L.; Lavoie, J.C.; Jenssen, H.; Friel, J.K. Novel Anti-Oxidative Peptides from Enzymatic Digestion of Human Milk. Food Chem. 2011, 126, 1138–1143. [Google Scholar] [CrossRef]
  145. Hernandez-Ledesma, B.; Quiros, A.; Amigo, L.; Recio, I. Identification of Bioactive Peptides after Digestion of Human Milk and Infant Formula with Pepsin and Pancreatin. Int. Dairy J. 2007, 17, 42–49. [Google Scholar] [CrossRef]
  146. Vukic, V.R.; Vukic, D.V.; Milanovic, S.D.; Ilicic, M.D.; Kanuric, K.G.; Johnson, M.S. In Silico Identification of Milk Antihypertensive Di-and Tripeptides Involved in Angiotensin I–Converting Enzyme Inhibitory Activity. Nutr. Res. 2017, 46, 22–30. [Google Scholar] [CrossRef]
  147. Malinowski, J.; Klempt, M.; Clawin-Rädecker, I.; Lorenzen, P.C.; Meisel, H. Identification of a NFκB Inhibitory Peptide from Tryptic β-Casein Hydrolysate. Food Chem. 2014, 165, 129–133. [Google Scholar] [CrossRef]
  148. Paiva, L.; Lima, E.; Neto, A.I.; Baptista, J. Isolation and Characterization of Angiotensin I-Converting Enzyme (ACE) Inhibitory Peptides from Ulva rigida C. Agardh Protein Hydrolysate. J. Funct. Foods 2016, 26, 65–76. [Google Scholar] [CrossRef]
  149. Berthou, J.; Migliore-Samour, D.; Lifchitz, A.; Delettré, J.; Floc’h, F.; Jollès, P. Immunostimulating Properties and Three-Dimensional Structure of Two Tripeptides from Human and Cow Caseins. FEBS Lett. 1987, 218, 55–58. [Google Scholar] [CrossRef]
  150. Coste, M.; Rochet, V.; Léonil, J.; Mollé, D.; Bouhallab, S.; Tomé, D. Identification of C-Terminal Peptides of Bovine β-Casein That Enhance Proliferation of Rat Lymphocytes. Immunol. Lett. 1992, 33, 41–46. [Google Scholar] [CrossRef] [PubMed]
  151. Sandré, C.; Gleizes, A.; Forestier, F.; Gorges-Kergot, R.; Chilmonczyk, S.; Léonil, J.; Moreau, M.-C.; Labarre, C. A Peptide Derived from Bovine β-Casein Modulates Functional Properties of Bone Marrow-Derived Macrophages from Germfree and Human Flora-Associated Mice. J. Nutr. 2001, 131, 2936–2942. [Google Scholar] [CrossRef]
  152. Rojas-Ronquillo, R.; Cruz-Guerrero, A.; Flores-Nájera, A.; Rodríguez-Serrano, G.; Gómez-Ruiz, L.; Reyes-Grajeda, J.P.; Jiménez-Guzmán, J.; García-Garibay, M. Antithrombotic and Angiotensin-Converting Enzyme Inhibitory Properties of Peptides Released from Bovine Casein by Lactobacillus casei Shirota. Int. Dairy J. 2012, 26, 147–154. [Google Scholar] [CrossRef]
  153. Lu, Y.; Govindasamy-Lucey, S.; Lucey, J. Angiotensin-I-Converting Enzyme-Inhibitory Peptides in Commercial Wisconsin Cheddar Cheeses of Different Ages. J. Dairy Sci. 2016, 99, 41–52. [Google Scholar] [CrossRef]
  154. Sowmya, K.; Bhat, M.I.; Bajaj, R.K.; Kapila, S.; Kapila, R. Buffalo Milk Casein Derived Decapeptide (YQEPVLGPVR) Having Bifunctional Anti-Inflammatory and Antioxidative Features under Cellular Milieu. Int. J. Pept. Res. Ther. 2019, 25, 623–633. [Google Scholar] [CrossRef]
  155. Zhou, J.; Ma, L.; Xu, H.; Gao, Y.; Jin, Y.; Zhao, L.; David, X.A.L.; Zhan, D.; Zhang, S. Immunomodulating Effects of Casein-Derived Peptides QEPVL and QEPV on Lymphocytes In Vitro and In Vivo. Food Funct. 2014, 5, 2061–2069. [Google Scholar] [CrossRef]
  156. Zhao, H.; Zhou, F.; Wang, L.; Bai, F.; Dziugan, P.; Walczak, P.; Zhang, B. Characterization of a Bioactive Peptide with Cytomodulatory Effect Released from Casein. Eur. Food Res. Technol. 2014, 238, 315–322. [Google Scholar] [CrossRef]
  157. Vermeirssen, V.; van der Bent, A.; Van Camp, J.; van Amerongen, A.; Verstraete, W. A Quantitative in Silico Analysis Calculates the Angiotensin I Converting Enzyme (ACE) Inhibitory Activity in Pea and Whey Protein Digests. Biochimie 2004, 86, 231–239. [Google Scholar] [CrossRef] [PubMed]
  158. Aihara, K.; Ishii, H.; Yoshida, M. Casein-Derived Tripeptide, Val-Pro-Pro (VPP), Modulates Monocyte Adhesion to Vascular Endothelium. J. Atheroscler. Thromb. 2009, 16, 594–603. [Google Scholar] [CrossRef] [PubMed]
  159. Gómez-Ruiz, J.Á.; Ramos, M.; Recio, I. Angiotensin-Converting Enzyme-Inhibitory Peptides in Manchego Cheeses Manufactured with Different Starter Cultures. Int. Dairy J. 2002, 12, 697–706. [Google Scholar] [CrossRef]
  160. Hashimoto, A.; Aoyagi, H.; Izumiya, N. Synthetic Identification of Bitter Heptapeptide in Tryptic Hydrolysate of Casein. Bull. Chem. Soc. Jpn. 1980, 53, 2926–2928. [Google Scholar] [CrossRef]
  161. Hernández-Ledesma, B.; Amigo, L.; Ramos, M.; Recio, I. Angiotensin Converting Enzyme Inhibitory Activity in Commercial Fermented Products. Formation of Peptides under Simulated Gastrointestinal Digestion. J. Agric. Food Chem. 2004, 52, 1504–1510. [Google Scholar] [CrossRef]
  162. Lee, H.S.; Lee, K.J. Cathepsin B Inhibitory Peptides Derived from β-Casein. Peptides 2000, 21, 807–809. [Google Scholar] [CrossRef]
  163. Gómez-Ruiz, J.A.; Taborda, G.; Amigo, L.; Recio, I.; Ramos, M. Identification of ACE-Inhibitory Peptides in Different Spanish Cheeses by Tandem Mass Spectrometry. Eur. Food Res. Technol. 2006, 223, 595–601. [Google Scholar] [CrossRef]
  164. Wessels, S.; Axelsson, L.; Hansen, E.B.; De Vuyst, L.; Laulund, S.; Lähteenmäki, L.; Lindgren, S.; Mollet, B.; Salminen, S.; von Wright, A. The Lactic Acid Bacteria, the Food Chain, and Their Regulation. Trends Food Sci. Technol. 2004, 15, 498–505. [Google Scholar] [CrossRef]
  165. Fuquay, J.W.; McSweeney, P.L.; Fox, P.F. Encyclopedia of Dairy Sciences; Academic Press: Cambridge, MA, USA, 2011; ISBN 0-12-374407-5. [Google Scholar]
  166. Ren, C.; Cheng, L.; Sun, Y.; Zhang, Q.; de Haan, B.J.; Zhang, H.; Faas, M.M.; de Vos, P. Lactic Acid Bacteria Secrete Toll like Receptor 2 Stimulating and Macrophage Immunomodulating Bioactive Factors. J. Funct. Foods 2020, 66, 103783. [Google Scholar] [CrossRef]
  167. Ren, C.; Dokter-Fokkens, J.; Figueroa Lozano, S.; Zhang, Q.; de Haan, B.J.; Zhang, H.; Faas, M.M.; de Vos, P. Lactic Acid Bacteria May Impact Intestinal Barrier Function by Modulating Goblet Cells. Mol. Nutr. Food Res. 2018, 62, 1700572. [Google Scholar] [CrossRef]
  168. Venegas-Ortega, M.G.; Flores-Gallegos, A.C.; Martínez-Hernández, J.L.; Aguilar, C.N.; Nevárez-Moorillón, G.V. Production of Bioactive Peptides from Lactic Acid Bacteria: A Sustainable Approach for Healthier Foods. Compr. Rev. Food Sci. Food Saf. 2019, 18, 1039–1051. [Google Scholar] [CrossRef]
  169. Haandrikman, A.J.; Kok, J.; Venema, G. Lactococcal Proteinase Maturation Protein PrtM Is a Lipoprotein. J. Bacteriol. 1991, 173, 4517–4525. [Google Scholar] [CrossRef]
  170. Holck, A.; Næs, H. Cloning, Sequencing and Expression of the Gene Encoding the Cell-Envelope-Associated Proteinase from Lactobacillus paracasei subsp. Paracasei NCDO 151. Microbiology 1992, 138, 1353–1364. [Google Scholar] [CrossRef]
  171. Siezen, R.J. Multi-Domain, Cell-Envelope Proteinases of Lactic Acid Bacteria. In Lactic Acid Bacteria: Genetics, Metabolism and Applications, Proceedings of the Sixth Symposium on Lactic Acid Bacteria: Genetics, Metabolism and Applications, Veldhoven, The Netherlands, 19–23 September 1999; Konings, W.N., Kuipers, O.P., In ’t Veld, J.H.J.H., Eds.; Springer: Dordrecht, The Netherlands, 1999; pp. 139–155. ISBN 978-94-017-2027-4. [Google Scholar]
  172. Kieliszek, M.; Pobiega, K.; Piwowarek, K.; Kot, A.M. Characteristics of the Proteolytic Enzymes Produced by Lactic Acid Bacteria. Molecules 2021, 26, 1858. [Google Scholar] [CrossRef]
  173. Doeven, M.K.; Kok, J.; Poolman, B. Specificity and Selectivity Determinants of Peptide Transport in Lactococcus lactis and Other Microorganisms. Mol. Microbiol. 2005, 57, 640–649. [Google Scholar] [CrossRef] [PubMed]
  174. Tellez, A.; Corredig, M.; Brovko, L.Y.; Griffiths, M.W. Characterization of Immune-Active Peptides Obtained from Milk Fermented by Lactobacillus helveticus. J. Dairy Res. 2010, 77, 129–136. [Google Scholar] [CrossRef]
  175. Kok, J.; van Gijtenbeek, L.A.; de Jong, A.; van der Meulen, S.B.; Solopova, A.; Kuipers, O.P. The Evolution of Gene Regulation Research in Lactococcus lactis. FEMS Microbiol. Rev. 2017, 41, S220–S243. [Google Scholar] [CrossRef] [PubMed]
  176. de Ruyter, P.G.; Kuipers, O.P.; de Vos, W.M. Controlled Gene Expression Systems for Lactococcus lactis with the Food-Grade Inducer Nisin. Appl. Environ. Microbiol. 1996, 62, 3662–3667. [Google Scholar] [CrossRef] [PubMed]
  177. Mu, D.; Montalbán-López, M.; Masuda, Y.; Kuipers, O.P. Zirex: A Novel Zinc-Regulated Expression System for Lactococcus lactis. Appl. Environ. Microbiol. 2013, 79, 4503–4508. [Google Scholar] [CrossRef]
  178. Miyoshi, A.; Jamet, E.; Commissaire, J.; Renault, P.; Langella, P.; Azevedo, V. A Xylose-Inducible Expression System for Lactococcus lactis. FEMS Microbiol. Lett. 2004, 239, 205–212. [Google Scholar] [CrossRef]
  179. Linares, D.M.; Alvarez-Sieiro, P.; del Rio, B.; Ladero, V.; Redruello, B.; Martin, M.C.; Fernandez, M.; Alvarez, M.A. Implementation of the Agmatine-Controlled Expression System for Inducible Gene Expression in Lactococcus lactis. Microb. Cell Factories 2015, 14, 208. [Google Scholar] [CrossRef]
  180. Leenhouts, K.; Buist, G.; Bolhuis, A.; Ten Berge, A.; Kiel, J.; Mierau, I.; Dabrowska, M.; Venema, G.; Kok, J. A General System for Generating Unlabelled Gene Replacements in Bacterial Chromosomes. Mol. Gen. Genet. MGG 1996, 253, 217–224. [Google Scholar] [CrossRef]
  181. Solem, C.; Defoor, E.; Jensen, P.R.; Martinussen, J. Plasmid pCS1966, a New Selection/Counterselection Tool for Lactic Acid Bacterium Strain Construction Based on the oroP Gene, Encoding an Orotate Transporter from Lactococcus lactis. Appl. Environ. Microbiol. 2008, 74, 4772–4775. [Google Scholar] [CrossRef] [PubMed]
  182. Van Pijkeren, J.-P.; Britton, R.A. High Efficiency Recombineering in Lactic Acid Bacteria. Nucleic Acids Res. 2012, 40, e76. [Google Scholar] [CrossRef]
  183. Barrangou, R.; van Pijkeren, J.-P. Exploiting CRISPR–Cas Immune Systems for Genome Editing in Bacteria. Curr. Opin. Biotechnol. 2016, 37, 61–68. [Google Scholar] [CrossRef]
  184. Jiang, Y.; Chen, B.; Duan, C.; Sun, B.; Yang, J.; Yang, S. Multigene Editing in the Escherichia coli Genome via the CRISPR-Cas9 System. Appl. Environ. Microbiol. 2015, 81, 2506–2514, Erratum in Appl. Environ. Microbiol. 2016, 82, e01181-16. [Google Scholar] [CrossRef]
  185. Selle, K.; Klaenhammer, T.R.; Barrangou, R. CRISPR-Based Screening of Genomic Island Excision Events in Bacteria. Proc. Natl. Acad. Sci. USA 2015, 112, 8076–8081. [Google Scholar] [CrossRef] [PubMed]
  186. Wang, Y.; Zhang, Z.-T.; Seo, S.-O.; Lynn, P.; Lu, T.; Jin, Y.-S.; Blaschek, H.P. Bacterial Genome Editing with CRISPR-Cas9: Deletion, Integration, Single Nucleotide Modification, and Desirable “Clean” Mutant Selection in Clostridium beijerinckii as an Example. ACS Synth. Biol. 2016, 5, 721–732. [Google Scholar] [CrossRef]
  187. Westbrook, A.W.; Moo-Young, M.; Chou, C.P. Development of a CRISPR-Cas9 Tool Kit for Comprehensive Engineering of Bacillus subtilis. Appl. Environ. Microbiol. 2016, 82, 4876–4895. [Google Scholar] [CrossRef]
  188. Oh, J.-H.; van Pijkeren, J.-P. CRISPR–Cas9-Assisted Recombineering in Lactobacillus reuteri. Nucleic Acids Res. 2014, 42, e131. [Google Scholar] [CrossRef] [PubMed]
  189. Berlec, A.; Škrlec, K.; Kocjan, J.; Olenic, M.; Štrukelj, B. Single Plasmid Systems for Inducible Dual Protein Expression and for CRISPR-Cas9/CRISPRi Gene Regulation in Lactic Acid Bacterium Lactococcus lactis. Sci. Rep. 2018, 8, 1009. [Google Scholar] [CrossRef]
  190. Framson, P.E.; Nittayajarn, A.; Merry, J.; Youngman, P.; Rubens, C.E. New Genetic Techniques for Group B Streptococci: High-Efficiency Transformation, Maintenance of Temperature-Sensitive pWV01 Plasmids, and Mutagenesis with Tn917. Appl. Environ. Microbiol. 1997, 63, 3539–3547. [Google Scholar] [CrossRef]
  191. Song, X.; Huang, H.; Xiong, Z.; Ai, L.; Yang, S. CRISPR-Cas9D10A Nickase-Assisted Genome Editing in Lactobacillus casei. Appl. Environ. Microbiol. 2017, 83, e01259-17, Erratum in Appl. Environ. Microbiol. 2018, 84, e00146-18. [Google Scholar] [CrossRef] [PubMed]
  192. Wang, P.; Yi, Y.; Lü, X. CRISPR/Cas9-Based Genome Editing Platform for Companilactobacillus crustorum to Reveal the Molecular Mechanism of Its Probiotic Properties. J. Agric. Food Chem. 2021, 69, 15279–15289. [Google Scholar] [CrossRef]
  193. Goh, Y.J.; Barrangou, R. Portable CRISPR-Cas9N System for Flexible Genome Engineering in Lactobacillus acidophilus, Lactobacillus gasseri, and Lactobacillus paracasei. Appl. Environ. Microbiol. 2021, 87, e02669-20. [Google Scholar] [CrossRef] [PubMed]
  194. Hartmann, H.A.; Wilke, T.; Erdmann, R. Efficacy of Bacteriocin-Containing Cell-Free Culture Supernatants from Lactic Acid Bacteria to Control Listeria Monocytogenes in Food. Int. J. Food Microbiol. 2011, 146, 192–199. [Google Scholar] [CrossRef] [PubMed]
  195. Jo, D.-M.; Park, S.-K.; Khan, F.; Kang, M.-G.; Lee, J.-H.; Kim, Y.-M. An Approach to Extend the Shelf Life of Ribbonfish Fillet Using Lactic Acid Bacteria Cell-Free Culture Supernatant. Food Control 2021, 123, 107731. [Google Scholar] [CrossRef]
  196. Mao, Y.; Wang, Y.; Luo, X.; Chen, X.; Wang, G. Impact of Cell-Free Supernatant of Lactic Acid Bacteria on Staphylococcus aureus Biofilm and Its Metabolites. Front. Vet. Sci. 2023, 10, 1184989. [Google Scholar] [CrossRef]
  197. Liu, R.; Zhang, Y.; Zhai, G.; Fu, S.; Xia, Y.; Hu, B.; Cai, X.; Zhang, Y.; Li, Y.; Deng, Z.; et al. A Cell-Free Platform Based on Nisin Biosynthesis for Discovering Novel Lanthipeptides and Guiding Their Overproduction In Vivo. Adv. Sci. 2020, 7, 2001616. [Google Scholar] [CrossRef]
  198. O’Donnell, S.T.; Ross, R.P.; Stanton, C. The Progress of Multi-Omics Technologies: Determining Function in Lactic Acid Bacteria Using a Systems Level Approach. Front. Microbiol. 2020, 10, 3084. [Google Scholar] [CrossRef]
  199. Sun, Z.; Liu, L.; Zhang, H.; Wang, Y.; Liu, J.; Wang, Z.; Mu, D.; Wu, X.; Li, X. Multi-Omics Analysis of Metabolic Differences in Rape Bee Pollen Fermented by Single and Mixed Lactic Acid Bacterial Strains. Food Biosci. 2024, 62, 105401. [Google Scholar] [CrossRef]
  200. Shi, H.; An, F.; Lin, H.; Li, M.; Wu, J.; Wu, R. Advances in Fermented Foods Revealed by Multi-Omics: A New Direction toward Precisely Clarifying the Roles of Microorganisms. Front. Microbiol. 2022, 13, 1044820. [Google Scholar] [CrossRef] [PubMed]
  201. Zhang, X.; Zheng, Y.; Liu, Z.; Su, M.; Wu, Z.; Zhang, H.; Zhang, C.; Xu, X. Insights into Characteristic Metabolites and Potential Bioactive Peptides Profiles of Fresh Cheese Fermented with Three Novel Probiotics Based Metabolomics and Peptidomics. Food Chem. X 2024, 21, 101147. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, T.; Wei, G.; Chen, F.; Ma, Q.; Huang, A. Integrated Metabolomics and Peptidomics to Delineate Characteristic Metabolites in Milk Fermented with Novel Lactiplantibacillus plantarum L3. Food Chem. X 2023, 18, 100732. [Google Scholar] [CrossRef] [PubMed]
  203. Pei, J.; Gao, X.; Pan, D.; Hua, Y.; He, J.; Liu, Z.; Dang, Y. Advances in the Stability Challenges of Bioactive Peptides and Improvement Strategies. Curr. Res. Food Sci. 2022, 5, 2162–2170. [Google Scholar] [CrossRef]
  204. Siddiqui, S.A.; Erol, Z.; Rugji, J.; Taşçı, F.; Kahraman, H.A.; Toppi, V.; Musa, L.; Di Giacinto, G.; Bahmid, N.A.; Mehdizadeh, M.; et al. An Overview of Fermentation in the Food Industry—Looking Back from a New Perspective. Bioresour. Bioprocess. 2023, 10, 85. [Google Scholar] [CrossRef]
Figure 2. (a) Pie chart showing the proportion of Angiotensin-I-Converting Enzyme inhibitory (ACE-I) peptides among all bovine β-casein-derived bioactive peptides. (b) Doughnut chart visualizing the proportion of the half maximal inhibitory concentration (IC50) of all bovine β-casein-derived ACE-I peptides. (c) Violin plot of the IC50 value distribution of all bovine β-casein-derived ACE-I peptides. (d) Alignment all bovine β-casein-derived ACE-I peptides with bovine β-casein. Color of each peptide is based on its IC50 value, as indicated in the inset.
Figure 2. (a) Pie chart showing the proportion of Angiotensin-I-Converting Enzyme inhibitory (ACE-I) peptides among all bovine β-casein-derived bioactive peptides. (b) Doughnut chart visualizing the proportion of the half maximal inhibitory concentration (IC50) of all bovine β-casein-derived ACE-I peptides. (c) Violin plot of the IC50 value distribution of all bovine β-casein-derived ACE-I peptides. (d) Alignment all bovine β-casein-derived ACE-I peptides with bovine β-casein. Color of each peptide is based on its IC50 value, as indicated in the inset.
Ijms 26 08119 g002
Figure 3. Flowchart of the curation process for β-casein-derived bioactive peptides. The diagram illustrates the screening process applied to peptides retrieved from three databases (BIOPEP, MBPDB, and EROP-Moscow). Four exclusion criteria were used: (i) lack of supporting references, (ii) only hypothetical or predicted activity, (iii) discrepancies between the peptide sequence and the original literature, and (iv) peptides associated solely with a bitter taste. The final selection resulted in 140 unique bioactive peptides, as listed in Table 1.
Figure 3. Flowchart of the curation process for β-casein-derived bioactive peptides. The diagram illustrates the screening process applied to peptides retrieved from three databases (BIOPEP, MBPDB, and EROP-Moscow). Four exclusion criteria were used: (i) lack of supporting references, (ii) only hypothetical or predicted activity, (iii) discrepancies between the peptide sequence and the original literature, and (iv) peptides associated solely with a bitter taste. The final selection resulted in 140 unique bioactive peptides, as listed in Table 1.
Ijms 26 08119 g003
Figure 4. Schematic representation of the peptide liberation system of Lc. lactis MG1363 process from β-casein β-casein hydrolysis is initiated by PrtP, after its autoproteolytic activation with the aid of PrtM. Subsequently, oligopeptides are transported into the cell by the oligopeptide permease Opp, while di-/tripeptides are internalized by the Dpp or DtpT transport system [173]. The peptides are then degraded into smaller peptides and free amino acids by the concerted action of 15 peptidases, which are classified and colored by their indicated cleavage specificity [15].
Figure 4. Schematic representation of the peptide liberation system of Lc. lactis MG1363 process from β-casein β-casein hydrolysis is initiated by PrtP, after its autoproteolytic activation with the aid of PrtM. Subsequently, oligopeptides are transported into the cell by the oligopeptide permease Opp, while di-/tripeptides are internalized by the Dpp or DtpT transport system [173]. The peptides are then degraded into smaller peptides and free amino acids by the concerted action of 15 peptidases, which are classified and colored by their indicated cleavage specificity [15].
Ijms 26 08119 g004
Figure 5. Engineered LAB-based strategies for enhanced peptide production: (a) Representative inducible expression systems commonly applied in LAB, such as nisin-controlled gene expression (NICE), xylose-, zinc-, and agmatine-inducible promoters. (b) Genome editing tools for LAB, including pORI- or pCS1966-based homologous recombination, RecT-mediated single-strand recombineering, and CRISPR(i)-based approaches. (c) Cell-free protein synthesis (CFPS) systems derived from LAB, enabling rapid peptide production and pathway prototyping without living cells. (d) Application of multi-omics approaches (genomics, transcriptomics, proteomics, and metabolomics) to uncover and optimize peptide biosynthesis pathways in LAB.
Figure 5. Engineered LAB-based strategies for enhanced peptide production: (a) Representative inducible expression systems commonly applied in LAB, such as nisin-controlled gene expression (NICE), xylose-, zinc-, and agmatine-inducible promoters. (b) Genome editing tools for LAB, including pORI- or pCS1966-based homologous recombination, RecT-mediated single-strand recombineering, and CRISPR(i)-based approaches. (c) Cell-free protein synthesis (CFPS) systems derived from LAB, enabling rapid peptide production and pathway prototyping without living cells. (d) Application of multi-omics approaches (genomics, transcriptomics, proteomics, and metabolomics) to uncover and optimize peptide biosynthesis pathways in LAB.
Ijms 26 08119 g005
Table 1. Selection of 140 certified bioactive peptides derived from bovine β-casein.
Table 1. Selection of 140 certified bioactive peptides derived from bovine β-casein.
NOSequence aStart bEnd bBioactivity cReferences
1KVLILA27ACE-I[61]
2RELEELNVPGEIVESLSSSEESITRINK1643Immuno-R[19,21,43,62,63,64,65,66,67]
3RELEELNVPGEIVESLSSSEESITR1640Immuno-R[18,20,22,68]
4LNVPGEIVE2129ACE-I[69]
5VPGEIVE2329DPP-IV-I[70]
6RINKK4044ACE-I; Anti-M[71,72]
7RINK4043Anti-M[71]
8INKKI4145Immuno-R; Anti-C[73,74]
9NKKI4245Anti-M[71]
10DELQDKIHPFAQTQSLVYPFPGPIPNS5884ACE-I[75]
11DKIHPF6267ACE-I[69]
12KIHPFAQTQSLVYP6376ACE-I[76]
13IHPFAQTQ6471PEP-I[77]
14IHPFAQTQSLVYP6476ACE-I[76]
15HPFAQTQSLVYP6576ACE-I[76]
16FAQTQSLVYP6776ACE-I[76]
17AQTQSLVYP6876ACE-I[76]
18QTQSLVYP6976ACE-I[76]
19TQSLVYP7076ACE-I[76]
20QSLVYP7176ACE-I[76]
21SLVYP7276ACE-I[76]
22LVYPFPGPIPNSLPQNIPP7391ACE-I[78,79,80]
23LVYPFPGPIPNSLPQN7388ACE-I[81]
24LVYPFPGPIPNSLPQ7387PEP-I[53]
25LVYPFPGP7380ACE-I[78]
26LVYPFP7378ACE-I[82]
27LVYP7376ACE-I[76]
28LVY7375ACE-I[83]
29VYPFPGPIP7482PEP-I[77]
30VYPFPGPI7481PEP-I[77]
31VYPFPGPIPN7483ACE-I[84]
32VYPFPGPIPNSLPQNIPP7491ACE-I[79]
33VYPFPG7479ACE-I[85]
34VYP7476ACE-I[76,85]
35YPFPGPIP7582Opioid; ACE-I[86,87]
36YPFPGPIPNSL7585Opioid[88,89]
37YPFPGPI7581Opioid; Satiety-Ic; Immuno-R; Anxiolytic; Anti-C[25,26,27,28,90,91,92,93,94,95,96,97,98,99]
38YPFPG7579Opioid; PNO; IM-Ic; Immuno-R; L&M-Ip[25,31,33,34,91,92]
39YPFPGP7580Opioid; DPP-IV-I[25,70,90,100]
40YPFP7578Opioid; Anti-C[90,99,100,101]
41YPFPGPIPN7583ACE-I; DPP-IV-I; Opioid[84,102,103,104]
42YPFPGPIPNSLPQ7587Opioid[102]
43YPFPGPIPNSLPQNIPPLTQT7595Opioid[102]
44PFPGPI7681Cathepsin B-I[90]
45FPGPIPN7783DPP-IV-I[104]
46PGPIPN7883Immuno-R; Anti-C[105,106,107,108,109,110]
47NSLP8386ACE-I[111]
48SLPQN8488ACE-I[72]
49LPQNIPPL8592DPP-IV-I[104]
50LPQNIPPLT8593DPP-IV-I[70]
51LPQNIPP8591DPP-IV-I[104]
52LPQNIPPLTQTPVVVPPFLQPEVMGVSK85112ACE-I[75]
53LPQ8587DPP-IV-I[104]
54PQNIPPL8692DPP-IV-I[104]
55NIPPLTQTPV8897ACE-I[69]
56IPPLTQT8995DPP-IV-I[70]
57IPP8991ACE-I[49]
58LTQTPVVVPPF92102ACE-I[80,112]
59TQTPVVVPPFLQPE93106Anti-O (DPPH)[52]
60TPVVVPPFLQP95105ACE-I[85]
61VVVPPF97102ACE-I[80]
62VVPP98101ACE-I[61]
63VPP99101ACE-I; Anti-Infla; Boneloss-R[49,72,113,114,115,116,117,118,119,120,121]
64FLQP102105ACE-I; DPP-IV-I[111,122]
65LQP103105ACE-I[51]
66LQPE103106Hypolipidemic[55]
67GVSKVKEAMAPKHKEMPFPKYPVEPFTESQ109138Opioid; MUC-Ic[123,124,125,126]
68VKEAMAPK113120Anti-O (LA/Lox; LA/AAPH; Hpode/Hb); Anti-M[40,127]
69EAMAPKHK115122Anti-M[40]
70EAMAPK115120Anti-M[40]
71MAP117119ACE-I[51,111]
72HKEMPFPK121128Anti-M[40,128,129]
73EMPFPK123128MUC-Ic; Anti-M; Anti-M; Brad-P[40,124,130,131]
74MPFPKYPVEP124133ACE-I[132]
75KYPVEPFTESQSLTL128142ACE-I[75]
76KYP128130ACE-I[111]
77YPVEPF129134Opioid; DPP-IV-I; MUC-Ic[70,102,124,133]
78YPVEPFTE129136ACE-I; Brad-P[131]
79VEP131133ACE-I[111]
80HLPLP140144ACE-I[76,134,135]
81LPLP143146ACE-I[76]
82VENLHLPLPLL145155ACE-I[136]
83ENLHLPLPLL146155ACE-I[136]
84NLHLP147151ACE-I[76]
85NLHLPLPLL147155ACE-I[136]
86LHLP148151ACE-I[76]
87LHLPLPL148154ACE-I[80]
88LHLPLP148153ACE-I[80]
89LPLPLL150155DPP-IV-I[70]
90LPLPL150154DPP-IV-I[70,137]
91LPL150152DPP-IV-I[137]
92LQSW155158ACE-I[138]
93QSWMHQPHQ156164ACE-I[139]
94HQP163165ACE-I[111]
95PLP165167ACE-I[76]
96LPP166168ACE-I[111]
97PPQSVLSLSQSKVLPVPQ173190ACE-I[75]
98SQSKVLPVPQ181190ACE-I[132]
99SQSKVLPVPQK181191Hypolipidemic[57]
100SKVLPVPQ183190ACE-I[75]
101KVLPVPQK184191Anti-O (LA/Lox; LA/AAPH; Hpode/Hb)[127]
102KVLPVPQ184190ACE-I[138]
103KVLPVP184189ACE-I[138,140]
104VLPVPQ185190Hypolipidemic[55]
105VLPVPQK185191Anti-O (LA/Lox; LA/AAPH; Hpode/Hb); Anti-M; Anti-apo[40,127,141]
106VLPVPQKAVPYPQR185198Anti-M[40]
107LPVPQ186190DPP-IV-I[70]
108LPVP186189DPP-IV-I[133]
109AVPYPQR192198ACE-I; Anti-O (LA/Lox; LA/AAPH; Hpode/Hb); Anti-M[40,127,130,142]
110AVPYP192196ACE-I[143]
111AVP192194ACE-I[143]
112VPYPQ193197Anti-O (ORAC)[144]
113PYPQ194197Anti-O (ABTS)[145]
114PYP194196ACE-I[143]
115PQR196198ACE-I[146]
116RDMPIQAF198205ACE-I[75]
117DMPIQAFLLYQEPVLGPVR199217Anti-Infla[147]
118IQA202204ACE-I[111]
119AFL204206ACE-I[148]
120LLYQEPVLGPVRGPFPIIV206224ACE-I[75]
121LLY206208Immuno-R[149]
122LYQEPVLGPVRGPFPIIV207224Immuno-R[150]
123YQEPVLGPVRGPFPI208222Anti-M[39]
124YQEPVLGPVRGPFPIIV208224ACE-I; Anti-M; Immuno-R[39,75,151,152]
125YQEPVLGPVR208217ACE-I; Anti-O (ABTS&ORAC); Anti-Infla; Anti-Co[54,153,154]
126YQEPVL208213ACE-I[72,130]
127QEPVL209213Anti-Infla; Immuno-R[44,155]
128QEPV209212Immuno-R[155]
129QEPVLGPVRGPFP209221Hypoglycemic[56]
130QEPVLGPVRGPFPIIV209224ACE-I[153]
131EPVLGPVRGP210219Cyto-M[156]
132EPVLGPVRGPFP210221ACE-I[132]
133VLGP212215ACE-I; DPP-IV-I[111,122]
134VLGPVRGPFP212221ACE-I[80]
135LGP213215ACE-I[157,158]
136VRGPFPIIV216224ACE-I[80]
137VRGPFP216221ACE-I[159]
138GPFPIIV218224ACE-I[160,161]
139GPFPI218222Cathepsin B-I[162]
140PFP219221ACE-I[163]
a The peptide sequences are presented in the one-letter amino acid code. b The start and end positions of the peptide from bovine β-casein sequence, including signal peptide, sequence from UniProt (www.uniprot.org (accessed on 15 May 2025)) with accession number P02666. c Bioactivities of the indicated peptide. Abbreviations: ACE-I, angiotensin-converting enzyme inhibitory; Anti-M, antimicrobial; Anti-O, antioxida-tive; Anti-apo, Anti-apoptotic; DPP-IV-I, dipeptidyl peptidase 4 inhibitory; Immuno-R, immunoregulatory; Anti-C, anticancer; PEP-I, prolyl endopeptidase-inhibitory; Satiety-Ic, Satiety Increasement; IM-Ic, Intestinal Mobility Increasement; PNO, Promoting Neurite Outgrowth; L&M-Ip, Learning and Memory Improvement; Cathepsin B-I, Cathepsin B inhibitory; Inflam-P, Inflammation Promotion; Boneloss-R, Bone loss reduction; Anti-Infla, Anti-inflammatory; MUC-Ic, MUC expression increasement; Brad-P, Bradykinin-Potentiating; Anti-Co, Anticoagulant; Cyto-M, Cytomodulatory.
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

Huang, C.; Cheng, L. Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release. Int. J. Mol. Sci. 2025, 26, 8119. https://doi.org/10.3390/ijms26178119

AMA Style

Huang C, Cheng L. Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release. International Journal of Molecular Sciences. 2025; 26(17):8119. https://doi.org/10.3390/ijms26178119

Chicago/Turabian Style

Huang, Chenxi, and Lianghui Cheng. 2025. "Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release" International Journal of Molecular Sciences 26, no. 17: 8119. https://doi.org/10.3390/ijms26178119

APA Style

Huang, C., & Cheng, L. (2025). Unlocking Casein Bioactivity: Lactic Acid Bacteria and Molecular Strategies for Peptide Release. International Journal of Molecular Sciences, 26(17), 8119. https://doi.org/10.3390/ijms26178119

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