Next Article in Journal
Migratory Wild Birds as Potential Long-Distance Transmitters of Toxoplasma gondii Infection
Next Article in Special Issue
Exposure to Biological Fluids in Dental Practice—Narrative Review on Appropriate Risk Assessment to Guide Post-Exposure Management
Previous Article in Journal
Syndrome of Transient Headache and Neurologic Deficits with Cerebrospinal Fluid Lymphocytosis (HaNDL): HHV-7 Finding in Cerebrospinal Fluid Challenges Diagnostic Criteria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Bioactive Antimicrobial Peptides from Food Proteins: Perspectives and Challenges for Controlling Foodborne Pathogens

by
Jessica Audrey Feijó Corrêa
1,
Tiago de Melo Nazareth
1,2,*,
Giovanna Fernandes da Rocha
1 and
Fernando Bittencourt Luciano
1
1
Laboratory of Agri-Food Research and Innovation, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, R. Imaculada Conceição 1155, Curitiba 80215-901, Brazil
2
Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Burjassot, Spain
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(3), 477; https://doi.org/10.3390/pathogens12030477
Submission received: 29 January 2023 / Revised: 26 February 2023 / Accepted: 16 March 2023 / Published: 17 March 2023
(This article belongs to the Special Issue Reviews of Infectious Diseases)

Abstract

:
Bioactive peptides (BAPs) derived from food proteins have been extensively studied for their health benefits, majorly exploring their potential use as nutraceuticals and functional food components. These peptides possess a range of beneficial properties, including antihypertensive, antioxidant, immunomodulatory, and antibacterial activities, and are naturally present within dietary protein sequences. To release food-grade antimicrobial peptides (AMPs), enzymatic protein hydrolysis or microbial fermentation, such as with lactic acid bacteria (LAB), can be employed. The activity of AMPs is influenced by various structural characteristics, including the amino acid composition, three-dimensional conformation, liquid charge, putative domains, and resulting hydrophobicity. This review discusses the synthesis of BAPs and AMPs, their potential for controlling foodborne pathogens, their mechanisms of action, and the challenges and prospects faced by the food industry. BAPs can regulate gut microbiota by promoting the growth of beneficial bacteria or by directly inhibiting pathogenic microorganisms. LAB-promoted hydrolysis of dietary proteins occurs naturally in both the matrix and the gastrointestinal tract. However, several obstacles must be overcome before BAPs can replace antimicrobials in food production. These include the high manufacturing costs of current technologies, limited in vivo and matrix data, and the difficulties associated with standardization and commercial-scale production.

1. Introduction

BAPs derived from food proteins have been extensively studied and described due to the recognition of their health-promoting properties. Protein hydrolysates are potential sources of BAPs since many are encrypted in the structure of the native/precursor protein. Once released, they can exert beneficial biological effects in the consumer, such as anti-inflammatory, antihypertensive, hypocholesterolemic, hypoglycemic, antioxidative, and antitumoral activities [1,2].
For instance, several BAPs with inhibitory activity towards angiotensin-converting enzymes (ACEs) have been identified in different food matrices, such as coffee [3], cheese [4], and dry-cured ham [1], demonstrating their potential use in the treatment of hypertension. It is noteworthy that recent studies have demonstrated that the cellular receptors for ACE2 are involved in infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen responsible for the global pandemic of COVID-19 (coronavirus disease 2019) [5,6]. Similar immunomodulatory effects may prevent the progression of foodborne diseases, as observed in the peptide Jelleine-I, from honeybee royal jelly, against Listeria monocytogenes infection (Lepidoptera as a model system) [7].
Another interesting biological effect observed in the consumption of BAPs is gut microbiome modulation, which is associated with alterations promoted in the balance of reactive oxygen species (ROS) in the gut [8,9,10]. Recent studies have associated gut microbiome modulation promoted by BAPs with preventing and combating neurodegenerative diseases, such as Parkinson’s disease, Alzheimer’s disease, and dementia [11,12]. The intricate mechanisms involved in the neuroprotective and cognitive-enhancing effects are not yet fully elucidated and seem to vary among the BAPs [11]. Either way, it is known that gut microbiome modulation is also an effective approach to mitigating gastrointestinal infections [13,14,15]. The relative abundances of probiotic and commensal bacteria, as well as their diversity, are strongly related to bioprotection against pathogenic bacteria [16].
In summary, such examples illustrate that studies on BAPs derived from food proteins are required in order to explore their full potential in the pharmacological and nutraceutical fields. Although less explored to date, the antimicrobial bioactivity exerted by these molecules has shown that they can be explored in many areas serving different purposes, with promising applications in the agri-food industry as they are potentially food grade [17,18]. The application of AMPs in this industry has been gaining attention due to their high efficacy against pathogenic and contaminant microorganisms without the hazards associated with antibiotic residues from human and animal use [19]. In addition, as consumers become more concerned about the indiscriminate use of chemical compounds, this industry seeks safe and effective alternatives to replace them [20]. For instance, the use of AMPs figures as a promising alternative to antibiotic growth promoters [21], crop protection agents [22], and preservatives in food [23].
From this perspective, this review gathers what is currently known on the use of hydrolysates and BAPs derived from food proteins for controlling foodborne pathogens, and discusses the prospects for and the challenges to implementation of these promising molecules in the agri-food industry.

2. Production of BAPs and Hydrolysates

BAPs can be obtained through protein hydrolysis. Figure 1 shows a schematic representation of the generation of BAPs. Food protein hydrolysis is usually done biologically through enzymes or microbial fermentation [24]. For instance, during the gastrointestinal digestion process, BAPs are released from the precursor food protein through intense enzymatic activity—generally involving pepsin, trypsin, and pancreatin [25,26]. Food proteins are also susceptible to hydrolysis during food processing, especially during ripening and fermentation, which can directly influence the profile of generated BAPs. This has been observed in both plant proteins (e.g., in different ripening stages of soybean (Glycine max L.) [27]) and animal-origin proteins (e.g., in different cheeses [28,29]).
Although the use of acid and alkaline chemical agents can combine high degrees of protein hydrolysis with low cost, they are not widely used for BAP generation since many alterations in the amino acids occur, presumptively hindering potential bioactivities [24]. Alkaline hydrolysis with hydroxides, for instance, can completely destroy most amino acids and is used for only a few applications in the food industry—mostly restricted to producing foaming agents [30]. On the other hand, many acid protein hydrolysates are used in the food industry as flavor enhancers, although, the process destroys tryptophan and impairs methionine, glutamine, and asparagine [24,31]. Such outcomes might affect the presumed bioactivities of the generated sequences, undermining the employment of chemical hydrolysis methods for BAP production. Notwithstanding, studies on brewer’s-spent-grain hydrolysates (a co-product of the brewing industry) have shown that pre-performing an alkaline protein extraction generally enhances the in vitro bioactivity of enzymatically produced hydrolysates [32].
Anyhow, biological methods are commonly preferred for the GRAS (generally recognized as safe) nature of the generated peptides, as well as for their eco-friendly status [33]. In microbial fermentation, the use of fermentative strains such as LAB is a cost-effective approach [34], considering the additional advantage of the potential removal of hyperallergic and antinutritional factors of the food matrix (e.g., phytate, saponins, and digestive enzyme inhibitors) [24]. It has also been demonstrated that the BAPs generated through microbial fermentation are produced either by protein hydrolysis or by the microorganism itself [35]. However, variations in microbial activity challenge the consistency and reproducibility of microbial hydrolysis processes.
In this sense, hydrolysis of food-sourced proteins by cell-free proteases gains attention, considering that, compared to microbial fermentation, more control can be exerted on the process. In addition, due to its high specificity, favorable environments, and no residual chemicals in the final peptide preparations, enzymatic hydrolysis for BAP production is favored [36]. A wide array of proteases can be used for protein hydrolysis, and these can be obtained from different sources, mainly animals, plants, and microbes [37]. In general, proteases can be classified as exopeptidases or endopeptidases depending on the type of reaction catalyzed by the enzyme. The exopeptidases hydrolyze a peptide bond in the terminal region (N- or C-terminus), as observed for aminopeptidases and carboxypeptidases. As the name suggests, the endopeptidases hydrolyze a peptide bond within an internal region of the protein/peptide. This is the case with pepsin and trypsin, for instance. Within these main classifications, others arise based on the specific targets for cleavage [24].
Another approach worth mentioning is the recombinant expression of BAPs. In the study of Liu and Pan [38], the gene of lunasin—a BAP derived from soybean, which is commercialized as a dietary supplement for health benefits—was synthesized and engineered into Escherichia coli for exogenous expression. Moreover, the chemical synthesis of BAPs is also attainable through soluble-phase and solid-phase processes using amino acid units [39]. These are interesting, yet costly, for producing well-characterized and valuable BAPs. In general, the previously described food-derived BAPs with antimicrobial activity were obtained through biological hydrolysis.
Considering further applications, the fractionation and purification processes are required to standardize and concentrate the active molecules from hydrolysates. Membrane filtration is used to recover peptides and amino acids based on specific molecular weight cut-offs. Chromatographic methods are usually employed for purification, including size-exclusion chromatography, ion-exchange chromatography, and reversed-phase high-performance liquid chromatography. In addition, for identification and characterization of BAP sequences, liquid chromatography has usually been combined with mass spectrometry with ionization methods (e.g., electrospray ionization [ESI] and matrix-assisted laser desorption/ionization [MALDI]) [40,41].

2.1. Sources of BAPs: A Route to Valorize By-Products

As previously mentioned, the biological activity of the generated peptides varies depending on several factors, such as the processing conditions and the protein source—with the latter being the determinant one. Hypothetically, any protein can generate a BAP through hydrolysis. Considering food proteins, different protein-rich foods, including animal-, plant-, and microbial-sourced (fungal and bacterial), have been associated with BAP production [42,43,44,45].
Notably, many proteins are exclusive of animal organisms, making food of animal origin a valuable source for the generation of specific BAPs. For instance, many of the most-studied BAPs, including lactotransferrin and lactoferricin, are derived from milk [43,46,47]. There has also been a trend of studies into seafood-derived BAPs, especially regarding by-products [34,48,49,50,51].
Exploiting different by-products of the food industry for generating BAPs allows their further valorization. Several studies have addressed the hydrolysis of protein-rich by-products, such as fish processing residues (backbone, skin, head, and abdominal cut-offs), crop and agricultural wastes, waste whey, and low-value meat by-products [52,53,54]. The findings suggest that when peptides are isolated and purified, they have promising applications such as ingredients or preservatives in foods and in the pharmaceutical and nutraceutical fields. They can also be applied as biostimulants or bioprotectors for plants and soil in environmental and agronomic approaches (both hydrolysates and isolated BAPs).
It is worth noting that the microbial fermentation process may also generate bioactive compounds other than peptides, such as organic and phenolic acids, bacteriocins, fatty acids, and peroxides—resulting from the metabolism of the microorganism [55]. The fermentation of by-products has proven itself to be a potential and feasible approach to obtaining BAPs. For instance, in the studies carried out by Ramírez et al. [56,57], spent coffee grounds fermented by Bacillus clausii showed an increased abundance of BAPs, and nejayote (maize wastewater) fermented by the same strain exhibited an increased total phenolic content and antioxidant activity. Even though the studies observed these effects separately, both activities are expected to occur considering the capacities of the fermenting bacteria.
Whey, a natural by-product of cheese production, is a frequent subject of study for BAP potential. Fermentation by LAB is usually preferred since they possess great potential to improve the bioactive properties of food combined with the advantages of displaying a high proteolytic activity and being classified as GRAS [58,59]. The topic of LAB will be resumed in Section 4.
Compared to animal protein sources, plant protein sources have more limited potential for the generation of commercial BAPs. Many plants are proven sources of prospective BAPs, in any case. Notable among these are the pulses, which are the dry seeds of annual legumes from the Fabaceae (or Leguminosae) family [60]. Pulses are a rich protein source, making them potential BAP sources. Seed storage proteins are the most abundant, but pulses also contain proteins considered antinutritional, such as lectins and protease inhibitors, which negatively affect diet quality. However, after hydrolysis, these antinutritional proteins may generate BAPs with potential health-promotion effects (e.g., anticancer activity) [60].
As many pulses, such as soybean and peanuts, are used for edible-oil extraction, there is a massive generation of protein-rich by-products, which can be used for bioactive compound recovery, including BAPs [40]. For instance, high content of proteins of low molecular weight and of peptides can be found in maize nixtamalization wastewater (known as nejayote), which has shown potential anti-inflammatory activity [61].
In short, using protein-rich by-products to generate BAPs is an advantageous and eco-friendly approach to seize and valorize under-utilized and low-cost materials.

2.2. Encrypted Peptides and How to Find Them: The Importance of In Silico Approaches

It is possible to investigate the putative antimicrobial activity of peptides in silico with the help of bioinformatics tools [62]. These tools can, for instance, analyze the sequence of food proteins in search of cryptic peptides that acquire this potential when released from the chain that holds them.
Predicting BAPs using in silico approaches is straightforward, as thousands of sequences are able to be screened in much less time than in vitro approaches allow. This, however, must be further combined with experimental assays to prove the predicted bioactivities, as many predictions might not reflect the reality. This was the case in a previous study published by our group [63], in which two whey-derived peptides did not inhibit important bacterial pathogens and mycotoxigenic fungi in vitro despite the prospective in silico predictions.
In silico predictions scan protein databases to identify peptides that match the input criteria. In the study of Brand et al. [64], a methodology for the identification of putative antimicrobial encrypted peptides was proposed based on physiochemical parameters (differential scanning calorimetry and circular dichroism) that may reflect the effects on target-membranes—an important mechanism of action of AMPs (see Section 3.1) [65]. In short, the developed exploratory software Kamal searches within the protein sequence for similarities to known AMPs. This approach complements the traditionally applied prediction methods, which majorly simulate enzymatic cleavages. In those, the action of specific proteases on the protein is simulated to obtain the hydrolysate sequences, which are further investigated for bioactivities based on a database, such as BIOPEP-UWM [66,67,68] or CAMP R3 [69].
The latter, CAMP R3 (Collection of Antimicrobial Peptides), comprises deduced and experimentally proved AMP sequences and also offers prediction tools. Encrypted AMPs were recovered from soybean meal aqueous extract after prediction through analyses using free algorithms provided by CAMP R3 [70]. In the study, hydrolysis was carried out by thermal activation of proteases already present in the extract, and the peptides were recovered through membrane ultrafiltration and chromatographic fractionation.
Apart from discovering encrypted BAPs within a protein sequence, in silico approaches can also be explored to design and optimize them. For instance, in the study by Porto et al. [71], a computational approach was described for the manipulation of a natural AMP from guava (Psidium guajava L.). The designed guavanin peptides showed a different mechanism of action to that of most naturally occurring AMPs (i.e., membrane hyperpolarization), which can be further investigated for the development of novel molecules to circumvent the antimicrobial resistance by microorganisms [65].

3. The Antimicrobial Activity of Food-Sourced Peptides

The growth in the incidence of foodborne infections poses a risk to the population’s health as well as to the economy. Food contaminated by microorganisms may contain pathogenic bacteria, fungi, parasites, viruses, and toxins, being associated with more than 200 different diseases [23].
Because of this, the use of preservatives is demanded in many different foods to assure safety while maintaining the quality and sensory attributes of the product. In addition, as previously mentioned, natural or minimally-processed antimicrobials are constantly sought to meet consumer trends and minimize the concerns regarding microbial resistance to the synthetic compounds traditionally used as antimicrobial agents [20]. The use of AMPs arises in this scenario.
Among the most used and well-characterized AMPs, the nisins produced by Lactococcus lactis subsp. lactis, are bacteriocins of the class of lantibiotics widely used as food preservatives. Nisins are classified as GRAS and are a food additive regulated in several countries [72]. In recent studies, nisin was tested as a possible chemotherapeutic for the treatment of bovine mastitis, in which Staphylococcus is one of the most important etiological agents, and which often results in prolonged, recurrent, and persistent infections [73,74]. Although not essentially a food-derived AMP, nisin production is often associated with whey valorization processes [75].
Another bioactive molecule studied for the treatment of bovine mastitis is lactoferrin, a protein found in milk. The proteolytic degradation of bovine lactoferrin (naturally occurring during stomach digestion) generates lactoferricin, the most-studied AMP derived from milk. With an amphipathic character and an antiparallel β-sheet structure, lactoferricin and its shorter derivatives display relevant antimicrobial activity. Svendsen et al. [76] suggested that they pose a dual mechanism of action—one related to membrane destabilization and the other to intracellular targets. The general mechanisms of action of AMPs are addressed in Section 3.1.
AMPs have also attracted the attention of the poultry and pork industries due to the growing antimicrobial resistance to conventional antibiotics and the consequent search for effective alternatives for disease control and growth promoters in animal production [77,78]. Studies with weaned piglets showed that feed supplementation with lactoferrin increased the efficiency of weight gain and average daily weight gain [79]. In addition, studies with an artificial peptide (lactoferricin–lactoferrampin fusion) improved growth performance and reduced the occurrence of diarrhea in piglets, with effects similar to those observed with the use of the antibiotic colistin sulfate [80]. These results show the potential use of AMPs as substitutes to antibiotic growth promoters which are usually associated with the rise of antimicrobial resistance worldwide.
Table 1 shows studies associated with using BAPs or hydrolysates from food proteins to control pathogens.
In the context of the AMPs addressed in this review, it should be noted that these molecules can present more than one bioactivity. One important bioactivity is the cytolytic activity against tumor cells, which can also define them as anticancer peptides (ACPs) [93]. Although not the focus of this review, studies with ACPs demonstrate their great chemotherapeutic potential as they generally are specifically toxic to cancer cells. This specificity is due to the existence of electrostatic interactions between cancer cells and ACPs, since these cells generally have a strong negative charge on their cell surface due to the high presence of anionic molecules (e.g., heparin sulfate, and mucins) [93,94]. This negative net charge found in cancer cells is similar to that of prokaryotic membranes, which justifies the dual antimicrobial and anticancer activities of certain peptides. Such mechanisms found for both AMPs and ACPs will be better defined in Section 3.1.

3.1. Mechanism of Antimicrobial Activity of BAPs

The knowledge of the molecular mechanisms involved in AMPs activity is essential for their effective application in the agri-food industry. Such information can guide the optimization of the bioactivities through the definition of the promising interactions in a matrix and in vivo (in the case of consumption) and even through genetic engineering of the molecules [95]. However, much remains to be explored and elucidated within this field.
When it comes to size, AMPs are very heterogeneous. Nevertheless, some features are usually shared, including net positive charge and hydrophobicity, which are associated with the ability of AMPs to interact with membrane and/or cytoplasmic components of the microorganisms [65]. Such interaction can occur through nonreceptor-mediated or receptor-mediated mechanisms [96]. In general, most of the known receptor-mediated AMPs are bacteriocins (i.e., produced by bacteria), such as the previously mentioned nisins. Briefly, they act towards a specific target, the receptor, present in the membrane or intracellular component (e.g., DNA and ribosomes), exhibiting a greater specificity compared to nonreceptor-mediated AMPs.
In general, the cell surface of microorganisms presents a net negative charge due to the presence of negatively charged phospholipids and other membrane components, such as teichoic acid (in Gram-positive bacteria) and lipopolysaccharides (in Gram-negative bacteria) [97]. The nonreceptor-mediated mechanism is based on the interaction of AMPs with the membrane, as the peptides generally exhibit a net positive charge. Considering this, the similarity in the activity of AMPs and ACPs is observed, as both prokaryotic cells and cancer cells are preferred targets for the net negative charge they possess. Eukaryotic cells, in contrast, present a more neutral net charge [65,98].
AMP activity is related to different structural determinants, which are based on a first step to the amino acid composition. From this, other important features of antimicrobial activity are defined, such as conformation, charge, domain presence, and hydrophobicity [65].
Focusing on the AMPs, which show a more generalist activity through membrane-targeting, the formation of pores and membrane destabilization, are the outcomes that lead to cell death [99]. Different mechanisms have been proposed for this to occur—AMP insertion into the lipid bilayer to form transmembrane pores and membrane permeabilization through electrostatic interactions—both leading to the collapse of the cell integrity. Additionally, some nonreceptor-mediated AMPs have been described to also present intracellular targets through mechanisms not fully elucidated to date [100]. Anyhow, the translocation of the AMP into the cell is required for this to happen.

Gut Microbiota Modulation and Immunomodulation as Mechanisms of Action: Gaps to Be Filled

The gastrointestinal tract comes into contact with several dietary proteins, expressing a wide variety of receptors and regulatory signals as a response to ingested bioactive compounds. Therefore, it has been shown that such modulation of digestive system physiology is essential for the maintenance and improvement of health [101].
The immunomodulatory mechanisms of AMPs, as well as the microbiota modulation they promote in the gut, are closely linked to the benefits to human and animal health that they promote [102].
Immunomodulation occurs when peptides bind to specific receptors in the consumer organism, promoting an immune response and affecting cellular functions. This results in the suppression or stimulation of specific effectors that may enhance the production of antibodies, cytokine expression, lymphocyte activation, or proliferation; and/or non-specific effectors that lead to the activation of macrophages, natural killer cells, and granulocytes [103,104].
AMPs with immunomodulatory activity are essentially produced by organisms with the purpose of acting on their own metabolism. For example, it has been reported in humans that keratinocytes (epidermal cells) produce AMPs that promote the expression of cytokines and chemokines, thus stimulating the immune response to viral infections [105]. However, in food, we ingest many AMPs from the most different sources. The involved immunomodulatory mechanisms are especially important when analyzing the bioactivities and benefits associated with the consumption of certain types of food [106].
As previously mentioned in Section 1, the AMP Jelleine-I affected the membrane integrity, disrupted some intracellular structures, induced the production of ROS, inhibited biofilm formation, and interacted with the DNA of L. monocytogenes. In vivo, the administration of Jelleine-I to Galleria mellonella infected with the pathogen has shown interesting immunomodulatory effects, including the increase of hemocyte counts, upregulation of other host AMPs, and inhibition of pro-inflammatory cytokines. As a result of the bacterial inhibition, the survival rate of the infected insects increased, and cell proliferation without hemolysis or cytotoxicity was also observed [7].
Beyond regulating pro-inflammatory reactions, AMPs, and BAPs are able to modulate the gut microbiota. The bioprotection of AMPs against pathogens benefits the proliferation of probiotic and commensal bacteria in the gut, which also prevents the colonization of the intestine by pathogenic bacteria. Different mechanisms may be involved in the latter, including competitive exclusion, the consumption of available nutrients, the upregulation of host defense genes, and the production of antimicrobial compounds [7,107,108].
The bioprotection promoted by AMPs towards pathogens is probably related to the disruption of membranes and intracellular targets, mechanisms already described. Additionally, AMPs and BAPs can influence the balance of ROS. In vivo, such influence is believed to occur in the intestine, and is connected to the reduction of oxidative stress. As mentioned in Section 1, such events have been associated with the prevention of neurodegenerative diseases [11,12], as oxidative stress is seen as one of the main villains in the progression of these conditions [109,110,111]. In addition to the repression of ROS generation, many BAPs have antioxidant activity, which may also reduce oxidative stress.
It is worth mentioning that we are considering the AMPs that do not hinder commensal and probiotic bacteria in the gut. This is observed, for instance, in the defensins and cathelicidins, AMPs specialized in host defense that are secreted by the Paneth cells located in the small intestine epithelium [112]. These AMPs are able to inhibit several foodborne pathogens without affecting probiotic and commensal bacteria. Indeed, there are intricate host–microbiota interactions that control the expression of AMPs and suppress pathogen colonization, assuring gut homeostasis [113,114]. This was observed in the study carried out by Cazorla et al. [115], in which microbial probiotics increased the number of Paneth cells and the secretion of AMPs, with consequent enhancement of the antimicrobial activity towards the pathogens S. aureus and Salmonella Typhimurium in vivo.
In short, there is still much to be elucidated within this field. Further studies will allow the molecular mechanisms involved in the immunomodulatory and gut-microbiota-modulation activities to be understood and explored in the near future.

4. The Endless Potential of a Vanguard: Lactic Acid Bacteria

LAB are Gram-positive bacteria responsible for producing a wide variety of bioactive compounds, including hydrogen peroxide, fatty acids, short-chain peptides, and bacteriocins. As many of these compounds exert bioprotective effect against pathogens and deteriorating agents, the significance of LAB in the food industry goes far beyond the production of fermented foods [116,117]. Additionally, these bacteria can adhere to and colonize the digestive system of mammals, exerting probiotic activity [16,118].
It is well known that LAB are important producers of BAPs and AMPs, resulting from the fermentation of products and through the proteolysis of food proteins. LAB strains generally display high proteolytic activity, being able to generate BAPs at a relatively low production cost. Since the proteolytic activity of LAB is strain-dependent, a great variety of proteolytic activities is expected, reflecting an even greater variety of generated BAPs [59,119].
Mostly explored in previous studies are the Lactococcus, Streptococcus, and Leuconostoc genera, as well as the lactobacilli, which are commonly found in foods such as butter, milk, and cheese [120]. The mechanisms of action from which LAB generate BAPs and AMPs are contained in Table 2. Most of what is currently known has come from studies regarding the AMPs produced by LAB through their own metabolic pathways—i.e., bacteriocins. Today, bacteriocins derived from LAB stand out in both commercial and academic spheres [121,122]. Because these bacteria are GRAS, their bacteriocins can be employed as natural preservatives in food. This presents a potential alternative to the synthetic chemicals that have been traditionally used.
However, little is known about the AMPs generated through LAB-promoted hydrolysis of food proteins, even though this is expected to naturally occur in the matrix—considering foods with the presence of LAB—and in the gastrointestinal tract—considering microbiota interactions.
A LAB known to produce AMPs is Lactiplantibacillus plantarum, which has been addressed in different studies to assess its potential to inhibit important foodborne pathogens [82,141,142]. Such studies have reported the direct inhibition promoted by L. plantarum, cell-free supernatant, and isolated bacteriocins. Additionally, L. plantarum has been described to generate AMPs and BAPs through the proteolysis of milk proteins [47,133,143]. BAPs from camel milk fermented with a starter culture of L. plantarum were assessed in the study of Muhialdin and Algboory (2018) to determine their benefits to human health, in view of the good health of Iraqi Bedouins who consume the drink regularly. It is believed that fermented camel milk offers intense protection against infections and diseases and can increase the general energy of its consumers [144]. It has also been shown to demonstrate antioxidant [145], antimicrobial [143], antihypertensive, anticancer [146] and antidiabetic [147] activities. The product is used to treat various diseases in Iran, such as jaundice, tuberculosis, anemia, and asthma [144]. Such effects are mainly due to the presence of BAPs derived from milk proteins which are released through the enzymatic proteolysis occurred during fermentation.
Fermented camel milk has shown inhibitory activity against Gram-positive and -negative foodborne pathogenic bacteria, including E. coli, L. monocytogenes, S. aureus, and Salmonella Typhimurium [47,148,149]. It has also been demonstrated that the product has a high concentration of AMPs with low molecular weight, and the most active fraction contained 32 AMPs derived from milk proteins [133].
In addition to L. plantarum and camel milk, other LAB strains and/or kinds of milk can be used to generate different peptides. For instance, AMPs derived from bovine and goat milk have been described [91,145,150], as well as AMPs obtained through fermentation with Lactobacillus acidophilus and Lactobacillus delbrueckii subsp. lactis [151,152].

5. Current Challenges to the Implementation of Bioactive Peptides in the Food Industry

When it comes to pharmaceutical-grade BAPs, the major method of industrial manufacture is chemical synthesis. Despite the high cost and the low output, this is a reliable method for obtaining AMPs for antibiotic substitution, for instance [24,39].
Hydrolysates, BAPs, and AMPs derived from food proteins hold great promise for application as bioactive ingredients or preservatives in the food industry. The preferred method for the release of pharmaceutical- and food-grade BAPs from precursor proteins is through enzymatic hydrolysis [36,153].
In enzymatic hydrolysis, many parameters influence the final product and yield, including the size of the desirable peptides, the employed enzyme, the enzyme-to-substrate ratio, and several physical–chemical conditions, including pressure, temperature, and duration of the process [47].
In addition, the high cost of enzymes, the restricted choice of assured food-grade proteases, and the presence of enzyme inhibitors in the raw content to be hydrolyzed (usually associated with poor yield) remain a challenge for the industry [36].
There has been increasing interest in novel technologies to substitute for or enhance the yield of conventional enzymatic methods for obtaining hydrolysates and BAPs. However, studies are still needed to clarify if the generated BAPs would maintain the specific features and activity, as well as the safety required for application [47]. Anyhow, alternatives to conventional methods are desperately required to ensure a sustainable approach, which is currently lacking, as well as to reduce the high costs.
Regarding the generation of BAPs and AMPs derived from food proteins in the gastrointestinal tract, digestibility and bioavailability are critical aspects to be considered. Although necessary for the liberation of the bioactive molecules from the precursor protein, the action of digestive enzymes may equally lead to the loss of bioactivity of a sequence, depending on where in the sequence the hydrolysis occurs [33]. The peptides that exert their bioactivities in the consumer organism may have resistance to digestion, penetrating through the intestine to reach other tissues. Therefore, simulated digestion assays are essential to predict the bioavailability of BAPs and AMPs after gastrointestinal digestion [154]. However, it is worth noting that in vivo conditions may differ from individual to individual, and such subjectivity hinders standardized application and the formulation of precise regulations [155].
Currently, there is a lack of in vivo studies into BAPs and hydrolysates, especially in human subjects. The studies would provide vital information on their putative interactions with other drugs, for instance. Equally, studies into food matrices are scarce but essential to evaluate the effectiveness and safety of their use as additives or preservatives, considering the possible reactions with food components that might generate undesirable adducts or complexes [19]. Besides, there is an additional factor to be considered when it comes to the use of AMPs—the protective effect of the food matrix in the microorganisms against the action of preservatives [156,157].
Moreover, many of the studies carried out so far give no precise sequences of the BAPs responsible for the antimicrobial activity, which hinders further assays for the determination of their half-maximal inhibitory concentrations (IC50) [23,47]. To top it all, little is known about the stability of the molecules during their manufacturing processes [155].
In short, further studies must address such lacunae to allow a future successful implementation of the use of BAPs and AMPs in the food industry.

6. Prospects

BAPs are promising molecules for many applications. Many studies have shown their applicability in the pharmacological and nutraceutical fields due to their antioxidant, antidiabetic, antihypertensive, anticancer, anti-inflammatory, hypocholesterolemic, anti-hyperpigmentation, intestine-modulatory, and antimicrobial activities, among others [47,155,158]. To a lesser extent, their potential use in cosmetology and crop improvement has also been described [22,159,160,161,162].
The agri-food industry can benefit from these bioactivities, including, in particular, the antimicrobial potential. The development of safe and efficient alternatives for preservatives and food additives is a constant requirement of this industry. The overuse of antibiotics in animal production is also a major global public health problem due to the emergence of antibiotic resistance. The use of BAPs with antimicrobial activity, i.e., AMPs, figures as an auspicious approach for both food safety and animal-growth-promotion matters [21,163,164].
In addition, recent studies have shown the protective effects of BAPs against the cytotoxicity of the Fusarium mycotoxins deoxynivalenol (DON) and T-2 toxin, mainly associated with the decrease of oxidative stress [165,166]. This is also interesting for both food and feed applications since Fusarium is a ubiquitous fungal genus that infects crops and produces several types of mycotoxins—which are often found contaminating cereals and cereal-based products [167,168].
Against foodborne pathogens, the use of AMPs derived from food proteins shows great potential, with proven activity against important bacteria, including L. monocytogenes, S. aureus, S. enterica, E. coli, P. aeruginosa, K. pneumoniae, B. subtilis, and V. parahaemolyticus (Check Table 1 for references). However, most studies have been carried out in vitro, and the in vivo interactions and the interactions with food matrices need to be further evaluated—when considering the use of AMPs as food preservatives or drug. Moreover, the optimal delivery method for food applications must be investigated. This may vary greatly based on the target microorganism, the food, the storage and processing conditions, the evaluated AMP, etc. For instance, the encapsulation of AMPs could be a good approach to enhancing their resistance to undesirable interactions, as well as the formulation of edible films and coatings with AMPs [169].

7. Conclusions

In this review, we have provided an overview of what is currently known regarding using BAPs derived from food proteins to control foodborne pathogens.
Despite their great potential, several challenges need to be overcome before the successful implementation of BAPs as antimicrobial alternatives in the food industry. The high production cost of the currently applied technologies, the limited in vivo and matrix data, and the problematic standardization of conditions hinder the industrial scale-up, restricting the potential of AMPs from being fully explored.

Author Contributions

Conceptualization, J.A.F.C. and T.d.M.N.; writing—original draft, J.A.F.C. and G.F.d.R.; writing—review and editing, J.A.F.C. and T.d.M.N.; supervision and funding acquisition, T.d.M.N. and F.B.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Brazilian National Council for Scientific and Technological Development, processes CNPq n° 437728/2018-8 and 308598/2020-2.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank the Coordination for the Improvement of Higher Education Personnel, processes CAPES 001 and 88887.512219/2020-00; the pre-Ph.D. program of the Universitat de València (Atracció de Talent UV-INV-PREDOC19F1-1006684); and the Pontifícia Universidade Católica do Paraná.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Toldrá, F.; Gallego, M.; Reig, M.; Aristoy, M.C.; Mora, L. Bioactive Peptides Generated in the Processing of Dry-Cured Ham. Food Chem. 2020, 321, 126689. [Google Scholar] [CrossRef] [PubMed]
  2. Kaur, A.; Kehinde, B.A.; Sharma, P.; Sharma, D.; Kaur, S. Recently Isolated Food-Derived Antihypertensive Hydrolysates and Peptides: A Review. Food Chem. 2021, 346, 128719. [Google Scholar] [CrossRef] [PubMed]
  3. Ribeiro, E.; de Souza Rocha, T.; Prudencio, S.H. Potential of Green and Roasted Coffee Beans and Spent Coffee Grounds to Provide Bioactive Peptides. Food Chem. 2021, 348, 129061. [Google Scholar] [CrossRef] [PubMed]
  4. Rafiq, S.; Gulzar, N.; Sameen, A.; Huma, N.; Hayat, I.; Ijaz, R. Functional Role of Bioactive Peptides with Special Reference to Cheeses. Int. J. Dairy Technol. 2021, 74, 1–16. [Google Scholar] [CrossRef]
  5. Wrapp, D.; Wang, N.; Corbett, K.S.; Goldsmith, J.A.; Hsieh, C.-L.; Abiona, O.; Graham, B.S.; McLellan, J.S. Cryo-EM Structure of the 2019-NCoV Spike in the Prefusion Conformation. Science 2020, 367, 1260–1263. [Google Scholar] [CrossRef] [Green Version]
  6. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.-H.; Nitsche, A.; et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell 2020, 181, 271–280.e8. [Google Scholar] [CrossRef]
  7. Shen, P.; Ding, K.; Wang, L.; Tian, J.; Huang, X.; Zhang, M.; Dang, X. In Vitro and in Vivo Antimicrobial Activity of Antimicrobial Peptide Jelleine-I against Foodborne Pathogen Listeria Monocytogenes. Int. J. Food Microbiol. 2023, 387, 110050. [Google Scholar] [CrossRef]
  8. Ma, X.; Cui, X.; Li, J.; Li, C.; Wang, Z. Peptides from Sesame Cake Reduce Oxidative Stress and Amyloid-β-Induced Toxicity by Upregulation of SKN-1 in a Transgenic Caenorhabditis Elegans Model of Alzheimer’s Disease. J. Funct. Foods 2017, 39, 287–298. [Google Scholar] [CrossRef]
  9. Zhang, Z.; He, S.; Cao, X.; Ye, Y.; Yang, L.; Wang, J.; Liu, H.; Sun, H. Potential Prebiotic Activities of Soybean Peptides Maillard Reaction Products on Modulating Gut Microbiota to Alleviate Aging-Related Disorders in D-Galactose-Induced ICR Mice. J. Funct. Foods 2020, 65, 103729. [Google Scholar] [CrossRef]
  10. Yardeni, T.; Tanes, C.E.; Bittinger, K.; Mattei, L.M.; Schaefer, P.M.; Singh, L.N.; Wu, G.D.; Murdock, D.G.; Wallace, D.C. Host Mitochondria Influence Gut Microbiome Diversity: A Role for ROS. Sci. Signal. 2019, 12, eaaw3159. [Google Scholar] [CrossRef]
  11. Wu, S.; Bekhit, A.E.-D.A.; Wu, Q.; Chen, M.; Liao, X.; Wang, J.; Ding, Y. Bioactive Peptides and Gut Microbiota: Candidates for a Novel Strategy for Reduction and Control of Neurodegenerative Diseases. Trends Food Sci. Technol. 2021, 108, 164–176. [Google Scholar] [CrossRef]
  12. Wang, S.; Sun-Waterhouse, D.; Neil Waterhouse, G.I.; Zheng, L.; Su, G.; Zhao, M. Effects of Food-Derived Bioactive Peptides on Cognitive Deficits and Memory Decline in Neurodegenerative Diseases: A Review. Trends Food Sci. Technol. 2021, 116, 712–732. [Google Scholar] [CrossRef]
  13. Vitetta, L.; Saltzman, E.; Nikov, T.; Ibrahim, I.; Hall, S. Modulating the Gut Micro-Environment in the Treatment of Intestinal Parasites. J. Clin. Med. 2016, 5, 102. [Google Scholar] [CrossRef] [Green Version]
  14. Swaggerty, C.; Callaway, T.; Kogut, M.; Piva, A.; Grilli, E. Modulation of the Immune Response to Improve Health and Reduce Foodborne Pathogens in Poultry. Microorganisms 2019, 7, 65. [Google Scholar] [CrossRef] [Green Version]
  15. Lee, H.; Ko, G. Antiviral Effect of Vitamin A on Norovirus Infection via Modulation of the Gut Microbiome. Sci. Rep. 2016, 6, 25835. [Google Scholar] [CrossRef] [Green Version]
  16. Evangelista, A.G.; Danielski, G.M.; Corrêa, J.A.F.; Cavalari, C.M.d.A.; Souza, I.R.; Luciano, F.B.; de Macedo, R.E.F. Carnobacterium as a Bioprotective and Potential Probiotic Culture to Improve Food Quality, Food Safety, and Human Health–a Scoping Review. Crit. Rev. Food Sci. Nutr. 2022, 62, 1–14. [Google Scholar] [CrossRef]
  17. Chauhan, V.; Kanwar, S.S. Bioactive Peptides. In Biotechnological Production of Bioactive Compounds; Elsevier: Amsterdam, The Netherlands, 2020; pp. 107–137. ISBN 9780444643230. [Google Scholar]
  18. Etemadian, Y.; Ghaemi, V.; Shaviklo, A.R.; Pourashouri, P.; Sadeghi Mahoonak, A.R.; Rafipour, F. Development of Animal/Plant-Based Protein Hydrolysate and Its Application in Food, Feed and Nutraceutical Industries: State of the Art. J. Clean. Prod. 2021, 278, 123219. [Google Scholar] [CrossRef]
  19. Wang, S.; Zeng, X.; Yang, Q.; Qiao, S. Antimicrobial Peptides as Potential Alternatives to Antibiotics in Food Animal Industry. Int. J. Mol. Sci. 2016, 17, 603. [Google Scholar] [CrossRef]
  20. Corrêa, J.A.F.; dos Santos, J.V.G.; Evangelista, A.G.; Pinto, A.C.S.M.; de Macedo, R.E.F.; Luciano, F.B. Combined Application of Phenolic Acids and Essential Oil Components against Salmonella Enteritidis and Listeria Monocytogenes in Vitro and in Ready-to-Eat Cooked Ham. LWT 2021, 149, 111881. [Google Scholar] [CrossRef]
  21. Mohammadrezaei, M.; Navidshad, B.; Gheisari, A.; Toghyani, M. Cottonseed Meal Bioactive Peptides as an Alternative to Antibiotic Growth Promoters in Broiler Chicks. Int. J. Pept. Res. Ther. 2021, 27, 329–340. [Google Scholar] [CrossRef]
  22. Pan, S.; Agyei, D.; Jeevanandam, J.; Danquah, M.K. Bio-Active Peptides: Role in Plant Growth and Defense. In Natural Bio-Active Compounds; Springer: Singapore, 2019; pp. 1–29. [Google Scholar]
  23. León Madrazo, A.; Segura Campos, M.R. Review of Antimicrobial Peptides as Promoters of Food Safety: Limitations and Possibilities within the Food Industry. J. Food Saf. 2020, 40, e12854. [Google Scholar] [CrossRef]
  24. Hou, Y.; Wu, Z.; Dai, Z.; Wang, G.; Wu, G. Protein Hydrolysates in Animal Nutrition: Industrial Production, Bioactive Peptides, and Functional Significance. J. Anim. Sci. Biotechnol. 2017, 8, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Kartal, C.; Kaplan Türköz, B.; Otles, S. Prediction, Identification and Evaluation of Bioactive Peptides from Tomato Seed Proteins Using in Silico Approach. J. Food Meas. Charact. 2020, 14, 1865–1883. [Google Scholar] [CrossRef]
  26. Hernández-Ledesma, B.; Quirós, 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]
  27. Alnuaimi, A.; Fisayo Ajayi, F.; Hamdi, M.; Mudgil, P.; Kamal, H.; Yuen Gan, C.; Maqsood, S. A Comparative Analysis of Anti-Lipidemic Potential of Soybean (Glycine Max) Protein Hydrolysates Obtained from Different Ripening Stages: Identification, and Molecular Interaction Mechanisms of Novel Bioactive Peptides. Food Chem. 2023, 402, 134192. [Google Scholar] [CrossRef]
  28. Martini, S.; Conte, A.; Tagliazucchi, D. Effect of Ripening and in Vitro Digestion on the Evolution and Fate of Bioactive Peptides in Parmigiano-Reggiano Cheese. Int. Dairy J. 2020, 105, 104668. [Google Scholar] [CrossRef]
  29. Atanasova, J.; Dalgalarrondo, M.; Iliev, I.; Moncheva, P.; Todorov, S.D.; Ivanova, I.V. Formation of Free Amino Acids and Bioactive Peptides During the Ripening of Bulgarian White Brined Cheeses. Probiotics Antimicrob. Proteins 2021, 13, 261–272. [Google Scholar] [CrossRef]
  30. Gao, J.; Wang, Y.; Yan, Y.; Li, Z.; Chen, M. Protein Extraction from Excess Sludge by Alkali-Thermal Hydrolysis. Environ. Sci. Pollut. Res. 2020, 27, 8628–8637. [Google Scholar] [CrossRef]
  31. Pasupuleti, V.K.; Holmes, C.; Demain, A.L. Applications of Protein Hydrolysates in Biotechnology. In Protein Hydrolysates in Biotechnology; Springer: Dordrecht, The Netherlands, 2008; pp. 1–9. [Google Scholar]
  32. Connolly, A.; Cermeño, M.; Crowley, D.; O’Callaghan, Y.; O’Brien, N.M.; FitzGerald, R.J. Characterisation of the in Vitro Bioactive Properties of Alkaline and Enzyme Extracted Brewers’ Spent Grain Protein Hydrolysates. Food Res. Int. 2019, 121, 524–532. [Google Scholar] [CrossRef]
  33. Nasri, R.; Abdelhedi, O.; Nasri, M.; Jridi, M. Fermented Protein Hydrolysates: Biological Activities and Applications. Curr. Opin. Food Sci. 2022, 43, 120–127. [Google Scholar] [CrossRef]
  34. Rajendran, S.R.C.K.; Mohan, A.; Khiari, Z.; Udenigwe, C.C.; Mason, B. Yield, Physicochemical, and Antioxidant Properties of Atlantic Salmon Visceral Hydrolysate: Comparison of Lactic Acid Bacterial Fermentation with Flavourzyme Proteolysis and Formic Acid Treatment. J. Food Process. Preserv. 2018, 42, e13620. [Google Scholar] [CrossRef]
  35. Sanjukta, S.; Rai, A.K. Production of Bioactive Peptides during Soybean Fermentation and Their Potential Health Bene Fi Ts. Trends Food Sci. Technol. 2016, 50, 1–10. [Google Scholar] [CrossRef]
  36. Ulug, S.K.; Jahandideh, F.; Wu, J. Novel Technologies for the Production of Bioactive Peptides. Trends Food Sci. Technol. 2021, 108, 27–39. [Google Scholar] [CrossRef]
  37. Chew, L.Y.; Toh, G.T.; Ismail, A. Application of Proteases for the Production of Bioactive Peptides. In Enzymes in Food Biotechnology; Elsevier: Amsterdam, The Netherlands, 2019; pp. 247–261. [Google Scholar]
  38. Liu, C.-F.; Pan, T.-M. Recombinant Expression of Bioactive Peptide Lunasin in Escherichia Coli. Appl. Microbiol. Biotechnol. 2010, 88, 177–186. [Google Scholar] [CrossRef]
  39. Akbarian, M.; Khani, A.; Eghbalpour, S.; Uversky, V.N. Bioactive Peptides: Synthesis, Sources, Applications, and Proposed Mechanisms of Action. Int. J. Mol. Sci. 2022, 23, 1445. [Google Scholar] [CrossRef] [PubMed]
  40. Karami, Z.; Akbari-adergani, B.; Duangmal, K. Recent Development on Recovering Bioactive Peptides and Phenolic Compounds from Under-utilised By-products during Production of Certain Edible Oil Plants: Current Situation and Future Perspectives. Int. J. Food Sci. Technol. 2022, 57, 4894–4905. [Google Scholar] [CrossRef]
  41. Rizzello, C.G.; Tagliazucchi, D.; Babini, E.; Sefora Rutella, G.; Taneyo Saa, D.L.; Gianotti, A. Bioactive Peptides from Vegetable Food Matrices: Research Trends and Novel Biotechnologies for Synthesis and Recovery. J. Funct. Foods 2016, 27, 549–569. [Google Scholar] [CrossRef]
  42. Ali, M.A.; Kamal, M.M.; Rahman, M.H.; Siddiqui, M.N.; Haque, M.A.; Saha, K.K.; Rahman, M.A. Functional Dairy Products as a Source of Bioactive Peptides and Probiotics: Current Trends and Future Prospectives. J. Food Sci. Technol. 2022, 59, 1263–1279. [Google Scholar] [CrossRef]
  43. Samtiya, M.; Samtiya, S.; Badgujar, P.C.; Puniya, A.K.; Dhewa, T.; Aluko, R.E. Health-Promoting and Therapeutic Attributes of Milk-Derived Bioactive Peptides. Nutrients 2022, 14, 3001. [Google Scholar] [CrossRef]
  44. Samtiya, M.; Aluko, R.E.; Dhewa, T.; Moreno-Rojas, J.M. Potential Health Benefits of Plant Food-Derived Bioactive Components: An Overview. Foods 2021, 10, 839. [Google Scholar] [CrossRef]
  45. Maestri, E.; Marmiroli, M.; Marmiroli, N. Bioactive Peptides in Plant-Derived Foodstuffs. J. Proteomics 2016, 147, 140–155. [Google Scholar] [CrossRef] [PubMed]
  46. López-García, G.; Dublan-García, O.; Arizmendi-Cotero, D.; Oliván, L.M.G. Antioxidant and Antimicrobial Peptides Derived from Food Proteins. Molecules 2022, 27, 1343. [Google Scholar] [CrossRef] [PubMed]
  47. Ali Redha, A.; Valizadenia, H.; Siddiqui, S.A.; Maqsood, S. A State-of-Art Review on Camel Milk Proteins as an Emerging Source of Bioactive Peptides with Diverse Nutraceutical Properties. Food Chem. 2022, 373, 131444. [Google Scholar] [CrossRef] [PubMed]
  48. Ahmed, R.; Chun, B.-S. Subcritical Water Hydrolysis for the Production of Bioactive Peptides from Tuna Skin Collagen. J. Supercrit. Fluids 2018, 141, 88–96. [Google Scholar] [CrossRef]
  49. Je, J.-Y.; Qian, Z.-J.; Byun, H.-G.; Kim, S.-K. Purification and Characterization of an Antioxidant Peptide Obtained from Tuna Backbone Protein by Enzymatic Hydrolysis. Process Biochem. 2007, 42, 840–846. [Google Scholar] [CrossRef]
  50. Neves, A.C.; Harnedy, P.A.; O’Keeffe, M.B.; Alashi, M.A.; Aluko, R.E.; FitzGerald, R.J. Peptide Identification in a Salmon Gelatin Hydrolysate with Antihypertensive, Dipeptidyl Peptidase IV Inhibitory and Antioxidant Activities. Food Res. Int. 2017, 100, 112–120. [Google Scholar] [CrossRef]
  51. Alemán, A.; Gómez-Guillén, M.C.; Montero, P. Identification of Ace-Inhibitory Peptides from Squid Skin Collagen after in Vitro Gastrointestinal Digestion. Food Res. Int. 2013, 54, 790–795. [Google Scholar] [CrossRef]
  52. Kobbi, S.; Bougatef, A.; Le Flem, G.; Balti, R.; Mickael, C.; Fertin, B.; Chaabouni, S.; Dhulster, P.; Nedjar, N. Purification and Recovery of RuBisCO Protein from Alfalfa Green Juice: Antioxidative Properties of Generated Protein Hydrolysate. Waste Biomass Valorization 2017, 8, 493–504. [Google Scholar] [CrossRef]
  53. Caballero, P.; Rodríguez-Morgado, B.; Macías, S.; Tejada, M.; Parrado, J. Obtaining Plant and Soil Biostimulants by Waste Whey Fermentation. Waste Biomass Valorization 2020, 11, 3281–3292. [Google Scholar] [CrossRef]
  54. Borrajo, P.; Pateiro, M.; Barba, F.J.; Mora, L.; Franco, D.; Toldrá, F.; Lorenzo, J.M. Antioxidant and Antimicrobial Activity of Peptides Extracted from Meat By-Products: A Review. Food Anal. Methods 2019, 12, 2401–2415. [Google Scholar] [CrossRef]
  55. Mariam, S.H.; Zegeye, N.; Aseffa, A.; Howe, R. Diffusible Substances from Lactic Acid Bacterial Cultures Exert Strong Inhibitory Effects on Listeria Monocytogenes and Salmonella Enterica Serovar Enteritidis in a Co-Culture Model. BMC Microbiol. 2017, 17, 35. [Google Scholar] [CrossRef] [Green Version]
  56. Ramírez, K.; Pineda-hidalgo, K.V.; Rochín-medina, J.J. Fermentation of Spent Coffee Grounds by Bacillus Clausii Induces Release of Potentially Bioactive Peptides. LWT 2021, 138, 110685. [Google Scholar] [CrossRef]
  57. Ramírez, K.; Quintero-Soto, M.F.; Rochín-Medina, J.J. Enhancement of the Antioxidant and Antimicrobial Activities of Maize Wastewater by an Eco-Friendly Process. J. Food Meas. Charact. 2020, 14, 1682–1689. [Google Scholar] [CrossRef]
  58. Khubber, S.; Marti-Quijal, F.J.; Tomasevic, I.; Remize, F.; Barba, F.J. Lactic Acid Fermentation as a Useful Strategy to Recover Antimicrobial and Antioxidant Compounds from Food and By-Products. Curr. Opin. Food Sci. 2022, 43, 189–198. [Google Scholar] [CrossRef]
  59. Worsztynowicz, P.; Białas, W.; Grajek, W. Integrated Approach for Obtaining Bioactive Peptides from Whey Proteins Hydrolysed Using a New Proteolytic Lactic Acid Bacteria. Food Chem. 2020, 312, 126035. [Google Scholar] [CrossRef]
  60. López-Barrios, L.; Gutiérrez-Uribe, J.A.; Serna-Saldívar, S.O. Bioactive Peptides and Hydrolysates from Pulses and Their Potential Use as Functional Ingredients. J. Food Sci. 2014, 79, 134–149. [Google Scholar] [CrossRef]
  61. Buitimea-Cantúa, N.E.; Antunes-Ricardo, M.; Gutiérrez-Uribe, J.A.; del Refugio Rocha-Pizaña, M.; de la Rosa-Millán, J.; Torres-Chávez, P.I. Protein-Phenolic Aggregates with Anti-Inflammatory Activity Recovered from Maize Nixtamalization Wastewaters (Nejayote). LWT 2020, 134, 109881. [Google Scholar] [CrossRef]
  62. Udenigwe, C.C. Bioinformatics Approaches, Prospects and Challenges of Food Bioactive Peptide Research. Trends Food Sci. Technol. 2014, 36, 137–143. [Google Scholar] [CrossRef]
  63. Corrêa, J.A.F.; Udenigwe, C.; Bittencourt Luciano, F. Avaliação in Vitro de Atividade Antimicrobiana de Peptídeos Anfipáticos Derivados de Proteínas Do Soro de Leite. Rev. Acadêmica Ciência Anim. 2018, 16, 1–6. [Google Scholar] [CrossRef]
  64. Brand, G.D.; Magalhães, M.T.Q.; Tinoco, M.L.P.; Aragão, F.J.L.; Nicoli, J.; Kelly, S.M.; Cooper, A.; Bloch, C. Probing Protein Sequences as Sources for Encrypted Antimicrobial Peptides. PLoS ONE 2012, 7, e45848. [Google Scholar] [CrossRef]
  65. Corrêa, J.A.F.; Evangelista, A.G.; de Melo Nazareth, T.; Luciano, F.B. Fundamentals on the Molecular Mechanism of Action of Antimicrobial Peptides. Materialia 2019, 8, 100494. [Google Scholar] [CrossRef]
  66. Minkiewicz, P.; Iwaniak, A.; Darewicz, M. BIOPEP-UWM Database of Bioactive Peptides: Current Opportunities. Int. J. Mol. Sci. 2019, 20, 5978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Minkiewicz, P.; Iwaniak, A.; Darewicz, M. BIOPEP-UWM Virtual—A Novel Database of Food-Derived Peptides with In Silico-Predicted Biological Activity. Appl. Sci. 2022, 12, 7204. [Google Scholar] [CrossRef]
  68. 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] [Green Version]
  69. Waghu, F.H.; Barai, R.S.; Gurung, P.; Idicula-Thomas, S. CAMP R3: A Database on Sequences, Structures and Signatures of Antimicrobial Peptides. Nucleic Acids Res. 2016, 44, D1094–D1097. [Google Scholar] [CrossRef] [Green Version]
  70. Freitas, C.S.; Vericimo, M.A.; da Silva, M.L.; da Costa, G.C.V.; Pereira, P.R.; Paschoalin, V.M.F.; Del Aguila, E.M. Encrypted Antimicrobial and Antitumoral Peptides Recovered from a Protein-Rich Soybean (Glycine Max) by-Product. J. Funct. Foods 2019, 54, 187–198. [Google Scholar] [CrossRef]
  71. Porto, W.F.; Irazazabal, L.; Alves, E.S.F.; Ribeiro, S.M.; Matos, C.O.; Pires, Á.S.; Fensterseifer, I.C.M.; Miranda, V.J.; Haney, E.F.; Humblot, V.; et al. In Silico Optimization of a Guava Antimicrobial Peptide Enables Combinatorial Exploration for Peptide Design. Nat. Commun. 2018, 9, 1490. [Google Scholar] [CrossRef] [Green Version]
  72. Khan, I.; Oh, D.-H. Integration of Nisin into Nanoparticles for Application in Foods. Innov. Food Sci. Emerg. Technol. 2016, 34, 376–384. [Google Scholar] [CrossRef]
  73. Huang, F.; Teng, K.; Liu, Y.; Wang, T.; Xia, T.; Yun, F.; Zhong, J. Nisin Z Attenuates Lipopolysaccharide-Induced Mastitis by Inhibiting the ERK1/2 and P38 Mitogen-Activated Protein Kinase Signaling Pathways. J. Dairy Sci. 2022, 105, 3530–3543. [Google Scholar] [CrossRef]
  74. Bennett, S.; Fliss, I.; Ben Said, L.; Malouin, F.; Lacasse, P. Efficacy of Bacteriocin-Based Formula for Reducing Staphylococci, Streptococci, and Total Bacterial Counts on Teat Skin of Dairy Cows. J. Dairy Sci. 2022, 105, 4498–4507. [Google Scholar] [CrossRef]
  75. Chourasia, R.; Phukon, L.C.; Abedin, M.M.; Padhi, S.; Singh, S.P.; Rai, A.K. Whey Valorization by Microbial and Enzymatic Bioprocesses for the Production of Nutraceuticals and Value-Added Products. Bioresour. Technol. Rep. 2022, 19, 101144. [Google Scholar] [CrossRef]
  76. Svendsen, J.S.M.; Grant, T.M.; Rennison, D.; Brimble, M.A.; Svenson, J. Very Short and Stable Lactoferricin-Derived Antimicrobial Peptides: Design Principles and Potential Uses. Acc. Chem. Res. 2019, 52, 749–759. [Google Scholar] [CrossRef]
  77. Xiao, H.; Shao, F.; Wu, M.; Ren, W.; Xiong, X.; Tan, B.; Yin, Y. The Application of Antimicrobial Peptides as Growth and Health Promoters for Swine. J. Anim. Sci. Biotechnol. 2015, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  78. Evangelista, A.G.; Corrêa, J.A.F.; Pinto, A.C.S.M.; Luciano, F.B. The Impact of Essential Oils on Antibiotic Use in Animal Production Regarding Antimicrobial Resistance—A Review. Crit. Rev. Food Sci. Nutr. 2021, 62, 5267–5283. [Google Scholar] [CrossRef]
  79. Wang, Y.; Shan, T.; Xu, Z.; Liu, J.; Feng, J. Effect of Lactoferrin on the Growth Performance, Intestinal Morphology, and Expression of PR-39 and Protegrin-1 Genes in Weaned Piglets1. J. Anim. Sci. 2006, 84, 2636–2641. [Google Scholar] [CrossRef]
  80. Tang, Z.; Yin, Y.; Zhang, Y.; Huang, R.; Sun, Z.; Li, T.; Chu, W.; Kong, X.; Li, L.; Geng, M.; et al. Effects of Dietary Supplementation with an Expressed Fusion Peptide Bovine Lactoferricin–Lactoferrampin on Performance, Immune Function and Intestinal Mucosal Morphology in Piglets Weaned at Age 21 D. Br. J. Nutr. 2008, 101, 998–1005. [Google Scholar] [CrossRef] [Green Version]
  81. de Oliveira Filho, J.G.; Rodrigues, J.M.; Valadares, A.C.F.; de Almeida, A.B.; Valencia-Mejia, E.; Fernandes, K.F.; Lemes, A.C.; Alves, C.C.F.; Sousa, H.A.d.F.; da Silva, E.R.; et al. Bioactive Properties of Protein Hydrolysate of Cottonseed Byproduct: Antioxidant, Antimicrobial, and Angiotensin-Converting Enzyme (ACE) Inhibitory Activities. Waste Biomass Valorization 2021, 12, 1395–1404. [Google Scholar] [CrossRef]
  82. Martín, I.; Rodríguez, A.; Alía, A.; Martínez-Blanco, M.; Lozano-Ojalvo, D.; Córdoba, J.J. Control of Listeria Monocytogenes Growth and Virulence in a Traditional Soft Cheese Model System Based on Lactic Acid Bacteria and a Whey Protein Hydrolysate with Antimicrobial Activity. Int. J. Food Microbiol. 2022, 361, 109444. [Google Scholar] [CrossRef]
  83. Demers-Mathieu, V.; Gauthier, S.F.; Britten, M.; Fliss, I.; Robitaille, G.; Jean, J. Antibacterial Activity of Peptides Extracted from Tryptic Hydrolyzate of Whey Protein by Nanofiltration. Int. Dairy J. 2013, 28, 94–101. [Google Scholar] [CrossRef]
  84. Mishra, B.; Felix, L.; Basu, A.; Kollala, S.S.; Chhonker, Y.S.; Ganesan, N.; Murry, D.J.; Mylonakis, E. Design and Evaluation of Short Bovine Lactoferrin-Derived Antimicrobial Peptides against Multidrug-Resistant Enterococcus Faecium. Antibiotics 2022, 11, 1085. [Google Scholar] [CrossRef]
  85. Memarpoor-Yazdi, M.; Asoodeh, A.; Chamani, J. A Novel Antioxidant and Antimicrobial Peptide from Hen Egg White Lysozyme Hydrolysates. J. Funct. Foods 2012, 4, 278–286. [Google Scholar] [CrossRef]
  86. Al-Mohammadi, A.-R.; Osman, A.; Enan, G.; Abdel-Shafi, S.; El-Nemer, M.; Sitohy, M.; Taha, M.A. Powerful Antibacterial Peptides from Egg Albumin Hydrolysates. Antibiotics 2020, 9, 901. [Google Scholar] [CrossRef] [PubMed]
  87. Zanutto-Elgui, M.R.; Vieira, J.C.S.; do Prado, D.Z.; Buzalaf, M.A.R.; Padilha, P.d.M.; Elgui de Oliveira, D.; Fleuri, L.F. Production of Milk Peptides with Antimicrobial and Antioxidant Properties through Fungal Proteases. Food Chem. 2019, 278, 823–831. [Google Scholar] [CrossRef] [PubMed]
  88. Fialho, T.L.; Carrijo, L.C.; Magalhães Júnior, M.J.; Baracat-Pereira, M.C.; Piccoli, R.H.; de Abreu, L.R. Extraction and Identification of Antimicrobial Peptides from the Canastra Artisanal Minas Cheese. Food Res. Int. 2018, 107, 406–413. [Google Scholar] [CrossRef] [PubMed]
  89. Castellano, P.; Mora, L.; Escudero, E.; Vignolo, G.; Aznar, R.; Toldrá, F. Antilisterial Peptides from Spanish Dry-Cured Hams: Purification and Identification. Food Microbiol. 2016, 59, 133–141. [Google Scholar] [CrossRef] [Green Version]
  90. Lima, C.A.; Campos, J.F.; Filho, J.L.L.; Converti, A.; da Cunha, M.G.C.; Porto, A.L.F. Antimicrobial and Radical Scavenging Properties of Bovine Collagen Hydrolysates Produced by Penicillium Aurantiogriseum URM 4622 Collagenase. J. Food Sci. Technol. 2015, 52, 4459–4466. [Google Scholar] [CrossRef] [Green Version]
  91. Osman, A.; Goda, H.A.; Abdel-Hamid, M.; Badran, S.M.; Otte, J. Antibacterial Peptides Generated by Alcalase Hydrolysis of Goat Whey. LWT 2016, 65, 480–486. [Google Scholar] [CrossRef]
  92. Wald, M.; Schwarz, K.; Rehbein, H.; Bußmann, B.; Beermann, C. Detection of Antibacterial Activity of an Enzymatic Hydrolysate Generated by Processing Rainbow Trout By-Products with Trout Pepsin. Food Chem. 2016, 205, 221–228. [Google Scholar] [CrossRef]
  93. Gaspar, D.; Veiga, A.S.; Castanho, M.A.R.B. From Antimicrobial to Anticancer Peptides. A Review. Front. Microbiol. 2013, 4, 294. [Google Scholar] [CrossRef] [Green Version]
  94. Hoskin, D.W.; Ramamoorthy, A. Studies on Anticancer Activities of Antimicrobial Peptides. Biochim. Biophys. Acta Biomembr. 2008, 1778, 357–375. [Google Scholar] [CrossRef] [Green Version]
  95. Cardoso, P.; Glossop, H.; Meikle, T.G.; Aburto-Medina, A.; Conn, C.E.; Sarojini, V.; Valery, C. Molecular Engineering of Antimicrobial Peptides: Microbial Targets, Peptide Motifs and Translation Opportunities. Biophys. Rev. 2021, 13, 35–69. [Google Scholar] [CrossRef]
  96. Lee, E.Y.; Lee, M.W.; Wong, G.C.L. Modulation of Toll-like Receptor Signaling by Antimicrobial Peptides. Semin. Cell Dev. Biol. 2019, 88, 173–184. [Google Scholar] [CrossRef]
  97. Bhattacharya, S.; Reddy, D.; Reddy, R.; Sharda, A.; Bose, K.; Gupta, S. Incorporation of a Tag Helps to Overcome Expression Variability in a Recombinant Host. Biotechnol. Rep. 2016, 11, 62–69. [Google Scholar] [CrossRef] [Green Version]
  98. Andoh, Y.; Kitou, S.; Okazaki, S. Difference in Molecular Mechanisms Governing Changes in Membrane Properties of Phospholipid Bilayers Induced by Addition of Nonionic and Zwitterionic Surfactants. J. Mol. Liq. 2018, 271, 933–941. [Google Scholar] [CrossRef]
  99. Shai, Y. Mechanism of the Binding, Insertion and Destabilization of Phospholipid Bilayer Membranes by a-Helical Antimicrobial and Cell Non-Selective Membrane-Lytic Peptides. Biochim. Biophys. Acta Biomembr. 1999, 1462, 55–70. [Google Scholar] [CrossRef] [Green Version]
  100. Li, T.; Liu, Q.; Wang, D.; Li, J. Characterization and Antimicrobial Mechanism of CF-14, a New Antimicrobial Peptide from the Epidermal Mucus of Catfish. Fish Shellfish. Immunol. 2019, 92, 881–888. [Google Scholar] [CrossRef]
  101. Fernández-Tomé, S.; Hernández-Ledesma, B. Gastrointestinal Digestion of Food Proteins under the Effects of Released Bioactive Peptides on Digestive Health. Mol. Nutr. Food Res. 2020, 64, 2000401. [Google Scholar] [CrossRef]
  102. Liébana-García, R.; Olivares, M.; Bullich-Vilarrubias, C.; López-Almela, I.; Romaní-Pérez, M.; Sanz, Y. The Gut Microbiota as a Versatile Immunomodulator in Obesity and Associated Metabolic Disorders. Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101542. [Google Scholar] [CrossRef]
  103. Meisel, H. Multifunctional Peptides Encrypted in Milk Proteins. BioFactors 2004, 21, 55–61. [Google Scholar] [CrossRef]
  104. Gauthier, S.F.; Pouliot, Y.; Saint-Sauveur, D. Immunomodulatory Peptides Obtained by the Enzymatic Hydrolysis of Whey Proteins. Int. Dairy J. 2006, 16, 1315–1323. [Google Scholar] [CrossRef]
  105. Chessa, C.; Bodet, C.; Jousselin, C.; Wehbe, M.; Lévêque, N.; Garcia, M. Antiviral and Immunomodulatory Properties of Antimicrobial Peptides Produced by Human Keratinocytes. Front. Microbiol. 2020, 11, 1155. [Google Scholar] [CrossRef] [PubMed]
  106. Théolier, J.; Hammami, R.; Labelle, P.; Fliss, I.; Jean, J. Isolation and Identification of Antimicrobial Peptides Derived by Peptic Cleavage of Whey Protein Isolate. J. Funct. Foods 2013, 5, 706–714. [Google Scholar] [CrossRef]
  107. Martín, R.; Langella, P. Emerging Health Concepts in the Probiotics Field: Streamlining the Definitions. Front. Microbiol. 2019, 10, 1047. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, J.; Zeng, Y.; Wang, S.; Liu, H.; Zhang, D.; Zhang, W.; Wang, Y.; Ji, H. Swine-Derived Probiotic Lactobacillus Plantarum Inhibits Growth and Adhesion of Enterotoxigenic Escherichia Coli and Mediates Host Defense. Front. Microbiol. 2018, 9, 1364. [Google Scholar] [CrossRef] [PubMed]
  109. Gandhi, S.; Abramov, A.Y. Mechanism of Oxidative Stress in Neurodegeneration. Oxid. Med. Cell. Longev. 2012, 2012, 428010. [Google Scholar] [CrossRef] [Green Version]
  110. Singh, A.; Kukreti, R.; Saso, L.; Kukreti, S. Oxidative Stress: A Key Modulator in Neurodegenerative Diseases. Molecules 2019, 24, 1583. [Google Scholar] [CrossRef] [Green Version]
  111. Kim, G.H.; Kim, J.E.; Rhie, S.J.; Yoon, S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp. Neurobiol. 2015, 24, 325–340. [Google Scholar] [CrossRef] [Green Version]
  112. Bevins, C.L.; Salzman, N.H. Paneth Cells, Antimicrobial Peptides and Maintenance of Intestinal Homeostasis. Nat. Rev. Microbiol. 2011, 9, 356–368. [Google Scholar] [CrossRef]
  113. Zong, X.; Fu, J.; Xu, B.; Wang, Y.; Jin, M. Interplay between Gut Microbiota and Antimicrobial Peptides. Anim. Nutr. 2020, 6, 389–396. [Google Scholar] [CrossRef]
  114. Muniz, L.R.; Knosp, C.; Yeretssian, G. Intestinal Antimicrobial Peptides during Homeostasis, Infection, and Disease. Front. Immunol. 2012, 3, 310. [Google Scholar] [CrossRef] [Green Version]
  115. Cazorla, S.I.; Maldonado-Galdeano, C.; Weill, R.; De Paula, J.; Perdigón, G.D.V. Oral Administration of Probiotics Increases Paneth Cells and Intestinal Antimicrobial Activity. Front. Microbiol. 2018, 9, 736. [Google Scholar] [CrossRef]
  116. García-Díez, J.; Saraiva, C. Use of Starter Cultures in Foods from Animal Origin to Improve Their Safety. Int. J. Environ. Res. Public Health 2021, 18, 2544. [Google Scholar] [CrossRef]
  117. Kadyan, S.; Rashmi, H.M.; Pradhan, D.; Kumari, A.; Chaudhari, A.; Deshwal, G.K. Effect of Lactic Acid Bacteria and Yeast Fermentation on Antimicrobial, Antioxidative and Metabolomic Profile of Naturally Carbonated Probiotic Whey Drink. Lwt 2021, 142, 111059. [Google Scholar] [CrossRef]
  118. Giraffa, G.; Chanishvili, N.; Widyastuti, Y. Importance of Lactobacilli in Food and Feed Biotechnology. Res. Microbiol. 2010, 161, 480–487. [Google Scholar] [CrossRef]
  119. Raveschot, C.; Cudennec, B.; Coutte, F.; Flahaut, C.; Fremont, M.; Drider, D.; Dhulster, P. Production of Bioactive Peptides by Lactobacillus Species: From Gene to Application. Front. Microbiol. 2018, 9, 2354. [Google Scholar] [CrossRef] [Green Version]
  120. Amiri, S.; Aghamirzaei, M.; Mostashari, P.; Sarbazi, M.; Tizchang, S.; Madahi, H. The Impact of Biotechnology on Dairy Industry. In Microbial Biotechnology in Food and Health; Elsevier: Amsterdam, The Netherlands, 2021; pp. 53–79. [Google Scholar]
  121. Paul Ross, R.; Morgan, S.; Hill, C. Preservation and Fermentation: Past, Present and Future. Int. J. Food Microbiol. 2002, 79, 3–16. [Google Scholar] [CrossRef] [Green Version]
  122. Johnson, E.M.; Jung, D.Y.-G.; Jin, D.Y.-Y.; Jayabalan, D.R.; Yang, D.S.H.; Suh, J.W. Bacteriocins as Food Preservatives: Challenges and Emerging Horizons. Crit. Rev. Food Sci. Nutr. 2017, 58, 2743–2767. [Google Scholar] [CrossRef]
  123. Acuña, L.; Corbalán, N.; Quintela-Baluja, M.; Barros-Velázquez, J.; Bellomio, A. Expression of the Hybrid Bacteriocin Ent35-MccV in Lactococcus Lactis and Its Use for Controlling Listeria Monocytogenes and Escherichia Coli in Milk. Int. Dairy J. 2020, 104, 104650. [Google Scholar] [CrossRef]
  124. Khelissa, S.; Chihib, N.-E.; Gharsallaoui, A. Conditions of Nisin Production by Lactococcus Lactis Subsp. Lactis and Its Main Uses as a Food Preservative. Arch. Microbiol. 2021, 203, 465–480. [Google Scholar] [CrossRef]
  125. Dal Bello, B.; Cocolin, L.; Zeppa, G.; Field, D.; Cotter, P.D.; Hill, C. Technological Characterization of Bacteriocin Producing Lactococcus Lactis Strains Employed to Control Listeria Monocytogenes in Cottage Cheese. Int. J. Food Microbiol. 2012, 153, 58–65. [Google Scholar] [CrossRef]
  126. Delorme, C. Safety Assessment of Dairy Microorganisms: Streptococcus Thermophilus. Int. J. Food Microbiol. 2008, 126, 274–277. [Google Scholar] [CrossRef] [PubMed]
  127. Iyer, R.; Tomar, S.K.; Uma Maheswari, T.; Singh, R. Streptococcus Thermophilus Strains: Multifunctional Lactic Acid Bacteria. Int. Dairy J. 2010, 20, 133–141. [Google Scholar] [CrossRef]
  128. Renye, J.A.; Somkuti, G.A.; Garabal, J.I.; Steinberg, D.H. Bacteriocin Production by Streptococcus Thermophilus in Complex Growth Media. Biotechnol. Lett. 2016, 38, 1947–1954. [Google Scholar] [CrossRef] [PubMed]
  129. Ahmed, Z.; Wang, Y.; Cheng, Q.; Imran, M. Lactobacillus Acidophilus Bacteriocin, from Production to Their Application: An Overview. African J. Biotechnol. 2010, 9, 2843–2850. [Google Scholar]
  130. Uzuner, A.E.; Kinik, O.; Korel, F.; Yildiz, G.; Yerlikaya, O. Usage of Rice Milk in Probiotic Yoghurt Production. Carpathian J. Food Sci. Technol. 2016, 8, 5–25. [Google Scholar]
  131. Hati, S.; Patel, N.; Sakure, A.; Mandal, S. Influence of Whey Protein Concentrate on the Production of Antibacterial Peptides Derived from Fermented Milk by Lactic Acid Bacteria. Int. J. Pept. Res. Ther. 2018, 24, 87–98. [Google Scholar] [CrossRef]
  132. Atanasova, J.; Moncheva, P.; Ivanova, I. Proteolytic and Antimicrobial Activity of Lactic Acid Bacteria Grown in Goat Milk. Biotechnol. Biotechnol. Equip. 2014, 28, 1073–1078. [Google Scholar] [CrossRef] [Green Version]
  133. Muhialdin, B.J.; Algboory, H.L. Identification of Low Molecular Weight Antimicrobial Peptides from Iraqi Camel Milk Fermented with Lactobacillus Plantarum. PharmaNutrition 2018, 6, 69–73. [Google Scholar] [CrossRef]
  134. Tenea, G.N.; Olmedo, D.; Ortega, C. Peptide-Based Formulation from Lactic Acid Bacteria Impairs the Pathogen Growth in Ananas Comosus (Pineapple). Coatings 2020, 10, 457. [Google Scholar] [CrossRef]
  135. Gupta, R.; Srivastava, S. Antifungal Effect of Antimicrobial Peptides (AMPs LR14) Derived from Lactobacillus Plantarum Strain LR/14 and Their Applications in Prevention of Grain Spoilage. Food Microbiol. 2014, 42, 1–7. [Google Scholar] [CrossRef]
  136. Weerkamp, A.H.; Klijn, N.; Neeter, R.; Smit, G. Properties of Mesophilic Lactic Acid Bacteria from Raw Milk and Naturally Fermented Raw Milk Products. Netherlands Milk Dairy J. 1996, 50, 319–332. [Google Scholar]
  137. Slattery, L.; O’Callaghan, J.; Fitzgerald, G.F.; Beresford, T.; Ross, R.P. Invited Review: Lactobacillus Helveticus—A Thermophilic Dairy Starter Related to Gut Bacteria. J. Dairy Sci. 2010, 93, 4435–4454. [Google Scholar] [CrossRef] [Green Version]
  138. Papathanasopoulos, M.A.; Krier, F.; Revol-Junelles, A.-M.; Lefebvre, G.; Le Caer, J.P.; von Holy, A.; Hastings, J.W. Multiple Bacteriocin Production by Leuconostoc MesenteroidesTA33a and Other Leuconostoc/Weissella Strains. Curr. Microbiol. 1997, 35, 331–335. [Google Scholar] [CrossRef]
  139. Kingcha, Y.; Tosukhowong, A.; Zendo, T.; Roytrakul, S.; Luxananil, P.; Chareonpornsook, K.; Valyasevi, R.; Sonomoto, K.; Visessanguan, W. Anti-Listeria Activity of Pediococcus Pentosaceus BCC 3772 and Application as Starter Culture for Nham, a Traditional Fermented Pork Sausage. Food Control 2012, 25, 190–196. [Google Scholar] [CrossRef]
  140. Vera Pingitore, E.; Todorov, S.D.; Sesma, F.; Gombossy de Melo Franco, B.D. Application of Bacteriocinogenic Enterococcus Mundtii CRL35 and Enterococcus Faecium ST88Ch in the Control of Listeria Monocytogenes in Fresh Minas Cheese. Food Microbiol. 2012, 32, 38–47. [Google Scholar] [CrossRef]
  141. Pinto, A.; Barbosa, J.; Albano, H.; Isidro, J.; Teixeira, P. Screening of Bacteriocinogenic Lactic Acid Bacteria and Their Characterization as Potential Probiotics. Microorganisms 2020, 8, 393. [Google Scholar] [CrossRef] [Green Version]
  142. Siroli, L.; Patrignani, F.; Serrazanetti, D.I.; Tabanelli, G.; Montanari, C.; Gardini, F.; Lanciotti, R. Lactic Acid Bacteria and Natural Antimicrobials to Improve the Safety and Shelf-Life of Minimally Processed Sliced Apples and Lamb’s Lettuce. Food Microbiol. 2015, 47, 74–84. [Google Scholar] [CrossRef]
  143. Lafta, H.; Jarallah, E.M.; Darwash, A. Antibacterial Activity of Fermented Camel Milk Using Two Lactic Acid Bacteria. J. Babylon Univ. Pure Appl. Sci. 2014, 22, 2377–2382. [Google Scholar]
  144. Mal, G.; Sena, D.S.; Jain, V.K.; Sahani, M.S. Therapeutic Value of Camel Milk as a Nutritional Supplement for Multiple Drug Resistant (MDR) Tuberculosis Patients. Isr. J. Vet. Med. 2006, 61, 88–91. [Google Scholar]
  145. Soleymanzadeh, N.; Mirdamadi, S.; Kianirad, M. Antioxidant Activity of Camel and Bovine Milk Fermented by Lactic Acid Bacteria Isolated from Traditional Fermented Camel Milk (Chal). Dairy Sci. Technol. 2016, 96, 443–457. [Google Scholar] [CrossRef] [Green Version]
  146. Ayyash, M.; Al-Nuaimi, A.K.; Al-Mahadin, S.; Liu, S.-Q. In Vitro Investigation of Anticancer and ACE-Inhibiting Activity, α-Amylase and α-Glucosidase Inhibition, and Antioxidant Activity of Camel Milk Fermented with Camel Milk Probiotic: A Comparative Study with Fermented Bovine Milk. Food Chem. 2018, 239, 588–597. [Google Scholar] [CrossRef] [PubMed]
  147. Ejtahed, H.S.; Niasari Naslaji, A.; Mirmiran, P.; Zraif Yeganeh, M.; Hedayati, M.; Azizi, F.; Moosavi Movahedi, A. Effect of Camel Milk on Blood Sugar and Lipid Profile of Patients With Type 2 Diabetes: A Pilot Clinical Trial. Int. J. Endocrinol. Metab. 2014, 13, e21160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Benkerroum, N.; Mekkaoui, M.; Bennani, N.; Hidane, K. Antimicrobial Activity of Camel’s Milk against Pathogenic Strains of Escherichia Coli and Listeria Monocytogenes. Int. J. Dairy Technol. 2004, 57, 39–43. [Google Scholar] [CrossRef]
  149. Abdel-Hamid, M.; Romeih, E.; Saporito, P.; Osman, A.; Mateiu, R.V.; Mojsoska, B.; Jenssen, H. Camel Milk Whey Hydrolysate Inhibits Growth and Biofilm Formation of Pseudomonas Aeruginosa PAO1 and Methicillin-Resistant Staphylococcus Aureus. Food Control 2020, 111, 107056. [Google Scholar] [CrossRef]
  150. Izquierdo-González, J.J.; Amil-Ruiz, F.; Zazzu, S.; Sánchez-Lucas, R.; Fuentes-Almagro, C.A.; Rodríguez-Ortega, M.J. Proteomic Analysis of Goat Milk Kefir: Profiling the Fermentation-Time Dependent Protein Digestion and Identification of Potential Peptides with Biological Activity. Food Chem. 2019, 295, 456–465. [Google Scholar] [CrossRef]
  151. Hayes, M.; Ross, R.P.; Fitzgerald, G.F.; Hill, C.; Stanton, C. Casein-Derived Antimicrobial Peptides Generated by Lactobacillus Acidophilus DPC6026. Appl. Environ. Microbiol. 2006, 72, 2260–2264. [Google Scholar] [CrossRef] [Green Version]
  152. Villegas, J.M.; Picariello, G.; Mamone, G.; Espeche Turbay, M.B.; Savoy de Giori, G.; Hebert, E.M. Milk-Derived Angiotensin-I-Converting Enzymeinhibitory Peptides Generated by Lactobacillus Delbrueckii Subsp. Lactis CRL 581. Peptidomics 2014, 1, 22–29. [Google Scholar] [CrossRef]
  153. Agyei, D.; Danquah, M.K. Industrial-Scale Manufacturing of Pharmaceutical-Grade Bioactive Peptides. Biotechnol. Adv. 2011, 29, 272–277. [Google Scholar] [CrossRef]
  154. Martinez-Villaluenga, C.; Peñas, E.; Frias, J. Bioactive Peptides in Fermented Foods. In Fermented Foods in Health and Disease Prevention; Elsevier: Amsterdam, The Netherlands, 2017; pp. 23–47. [Google Scholar]
  155. Chalamaiah, M.; Keskin Ulug, S.; Hong, H.; Wu, J. Regulatory Requirements of Bioactive Peptides (Protein Hydrolysates) from Food Proteins. J. Funct. Foods 2019, 58, 123–129. [Google Scholar] [CrossRef]
  156. Witkowska, A.M.; Hickey, D.K.; Alonso-Gomez, M.; Wilkinson, M.G. The Microbiological Quality of Commercial Herb and Spice Preparations Used in the Formulation of a Chicken Supreme Ready Meal and Microbial Survival Following a Simulated Industrial Heating Process. Food Control 2011, 22, 616–625. [Google Scholar] [CrossRef]
  157. Pérez-Rodríguez, F.; Carrasco, E.; Valero, A. Impact of Dehydration and Drying Operations on the Microbial Ecology of Foods. In Quantitative Microbiology in Food Processing; John Wiley & Sons, Ltd.: Chichester, UK, 2016; pp. 160–175. [Google Scholar]
  158. Udenigwe, C.C.; Aluko, R.E. Food Protein-Derived Bioactive Peptides: Production, Processing, and Potential Health Benefits. J. Food Sci. 2012, 77, R11–R24. [Google Scholar] [CrossRef]
  159. Martín, M.-H.J.; Ángel, M.-M.M.; Aarón, S.-L.J.; Israel, B.-G. Protein Hydrolysates as Biostimulants of Plant Growth and Development. In Biostimulants: Exploring Sources and Applications; Springer: Singapore, 2022; pp. 141–175. [Google Scholar]
  160. Zhao, X.; Zhang, X.; Liu, D. Collagen Peptides and the Related Synthetic Peptides: A Review on Improving Skin Health. J. Funct. Foods 2021, 86, 104680. [Google Scholar] [CrossRef]
  161. Cai, G.; Moffitt, K.; Navone, L.; Zhang, Z.; Robins, K.; Speight, R. Valorisation of Keratin Waste: Controlled Pretreatment Enhances Enzymatic Production of Antioxidant Peptides. J. Environ. Manag. 2022, 301, 113945. [Google Scholar] [CrossRef]
  162. Ashaolu, T.J. Nanoemulsions for Health, Food, and Cosmetics: A Review. Environ. Chem. Lett. 2021, 19, 3381–3395. [Google Scholar] [CrossRef]
  163. Silveira, R.F.; Roque-Borda, C.A.; Vicente, E.F. Antimicrobial Peptides as a Feed Additive Alternative to Animal Production, Food Safety and Public Health Implications: An Overview. Anim. Nutr. 2021, 7, 896–904. [Google Scholar] [CrossRef]
  164. Nazeer, N.; Uribe-Diaz, S.; Rodriguez-Lecompte, J.C.; Ahmed, M. Antimicrobial Peptides as an Alternative to Relieve Antimicrobial Growth Promoters in Poultry. Br. Poult. Sci. 2021, 62, 672–685. [Google Scholar] [CrossRef]
  165. Deng, Z.; Yu, H.; Yang, Z.; Hu, L.; Liu, Q.; Wang, Y.; Wei, H.-K.; Peng, J. Gly-Pro-Ala Peptide and FGSHF3 Exert Protective Effects in DON-Induced Toxicity and Intestinal Damage via Decreasing Oxidative Stress. Food Res. Int. 2021, 139, 109840. [Google Scholar] [CrossRef]
  166. Taroncher, M.; Rodríguez-Carrasco, Y.; Aspevik, T.; Kousoulaki, K.; Barba, F.J.; Ruiz, M.-J. Cytoprotective Effects of Fish Protein Hydrolysates against H2O2-Induced Oxidative Stress and Mycotoxins in Caco-2/TC7 Cells. Antioxidants 2021, 10, 975. [Google Scholar] [CrossRef]
  167. Ferrigo, D.; Raiola, A.; Causin, R. Fusarium Toxins in Cereals: Occurrence, Legislation, Factors Promoting the Appearance and Their Management. Molecules 2016, 21, 627. [Google Scholar] [CrossRef] [Green Version]
  168. Islam, M.N.; Tabassum, M.; Banik, M.; Daayf, F.; Fernando, W.G.D.; Harris, L.J.; Sura, S.; Wang, X. Naturally Occurring Fusarium Species and Mycotoxins in Oat Grains from Manitoba, Canada. Toxins 2021, 13, 670. [Google Scholar] [CrossRef]
  169. Tkaczewska, J. Peptides and Protein Hydrolysates as Food Preservatives and Bioactive Components of Edible Films and Coatings—A Review. Trends Food Sci. Technol. 2020, 106, 298–311. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the generation of BAPs. A generic globular protein is hydrolyzed, releasing the encrypted peptides from the protein matrix. Some of the released peptides may present interesting bioactivities, such as antimicrobial effects towards foodborne pathogens—the focus of this review.
Figure 1. Schematic representation of the generation of BAPs. A generic globular protein is hydrolyzed, releasing the encrypted peptides from the protein matrix. Some of the released peptides may present interesting bioactivities, such as antimicrobial effects towards foodborne pathogens—the focus of this review.
Pathogens 12 00477 g001
Table 1. Use of BAPs/hydrolysates derived from food proteins for microbiological control.
Table 1. Use of BAPs/hydrolysates derived from food proteins for microbiological control.
BAP/HydrolysateTreatment/Evaluation MethodObserved EffectsReference
Enzymatic hydrolyzed cottonseed mealIn vivo evaluation of supplementation in feed for broiler chicks (15 or 20 g/kg)Decreased population of Escherichia coli and increased Lactobacilli counts in ileum; positive effects on zootechnical parameters.[21]
Enzymatic hydrolyzed cottonseed mealChallenge in vitro test with Colletotrichum gloeosporioides, E. coli O157:H7, and Staphylococcus aureus.Inhibition of C. gloeosporioides and S. aureus growth; no inhibition on E. coli was observed.[81]
Whey protein hydrolysateChallenge test with Listeria monocytogenes; evaluation in soft cheese-based agar (5 μg/plate), combined or not with LAB strains.In combination with LAB strains, reduction of L. monocytogenes was achieved, although no inhibition was observed when alone.[82]
Anionic peptide-enriched extract derived from whey proteinsChallenge test with Listeria innocua and L. monocytogenes; application in reconstituted Cheddar cheese (10 or 20 mg/g) incorporated with lactococci.Higher anti-listerial activity under higher temperatures and/or low salt content; L. monocytogenes more susceptible than L. innocua; no inhibition on LAB.[83]
Encrypted peptides recovered from soybean meal by-productIn vitro evaluation of aqueous extract fractions towards Gram-positive and -negative pathogens; in silico prediction of the antimicrobial sequences.Inhibition of S. aureus, Acinetobacter genomospecies, Aeromonas hydrophila, E. coli, Salmonella enterica, and Vibrio parahaemolyticus; 83 peptide sequences classified as AMP candidates.[70]
Bovine lactoferrin-derived AMPsShort (~12 residues-long) AMPs derived from the protein were designed and evaluated towards Enterococcus faecium in vivo and ex vivo.Designed AMPs showed high antimicrobial activity on free cells and biofilm, low mammalian cytotoxicity, and membrane-activating mechanisms.[84]
Hen egg-white lysozyme-derived AMPs (enzymatic hydrolysis)Evaluation through a radial diffusion assay.Antibacterial activity against Leuconostoc mesenteroides and E. coli, the latter showing greater susceptibility.[85]
Egg albumin hydrolysates (enzymatic hydrolysis)Evaluation of the antimicrobial activity of hydrolysates through the disc diffusion and tube dilution method.Antibacterial activity against L. monocytogenes, Bacillus cereus, S. aureus, Salmonella Typhimurium, Streptococcus pyogenes, Klebsiella oxytoca, Pseudomonas aeruginosa, Bacillus subtilis, Listeria ivanovii, and E. coli.[86]
Goat- and bovine-milk-derived BAPs (enzymatic hydrolysis)After fungal proteolysis, evaluation through disk diffusion towards bacterial and fungal microorganisms.Antimicrobial activity towards L. monocytogenes, S. aureus, E. coli, S. enterica, P. aeruginosa, Fusarium oxysporum, Penicillium expansum, and Candida albicans; no inhibition against Aspergillus fumigatus was observed.[87]
Water-soluble AMPs recovered from the ripened Brazilian Canastra artisanal Minas cheeseEvaluation of the promoted inhibition on E. coli, comparing different ripening stages and cheese producers; identification of peptide sequences.Observed variations influenced by temperature, pH, and other manufacturing characteristics; identification of six validated AMPs, 8–14 residues long, derived from caseins.[88]
Peptide-rich fractions extracted from Spanish dry-cured ham128 fractions chromatographically purified were evaluated through agar-well-diffusion assay for the inhibition of L. monocytogenes and L. innocua; peptidomic study on the naturally generated BAPs.Two fractions showed inhibitory effects towards Listeria strains; identification of 105 BAPs in the two bioactive fractions, 10 with anti-listerial activity.[89]
Bovine collagen hydrolysates (enzymatic hydrolysis)Antimicrobial activity was evaluated for the hydrolysates (0.5 to 5 mg/mL); peptide profiling of hydrolysates.Hydrolysates showed inhibitory activity towards E. coli, S. aureus, and B. subtilis; no inhibition was achieved against E. faecalis, P. aeruginosa, or Klebsiella pneumoniae. Identification of several peptides with low molar mass (<2 kDa).[90]
Goat-whey hydrolysates (enzymatic hydrolysis)Evaluation of the antimicrobial activity of hydrolysates through disc diffusion method; peptide profiling of fractions.The hydrolysate showed bactericidal effects towards B. cereus, Salmonella Typhimurium, and E. coli; and bacteriostatic activity against S. aureus. Two peptides accounted for the bioactivity.[91]
Rainbow trout by-product hydrolysates (enzymatic hydrolysis)Assessment of inhibitory activity against several bacterial strains.Inhibitory activity was detected towards all tested strains, with the highest activity against Flavobacterium species; prolonged lag phase of bacterial growth.[92]
Table 2. Lactic acid bacteria and their mechanisms to produce AMPs.
Table 2. Lactic acid bacteria and their mechanisms to produce AMPs.
Genus/SpeciesStudied Food ProductMechanism of AMP ProductionReferences
Lactococcus
Lactococcus lactis lactisSkim milkRibosomal synthesis of bacteriocins[123,124]
Lactococcus lactis lactisCottage cheeseProtein hydrolysis, releasing BAPs[125]
Streptococcus
Streptococcus thermophilusMilk, yogurt, soft and hard cheesesRibosomal synthesis of bacteriocins[126,127,128]
Lactobacillus
Lactobacillus acidophilusCheese, yogurtProtein hydrolysis, releasing free amino acids and BAPs[119,129]
Lactobacillus gasseriYogurtProtein hydrolysis, releasing free amino acids and BAPs[119,130]
Lactobacillus helveticusSkim milk supplemented with whey proteinProtein hydrolysis, releasing free BAPs[131]
Lactobacillus delbrueckii bulgaricusSkimmed goat milkProtein hydrolysis, releasing free amino acids and BAPs[132]
Lactiplantibacillus
Lactiplantibacillus plantarumFermented camel milkProtein hydrolysis, releasing free amino acids and BAPs[119,133]
Lactiplantibacillus plantarumPineappleProtein hydrolysis, releasing free amino acids and BAPs[134]
Lactiplantibacillus plantarumWheat grainProtein hydrolysis, releasing free amino acids and BAPs[135]
Leuconostoc
Leuconostoc mesenteroides cremorisCheese, butter, heavy creamRibosomal synthesis of bacteriocins[136,137,138]
Pediococcus
Pediococcus pentosaceusFermented pork sausageProduction of pediocin PA-1/AcH by protein hydrolysis[139]
Enterococcus
Enterococcus faeciumMinas cheeseRibosomal synthesis of bacteriocins[140]
Enterococcus mundtiiMinas cheeseRibosomal synthesis of bacteriocins[140]
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

Corrêa, J.A.F.; de Melo Nazareth, T.; Rocha, G.F.d.; Luciano, F.B. Bioactive Antimicrobial Peptides from Food Proteins: Perspectives and Challenges for Controlling Foodborne Pathogens. Pathogens 2023, 12, 477. https://doi.org/10.3390/pathogens12030477

AMA Style

Corrêa JAF, de Melo Nazareth T, Rocha GFd, Luciano FB. Bioactive Antimicrobial Peptides from Food Proteins: Perspectives and Challenges for Controlling Foodborne Pathogens. Pathogens. 2023; 12(3):477. https://doi.org/10.3390/pathogens12030477

Chicago/Turabian Style

Corrêa, Jessica Audrey Feijó, Tiago de Melo Nazareth, Giovanna Fernandes da Rocha, and Fernando Bittencourt Luciano. 2023. "Bioactive Antimicrobial Peptides from Food Proteins: Perspectives and Challenges for Controlling Foodborne Pathogens" Pathogens 12, no. 3: 477. https://doi.org/10.3390/pathogens12030477

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