Next Article in Journal
Effects of Litsea cubeba Essential Oil–Chitosan/Corn Starch Composite Films on the Quality and Shelf-Life of Strawberry (Fragaria × ananassa)
Previous Article in Journal
Electrostatic Fermentation: Molecular Response Insights for Tailored Beer Production
Previous Article in Special Issue
Recent Advances in the Determination of Milk Adulterants and Contaminants by Mid-Infrared Spectroscopy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides

by
Laura Quintieri
,
Francesca Fanelli
*,
Linda Monaci
and
Vincenzina Fusco
National Research Council of Italy, Institute of Sciences of Food Production (CNR-ISPA), 70126 Bari, Italy
*
Author to whom correspondence should be addressed.
Foods 2024, 13(4), 601; https://doi.org/10.3390/foods13040601
Submission received: 21 December 2023 / Revised: 31 January 2024 / Accepted: 14 February 2024 / Published: 16 February 2024
(This article belongs to the Special Issue Nutrition Quality and Microbiology of Milk)

Abstract

:
Milk is a source of many valuable nutrients, including minerals, vitamins and proteins, with an important role in adult health. Milk and dairy products naturally containing or with added probiotics have healthy functional food properties. Indeed, probiotic microorganisms, which beneficially affect the host by improving the intestinal microbial balance, are recognized to affect the immune response and other important biological functions. In addition to macronutrients and micronutrients, biologically active peptides (BPAs) have been identified within the amino acid sequences of native milk proteins; hydrolytic reactions, such as those catalyzed by digestive enzymes, result in their release. BPAs directly influence numerous biological pathways evoking behavioral, gastrointestinal, hormonal, immunological, neurological, and nutritional responses. The addition of BPAs to food products or application in drug development could improve consumer health and provide therapeutic strategies for the treatment or prevention of diseases. Herein, we review the scientific literature on probiotics, BPAs in milk and dairy products, with special attention to milk from minor species (buffalo, sheep, camel, yak, donkey, etc.); safety assessment will be also taken into consideration. Finally, recent advances in foodomics to unveil the probiotic role in human health and discover novel active peptide sequences will also be provided.

1. Introduction

Due to their content in numerous biologically active components that provide benefits to the host health, milk and milk-derived products can be considered functional foods. Indeed, functional foods are components of the diet that not only provide energy and nutrients but also positively modulate body functions, thus boosting health by reducing the risk of disease and/or by improving a given physiological response [1]. Among the nutritional and functional components of milk and its derivatives, probiotics and biologically active peptides (BAPs) play a pivotal role. Probiotics, as declared by the FAO and the WHO and confirmed by Hill et al. [2], are defined as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host”. Recently, the implications and healthy activities of probiotics against diseases such as irritable bowel syndrome, Parkinson’ disease, and prevention and treatment of allergies have also been recognized [3,4,5]. The mechanisms of action include (i) anti-inflammatory effects via suppression of proinflammatory cytokines; (ii) the modulation of gut microbiota through antagonism and inhibition of pathogen adhesion to the intestinal epithelia via production of bacteriocins, biosurfactants and short-chain fatty acids (SCFAs); (iii) enhancement of the gut barrier function of the intestinal mucosa by downregulation of low-grade mucosal immune activation, production of proteins of tight junctions and expansion of the mucus layer; (iv) development and improvement of the immunity system [3,4,5,6].
BPAs are specific protein fragments, mainly consisting of fewer than 50 amino acids, that positively affect body functions or conditions, thus influencing health. Common bioactivities of BPAs comprise several beneficial effects such as antihypertension, antioxidant, antimicrobial, antidiabetic, and anti-inflammation activities; antihypertensive properties are exerted due to the inhibition of angiotensin I-converting enzyme (ACE) and renin activities as well as the induction of vasodilation via upregulation of cyclo-oxygenase (COX), prostaglandin receptor, endothelial nitric oxide synthase expression and L-type Ca2+ channel blockade [7]; the relaxation of the mesenteric artery and the reduction in blood pressure in a cholecystokinin (CCK)-dependent manner has also been demonstrated for the peptide KFWGK released from bovine serum albumin (BSA) after subtilisin digestion [8]. In CaCo-2 cells, the antioxidant activity of milk-derived peptides has been attributed to the activation of the Keap1/Nrf2 pathway responsible for the overexpression of antioxidant enzymes such as glutathione reductase (GR), NADPH quinone oxidoreductase (NQO1), superoxide dismutase (SOD1) and thioredoxin reductase 1 [9]. The main mechanisms of antimicrobial peptides are instead related to changes in the physiological function of membranes and extravasation of cytoplasmic content [10]. Milk proteins can release BAPs during food processing and gastrointestinal digestion through enzymatic hydrolysis and fermentation. BPAs can also be obtained via chemical synthesis or recombinant deoxyribonucleic acid (DNA) technology of predicted active sequences [11,12]; shotgun proteomics and protein-based bioinformatics represent only an example of the current workflow for the identification and characterization of new potential food-derived bioactive peptides [13]. Likewise, the holistic effects of probiotic supplementation on inter- and extra-intestinal diseases are being demonstrated due to multiomics approaches in probiotic studies, coined “pro-biomics” [14].
The growing interest in milk-derived bioactive components, BPAs and probiotics is determined by the rising demand for sustainable nutraceuticals, i.e., produced by processes with high efficiency and low environmental impact, which are also safe, i.e., having a high bioavailability and none or few unwanted side effects. As consequences of this trend, the investigation of minor dairy species (buffalo, goat, sheep, mithun (Bos frontalis), yak (Poephagus grunniens) camel, donkey, and mare), counting from 11 to 0.2% of worldwide milk production, is increasing in recent years. These latter species show notable differences in composition: ruminant milk (cattle, sheep, and goats) is characterized by a high fat content and more caseins among protein fractions, while non-ruminant milk (mare, donkey) has more lactose and whey proteins content. Also the non-protein nitrogen (NPN) content (free amino acids, peptides, creatine, urea, ammonia, uric acid, orotic acid) is highly variable; for instance, the NPN content in mare milk is approximately 10–15% of the total milk nitrogen content, in cow milk, it is 5%, whereas ruminant milk has approximately 3–5% NPN [15]. Buffalo milk also has higher levels of fats, proteins, lactose, vitamin A, vitamin C and calcium than bovine milk. However, buffalo milk has a lower vitamin E and cholesterol; in addition, buffalo milk exhibits a higher buffering capacity (acidification capacity) than bovine milk. Further differences in the nutritional composition of non-bovine milk are reported by several recent studies [15,16,17].
Consequently, different functional health benefits have been found, e.g., compared to bovine milk, some studies suggest that milks from small ruminants (e.g., goat) cause fewer allergenic reactions due to the protein concentration and polymorphism [16,17]; similarly, conjugated linoleic acid and orotic acid in sheep milk aid the treatment and prevention of type 2 diabetes, cancer, and other diseases [16]. Non-bovine milk products are also good for isolating novel potential probiotics and probiotic carrier candidates [17]. Similarly, novel peptide sequences or peptides with improved stability, bioavailability, and efficiency could be obtained [15]. Taking into account this context, we firstly provide an overview of the detection and characterization of probiotics and BPAs in milk and milk-derived products as well as an updated review of their health-promoting properties; a careful consideration on the safety of probiotics and BPAs will also be given. In addition, we will provide an overview of the current state of the art, forthcoming challenges and tendencies of probiotics and BPAs in milk and its derivatives as well as their role in human health. Moreover, an overview of foodomics used to detect and characterize these healthy products/microorganisms will be provided.

2. Probiotics in Milk and Milk-Derived Products

A growing interest towards healthy foods is emerging worldwide in recent decades, with probiotic foods attracting the highest interest for their beneficial properties exerted on human health. As a result, the research on probiotic microorganisms is growing accordingly. Although probiotics isolated from humans should be more resistant to gastrointestinal conditions, the FAO/WHO [18] reported that the action rather than the source of microorganisms makes them probiotics. Therefore, probiotics may be found not only in humans but also in other ecological niches. Milk, with its high content of nutritious compounds, is a good medium for both beneficial and detrimental microorganisms [19]. Numerous lactic acid bacteria, which are among the most used probiotic microorganisms, have been isolated from milk and their safety and probiotic potential have been assessed (Table 1). Sieladie et al. [20] assessed the safety, cholesterol-lowering properties, and antimicrobial activity of 107 lactobacilli isolated from raw milk in the Western highlands of Cameroon. Fifteen isolates were selected for bile and acid tolerance, and all showed the ability to assimilate cholesterol in vitro and bile salt hydrolase activity [20]. Almost all isolates were sensitive to eight of the nine antibiotics tested, while all showed no hemolytic and gelatinase activity [20]. Only one strain, namely isolate 29V, showed antimicrobial activity against the target pathogens. All isolates were identified as Lactobacillus (Lb.) plantarum (recently amended to Lactiplantibacillus plantarum by phenotypic methods and typed by RAPD-PCR [20]). According to the overall results, the best potential probiotic strains were Lb. plantarum strains 1Rm, 11Rm and 29V [20]. Banwo et al. [21] isolated and identified two Enterococcus faecium strains from raw milk. The strains were characterized for their technological and probiotic features and a safety assessment was carried out, finding them suitable as starters for the production of fermented foods [21]. Eid et al. [22] isolated several lactobacilli from raw cow, buffalo and goat milk and demonstrated their antimicrobial activity against mastitis pathogens. Bin Masalam et al. [23] isolated 46 lactic acid bacteria strains and assessed their safety and probiotic potential. Two Lb. casei, one Lb. plantarum and one E. faecium strains showed the best probiotic potential [23]. Fourteen Lactococcus lactis strains isolated from raw milk and kefir grains were characterized for their technological and probiotic potential, finding that the strains isolated from kefir had a higher probiotic potential than those isolated from milk, which showed the best biochemical and technological features [24]. Reuben et al. [25] assessed the probiotic potential of lactic acid bacteria isolated from indigenous Bangladeshi raw milk, investigating antagonistic activity against pathogenic bacteria, survivability in simulated gastric juice, tolerance to phenol and bile salts, auto- and co-aggregation, adhesion to ileum epithelial cells, α-glucosidase inhibitory activity, hydrophobicity, and antibiotic susceptibility, finding Lb. casei C3, Lb. plantarum C16, Lb. fermentum G9, and Lb. paracasei G10 to be the most promising probiotic bacteria.
Daneshazari et al. [26] carried out a probiotic characterization and safety assessment of Bacillus spp. isolated from camel milk. In particular, tolerance to acid, bile salts and artificial gastric juice was assessed, followed by auto-aggregation, cell surface hydrophobicity, antioxidant characteristics, and ability to adhere to HT-29 cells. Hemolytic and lecithinase activities were also evaluated. The Bacillus subtilis CM1 and CM2 strains were found to be the most promising probiotics [26].
The probiotic properties of Bacillus subtilis GM1, a strain isolated from goat milk, were assessed in vitro [27].
An ancient method to avoid the spoilage of milk, thus preserving it, is fermentation. Raw milk or thermized/boiled milk may be subjected to (i) natural fermentation, (ii) black-slopping or (iii) adjunct of commercial starter or single/multiple autochthonous microbial cultures. Depending on the raw materials used, the production step, the equipment and the manufacturing environment involved, the metabolic activity of the resulting specific microbiota is responsible for the final textural, sensorial, and probiotic features of each fermented milk and dairy product [28,29,30,31,32,33,34,35,36,37,38,39]. Within the microbiota responsible for the transformation of milk into fermented milk and dairy products, a pivotal role is played by lactic acid bacteria (LABs) while yeasts are arising as important contributors for their technological and probiotic attributes [39]. As reported in Table 1, fermented milks from cows as well as minor dairy species are an important source of probiotic microorganisms.
Table 1. Potential probiotic microorganisms isolated from fermented milks b (updated from Fusco et al. [34]).
Table 1. Potential probiotic microorganisms isolated from fermented milks b (updated from Fusco et al. [34]).
StrainSpecies aOriginProbiotic Features Tested Reference
CYC 10058Lactobacillus (Lb.) kefiranofaciensKefirIn vitro functional characterization: acid and bile salt tolerance, adhesion, antimicrobial activity, production, purification, and structural characterization of kefiran, inhibition of Salmonella typhimurium adhesion to Caco-2 cells.[40]
SR246S. cerevisiaeSpontaneous fermented cow milk, SudanIn vitro functional characterization: low pH and bile tolerance, cytokine assay, adhesion to the non-tumorigenic porcine jejunal epithelial cell line.[41]
IS-10506
IS-20506
IS-27526
IS-23427
IS-16183
Lb. plantarum
Lb. plantarum
Enterococcus (E.) faecium
E. faecium
E. faecium
Dadih, Indonesian fermented buffalo milkIn vitro functional characterization: mucus adhesion, inhibition of pathogen adhesion, displacement of pathogens, competition between pathogens and LAB strains.[42]
CDCA 8348Lb. kefiriKefirIn vitro ability to produce antimicrobial compounds. In vitro protective action against the invasion of Salmonella enterica serovar Enteritidis (Salmonella enteritidis). In vitro safety characterization (antibiotic susceptibility and hemolytic activity). In vitro ability to antagonize the cytotoxic effects of clostridial toxins on Vero cells. In vivo impact of CDCA 8348 oral administration for 21 days on the mucosal immune response and gut microbiota in mice. In vitro adhesion properties. In vivo proinflammatory cytokine secretion and the absence of translocation of microorganisms to blood, spleen, and liver in mice orally administered with and without CDCA 8348. [43,44,45,46,47]
BFE6058
BFE6059
Lb. acidophilus
Lb. acidophilus
Maasai fermented milk productsIn vitro functional and safety characterization: resistance to low pH, bile salts deconjugation, tolerance to simulated GIT, auto-aggregation, adhesion properties to human cell lines, cholesterol assimilation, antibiotic susceptibility, DNAse activity, biogenic amine production, mucin degradation, hydrophobicity, hemolysis, antigenotoxic properties (ability to protect cells from DNA damage).[48]
Zhang
ZL12-1
BX6-6
Lb. casei
Lb. helveticus
Lb. plantarum
Koumiss In vitro functional characterization: acid tolerance, antimicrobial activities, ability to grow in MRS with bile salts, and viability in prolonged cold storage of fermented milk.[49]
05AM23
06TCa8
06Tca19
06Tca22
06Tca39
06Tca40
06Tca43
06TC3
Lb. plantarum
Lb. plantarum
Lb. paracasei subsp. paracasei
Lb. paracasei subsp. paracasei
Lb. paracasei subsp. tolerance
Lb. plantarum
Lb. paracasei subsp. paracasei
Lb. delbruekii subsp. lactis
Airag (alcoholic Mongolian fermented mare milk)
Tarag (Mongolian yogurt made from camel milk)
Tarag (Mongolian yogurt made from cow milk)
In vitro functional characterization: adhesion to Caco-2 cells, resistance to acid and bile salts. [50]
T3LLb. coryniformis subsp. torquensTibetan fermented yak milkIn vitro immunomodulating activity.[51]
NS8Lb. helveticusMongolian fermented koumissIn vivo and ex vivo assessment of the anti-inflammatory attributes. In vitro functional characterization: resistance to acid and bile salts, aggregation, adhesion to Caco-2 cells, cell surface hydrophobicity.[52]
KII13Lb. helveticusFermented cow milkIn vitro functional characterization: tolerance to simulated orogastointestinal tract, antimicrobial activity, bile salt hydrolase activity, adhesion, and cholesterol assimilation. In vivo cholesterol-lowering activity (in mice).[53]
S1K3 (MTCC5957)Lb. rhamnosusIndian fermented milkIn vitro functional characterization: antagonistic activity, adhesion, protective effect against Salmonella enterica. In vivo assessment of the protective effect against Salmonella enterica in mice after long-term (30 days) consumption of S1K3-fermented milk. [54]
M1Lb. kefiranofaciensTaiwanese kefir grainsIn vivo and in vitro anti-colitis, anti-asthmatic, antiallergic, and immunomodulatory effect; in vivo oral toxicity. Effects of heat, cold, acid bile salt adaptations on stress tolerance and protein expression.[55,56,57,58,59,60]
DN1Lb. kefiranofaciensKefirIn vivo modulation of gut microbiota and increase in fecal water content in mice induced by administration of DN1.[61]
Lp3Lb. plantarumFermented Tibetan yak milkIn vitro functional characterization: cell surface hydrophobicity, bile tolerance, and survival in simulated GIT. In vitro cholesterol-lowering test. In vivo cholesterol-lowering potential in rats.[62]
Jlus66Lb. paracaseiChinese naturally fermented milkIn vitro functional and safety characterization: tolerance to simulated GIT, in vitro cholesterol-lowering activity, cell surface hydrophobicity, and hemolytic activity. In vivo probiotic characterization: effect of the probiotic strain on non-alcoholic fatty liver in rats.[63]
Pro4
Pro7
Lb. paracasei
Lb. rhamnosus
Raw and fermented camel milk In vitro probiotic and safety characterization: phenol, bile salts, sodium chloride and acid tolerance, antimicrobial activity, antibiotic susceptibility, and hemolysis. In vivo assessment of the mucosal immune response in mice.[64]
DD2Lb. kefiranofaciensKefirIn vitro survivability in an experimental oral environment, antimicrobial activity, and antibiofilm formation activity against S. mutans and S. sobrinus.[65]
C4Lb. plantarumKefirIn vitro evaluation of the fermentation properties and potential probiotic activity in batch culture systems. Physicochemical, nutritional, and organoleptic characterization of a skimmed goat milk fermented with C4. In vivo immunomodulatory effect in mice.[66,67,68]
KCTC 5075Lb. kefiranofaciensKefirIn vivo assessment of the therapeutic effects of extracellular vesicles derived from KCTC 5075 in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease.[69]
K2E. faecalisKalarei, Indian fermented milkIn vitro probiotic and safety characterization: bile salt deconjugation, cholesterol assimilation ability, cholesterol-removing potential, SEM analysis for adsorption of cholesterol to probiotic cell surface, antioxidant potential, hemolytic activity, gelatin-hydrolyzing activity, biogenic amine production, and antibiotic susceptibility.[70]
YS5Lb. plantarumyogurtIn vitro and in vivo cholesterol assimilation. In vitro probiotic and safety characterization: acid and bile salt tolerance, bile salt hydrolase activity, antimicrobial activity, hemolysis, and antibiotic susceptibility.[71]
MSR101Lb. kefiriChinese kefir grainsCharacterization and in vitro antitumor activity of exopolysaccharide produced by the strain.[72]
N16Lactobacillus spp. aIndonesian dadih (fermented buffalo milk) In vitro functional characterization: acid and bile salt tolerance, adhesion to mucosal surface and antagonism against enteric pathogens. [73]
SL2, SL3 and SL5
SN1, SN7, SN8 and SN9
Saccharomyces cerevisiae
S. cerevisiae
Lait caille
Nunu
(African fermented cattle milks)
In vitro functional characterization: resistance to acid and bile salts, effect of yeasts on integrity of the Caco-2 cell monolayers, adhesion, and changes in pH in yeast cells during perfusion with the gastrointestinal pH.[74]
GCC_19M1Lb. plantarumIndian fermented raw milk In vitro probiotic and safety characterization: resistance to acid, sodium chloride, bile salts and pancreatin, tolerance to gastric juice model auto-aggregation, cell surface hydrophobicity, glucose fermentation, antimicrobial activity, antibiotic susceptibility, and hemolysis.[75]
SD11Lb. rhamnosusFermented milkIn vivo functional characterization: effect of fermented milk containing L. rhamnosus SD11 and maltitol on S. mutans in a double-blind, randomized, controlled study in humans.[76]
IS-10506Lb. plantarumDadih (Indonesian fermented water buffalo milk)In vivo probiotic characterization: intestinal stem cell activation to counter inflammation in a rodent model. Level of blood lipopolysaccharide and immune response in HIV-infected children. Faecal secretory immunoglobulin A level and immune response in children younger than two years. Effect of IS-10506 and zinc supplementation on humoral immune response and zinc status of Indonesian pre-school children. Effect on the Scoring Atopic Dermatitis Index (SCORAD) in children with atopic dermatitis. Renal tubular regeneration in pyelonephritic rats.[77,78,79,80,81,82]
BIOTEC006
BIOTEC007
BIOTEC008
BIOTEC011
BIOTEC012
BIOTEC013
BIOTEC014
BIOTEC015
BIOTEC016
Lactococcus (Lc.) lactis
Lc. lactis
Lc. lactis
Leuconostoc (Leuc.) pseudomesenteroides
Leuc. pseudomesenteroides
Lentilactobacillus (Lentil.) kefiri
Lentil. kefiri
Lentil. parakefiri
Lc. lactis
Mexican milk kefir grainsIn vitro probiotic characterization: resistance to simulated GIT, antimicrobial activity, aggregation, antibiotic susceptibility, and GABA production fermentability with commercial prebiotics.[83]
MK Y55
MK L1
Kluyveromyces marxianus
Lc lactis
Raw fermented milkIn vitro functional characterization: acid and bile salt tolerance, lactose utilization, resistance to simulated GIT, manufacture of fermented milk using the probiotic strains and microbial viability during storage, and resistance to simulated GIT of microorganisms in fermented milk.[84]
NWAFU-BIO-BS29Lactiplantibacillus plantarumChinese fermented cow milkIn vitro and in vivo probiotic and safety characterization: antibacterial activity, acid and bile salt tolerance, auto- and co-aggregation, cholesterol-lowering ability, antibiotic susceptibility, hemolysis, detection of resistance and virulence genes, antioxidant activity, short-chain fatty acid production, effects of L. plantarum NWAFU-BIO-BS29 on body weight, organs index, and colon H&E in BALB/c mice.[85]
NWAFU-BIO-BS29
NWAFU-BIO-AS16
NWAFU-BIO-D-S7
Lactiplantibacillus (Lactip.) plantarum
Companilactobacillus crustorum
Lb. gallinarum
Chinese fermented milkIn vitro functional and safety characterization: acid and bile salt and hydrogen peroxide tolerance, antimicrobial activity, auto- and co-aggregation, antioxidant activity, cholesterol-lowering ability, antibiotic susceptibility, hemolysis, detection of resistance and virulence genes, short-chain fatty acids analysis.[86]
EGER41Lactip. plantarumAmabere amaruranu, Kenyan fermented milk In vitro functional and safety characterization: phenol and acid tolerance, antagonistic activity, antibiotic susceptibility, and hemolytic activity. [87]
As21Lactip. plantarumFermented yak milkIn vitro probiotic characterization: hydrogen peroxide tolerance, scavenging activity of diphenylpicrylhydrazyl (DPPH) free radicals, hydroxyl radical scavenging activity, superoxide anion clearance, total antioxidant capacity and superoxide dismutase activity, cell surface hydrophobicity, bile tolerance, tolerance to simulated GIT, antioxidation and ageing experiments in worms, and probiotic effects on the oxidative senescence of Caenorhabditis elegans.[88]
BB101
02
Lb. fermentum
Lb. casei
Fermented milkIn vitro functional and safety characterization: acid and bile tolerance, hemolysis and antibiotic susceptibility. In vivo antiulcerogenic potentials of the probiotic strains in ethanol-induced gastric lesions model in mice.[89]
3 strains
2 strains
15 strains
Lc lactis subsp. cremoris
Leuc. mesenteroides subsp. jonggajibkimchii
Leuc. mesenteroides
Indian fermented cow milk dadih
Indian fermented yak milk dadih
Indian fermented cow milk soft churpi
Hard-variety Indian fermented yak milk chhurpi
Indian fermented cow milk mohi
Indian fermented yak milk philu
In vitro functional characterization: acid, bile and lysozyme tolerance, β-galactosidase activity, hydrophobicity, deconjugation of bile salts, and probiotic genes screening.[90]
LB12Lacticaseibacillus paracaseiDoogh, Iranian fermented milkIn vitro probiotic and safety characterization: resistance to simulated GIT, acid and bile salt tolerance, auto- and co-aggregation, cell surface hydrophobicity, adhesion capacity, antagonistic activity, DNase activity, antibiotic susceptibility, and hemolytic activity.[91]
QAUBL19
QAUBSS1
Bacillus licheniformis
Bacillus subtilus
Dadih, Pakistan fermented milkIn vitro probiotic and safety characterization: survival in simulated GIT, tolerance to acid and bile salts, antibacterial activity, antibiotic susceptibility, cell surface hydrophobicity, auto-aggregation, exopolysaccharide production, bile salt hydrolase activity, in vitro cholesterol removal activity, carbohydrate utilization ability, decarboxylase activity, antioxidant activity, hemolysis, and DNase activity.[92]
AcCh91Levilactobacillus brevisIndian fermented milkIn vitro functional and safety characterization: acid and bile tolerance, auto- and co-aggregation, antimicrobial activity, microbial attachments to hydrocarbons, bile salt hydrolysis. β-galactosidase activity, exopolysaccharide production, cholesterol reduction, hemolysis, screening of probiotic and functional genes, and in silico analysis for safety evaluation.[93]
a The genus Lactobacillus has been reclassified into 25 genera Zheng et al. [94]. In this table, both the old and the new nomenclature are provided. b Where not specified, the milk used for the production of dairy products is cow’s milk.
The probiotic lactic acid bacteria most frequently found in fermented milks are strains of the recently amended Lactobacillaceae family [94]. Apart from lactobacilli, strains of the Pediococcus genus mainly belonging to P. pentosaceus and P. acidilactici species are arising for their probiotic attributes (Table 1) [95]. Among yeasts, certain strains of Saccharomyces cerevisiae have been found to possess probiotic attributes (Table 1). However, certain clinical and foodborne S. cerevisiae strains are arising as opportunistic pathogens so that, as established by the EFSA (who confirmed its Qualified Presumption of Safety, QPS status), the inability to grow above 37 °C and to resist to antimycotics compounds used in human medicine must be demonstrated prior to adding viable cells of strains of this species in the food and feed chain [39].
In Table 2 are listed the probiotic microorganisms isolated from milk-derived products other than fermented milks.
Once again, lactobacilli are the most prevalent followed by strains of Enterococcus spp. However, mainly due to their ability to acquire virulence factors and resistance to several classes of antibiotics as well as their occurrence as opportunistic pathogens, enterococci are not generally recognized as safe (GRAS) microorganism and neither do they have QPS status [165,166]. Thus, their use as probiotics is highly controversial. A similar discussion can be had for yeasts such as Kluiveromyces marxianus (whose anamorph name is Candida kefir), which was declared as a significant opportunistic pathogen by the EFSA [39] despite the probiotic attributes found in certain strains isolated from dairy products (Table 2); however, its QPS status has been confirmed [167].
As depicted in Table 1 and Table 2, studies dealing with the probiotic and safety assessment of microorganisms isolated from fermented milk and dairy products are increasing in recent years; and apart from cow milk, non-bovine milks such as buffalo, ewe, goat, yak and camel milk derivatives are important sources of probiotics and promising carriers of probiotics [168,169,170,171,172,173]. Indeed, although bovine milk still dominates the probiotic market worldwide, there is an increasing trend towards the use of milk from species other than cows to deliver probiotics. This is mainly due to the adequate shelf-life viability of probiotics as well as the intrinsic functionality of non-bovine milk. However, given the absence or low amount of kappa casein, which negatively affects their coagulation capability, camel and donkey milk are mainly used as such for probiotic delivery and only seldom for the production of probiotic cheese, whereas ewe, goat, yak and buffalo dairy products are being frequently used as carriers of probiotics.
As for the beneficial effect of probiotics isolated from milk and milk-derived products, it should be highlighted that, although mandatory, only few articles assessed the probiotic features by in vivo studies focusing only on in vitro tests. Moreover, although a thorough safety assessment should target aspects such as antimicrobial susceptibility, metabolic activity, toxin production, side effects in humans, hemolytic activity, adverse outcomes in consumers, and infectivity in immunocompromised animal models [174], too high a number of the studies listed in Table 1 and Table 2 only carried out the antibiotic susceptibility assessment or did not perform any safety assessment at all. In addition, considering that the beneficial effect of a given probiotic is strain and dose dependent, the species- and strain-level identification of the potential probiotic as well as the enumeration of viable probiotics in a given probiotic food or supplement are mandatory to characterize probiotic microorganisms and authenticate probiotic food/beverage/supplements [175,176,177]. Moreover, the technological features of the potential probiotic should be characterized and their survival in the processing, storage, distribution, and shelf-life within the probiotic food/beverage/supplement should be assessed [176]. Nevertheless, as reported in Table 1 and Table 2, few studies made these assessments. Moreover, apart from a few articles that carried out in vivo studies in rats or in mice (Table 1 and Table 2), only one article [76] (Table 1) assessed the beneficial effects of probiotics via double-blind, randomized, controlled study in humans.

3. Beneficial Effects of Probiotic Milk and Milk-Derived Products

As we mentioned in the previous paragraph, no clinical trials in humans but studies using in vitro cell cultures or animal models have so been far carried out for probiotic milk and milk-derived products containing autochthonous potentially probiotic microorganisms. The pivotal role of probiotics in human health is well known [178] but controlled validated clinical trials are mandatory to verify that the health benefits are not altered or lost when the probiotic is incorporated into the food matrix due to the technological stresses it undergoes during manufacturing. The efficacy of probiotic milk and milk-derived products must be demonstrated in controlled validated clinical trials to prove that the probiotic features are not altered or lost passing from in vitro to in vivo studies. But even animal studies maybe not be adequate predictors of human experiences, humans being quite diverse from animals in terms of lifestyles, diet, and gut microbiome. However, clinical trials of probiotic milk and milk-derived products containing commercial probiotics or probiotics isolated from food matrices other than milk-based products have been carried out demonstrating numerous health benefits [179,180,181,182,183,184], leading to hypothesize that milk and dairy food/beverages containing autochthonous probiotic microorganisms would also positively impact human health. However, it should be highlighted that even numerous health claims associated with many probiotic strains already available on the market have been rejected by the EU due to either (i) insufficient characterization, (ii) invalidity of claims/unproven claims, (iii) the absence of beneficial effects on nutrition and or lack of progress on the physiological state of the body, (iv) lack of scientific basis and/or low quality of studies, and (v) the absence of placebo-controlled, double-blind clinical trials.

4. Bioactive Peptides from Milk Proteins: Overview of Health Benefits and Their Applications

Milk proteins are valuable sources of BPAs, exerting several health-promoting activities; well-known biological activities includes antimicrobial, antibiofilm [185,186,187], anticancer [188] anti-inflammatory [9], antihypertensive [189], antioxidative [190], opioid-like [191], dipeptidyl peptidase IV (DPP-IV), antiviral [192], and mineral-binding ability [190]). Antimicrobial and antibiofilm peptides from milk proteins display a wide spectrum of activity against bacteria and fungi with slower development of resistance mechanisms [187]; anticancer peptides induce distortion of proteins responsible for cancer cell proliferation, a reduction in the enzymatic activities associated with cancer growth, inhibition of the angiogenesis and initiation of necrosis or apoptosis processes. These mechanisms were proved for bovine or non-bovine milk-derived BPAs [193]. Opioid peptides are selective to μ-type (e.g., β-casein f (60–70) YPFPGPIPNSL), δ-type (e.g., αS1-casein f (90–96), RYLGYLE) and κ-type opioid receptors [191]. Recently, Wu et al. [194] also reviewed the potential of milk-derived BAPs affecting the gut microbiome, in turn regulating gut–brain functioning, gut health, and immune system activity.
BPA sequences, lower than 6 kDa, consist of 2 and 20 amino acid residues; BPAs remain inactive in the native protein and exhibit their activity after release and transport to the active site [190]. The biological properties of BPAs are affected by several factors: sequence length, amino acid motifs, and structure (α-helix, β-sheet, β-turn, and random coil); for example, the presence of alanine, cysteine, histidine, lysine, leucine, methionine, proline, valine, tryptophan, and tyrosine may contribute to antioxidant properties [195], while proline, lysine and aromatic amino acid residues contribute to ACEI activity [189,195]; branched-chain amino acid (BCAA)-rich motif sequences show high stability and antimicrobial and anticancer activities [195]. Secondary and tertiary structures are instead responsible for BPA antioxidant properties [196].
Some BPAs, in particular tripeptides, shared sequences among the same protein from different milks (goat, sheep, or camel) [197]; however, changes in BPA activity derived from proteins of different mammals can also be expected depending on the degree of homology between parent proteins [190,198].
Chronic diseases, such as metabolic diseases, have become a worldwide public health issue and several efforts are ongoing to discover novel therapeutic strategies. Overall, most published studies on rodent and cells or including clinical trials to evaluate the effects of BAPs on different pathological conditions were reported for bovine milk-derived BAPs [199,200,201,202]. To the best of our knowledge, there is, instead, limited literature about the biological effects of milk from minor species [203]. Thus, in an attempt to shed light on novel active sequences, Table 3 reports BPAs identified from native proteins of milk from minor dairy species; effective dosages determined in experimental trials, in vitro or in vivo models, were also reported. By contrast, presumptive active sequences by in silico approaches and or not validated in experimental trials were not included [204,205,206]. Overall, most identified bioactive peptide sequences from milk proteins of minor species belonged to caseins (CNs), in particular from αS2-casein (αS2-CN) and β-casein (β-CN); among whey proteins, bioactive peptides were instead released from β-lactoglobulin (β-LG).
The inhibition of the ACE, which converts angiotensin I to the vasoconstrictor agent angiotensin II by lowering blood pressure in cardiovascular disease, is widely reported for the listed peptides (Table 3). Among biological effects, peptides from camel milk affected both glucose transport and metabolism and the structural and functional properties of the pancreatic β-cells and insulin secretion. The main targeted enzymes are major carbohydrate digestive enzymes, i.e., the pancreatic α-amylase and the intestinal α-glucosidase responsible for the digestion of almost 90% of dietary carbohydrates, in turn releasing an abundance of glucose, causing postprandial hyperglycemia. Another enzyme that has been targeted for its inhibition by BAPs is the DPP-IV enzyme, which degrades major incretin hormones involved in the release of insulin in response to glucose [207].
After their release from native protein, BAPs were usually purified, identified, and chemically synthesized to validate the presumptive activity; effective doses were also determined by assaying HPLC fractions of hydrolyzed proteins [208,209,210]. Although BAPs have attracted increasing interest in the development of functional foods or novel drugs, the production of milk-derived BAPs as well as their purification has been limited by their high cost and lack of suitable large-scale technologies. This gap has heavily compromised the application of peptides in commercial products; the crude hydrolysate solution, indeed, is a complex mixture consisting of BPAs and many non-bioactive hydrolyzed compounds. Each peptide shows different charges, physicochemical properties, and molecular weight, making purification an ongoing challenge [211]. Several separation techniques have been employed to purify such peptide mixtures (ultrafiltration, chromatography-based methods, capillary electrophoresis and recently electrodialysis–ultrafiltration) that, however, are mainly used at the lab scale [211]. Thus, commercial food products supplemented with milk protein-derived BPAs are few and mainly consist of bovine milk-derived peptides (e.g., Calpis, Calpis Co., Ltd., Tokyo, Japan; EVOLUS, Valio, Ltd., Helsinki, Finland; Lowpept, nnaves S.A., Pontevedra, Spain; BioZate 1, Davisco Foods, LeSueur, MN, USA; [212,213,214]). Ayyash et al. [215] performed a comparative study of the healthy properties of camel milk and bovine milk fermented with camel milk probiotic strain. Although the authors did not investigate the proteolytic pathways of the indigenous LABs or identify the BAPs derived from the fermentation process, the study demonstrated that the health-promoting benefits (antioxidant, ACE inhibition, and antiproliferative activities) of water-soluble extracts in fermented camel milk were markedly higher than those in fermented bovine milk [215]. A novel food application of peptides from minor dairy species was investigated by Hajian et al. [216]. In this study, a camel milk low-fat ice cream with various concentrations of antioxidant casein hydrolysates was produced; the modified recipe of ice cream improved the melting resistance of the products and registered good acceptability by panelists.
Thus, even though much is known about the peptide sequences with biological effects from milk proteins, more research is still required on novel BAPs from minor dairy species and validation of their mechanisms of action. After digestion, BPAs can be absorbed in the intestine and enter the blood stream directly by ensuring their bioavailability in vivo and their activity at the target site or can be further hydrolyzed, thus reducing their activity; for these reasons, both for BAPs from bovine and other milk species, preclinical and clinical studies are needed to determine which levels are beneficial for health, their dose–response relation, stability, bioavailability and regulatory factors, pharmacokinetics, and residual activity after ingestion in foods.
Table 3. List of bioactive peptides (BPAs) from milk of minor dairy species and related beneficial effects.
Table 3. List of bioactive peptides (BPAs) from milk of minor dairy species and related beneficial effects.
OriginProcessPeptide SequenceNative
Protein
BioactivityDoseReferences
CamelHydrolysis
with
alcalase or papain
KDLWDDFKGL
MPSKPPLL
n.s.α-amylase inhibitory peptidesIC50: 0.027,
0.025 mg/mL
(as RP-HPLC fractions)
[210]
Hydrolysis with pepsin–pancreatinLEEQQQTEDEQQDQL
YLEELHRLNAGY
RGLHPVPQ
n.s.AntioxidantIC50: 0.08–0.19 mg/mL[217]
Hydrolysis with pepsin–pancreatinNEDNHPGALGEPV
KVLPVPQQMVPYPRQ
αs1-CN f (153–164)
β-CN f (185–197)
AntioxidantDPPH radical activity:
0.04, 0.02 mg/mL;
ABTS radical scavenging: 0.1, 0.01 mg/mL
[218]
Trypsin digestRLDGQGRPRVWLGR TPDNIDIWLGGIAEPQVKR VAYSDDGENWTEYRDQGAVEGKn.s.Antioxidant0.6–2.4
mg/mL
[219]
In vitro gastrointestinal digestionLPQVarious
proteins
DPP-IV inhibitory peptidesIC50: 82 µM[197]
HydrolysisLPVPQ
WK
β-CN f (172–177)
αs2-CN f (69–70)
DPP-IV inhibitory peptidesIC50: 43.69 µM
40.6 µM
[220]
CamelHydrolysis with trypsinLPVP,
MPVQA
β-CN
f (172–176)
β-CN
f (186–190)
DPP-IV inhibitory peptides87.0 ± 3.2, 93.3 ± 8.0 µM[205]
Hydrolysis with trypsinVPV, YPI and VPFβ-CNDPP-IV inhibitory peptidesIC50: 6.6, 35.0, 55.1 µM[205]
FermentationMVPYPQRβ-CN f (177–183)Antioxidant
ACEI
IC50: 89, 33 µM
IC50: 30 µM
[205,221]
CheeseLQKW
LLF
β-LG f (58–61)
β-LG f (103–105)
ACEIIC50: 3.5 μM
IC50: 82.4 μM
[222]
Milk fermentationAIPPKKNQDκ-CN
f (107–115)
ACEIIC50:  19.9 μM[223]
In vitro gastrointestinal digestion of camel milkIPP
LPP
VIP
k-CN
β -CN
αS2-CN
ACEIIC50: 5 μM
IC50: 9.6 μM
IC50: 26 μM
[197]
GoatIn silico prediction
and
in vitro validation
SWMHQPP
QSLVYPFTGPIPNSL
YPYQGPIVL
n.s.AntioxidantIC50: 16.83,
24.79,
20.94 µg/mL
[224]
KefirLHLPLP
HLPLP
DKIHP
αs2-CN f (26–31)
αs2-CN f (27–31)
β-CN f (47–51)
ACEIIC50: 5.5 μM
21 μM
233 μg/mL
[225]
Milk and cheeseTVDQHQαS2-CN
f (182–187)
ACEI210 μg/mL[198,226]
CheeseLVYPFPGPIPN
LVYPFPGPIPNSLPQNIPP
β-CN f (58–65)
β-CN f (58–73)
ACEI27.9 μM
5 μM
[226]
Hydrolysate
kefir
TGPIPN
LVYPFTGPIPN
β-CN f (78–83)
β-CN f (73–83)
ACEI316 μM
27.9 μM
[227]
Hydrolysate and cheeseSLPQβ-CN f (84–87)ACEI330 μM[227]
Hydrolysate and cheeseSQPKβ-CN f (181–184)ACEI354 μM[227]
Gastric digestion of isolated whey proteins and caseins of milkPEQSLACQCL
QSLVYPFTGPI
ARHPHPHLSFM
β-LG f (113–122),
β-CN f (56–66)
κ-CN f (96–106)
ACEI4.45 μM
4.27 μM (as mixture)
[228]
In vitro gastrointestinal digestionIAV
VPP
GPV
αS1-CN
β -CN
β -CN
ACEI27 μM
9 μM
4.7 μM
[197]
GoatIn vitro gastrointestinal digestionIPIk-CNDPP-IV-inhibitory activity3.5 μM[197]
Milk casein
hydrolysates
SDIPNPIGSE
NPWDQVKR,
SLSSSEESITH,
QEPVLGPVRGPFP
αS1-CN f (195–204)
αS2-CN f 123–130)
β-CN f (30–40)
β-CN f (207–219).
Anti-diabetic activity0.50 mg/mL
(as hydrolysate fractions)
[229]
Trypsin/chymotrypsin hydrolysatesINNQFLPYPY
MHQPPQPL
κ-CN f (51–60)
β-CN f (144–151)
DPP-IV-inhibitory activity40 μM
350 μM
[230]
Hydrolysis with
neutral and
alkaline proteases
VYPF
FPYCAP
FGGMAH
YPPYETY
YVPEPF
n.s.Antioxidant83–107 µg/mL
(as GFC fractions)
[209]
DonkeyEndogenous
peptides
REWFTFLK
MPFLKSPIVPF
Serum amyloid A protein
f (19–26)
Equus caballus (horse)
β-CN f (130–140)
ACEI0.71
10.3 μM
[15,231]
Endogenous
peptides
WFTFLKEAGQGAKDMWR
GQGAKDMWR
Serum amyloid A protein
f (11–29)
Equus caballus (horse)
Serum amyloid A protein
f (20–29)
Equus caballus (horse)
Antioxidant16.8 μM
22.7 μM
[15,231]
YakHydrolysis with trypsin and alcalaseTPVVVPPFLβ-CN f (90–100)Anti-breast cancercell viability inhibition
rate
IC50: 250–500 μg/mL
[232]
Pepsin hydrolysisKVISMI
RVMFKWA
n.s.Antimicrobial against Bacillus subtilis, Staphylcoccus aureus, Listeria innocua, Escherichia coli, Enterobacter cloacae, Salmonella paratyphi and
Candida albicans, Saccharomyces cerevisiae
MIC: 4–32 μg/mL
MIC: 8–32 μg/mL
[233]
YakTwo-step enzymatic hydrolysis
with trypsin and pepsin
KALNEINQFαS2-CNAntioxidant25–100 μg/mL[234]
Two-step enzymatic hydrolysis with
trypsin and pepsin
MHQPHQPLPPTVMFβ-CNAntioxidant6.25, 12.5,
25, 50 μg/mL
[235]
Fermentation
by L. plantarum JLAU103
LYLKPRunknownNeuroprotective effects on H2O2-injured HT-22 cells100–200 μM[236]
Hydrolysis
with alcalase
RELEELβ-CN f (1–6)Superoxide anion and hydroxyl radical scavenging activityIC50: 0.52
0.69 mg/mL
[237]
Hydrolysis with trypsin, pepsin, alcalase,
flavourzyme,
papain and neutrase
YQKFPQY
LPQNIPPL
SKVLPVPQK
LPYPYY
FLPYPYY
αS2-CN f (89–95)
β-CN f (70–77)
β-CN f (168–176)
κ-CN f (56–61)
κ-CN f (55–61)
ACEIIC50: 380 μM[238]
Hydrolysis with alcalasePPEIN
PLPLL
κ-CN f (156–160)
β-CN f (136–140)
ACEIIC50: 290 μM
IC50: 250 μM
[238]
yakTrypsin digestion combined with
QSAR molecular docking
KYIPIQκ-CN f (45–50)ACEIIC50: 7.28 μM[239]
Hydrolysis with
thermolysin/alcalase and thermolysin/proteinase K
FPQY
MPFPKYP
MFPPQ
QWQVL
αS2-CN f (91–95)
β-CN f (109–115)
β-CN f (156–160)
κ-CN f (75–79)
ACEIIC50: 12.4
2.9
40.8,
112 μM
[240]
SheepCheeseLKKISQ
VRYL
YIPIQY
LVYPFTGPIPN
αs2-CN f (165–170)
αs2-CN f (205–208)
κ-CN f (25–30)
β-CN f (58–68)
ACEIIC50: 2.6 μM
24.1 μM
10 μM
27.9 μM
[241,242]
Milk, kefirPYVRYLαs2-CN f (203–208)ACEI,
antioxidant
2.4 μM
nd
[242,243]
CheeseALPMHIR
IIVTQTMK
IDALNENK
β-LG f (142–148)
β-LG f (1–8)
β-LG f (84–91)
ACEIIC50: 62.6 μM
IC50: 70.8 μM IC50: 71.2 μM
[198]
CheeseVMFPPQSVL
VVAPFPEV
β-CN f (155–163)
αs1-CN f (24–31)
Antimicrobial activity against Gram-positive and Gram-negative bacteria 25–110 µg/mL[244]
In vitro gastrointestinal digestionIPAvarious
proteins
DPP-IV-inhibitory activity49 μM[197]
BuffaloHydrolysis of whey with Dregea sinensis Hemsl. proteaseDQPFFHYN
YSPFSSFPR
α-2-glycoprotein 1,
zinc-binding
clustering
Anti-inflammatory activity50–200 µg/mL[245]
Hydrolysis with
papain
GPFPIIV
YPVEPFT
β-CNACEI9.1 µg/mL
(as fraction)
[208]
Hydrolysis with trypsin and pepsinEDVPSER
NAVPITPTL
VLPVPQK
HPHPHLSF
αs1-CN f (84–90)
αs2-CN f (115–123)
β-CN f (170–176)
k-CN f (98–105)
Osteoblast proliferation activity>10 ng/mL[246].
Chemical
synthesis
VLPVPQKβ-CN f (170–176)Antiapoptotic effect>30 ng/mL[247]
Mare (equine)Koumiss YQNPRLGPTGELDPATQPIVAVHNPVIVβ-CN f (217–241)ACEI14.53 μM[15]
Hydrolysis of whey with papainNLEIILR
TQMVDEEIMEKFR
β-LG f (71–77)
β-LG f (143–155)
DPP-IV-inhibitory activity86.34 μM
69.84 μM
[248]
MilkVAPFPQPVVPYPQRβ-CN f (176–189Antioxidant<500 μg/mL[249]
n.s. = not specified; IC50: half maximal inhibitory concentration; MIC: minimal inhibitory concentration; nd: not determined.

5. Safety of Milk-Derived Bioactive Peptides

Commercialization of BPAs from milk proteins is still limited [193]. In addition to the lack of a cost-effective method of production, scarce information regarding their efficacy, safety, and bitter taste are the reasons heavily hindering their application in food and pharmaceutical sectors. Despite promising beneficial activities in humans, claims of BPAs are not supported by convincing evidence, appropriate clinical studies methodology, robust findings and, therefore, are usually rejected by the European Food Safety Authority (EFSA). In particular, human clinical trials should have an affordable experimental design including the use of appropriate control/placebo, eligibility criteria, and robust statistics to escape rejection of the obtained results. As regards BPAs as a supplement of food products, the EFSA also requests information on peptide quantity, peptide sequence and length, amino acid composition, molecular weight distribution, manufacturing process, physicochemical characteristics, and conditions of use. All of this combined with information on peptide bioavailability in humans, the understanding of mechanism of action in preclinical studies, and the safety of BPA administered in a chronic manner are areas in need of attention [193,250]. Overall, health claims of bioactive peptides can be divided into three types, “general function claims” (e.g., “Milk peptides help support the normal function of immune system”), “disease risk reduction claims” (e.g., “Egg peptides have shown to reduce cholesterol levels”), and “claims relating to children’s development or health” (e.g., fish peptides help children’s cognitive development). Researchers should submit an application to the EFSA through an EU country’s competent authority. After evaluation by the EFSAs’ Dietetic products, Nutrition and Allergies panel (EFSA NDA Panel), approval of a claim could be mainly based on the scientific substantiation and ability of an average consumer to understand the beneficial effects of the claims [251]. The application process usually takes from 6 to 8 months [252].
BAP safety is at the basis of clinical studies and food applications. Although their consumption is thought to be harmless, toxicological investigations must be increased. To date, no standard guidelines have been established for the safety assessment of milk-derived peptides, thus just a few investigations on BPA toxicity on humans have been undertaken. The toxicity of peptides on eukaryotic cells is usually measured by assaying their cytotoxicity or hemolytic activity on red blood cells before preclinical studies [192]. An in vitro work conducted by Ponstein-Simarro Doorten et al. [253] showed that daily ingestion of a hydrolysate derived from bovine milk (2 g/kg body weight) and containing the antihypertensive fragments IPP and VPP did not cause mutagenic or clastogenic effects [253]. Similarly, acute (2000 mg/kg) and daily (1000 mg/kg for 4 weeks) ingestion of casein hydrolysate [rich in ACEI RYLGY and AYFYPEL from αs1-casein fragments (90–94) and (143–149)] neither had any histological impact nor caused mortality in mice [254]. Also, intake of IPP-rich milk protein hydrolysates (7.5 mg IPP) for 1 month did not show adverse effects on hematology and clinical laboratory parameters with the exception of a significant increase in flatulence [199]. Other studies evaluating the toxicological aspects of milk-derived BPAs have been determined and the results to date suggest that they are safe [192].
Given the likelihood of the formation of allergenic peptides from food proteins, before their launch on the market, bioactive peptides should also be tested for allergenicity response. BAPs such as NSAEPEQSLC and VRTPGVDDGAL (from milk β-lactoglobulin) exhibit allergic reactions in humans. Similarly, the ACE inhibitor FFVAPFPE VFGK obtained from the casein hydrolysate was predicted to be an allergic peptide by the BIOPEP-UWM database [255], highlighting the need to provide allergenic warnings for products containing the related BAP.

6. The Role of Foodomics in the Detection and Characterization of Bioactive Peptides and Probiotics

Foodomics has recently emerged as an innovative and promising discipline aiming at studying the relationship between food safety and quality and food nutrition and composition by complementing omics techniques such as proteomics, genomics, metabolomics and transcriptomics with chemometric and bioinformatics tools. It has expanded the application fields, proving to be a valuable approach to improve the food safety and quality of products for the health and well-being of consumers, and for a better understanding of the underlying mechanisms and pathways behind the development of the sensory and technological properties of foods [256,257,258]. Foodomics can help to improve the quality of dairy products [259] and in understanding technological processes of complex matrix requiring sophisticated technology able to extract large amounts of information for the detection of fraud and/or adulterations, microbiological safety, and the assessment and improvement of transformation industrial processes (e.g., fermentation and ripening). For a better and more exhaustive characterization of the complexity of the biological system of a food, omic technologies can be used at a multilevel mode, which is known as the multiomic approach, as demonstrated by its application in the control and quality improvement of milk-based products [259]. Due to the complexity of samples, no individual omic approach alone is able to unravel the complexity of the response and demonstrate a specific health claim of a potential probiotic formulation. To this aim, the multiomic approach and data integration are the novel frontiers for studying probiotics [260]. A system biology approach enables the integration of large datasets from experimental and theoretical models derived from genomics, metagenomics, transcriptomics and metatranscriptomics, proteomics, metabolomics, peptidomics and lipidomics, providing a comprehensive picture of the complex network of biological processes necessary to evaluate the beneficial effect of probiotic strains to the host in the treatment of specific diseases [261].
These high-throughput approaches generating big data complemented with different cutting-edge analytical platforms, big data output, and bioinformatic tools have paved the way for a new concept of high-throughput analysis for elucidation of cellular mechanisms and pathways or for providing new knowledge on food safety and quality [262]. The utilization of such a multiomic approach generates data at different expression levels such as gene, transcript, protein, and finally metabolite. In this context, omics technologies target the expression of genes (genomes), transcription (transcriptomes) into mRNA, translation into proteins (proteomes) and secretion of metabolic by-products (metabolomes) within a specific sample. In Figure 1, the involvement of omics technologies in the identification and exploitation of probiotics and bioactive peptides is schematized.

6.1. Genomics and Transcriptomics

The first complete genome of a probiotic strain, the Lactococcus lactis ssp. lactis IL1403, was published in 2001 [263]. Since then, the contribution of omics technologies in the detection and characterization of probiotics strains has been pivotal and revolutionary, enabling the understanding of the biology of individual strains, deciphering the composition, evolution, dynamics, sensing and communication of complex microbial populations, their interaction with the host and activities related to their probiotic function [264].
Ventura et al. in 2009 [265] defined with the term propiogenomics the area of genomic-based studies of probiotic bacteria conferring health benefits to the host. The availability of hundreds of genomic sequences of potential probiotic strains and pan-genome analysis has enabled depicting the genetic signature of features associated with the probiotic functions: (i) the adaptation and colonization of the gut environment [266,267], which involved the presence of genes coding for human mucus-binding protein [268] conferring adherence capability and immunomodulatory effect, the presence of cell surface proteins, permeases, hydrolases and transporters [269]; (ii) salt and bile tolerance: the bile salt hydrolase (BSH) activity is responsible for the cholesterol-lowering effect of the probiotic strains. Survival in the low-pH environment of the gut is also important as a technological property, especially in the food sector, considering that yogurt and fermented milks are the most used carriers for probiotics administration. Despite the importance of efflux pumps and changes in the cell envelope [270,271,272], genomic analysis supported the identification of argC, argH, and dapA, as acid-tolerance-associated genes, which may play an essential role in high acid tolerance [273,274] in the probiotic Bifidobacterium longum NCIM 5672; (iii) antagonism against pathogens through production of antimicrobial compounds and short-chain lactic and fatty acids [275] with anti-inflammatory properties, gut barrier protection, and immunomodulation [276]. The bacteriocins are proteinaceous compounds that are able to damage the outer membrane of pathogenic bacteria, act as a protective barrier against biofilm development and have also been linked to probiotic immunomodulatory, anticancer, antioxidant, antibacterial, and cholesterol-lowering activities [277].
Genomic analysis also enables the preliminary in silico evaluation of the absence of undesirable traits, such as the presence of multidrug resistance or virulence genes [278,279,280,281]. This analysis is an essential requisite for pre-selection and utilization of strains in industrial formulation, which is regulated by the EFSA [282].
Furthermore, studying the genomes of probiotic microorganisms enables tracing the origin and evolution of strains and their relationship with gut microbiome, the genomic plasticity and the acquisition of genomic traits favoring their adaptation to the gut environment or the exertion of metabolic activities through horizontal gel transfer, such as in the L. lactis domestication [283].
The availability of the complete genomic sequences of probiotic microorganisms has contributed to the clarification of the correct taxonomy of a strain, which is pivotal to determining the authenticity of the probiotic authenticity [174,177], in particular in commercial broadly used strains (providing information for the development of genome-based methods to assess protocols to define the formulation of complex blends of multiple probiotics) [284]. To this aim, metagenomic approaches have been recently developed [285,286] and also combined with other omics technologies.
Metagenomic sequencing has enabled the taxonomic reconstruction of the human gut microbiome [287] and is crucial for evaluating the gut dysbiosis underlying diseases and the effect of probiotics administration on this ecosystem.
In addition, while amplicon-based metagenomic analysis provides a complete picture of bacterial population in a particular food product, shotgun-based metagenomics is also able to provide knowledge related to the potential probiotic features embedded, which can be exploited by these products or by the microorganisms in them.
Considering their potential role in modulating gut microbiota, this approach has recently been commonly used to characterize traditionally fermented foods, also with respect to safety aspects, such as the presence of antibiotic resistance genes. Kefir is one of the most studied milk-based fermented food, and recent works [288,289,290] applied metagenomics to evaluate the presence of probiotic species and predict their functionalities and identify isolates with antibacterial properties. A similar approach was used by Yasir et al. [291] to describe antimicrobial resistance genes in pasteurized and homemade fermented Arabian laban. Gioddu, which is the only Italian fermented milk product, was characterized by Maoloni et al. [292], who revealed a complex microbial population, dominated by Lactobacillus delbrueckii, but also including L. kefirii, and thus suggesting the presence of bioactive compounds similar to those of kefir.
Shangpliang et al. [293] applied a metagenomic approach to decipher the biomarkers for genes encoding different health-promoting functionalities in laal dadhi, an Indian fermented milk-based product. Genome mining of the metagenome-assembled genome of the five major abundant species (Lactococcus lactis, Lactococcus chungangensis, Lactobacillus helveticus, Streptococcus thermophilus and L. delbrueckii) enabled the identification of different probiotic and prebiotic functions, such as immunomodulation, short-chain fatty acid production, essential amino acid, antitumor genes, antimicrobial peptides, and vitamin biosynthesis.

6.2. Metabolomics

NMR- and MS-based technologies, particularly in the case of metabolomics, are essential tools for metabolome profiling [294]. Metabonomics is a subset of metabolomics and is defined as the quantitative measurement of the multiparametric metabolic responses of living systems to pathophysiological stimuli or genetic modification [295]; it can be considered as chemical profiling of a biological matrix (fluid, tissue, cell, etc.) since it aims at systematically profiling the widest range of small metabolites, ideally the complete collection (metabolome), present in that matrix, all this following an untargeted, comprehensive, and quantitative approach. UPLC-MS-based metabolomics has been exploited to track changes in the milk metabolomes during fermentation by two probiotic strains, Lacticaseibacillus paracasei PC-01 and Bifidobacterium adolescentis B8589 [296]. Such investigations aimed at analyzing probiotic-specific fermentative changes in milk providing information on the probiotic metabolism in that matrix and the potential beneficial mechanism of probiotic fermented milk, or investigating changes in the metabolites of fermented milk with novel probiotic Lactobacillus plantarum P9 [297]. The mentioned strategy can be applied to similar studies to characterize the fermentation process operated by probiotic strains in milk-based products.

6.3. Proteomics and Peptidomics

Among the numerous technologies available, HPLC coupled to mass spectrometry (HPLC-MS) or capillary electrophoresis (CE) is very useful in peptidomics, proteomics, and metabolomics [298,299].
In proteomic studies, chromatography coupled with mass spectrometric detection enabled the identification of many bioactive peptides obtained in fermented milk and its hydrolysate thus proved to be a promising procedure in reducing analysis time and streamlining the whole process, skipping the traditional steps of peptide isolation and purification [300]. Proteomics is typically employed for qualitative analyses and characterization of protein/peptides [301] in the early detection of the foodborne parasite, etc. [302]. Proteomics is also an important and robust tool in nutrition and screening metabolic pathways.
Metaproteomics is also a recent term and can be described as proteomics at the microbial level [303], and it is applicable in microbial studies for a better understanding of beneficial and spoilage or pathogenic microorganisms in foods under growing under different conditions [304]. Siragusa et al. [305] investigated the proteomics of L. plantarum in food materials providing some insights into growth behavior and microbial performance, making the adaptation of the strains possible in food biotechnology. De Angelis et al. [306] gathered numerous works in which the metabolic pathways, biotechnological characteristics, and interactions between different ecosystems of Lactobacillus sp., microorganisms widely used for the production of fermented meat, dairy and vegetables, are unveiled by using metaproteomics. The reconstruction of the metabolic pathways by means of bioinformatics may help in understanding these interactions. Combining metabolomics, transcriptomics, and proteomics as well as analyzing the resulting data enable a better understanding of the microbial metabolism underlaying the transformation of milk in its derivative products.
Proteomics has been used in parallel with metabolomics to characterize the metabolism of different strains of L. delbrueckii subsp. lactis and L. delbrueckii subsp. bulgaricus, describing differentiated metabolic pathways for amino acids, folate, and sugar [133,307,308,309] and quantitatively detecting metabolites such as organic acids (e.g., acetate, lactate and formate) and fatty acids [310]. Metabolomic analysis has been used in numerous recent studies to characterize the activity of microorganisms involved in dairy biotechnology [311].
The recent development of ‘omics’ technologies has also enabled performing an investigation on the fate of food in the gastrointestinal tract and characterization of bioavailable food components. It has also provided a better understanding of the way that foods are metabolized by the human body [312]. More work will be necessary in the future to integrate current knowledge with the human physiological response upon food consumption [312].
Peptidomics is also gaining steadily increasing attention in the last two decades [313]. In the past, wet chemistry experiments and in vitro and in vivo characterization were conducted to identify and characterize BPAs, whereas mass spectrometry has been combined with machine learning to discover bioactive peptides nowadays [313]. In general, to circumvent the high cost for conducting in vitro and in vivo assays, bioinformatics is helpful in: (i) screening BPAs bioactivities, (ii) evaluating their bioavailability, (iii) exploring interaction mechanisms, (iv) assessing ADMET (absorption, distribution, metabolism, excretion, and toxicity) and allergenicity properties, etc. [314]. Although, bioinformatics has also highly progressed in this regard with newly developed models, algorithms, software, docking strategies and web servers, its potential use in the discovery of BPAs has yet to be fully explored. There is a broad spectrum of bioinformatics-aided procedures from BPA preparation to the evaluation of its bioavailability, bioactivity, allergenicity, taste and ADMET properties, which has contributed to the creation of bioactive peptide databases for efficiently retrieving in-depth information. To date, there are dozens of bioactive peptide databases available, each documenting one or more bioactivities (e.g., BIOPEP), so that users can gather a wide range of information (bioactivities, sources, articles, etc.) about a given peptide [315]. In addition, peptides are classified into different categories (source of origin, bioactivity, etc.), thus facilitating the data mining process before undertaking other bioinformatics studies (e.g., QSAR analysis).

7. Conclusions and Future Perspectives

Milk and its derivatives, above all fermented milk-based foods, have represented a main staple of the human diet throughout history and have gained considerable attention in recent years due to their health and nutritional benefits. Macro- and micronutrients provide nutritional properties, whereas BPAs and probiotics, which can be released during manufacturing processes or colonize milk and its derivatives, are responsible for the nutraceutical properties of these products. Several genera of probiotic bacteria such as bifidobacteria and lactobacilli have been isolated from milk and used to produce dairy products with a high standard of safety, quality and high nutritional and functional properties. Likewise, a long list of BPAs has been identified in native proteins from cow milk or milk from minor species; more from the latter have been obtained due to the efforts of recent studies aimed at discovering novel active sequences and developing innovative functional foods. Recent technologies in foodomics coupled with bioinformatic tools have efficiently contributed to gaining more insights into the structural elucidation of bioactive peptides and gaining an understanding of their functional properties as well as promoting and accelerating the understanding of host–probiotics/peptides interactions. This has contributed to revealing the genetic information, evolutionary relationship, physiological properties, metabolic network, and mechanism of action of probiotics and peptides. However, despite wide interest and the acquired knowledge, more investigations need to be performed for in-depth safety assessment and to elucidate the mechanisms of actions at the basis of the prevention or treatment of some diseases (allergies, metabolic disorders, etc.). Consequently, this summary of the current knowledge on probiotics and peptides from milk will help to further promote the intake of milk and derivatives produced from probiotic microbes able to improve the digestive health and overall nutritional well-being of consumers. Meanwhile, obstacles such as the need to implement efficient and cost-effective strategies for industrial-scale production, good manufacturing practices as well as well-designed clinical trials have been highlighted as challenges to be overcome in order to achieve commercialization for daily use in the human diet, on approval of the claims made.

Author Contributions

Conceptualization L.Q., L.M., F.F., V.F. and L.Q. wrote the part of this manuscript dealing with bioactive peptides, while V.F. wrote all the parts on probiotics and was responsible for the overall review and quality control. F.F. and L.M. wrote the paragraph on foodomics for probiotics and bioactive peptides, respectively. F.F. made Figure 1. All authors have read and agreed to the published version of the manuscript.

Funding

This review was prepared within the National project “l’Evoluzione delle Produzioni Lattiero-Casearie: le Biotecnologie valorizzano la Tradizione”—ELEVATO, n. F/200112/03/X45, Fondo per la Crescita Sostenibile—Sportello “Agrifood” PON I&C 2014–2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest. The authors are not aware of any affiliations, memberships, funding, or financial holdings that might be perceived as affecting the objectivity of this review.

References

  1. Donato-Capel, L.; Garcia-Rodenas, C.L.; Pouteau, E.; Lehmann, U.; Srichuwong, S.; Erkner, A.; Kolodziejczyk, E.; Hughes, E.; Wooster, T.J.; Sagalowicz, L. Chapter 14—Technological means to modulate food digestion and physiological response. In Food Structures, Digestion and Health; Boland, M., Golding, M., Singh, H., Eds.; Academic Press: Cambridge, MA, USA, 2014; pp. 389–422. [Google Scholar] [CrossRef]
  2. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
  3. Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics may have beneficial effects in Parkinson’s disease: In vitro evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef]
  4. Lopez-Santamarina, A.; Gonzalez, E.G.; Lamas, A.; Mondragon, A.D.C.; Regal, P.; Miranda, J.M. Probiotics as a possible strategy for the prevention and treatment of allergies. a narrative review. Foods 2021, 10, 701. [Google Scholar] [CrossRef]
  5. Simon, E.; Călinoiu, L.F.; Mitrea, L.; Vodnar, D.C. Probiotics, prebiotics, and synbiotics: Implications and beneficial effects against irritable bowel syndrome. Nutrients 2021, 13, 2112. [Google Scholar] [CrossRef]
  6. Du, Z.; Comer, J.; Li, Y. Bioinformatics approaches to discovering food-derived bioactive peptides: Reviews and perspectives. TrAC Trends Anal. Chem. 2023, 162, 117051. [Google Scholar] [CrossRef]
  7. Okagu, I.U.; Ezeorba, T.P.C.; Aham, E.C.; Aguchem, R.N.; Nechi, R.N. Recent findings on the cellular and molecular mechanisms of action of novel food-derived antihypertensive peptides. Food Chem. 2022, 4, 100078. [Google Scholar] [CrossRef] [PubMed]
  8. Koyama, D.; Sasai, M.; Matsumura, S.; Inoue, K.; Ohinata, K. A milk-derived pentapeptide reduces blood pressure in advanced hypertension in a CCK system-dependent manner. Food Funct. 2020, 11, 9489–9494. [Google Scholar] [CrossRef] [PubMed]
  9. Tonolo, F.; Fiorese, F.; Moretto, L.; Folda, A.; Scalcon, V.; Grinzato, A.; Ferro, S.; Arrigoni, G.; Bindoli, A.; Feller, E.; et al. Identification of new peptides from fermented milk showing antioxidant properties: Mechanism of action. Antioxidants 2020, 9, 117. [Google Scholar] [CrossRef] [PubMed]
  10. Seyfi, R.; Kahaki, F.A.; Ebrahimi, T.; Montazersaheb, S.; Eyvazi, S.; Babaeipour, V.; Tarhriz, V. Antimicrobial peptides (AMPs): Roles, functions and mechanism of action. Int. J. Pept. Res. Ther. 2020, 26, 1451–1463. [Google Scholar] [CrossRef]
  11. 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]
  12. Losurdo, L.; Quintieri, L.; Caputo, L.; Gallerani, R.; Mayo, B.; De Leo, F. Cloning and expression of synthetic genes encoding angiotensin-I converting enzyme (ACE)-inhibitory bioactive peptides in Bifidobacterium pseudocatenulatum. FEMS Microbiol. Lett. 2013, 340, 24–32. [Google Scholar] [CrossRef] [PubMed]
  13. Carrera, M.; Pazos, M.; Aubourg, S.P.; Gallardo, J.M. Shotgun proteomics and protein-based bioinformatics for the characterization of food-derived bioactive peptides. Methods Mol. Biol. 2021, 2259, 215–223. [Google Scholar] [PubMed]
  14. Kiousi, D.E.; Rathosi, M.; Tsifintaris, M.; Chondrou, P.; Galanis, A. Pro-biomics: Omics technologies to unravel the role of probiotics in health and disease. Adv. Nutr. 2021, 12, 1802–1820. [Google Scholar] [CrossRef] [PubMed]
  15. Guha, S.; Sharma, H.; Deshwal, G.K.; Rao, P.S. A comprehensive review on bioactive peptides derived from milk and milk products of minor dairy species. Food Prod. Process. Nutr. 2021, 3, 1–21. [Google Scholar]
  16. Kanetkar, P.; Paswan, V.K.; Rose, H.; Shehata, A.M.; Felix, J.; Bunkar, D.S.; Rathaur, A.; Yamini, S.; Bhinchhar, B.K. Appraisal of some ethnic milk products from minor milch animal species around the world: A review. J. Ethn. Food 2023, 10, 40. [Google Scholar] [CrossRef]
  17. Wang, D.; Zhou, Y.; Zheng, X.; Guo, J.; Duan, H.; Zhou, S.; Yan, W. Yak milk: Nutritional value, functional activity, and current applications. Foods 2023, 12, 2090. [Google Scholar] [CrossRef] [PubMed]
  18. FAO/WHO. Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Liver Lactic Acid Bacteria; Food and Agriculture Organization and World Health Organization Joint Report; WHO: Geneva, Switzerland, 2001; 34p.
  19. Fusco, V.; Quero, G.M. Culture-dependent and -independent nucleic acid-based methods used in the microbial safety assessment of milk and dairy products. Comp. Rev. Food Sci. Food Saf. 2014, 13, 493–537. [Google Scholar] [CrossRef] [PubMed]
  20. Sieladie, V.; Zambou, F.; Kaktcham, M.; Cresci, A.; Fonteh, F. Probiotic Properties of Lactobacilli Strains Isolated from Raw Cow Milk in the Western Highlands of Cameroon. Rom. Food Biotechnol. 2011, 9, 12–28. [Google Scholar]
  21. Banwo, K.; Sanni, A.; Tan, H. Technological properties and probiotic potential of Enterococcus faecium strains isolated from cow milk. J. Appl. Microbiol. 2012, 114, 229–241. [Google Scholar] [CrossRef]
  22. Eid, R.; El Jakee, J.; Rahidy, A.; Asfour, H.; Omara, S.; Kandil, M.M.; Mahmood, Z.; Hahne, J.; Seida, A.A. Potential antimicrobial activities of probiotic Lactobacillus strains isolated from raw milk. Probiotics Health 2016, 4, 1000138. [Google Scholar]
  23. Bin Masalam, M.S.; Bahieldin, A.; Alharbi, M.G.; Al-Masaudi, S.; Al-Jaouni, S.K.; Harakeh, S.M.; Al-Hindi, R.R. Isolation, molecular characterization and probiotic potential of lactic acid bacteria in Saudi Raw and fermented milk. Evid.-Based Complement. Altern. Med. 2018, 25, 7970463. [Google Scholar] [CrossRef]
  24. Yerlikaya, O. Probiotic potential and biochemical and technological properties of Lactococcus lactis spp. lactis strains isolated from raw milk and kefir grains. J. Dairy Sci. 2018, 102, 124–134. [Google Scholar] [CrossRef] [PubMed]
  25. Reuben, R.C.; Roy, P.C.; Sarkar, S.L.; Rubayet Ul Alam, A.S.M.; Jahid, I.K. Characterization and evaluation of lactic acid bacteria from indigenous raw milk for potential probiotic properties. J. Dairy Sci. 2020, 103, 1223–1237. [Google Scholar] [CrossRef] [PubMed]
  26. Daneshazari, R.; Rabbani Khorasgani, M.; Hosseini-Abari, A.; Kim, J.H. Bacillus subtilis isolates from camel milk as probiotic candidates. Sci. Rep. 2023, 13, 3387. [Google Scholar] [CrossRef] [PubMed]
  27. Daneshazari, R.; Rabbani Khorasgani, M.; Hosseini-Abari, A. Preliminary in vitro assessment of probiotic properties of Bacillus subtilis GM1, a spore forming bacteria isolated from goat milk. Iran. J. Vet. Res. 2023, 24, 65–73. [Google Scholar] [CrossRef] [PubMed]
  28. Leroy, F.; De Vuyst, L. Lactic acid bacteria as functional starter cultures for the food fermentation industry. Trends Food Sci. Technol. 2004, 15, 67–78. [Google Scholar] [CrossRef]
  29. Coppola, S.; Fusco, V.; Andolfi, R.; Aponte, M.; Blaiotta, G.; Ercolini, D.; Moschetti, G. Evaluating microbial diversity during the manufacture of “fior di latte di Agerola”, a traditional raw milk cheese of Naples area. J. Dairy Res. 2006, 73, 264–272. [Google Scholar] [CrossRef]
  30. Aponte, M.; Fusco, V.; Andolfi, R.; Coppola, S. Lactic acid bacteria occurring during manufacture and ripening of Provolone del Monaco cheese: Detection by different analytical approaches. Int. Dairy J. 2008, 18, 403–413. [Google Scholar] [CrossRef]
  31. Fusco, V.; Fanelli, F.; Chieffi, D. Recent and advanced DNA-based technologies for the authentication of probiotic, Protected Designation of Origin (PDO) and Protected Geographical Indication (PGI) fermented foods and beverages. Foods 2023, 12, 3782. [Google Scholar] [CrossRef]
  32. Fusco, V.; Chieffi, D.; Fanelli, F.; Montemurro, M.; Rizzello, C.G.; Franz, C.M.A.P. The Weissella and Periweissella genera: Up-to-date taxonomy, ecology, safety, biotechnological, and probiotic potential. Front. Microbiol. 2023, 14, 1289937. [Google Scholar] [CrossRef]
  33. Fusco, V.; Chieffi, D.; De Angelis, M. Invited review: Fresh pasta filata cheeses: Composition, role, and evolution of the microbiota in their quality and safety. J. Dairy Sci. 2022, 105, 9347–9366. [Google Scholar] [CrossRef]
  34. Fusco, V.; Chieffi, D.; Benomar, N.; Abriouel, H. Indigenous probiotic microorganisms in fermented foods. In Probiotics for Human Nutrition in Health and Disease; Leite de Souza, E., de Brito Alves, J.L., Fusco, V., Eds.; Academic Press: Cambridge, MA, USA, 2022; ISBN 9780323899086. [Google Scholar] [CrossRef]
  35. Fusco, V.; Chieffi, D.; Fanelli, F.; Logrieco, A.F.; Cho, G.-S.; Kabisch, J.; Boehnlein, C.; Franz, C.M.A.P. Microbial quality and safety of milk and milk products in the 21st century. Comp. Rev. Food Sci. Food Saf. 2020, 19, 2013–2049. [Google Scholar] [CrossRef]
  36. Fusco, V.; Quero, M.G.; Poltronieri, P.; Morea, M.; Baruzzi, F. Autochthonous and probiotic lactic acid bacteria employed for production of “advanced traditional cheeses”. Foods 2019, 8, 412. [Google Scholar] [CrossRef]
  37. Fusco, V.; Quero, G.M.; Cho, G.; Kabisch, J.; Meske, D.; Neve, H.; Bockelmann, W.; Franz, C.M. The genus Weissella: Taxonomy, ecology and biotechnological potential. Front. Microbiol. 2015, 6, 155. [Google Scholar] [CrossRef]
  38. Quero, M.G.; Fusco, V.; Cocconcelli, P.S.; Owczarek, L.; Borcakli, M.; Fontana, C.; Skapska, S.; Jasinska, U.T.; Ozturk, T.; Morea, M. Microbiological, physico-chemical, nutritional and sensory characterization of traditional Matsoni: Selection and use of autochthonous multiple strain cultures to extend its shelf-life. Food Microbiol. 2014, 38, 179–191. [Google Scholar] [CrossRef]
  39. Tofalo, R.; Fusco, V.; Böhnlein, C.; Kabisch, J.; Logrieco, A.F.; Habermann, D.; Cho, G.S.; Benomar, N.; Abriouel, H.; Schmidt-Heydt, M.; et al. The life and times of yeasts in traditional food fermentations. Crit. Rev. Food Sci. Nutr. 2020, 60, 3103–3132. [Google Scholar] [CrossRef]
  40. Santos, M.; San Mauro, A.; Sanchez, A.; Torres, J.M.; Marquina, D. The antimicrobial properties of different strains of Lactobacillus spp. isolated from kefir. Syst. Appl. Microbiol. 2003, 26, 434–437. [Google Scholar] [CrossRef]
  41. van der Aa Kühle, A.; Skovgaard, K.; Jespersen, L. In vitro screening of probiotic properties of Saccharomyces cerevisiae var. boulardii and food-borne Saccharomyces cerevisiae strains. Int. J. Food Microbiol. 2005, 101, 29–39. [Google Scholar] [CrossRef]
  42. Collado, M.C.; Surono, I.S.; Meriluoto, J.; Salminen, S. Potential probiotic characteristics of Lactobacillus and Enterococcus strains isolated from traditional dadih fermented milk against pathogen intestinal colonization. J. Food Prot. 2007, 70, 700–705. [Google Scholar] [CrossRef]
  43. Golowczyc, M.A.; Mobili, P.; Garrote, G.L.; Abraham, A.G.; de Antoni, G.L. Protective action of Lactobacillus kefir carrying S-layer protein against Salmonella enterica serovar Enteritidis. Int. J. Food Microbiol. 2007, 118, 264–273. [Google Scholar] [CrossRef] [PubMed]
  44. Carasi, P.; Racedo, S.M.; Jacquot, C.; Romanin, D.E.; Serradell, M.A.; Urdaci, M.C. Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J. Immunol. Res. 2015, 2015, 361604. [Google Scholar] [CrossRef] [PubMed]
  45. Carasi, P.; Ambrosis, N.M.; De Antoni, G.L.; Bressollier, P.; Urdaci, M.C.; del Serradell, M.A. Adhesion properties of potentially probiotic Lactobacillus kefiri to gastrointestinal mucus. J. Dairy Res. 2014, 81, 16–23. [Google Scholar] [CrossRef] [PubMed]
  46. Carasi, P.; Díaz, M.; Racedo, S.M.; De Antoni, G.; Urdaci, M.C.; del Serradell, M.A. Safety characterization and antimicrobial properties of kefir-isolated Lactobacillus kefiri. BioMed Res. Int. 2014, 2014, 208974. [Google Scholar] [CrossRef]
  47. Carasi, P.; Trejo, F.M.; Pérez, P.F.; De Antoni, G.L.; de Serradell, M.L.A. Surface proteins from Lactobacillus kefir antagonize in vitro cytotoxic effect of Clostridium difficile toxins. Anaerobe 2012, 18, 135–142. [Google Scholar] [CrossRef] [PubMed]
  48. Mathara, J.M.; Schillinger, U.; Guigas, C.; Franz, C.; Kutima, P.M.; Mbugua, S.K.; Shin, H.K.; Holzapfel, W.H. Functional characteristics of Lactobacillus spp. from traditional Maasai fermented milk products in Kenya. Int. J. Food Microbiol. 2008, 126, 57–64. [Google Scholar] [CrossRef]
  49. Wu, R.; Wang, L.; Wang, J.; Li, H.; Menghe, B.; Wu, J.; Guo, M.; Zhang, H. Isolation and preliminary probiotic selection of lactobacilli from koumiss in Inner Mongolia. J. Basic Microbiol. 2009, 49, 318–326. [Google Scholar] [CrossRef]
  50. Takeda, S.; Yamasaki, K.; Takeshita, M.; Kikuchi, Y.; Tsend-Ayush, C.; Dashnyam, B.; Ahhmed, A.M.; Kawahara, S.; Muguruma, M. The investigation of probiotic potential of lactic acid bacteria isolated from traditional Mongolian dairy products. Anim. Sci. J. 2011, 82, 571–579. [Google Scholar] [CrossRef]
  51. Tuo, Y.; Zhang, L.; Han, X.; Du, M.; Zhang, Y.; Yi, H.; Zhang, W.; Jiao, Y. In vitro assessment of immunomodulating activity of the two Lactobacillus strains isolated from traditional fermented milk. World J. Microbiol. Biotechnol. 2011, 27, 505–511. [Google Scholar] [CrossRef]
  52. Rong, J.; Zheng, H.; Liu, M.; Hu, X.; Wang, T.; Zhang, W.; Jin, F.; Wang, L. Probiotic and anti-inflammatory attributes of an isolate Lactobacillus helveticus NS8 from Mongolian fermented koumiss. BMC Microbiol. 2015, 15, 196. [Google Scholar] [CrossRef]
  53. Damodharan, K.; Palaniyandi, S.A.; Yang, S.H.; Suh, J.W. Functional probiotic characterization and in vivo cholesterol-lowering activity of Lactobacillus helveticus isolated from fermented cow milk. J. Microbiol. Biotechnol. 2016, 26, 1675–1686. [Google Scholar] [CrossRef]
  54. Kemkang, T.S.; Kapila, S.; Shanmugam, V.P.; Reddi, S.; Kapila, R. Fermented milk with probiotic Lactobacillus S1K3 (MTCC5957) protects mice from salmonella by enhancing immune and nonimmune protection mechanisms at intestinal mucosal level. J. Nutr. Biochem. 2016, 30, 62–73. [Google Scholar] [CrossRef]
  55. Chen, M.J.; Tang, H.Y.; Chiang, M.L. Effects of heat, cold, acid and bile salt adaptations on the stress tolerance and protein expression of kefir-isolated probiotic Lactobacillus kefiranofaciens M1. Food Microbiol. 2017, 66, 20–27. [Google Scholar] [CrossRef] [PubMed]
  56. Chen, Y.P.; Chen, M.J. Effects of Lactobacillus kefiranofaciens M1 isolated from kefir grains on germ-free mice. PLoS ONE 2013, 8, e78789. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, H.C.; Wang, S.Y.; Chen, M.J. Microbiological study of lactic acid bacteria in kefir grains by culture-dependent and culture-independent methods. Food Microbiol. 2008, 25, 492–501. [Google Scholar] [CrossRef] [PubMed]
  58. Chen, Y.P.; Hsiao, P.J.; Hong, W.S.; Dai, T.Y.; Chen, M.J. Lactobacillus kefiranofaciens M1 isolated from milk kefir grains ameliorates experimental colitis in vitro and in vivo. J. Dairy Sci. 2012, 95, 63–74. [Google Scholar] [CrossRef]
  59. Chen, Y.P.; Lee, T.Y.; Hong, W.S.; Hsieh, H.H.; Chen, M.J. Effects of Lactobacillus kefiranofaciens M1 isolated from kefir grains on enterohemorrhagic Escherichia coli infection using mouse and intestinal cell models. J. Dairy Sci. 2013, 96, 7467–7477. [Google Scholar] [CrossRef]
  60. Hong, W.S.; Chen, Y.P.; Chen, M.J. The antiallergic effect of kefir lactobacilli. J. Food Sci. 2010, 75, 244–253. [Google Scholar] [CrossRef]
  61. Jeong, D.; Kim, D.H.; Kang, I.B.; Kim, H.; Song, K.Y.; Kim, H.S.; Seo, K.H. Modulation of gut microbiota and increase in fecal water content in mice induced by administration of Lactobacillus kefiranofaciens DN1. Food Funct. 2017, 8, 680–686. [Google Scholar] [CrossRef]
  62. Ding, W.; Shi, C.; Chen, M.; Zhou, J.; Long, R.; Guo, X. Screening for lactic acid bacteria in traditional fermented Tibetan yak milk and evaluating their probiotic and cholesterol-lowering potentials in rats fed a high-cholesterol diet. J. Funct. Foods 2017, 32, 324–332. [Google Scholar] [CrossRef]
  63. Ye, H.; Li, Q.; Zhang, Z.; Sun, M.; Zhao, C.; Zhang, T. Effect of a novel potential probiotic Lactobacillus paracasei Jlus66 isolated from fermented milk on nonalcoholic fatty liver in rats. Food Funct. 2017, 8, 4539–4546. [Google Scholar] [CrossRef]
  64. Elbanna, K.; El Hadad, S.; Assaeedi, A.; Aldahlawi, A.; Khider, M.; Alhebshi, A. In vitro and in vivo evidences for innate immune stimulators lactic acid bacterial starters isolated from fermented camel dairy products. Sci. Rep. 2018, 8, 12553. [Google Scholar] [CrossRef]
  65. Jeong, D.; Kim, D.H.; Song, K.Y.; Seo, K.H. Antimicrobial and anti-biofilm activities of Lactobacillus kefiranofaciens DD2 against oral pathogens. J. Oral Microbiol. 2018, 10, 1472985. [Google Scholar] [CrossRef]
  66. Moreno-Montoro, M.; Navarro-Alarcón, M.; Bergillos-Meca, T.; Giménez-Martínez, R.; Sánchez-Hernández, S.; Olalla-Herrera, M. Physicochemical, nutritional, and organoleptic characterization of a skimmed goat milk fermented with the probiotic strain Lactobacillus plantarum C4. Nutrients 2018, 10, 633. [Google Scholar] [CrossRef] [PubMed]
  67. Bergillos-Meca, T.; Costabile, A.; Walton, G.; Moreno-Montoro, M.; Ruiz-Bravo, A.; Ruiz-López, M.D. In vitro evaluation of the fermentation properties and potential probiotic activity of Lactobacillus plantarum C4 in batch culture systems. LWT-Food Sci. Technol. 2014, 60, 420–426. [Google Scholar] [CrossRef]
  68. Bujalance, C.; Moreno, E.; Jimenez-Valera, M.; Ruiz-Bravo, A. A probiotic strain of Lactobacillus plantarum stimulates lymphocyte responses in immunologically intact and immunocompromised mice. Int. J. Food Microbiol. 2007, 113, 28–34. [Google Scholar] [CrossRef] [PubMed]
  69. Seo, M.K.; Park, E.J.; Ko, S.Y.; Choi, E.W.; Kim, S. Therapeutic effects of kefir grain Lactobacillus-derived extracellular vesicles in mice with 2,4,6-trinitrobenzene sulfonic acid-induced inflammatory bowel disease. J. Dairy Sci. 2018, 101, 8662–8671. [Google Scholar] [CrossRef] [PubMed]
  70. Bhat, B.; Habib, N.; Bhagat, B.; Bajaj, B.K. Cholesterol lowering and antioxidant potential of probiotic bacteria isolated from locally fermented milk product kalarei. Indian J. Biochem. Biophys. 2019, 56, 363–372. [Google Scholar]
  71. Nami, Y.; Bakhshayesh, R.V.; Manafi, M.; Hejazi, M.A. Hypocholesterolaemic activity of a novel autochthonous potential probiotic Lactobacillus plantarum S5 isolated from yogurt. LWT-Food Sci. Technol. 2019, 111, 876–882. [Google Scholar] [CrossRef]
  72. Rajoka, M.S.R.; Mehwish, H.M.; Fang, H.; Padhiar, A.A.; Zeng, X.; Khurshid, M.; He, Z.; Zhao, L. Characterization and anti-tumor activity of exopolysaccharide produced by Lactobacillus kefiri isolated from Chinese kefir grains. J. Funct. Foods 2019, 63, 103588. [Google Scholar] [CrossRef]
  73. Harnentis, H.; Marlida, Y.; Nur, Y.S.; Qizna, W.; Santi, M.A.; Septiani, N.; Adzitey, F.; Huda, N. Novel probiotic lactic acid bacteria isolated from indigenous fermented foods from West Sumatera, Indonesia. Vet. World 2020, 13, 1922–1927. [Google Scholar] [CrossRef]
  74. Motey, G.A.; Johansen, P.G.; Owusu-Kwarteng, J.; Ofori, L.A.; Obiri-Danso, K.; Siegumfeldt, H.; Larsen, N.; Jespersen, L. Probiotic potential of Saccharomyces cerevisiae and Kluyveromyces marxianus isolated from West African spontaneously fermented cereal and milk products. Yeast 2020, 37, 403–412. [Google Scholar] [CrossRef]
  75. Nath, S.; Sikidar, J.; Roy, M.; Deb, B. In vitro screening of probiotic properties of Lactobacillus plantarum isolated from fermented milk product. Food Qual. Saf. 2020, 4, 213–223. [Google Scholar] [CrossRef]
  76. Pahumunto, N.; Piwat, S.; Chanvitan, S.; Ongwande, W.; Uraipan, S.; Teanpaisan, R. Fermented milk containing a potential probiotic Lactobacillus rhamnosus SD11 with maltitol reduces Streptococcus mutans: A double-blind, randomized, controlled study. J. Dent. Sci. 2020, 15, 403–410. [Google Scholar] [CrossRef]
  77. Prasetyo, R.V.; Surono, I.; Soemyarso, N.A.; Djojodimedjo, T.; Rauf, S.; Noer, M.S.; Sudarmo, S.M. Lactobacillus plantarum IS-10506 promotes renal tubular regeneration in pyelonephritic rats. Benef. Microbes 2020, 11, 59–66. [Google Scholar] [CrossRef]
  78. Prakoeswa, C.R.S.; Herwanto, N.; Prameswari, R.; Astari, L.; Sawitri, S.; Hidayati, A.N.; Indramaya, D.M.; Kusumowidagdo, E.R.; Surono, I.S. Lactobacillus plantarum IS-10506 supplementation reduced SCORAD in children with atopic dermatitis. Benef. Microbes 2017, 8, 833–840. [Google Scholar] [CrossRef]
  79. Surono, I.S.; Martono, P.D.; Kameo, S.; Suradji, E.W.; Koyama, H. Effect of probiotic L. plantarum IS-10506 and zinc supplementation on humoral immune response and zinc status of Indonesian pre-school children. J. Trace Elem. Med. Biol. 2014, 28, 465–469. [Google Scholar] [CrossRef]
  80. Kusumo, P.D.; Bela, B.; Wibowo, H.; Munasir, Z.; Surono, I.S. Lactobacillus plantarum IS-10506 supplementation increases faecal sIgA and immune response in children younger than two years. Benef. Microbes 2019, 10, 245–252. [Google Scholar] [CrossRef]
  81. Athiyyah, A.F.; Darma, A.; Ranuh, R.; Riawan, W.; Endaryanto, A.; Rantam, F.A.; Surono, I.S.; Sudarmo, S.M. Lactobacillus plantarum IS-10506 activates intestinal stem cells in a rodent model. Benef. Microbes 2018, 9, 755–760. [Google Scholar] [CrossRef] [PubMed]
  82. Athiyyah, A.F.; Brahmantya, H.; Dwiastuti, S.; Darma, A.; Puspitasari, D.; Husada, D.; Ranuh, R.; Endaryanto, A.; Surono, I.; Sudarmo, S.M. Effect of Lactobacillus plantarum IS-10506 on blood lipopolysaccharide level and immune response in HIV-infected children. Iran. J. Microbiol. 2019, 11, 137–144. [Google Scholar]
  83. Hurtado-Romero, A.; Del Toro-Barbosa Barbosa, M.; Gradilla-Hernández, M.S.; Garcia-Amezquita, L.E.; García-Cayuela, T. Probiotic properties, prebiotic fermentability, and GABA-producing capacity of microorganisms isolated from mexican milk kefir grains: A clustering evaluation for functional dairy food applications. Foods 2021, 10, 2275. [Google Scholar] [CrossRef]
  84. Galli, V.; Venturi, M.; Mari, E.; Guerrini, S.; Granchi, L. Selection of yeast and lactic acid bacteria strains, isolated from spontaneous raw milk fermentation, for the production of a potential probiotic fermented milk. Fermentation 2022, 8, 407. [Google Scholar] [CrossRef]
  85. Ismael, M.; Gu, Y.; Cui, Y.; Wang, T.; Yue, F.; Qin, Y.; Lü, X. Probiotic of Lactiplantibacillus plantarum NWAFU-BIO-BS29 isolated from Chinese traditional fermented milk and its potential therapeutic applications based on gut microbiota regulation. Foods 2022, 11, 3766. [Google Scholar] [CrossRef]
  86. Ismael, M.; Gu, Y.; Cui, Y.; Wang, T.; Yue, F.; Yantin, Q.; Lü, X. Lactic acid bacteria isolated from Chinese traditional fermented milk as novel probiotic strains and their potential therapeutic applications. 3 Biotech 2022, 12, 337. [Google Scholar] [CrossRef]
  87. Katiku, M.M.; Matofari, J.W.; Nduko, J.M. Preliminary evaluation of probiotic properties and safety profile of Lactiplantibacillus plantarum isolated from spontaneously fermented milk, Amabere amaruranu. Heliyon 2022, 8, e10342. [Google Scholar] [CrossRef]
  88. Li, W.; Gao, L.; Huang, W.; Ma, Y.; Muhammad, I.; Hanif, A.; Ding, Z.; Guo, X. Antioxidant properties of lactic acid bacteria isolated from traditional fermented yak milk and their probiotic effects on the oxidative senescence of Caenorhabditis elegans. Food Funct. 2022, 13, 3690–3703. [Google Scholar] [CrossRef] [PubMed]
  89. Ngwa, F.; Fossi, B.; Taiwe, G.; Tatsinkou, L.; Bobga, P.; Ndalloka, M.; Lea, D.; Wanyu, B.; Wanji, S. Isolation, characterisation and evaluation of antiulcerogenic potentials of probiotic lactic acid bacteria isolated from fermented milk and palm wine against ethanol-induced gastric ulcer in mice. J. Biosci. Med. 2022, 10, 311–331. [Google Scholar] [CrossRef]
  90. Rai, R.; Tamang, J.P. In vitro and genetic screening of probiotic properties of lactic acid bacteria isolated from naturally fermented cow-milk and yak-milk products of Sikkim, India. World J. Microbiol. Biotechnol. 2022, 38, 25. [Google Scholar] [CrossRef]
  91. Dehghani Champiri, I.; Bamzadeh, Z.; Rahimi, E.; Rouhi, L. Lacticaseibacillus paracasei LB12, a potential probiotic isolated from traditional Iranian fermented milk (doogh). Curr. Microbiol. 2023, 80, 333. [Google Scholar] [CrossRef]
  92. Khan, M.N.; Bashir, S.; Imran, M. Probiotic characterization of Bacillus species strains isolated from an artisanal fermented milk product Dahi. Folia Microbiol. 2023, 68, 757–769. [Google Scholar] [CrossRef] [PubMed]
  93. Shangpliang, H.N.J.; Tamang, J.P. Genome analysis of potential probiotic Levilactobacillus brevis AcCh91 isolated from Indian home-made fermented milk product (Chhurpi). Probiotics Antimicrob. Proteins 2023. [Google Scholar] [CrossRef] [PubMed]
  94. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
  95. Todorov, S.D.; Dioso, C.M.; Liong, M.T.; Nero, L.A.; Khosravi-Darani, K.; Ivanova, I.V. Beneficial features of Pediococcus: From starter cultures and inhibitory activities to probiotic benefits. World J. Microbiol. Biotechnol. 2022, 39, 4. [Google Scholar] [CrossRef]
  96. Psomas, E.I.; Fletouris, D.J.; Litopoulou-Tzanetaki, E.; Tzanetakis, N. Assimilation of cholesterol by yeast strains isolated from infant feces and feta cheese. J. Dairy Sci. 2003, 86, 3416–3422. [Google Scholar] [CrossRef]
  97. Bao, Y.; Zhang, Y.; Zhang, Y.; Liu, Y.; Wang, S.; Dong, X.; Wang, Y.; Zhang, H. Screening potential probiotic properties of Lactobacillus fermentum isolated from traditional dairy products. Food Control 2010, 21, 695–701. [Google Scholar] [CrossRef]
  98. Meira, S.M.; Helfer, V.E.; Velho, R.V.; Lopes, F.C.; Brandelli, A. Probiotic potential of Lactobacillus spp. isolated from Brazilian regional ovine cheese. J. Dairy Res. 2012, 79, 119–127. [Google Scholar] [CrossRef]
  99. Ahmadova, A.; Todorov, S.D.; Choiset, Y.; Rabesona, H.; Mirhadi Zadi, T.; Kuliyev, A.; de Melo Franco, B.D.G.; Chobert, G.-M.; Haertlé, T. Evaluation of antimicrobial activity, probiotic properties and safety of wild strain Enterococcus faecium AQ71 isolated from Azerbaijani Motal cheese. Food Control 2013, 30, 631–641. [Google Scholar] [CrossRef]
  100. Belicovà, A.; Mikulasova, M.; Dusinsky, R. Probiotic potential and safety properties of Lactobacillus plantarum from Slovak Bryndza cheese. BioMed Res. Int. 2013, 2013, 760298. [Google Scholar] [CrossRef] [PubMed]
  101. Binetti, A.; Carrasco, M.; Reinheimer, J.; Suàrez, V. Yeasts from autochthonal cheese starters: Technological and functional properties. J. Appl. Microbiol. 2013, 115, 434–444. [Google Scholar] [CrossRef] [PubMed]
  102. Lavilla-Lerma, L.; Pérez-Pulido, R.; Martínez-Bueno, M.; Maqueda, M.; Valdivia, E. Characterization of functional, safety, and gut survival related characteristics of Lactobacillus strains isolated from farmhouse goat’s milk cheeses. Int. J. Food Microbiol. 2013, 163, 136–145. [Google Scholar] [CrossRef] [PubMed]
  103. Melgar-Lalanne, G.; Rivera-Espinoza, Y.; Reyes Méndez, A.I.; Hernández-Sánchez, H. In vitro evaluation of the probiotic potential of halotolerant lactobacilli isolated from a ripened tropical Mexican cheese. Probiotics Antimicrob. Proteins 2013, 5, 239–251. [Google Scholar] [CrossRef] [PubMed]
  104. Tulini, F.L.; Winkelströter, L.K.; De Martinis, E.C. Identification and evaluation of the probiotic potential of Lactobacillus paraplantarum FT259, a bacteriocinogenic strain isolated from Brazilian semi-hard artisanal cheese. Anaerobe 2013, 22, 57–63. [Google Scholar] [CrossRef] [PubMed]
  105. Jeronymo-Ceneviva, A.B.; de Paula, A.T.; Silva, L.F.; Todorov, S.D.; Franco, B.D.; Penna, A.L. Probiotic properties of lactic acid bacteria isolated from water-buffalo mozzarella cheese. Probiotics Antimicrob. Proteins 2014, 6, 141–156. [Google Scholar] [CrossRef]
  106. Solieri, L.; Bianchi, A.; Mottolese, G.; Lemmetti, F.; Giudici, P. Tailoring the probiotic potential of non-starter Lactobacillus strains from ripened Parmigiano Reggiano cheese by in vitro screening and principal component analysis. Food Microbiol. 2014, 38, 240–249. [Google Scholar] [CrossRef]
  107. Tulumoğlu, Ş.; Kaya, H.İ.; Şimşek, Ö. Probiotic characteristics of Lactobacillus fermentum strains isolated from tulum cheese. Anaerobe 2014, 30, 120–125. [Google Scholar] [CrossRef] [PubMed]
  108. Uroić, K.; Milica, N.; Blazenka, K.; Pavunc, A.; Beganovic, J.; Lukic, J.; Branko, J.; Brankica, F.; Miljkovic, M.; Golić, N.; et al. Probiotic properties of lactic acid bacteria isolated from Croatian fresh soft cheese and Serbian white pickled cheese. Food Technol. Biotechnol. 2014, 52, 232–241. [Google Scholar]
  109. Caggia, C.; De Angelis, M.; Pitino, I.; Pino, A.; Randazzo, C.I. Probiotic features of Lactobacillus strains isolated from Ragusano and Pecorino Siciliano cheeses. Food Microbiol. 2015, 50, 109–117. [Google Scholar] [CrossRef]
  110. De Paula, A.; Jeronymo-Ceneviva, A.B.; Silva, L.F.; Todorov, S.D.; Franco, B.D.G.M.; Penna, A.L.B. Leuconostoc mesenteroides SJRP55: A potential probiotic strain isolated from Brazilian water buffalo mozzarella cheese. Ann. Microbiol. 2015, 65, 899–910. [Google Scholar] [CrossRef]
  111. Živković, M.; Čadež, N.; Uroić, K.; Miljković, M.; Tolinački, M.; Doušova, P.; Kos, B.; Šušković, J.; Raspor, P.; Topisirović, L.; et al. Evaluation of probiotic potential of yeasts isolated from traditional cheeses manufactured in Serbia and Croatia. J. Intercult. Ethnopharmacol. 2015, 4, 12–18. [Google Scholar] [CrossRef] [PubMed]
  112. Amaral, D.M.F.; Silva, L.F.; Casarotti, S.N.; Nascimento, L.C.S.; Penna, A.L.B. Enterococcus faecium and Enterococcus durans isolated from cheese: Survival in the presence of medications under simulated gastrointestinal conditions and adhesion properties. J. Dairy Sci. 2016, 100, 933–949. [Google Scholar] [CrossRef]
  113. Angmo, K.; Kumari, A.; Savitri, B.T. Probiotic characterization of lactic acid bacteria isolated from fermented foods and beverage of Ladakh. LWT-Food Sci. Technol. 2015, 66, 428–435. [Google Scholar] [CrossRef]
  114. Bian, X.; Evivie, S.E.; Muhammad, Z.; Luo, G.W.; Liang, H.Z.; Wang, N.N.; Huo, G.C. In vitro assessment of the antimicrobial potentials of Lactobacillus helveticus strains isolated from traditional cheese in Sinkiang China against food-borne pathogens. Food Funct. 2016, 7, 789–797. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, B.; Wang, Y.; Tan, Z.; Li, Z.; Jiao, Z.; Huang, Q. Screening of probiotic activities of lactobacilli strains isolated from traditional Tibetan qula, a raw yak milk cheese. Asian-Australas. J. Anim. Sci. 2016, 29, 1490–1499. [Google Scholar] [CrossRef] [PubMed]
  116. Acurcio, L.B.; Bastos, R.W.; de Cicco Sandes, S.H.; de Carvalho Guimarães, A.C.; Gregório Alves, C.; dos Reis, D.C.; Wuyts, S.; Cantini Nunes, A.; Dantas Cassali, G.; Lebeer, S.; et al. Protective effects of milk fermented by Lactobacillus plantarum B7 from Brazilian artisanal cheese on a Salmonella enterica serovar Typhimurium infection in BALB/c mice. J. Funct. Foods 2017, 33, 436–445. [Google Scholar] [CrossRef]
  117. Aguilar-Toalá, J.E.; Santiago-López, L.; Peres, C.; Garcia, H.; Vallejo-Cordoba, B.; González-Córdova, A.F.; Hernandez-Mendoza, A. Assessment of multifunctional activity of bioactive peptides derived from fermented milk by specific Lactobacillus plantarum strains. J. Dairy Sci. 2016, 100, 65–75. [Google Scholar] [CrossRef]
  118. Casarotti, S.N.; Carneiro, B.M.; Todorov, S.D.; Nero, L.A.; Rahal, P.; Barretto Penna, A.L. In vitro assessment of safety and probiotic potential characteristics of Lactobacillus strains isolated from water buffalo mozzarella cheese. Ann. Microbiol. 2017, 67, 289–301. [Google Scholar] [CrossRef]
  119. de Sant’Anna, F.M.; Acurcio, L.B.; Alvim, L.B.; de Castro, R.D.; de Oliveira, L.G.; da Silva, A.M.; Nunes, Á.C.; Nicoli, J.R.; Souza, M.R. Assessment of the probiotic potential of lactic acid bacteria isolated from Minas artisanal cheese produced in the Campo das Vertentes region, Brazil. Int. J. Dairy Technol. 2017, 70, 592–601. [Google Scholar] [CrossRef]
  120. Fadda, M.E.; Mossa, V.; Deplano, M.; Pisano, M.B.; Cosentino, S. In vitro screening of Kluyveromyces strains isolated from Fiore Sardo cheese for potential use as probiotics. LWT-Food Sci. Technol. 2017, 75, 100–106. [Google Scholar] [CrossRef]
  121. Plessas, S.; Nouska, C.; Karapetsas, A.; Kazakos, S.; Alexopoulos, A.; Mantzourani, I.; Chondrou, P.; Fournomiti, M.; Galanis, A.; Bezirtzoglou, E. Isolation, characterization and evaluation of the probiotic potential of a novel Lactobacillus strain isolated from Feta-type cheese. Food Chem. 2017, 226, 102–108. [Google Scholar] [CrossRef]
  122. Nami, Y.; Haghshenas, B.; Bakhshayesh, R.V.; Jalaly, H.M.; Lotfi, H.; Eslami, S.; Hejazi, M.A. Novel autochthonous lactobacilli with probiotic aptitudes as a main starter culture for probiotic fermented milk. LWT-Food Sci. Technol. 2018, 98, 85–93. [Google Scholar] [CrossRef]
  123. Ribeiro, S.C.; Stanton, C.; Yang, B.; Ross, R.P.; Silva, C.C.G. Conjugated linoleic acid production and probiotic assessment of Lactobacillus plantarum isolated from Pico cheese. LWT-Food Sci. Technol. 2018, 90, 403–411. [Google Scholar] [CrossRef]
  124. Santiago-López, L.; Hernández-Mendoza, A.; Mata-Haro, V.; Vallejo-Cordoba, B.; González-Córdova, A.F. Immune response induced by fermented milk with potential probiotic strains isolated from artisanal cocido cheese. Food Agric. Immunol. 2018, 29, 911–929. [Google Scholar] [CrossRef]
  125. Zoumpopoulou, G.; Tzouvanou, A.; Mavrogonatou, E.; Alexandraki, V.; Georgalaki, M.; Anastasiou, R.; Papadelli, M.; Manolopoulou, E.; Kazou, M.; Kletsas, D.; et al. Probiotic features of lactic acid bacteria isolated from a diverse pool of traditional Greek dairy products regarding specific strain-host interactions. Probiotics Antimicrob. Proteins 2018, 10, 313–322. [Google Scholar] [CrossRef] [PubMed]
  126. Baccouri, O.; Boukerb, A.M.; Farhat, L.B.; Zébré, A.; Zimmermann, K.; Domann, E.; Cambronel, M.; Barreau, M.; Maillot, O.; Rincé, I.; et al. Probiotic potential and safety evaluation of Enterococcus faecalis OB14 and OB15, isolated from traditional Tunisian testouri cheese and rigouta, using physiological and genomic analysis. Front. Microbiol. 2019, 10, 881. [Google Scholar] [CrossRef] [PubMed]
  127. de Souza, B.M.S.; Borgonovi, T.F.; Casarotti, S.N.; Todorov, S.D.; Penna, A.L.B. Lactobacillus casei and Lactobacillus fermentum strains isolated from mozzarella cheese: Probiotic potential, safety, acidifying kinetic parameters and viability under gastrointestinal tract conditions. Probiotics Antimicrob. Proteins 2019, 11, 382–396. [Google Scholar] [CrossRef] [PubMed]
  128. Mancini, A.; Carafa, I.; Franciosi, E.; Nardin, T.; Bottari, B.; Larcher, R.; Tuohy, K.M. In vitro probiotic characterization of high GABA producing strain Lactobacilluas brevis DSM 32386 isolated from traditional “wild” Alpine cheese. Ann. Microbiol. 2019, 69, 1435–1443. [Google Scholar] [CrossRef]
  129. Mangia, N.P.; Saliba, L.; Deiana, P. Functional and safety characterization of autochthonous Lactobacillus paracasei FS103 isolated from sheep cheese and its survival in sheep and cow fermented milks during cold storage. Ann. Microbiol. 2019, 69, 161–170. [Google Scholar] [CrossRef]
  130. Pino, A.; Russo, N.; Van Hoorde, K.; De Angelis, M.; Sferrazzo, G.; Randazzo, C.L.; Caggia, C. Piacentinu Ennese PDO cheese as reservoir of promising probiotic bacteria. Microorganisms 2019, 7, 254. [Google Scholar] [CrossRef]
  131. Ruiz-Moyano, S.; Gonçalves dos Santos, M.T.P.; Galván, A.I.; Merchán, A.V.; González, E.; de Guía Córdoba, M.; José Benito, M. Screening of autochthonous lactic acid bacteria strains from artisanal soft cheese: Probiotic characteristics and prebiotic metabolism. LWT-Food Sci. Technol. 2019, 114, 108388. [Google Scholar] [CrossRef]
  132. Schittler, L.; Perin, L.M.; de Lima Marques, J.; Lando, V.; Todorov, S.D.; Nero, L.A.; da Silva, W.P. Isolation of Enterococcus faecium, characterization of its antimicrobial metabolites and viability in probiotic Minas Frescal cheese. J. Food Sci. Technol. 2019, 56, 5128–5137. [Google Scholar] [CrossRef]
  133. Silva, W.M.; Sousa, C.S.; Oliveira, L.C.; Soares, S.C.; Souza, G.F.M.H.; Tavares, G.C.; Resende, C.P.; Folador, E.L.; Pereira, F.L.; Figueiredo, H.; et al. Comparative proteomic analysis of four biotechnological strains Lactococcus lactis through label-free quantitative proteomics. Microb. Biotechnol. 2019, 12, 265–274. [Google Scholar] [CrossRef]
  134. Tezel, B.U. Preliminary in vitro evaluation of the probiotic potential of the bacteriocinogenic strain Enterococcus lactis PMD74 isolated from Ezine cheese. J. Food Qual. 2019, 2019, 4693513. [Google Scholar] [CrossRef]
  135. Valente, G.L.C.; Acurcio, L.B.; Freitas, L.P.V.; Nicoli, J.R.; Silva, A.M.; Souza, M.R.; Penna, C.F.A.M. Short communication: In vitro and in vivo probiotic potential of Lactobacillus plantarum B7 and Lactobacillus rhamnosus D1 isolated from Minas artisanal cheese. J. Dairy Sci. 2019, 102, 5957–5961. [Google Scholar] [CrossRef]
  136. Bhagat, D.; Raina, N.; Kumar, A.; Katoch, M.; Khajuria, Y.; Slathia, P.S.; Sharma, P. Probiotic properties of a phytase producing Pediococcus acidilactici strain SMVDUDB2 isolated from traditional fermented cheese product, Kalarei. Sci. Rep. 2020, 10, 1926. [Google Scholar] [CrossRef]
  137. Domingos-Lopes, M.F.P.; Stanton, C.; Riss, R.P.; Silva, C.C.G. Histamine and cholesterol lowering abilities of lactic acid bacteria isolated from artisanal Pico cheese. J. Appl. Microbiol. 2020, 129, 1428–1440. [Google Scholar] [CrossRef]
  138. Hussein, W.E.; Abdelhamid, A.G.; Rocha-Mendoza, D.; García-Cano, I.; Yousef, A.E. Assessment of safety and probiotic traits of Enterococcus durans OSY-EGY, isolated from Egyptian artisanal cheese, using comparative genomics and phenotypic analyses. Front. Microbiol. 2020, 11, 608314. [Google Scholar] [CrossRef]
  139. Mahmoudi, M.; Khomeiri, M.; Saeidi, M.; Davoodi, H. Lactobacillus species from Iranian jug cheese: Identification and selection of probiotic based on safety and functional properties. Appl. Food Biotechnol. 2020, 8, 47–56. [Google Scholar] [CrossRef]
  140. Olajugbagbe, T.E.; Elugbadebo, O.E.; Omafuvbe, B.O. Probiotic potentials of Pediococuss acidilactici isolated from wara; a Nigerian unripened soft cheese. Heliyon 2020, 6, e04889. [Google Scholar] [CrossRef]
  141. Ołdak, A.; Zielińska, D.; Łepecka, A.; Długosz, E.; Kołożyn-Krajewska, D. Lactobacillus plantarum strains isolated from Polish regional cheeses exhibit anti-staphylococcal activity and selected probiotic properties. Probiotics Antimicrob. Proteins 2020, 12, 1025–1038. [Google Scholar] [CrossRef]
  142. Ait Chait, Y.; Gunenc, A.; Hosseinian, F.; Bendali, F. Antipathogenic and probiotic potential of Lactobacillus brevis strains newly isolated from Algerian artisanal cheeses. Folia Microbiol. 2021, 66, 429–440. [Google Scholar] [CrossRef] [PubMed]
  143. Barzegar, H.; Alizadeh Behbahani, B.; Falah, F. Safety, probiotic properties, antimicrobial activity, and technological performance of Lactobacillus strains isolated from Iranian raw milk cheeses. Food Sci. Nutr. 2021, 9, 4094–4107. [Google Scholar] [CrossRef] [PubMed]
  144. Hajikhani, R.; Onal Darilmaz, D.; Yuksekdag, Z.N.; Beyatli, Y. Assessment of some metabolic activities and potential probiotic properties of eight Enterococcus bacteria isolated from white cheese microbiota. Antonie Van Leeuwenhoek 2021, 114, 1259–1274. [Google Scholar] [CrossRef]
  145. Kouhi, F.; Hamid, M.; Yousef, N.; Jalil, K.; Afshin, J. Potential probiotic and safety characterisation of Enterococcus bacteria isolated from indigenous fermented Motal cheese. Int. Dairy J. 2021, 126, 105247. [Google Scholar] [CrossRef]
  146. Margalho, L.P.; Jorge, G.P.; Noleto, D.A.P.; Silva, C.E.; Abreu, J.S.; Piran, M.V.F.; Brocchi, M.; Sant’Ana, A.S. Biopreservation and probiotic potential of a large set of lactic acid bacteria isolated from Brazilian artisanal cheeses: From screening to in product approach. Microbiol. Res. 2021, 242, 126622. [Google Scholar] [CrossRef]
  147. Mohammed, S.; Hilmi Çon, A. Isolation and characterization of potential probiotic lactic acid bacteria from traditional cheese. LWT-Food Sci. Technol. 2021, 152, 112319. [Google Scholar] [CrossRef]
  148. Özkan, E.R.; Demirici, T.; Akin, N. In vitro assessment of probiotic and virulence potential of Enterococcus faecium strains derived from artisanal goatskin casing Tulum cheeses produced in central Taurus Mountains of Turkey. LWT-Food Sci. Technol. 2021, 141, 110908. [Google Scholar] [CrossRef]
  149. Zago, M.; Massimiliano, L.; Bonvini, V.; Penna, G.; Giraffa, G.; Rescigno, M. Functional characterization and immunomodulatory properties of Lactobacillus helveticus strains isolated from Italian hard cheeses. PLoS ONE 2021, 16, e0245903. [Google Scholar] [CrossRef]
  150. Afshari, A.; Hashemi, M.; Tavassoli, M.; Eraghi, V.; Noori, S.M.A. Probiotic bacteria from 10 different traditional Iranian cheeses: Isolation, characterization, and investigation of probiotic potential. Food Sci. Nutr. 2022, 10, 2009–2020. [Google Scholar] [CrossRef]
  151. Falfán-Cortés, R.N.; Mora-Peñaflor, N.; Gómez-Aldapa, C.A.; Rangel-Vargas, E.; Acevedo-Sandoval, O.A.; Franco-Fernández, M.J.; Castro-Rosas, J. Characterization and evaluation of the probiotic potential in vitro and in situ of Lacticaseibacillus paracasei isolated from Tenate cheese. J. Food Prot. 2022, 85, 112–121. [Google Scholar] [CrossRef] [PubMed]
  152. Metrouh, R.; Fares, R.; Mechai, A.; Debabza, M.; Menasria, T. Technological properties and probiotic potential of Lactiplantibacillus plantarum SJ14 isolated from Algerian traditional cheese “Jben”. J. Food Process. Preserv. 2022, 46, e16482. [Google Scholar] [CrossRef]
  153. Mohamad, G.; Abiad, C.A.; Zeina, K.; Christelle, F.I.; Reem, H.; Hussein, F.H. Preliminary physiological, phenotypic, and probiotic characterization of Lactobacillus strains isolated from Anbaris-traditional Lebanese fermented dairy product. Int. J. Food Prop. 2022, 25, 1266–1278. [Google Scholar] [CrossRef]
  154. Ngamsomchat, A.; Kaewkod, T.; Konkit, M.; Tragoolpua, Y.; Bovonsombut, S.; Chitov, T. Characterisation of Lactobacillus plantarum of dairy-product origin for probiotic Chèvre cheese production. Foods 2022, 11, 934. [Google Scholar] [CrossRef]
  155. Adugna, M.; Andualem, B. Isolation, characterization and safety assessment of probiotic lactic acid bacteria from Metata Ayib (traditional spiced cottage cheese). Food Humanit. 2023, 1, 85–91. [Google Scholar] [CrossRef]
  156. de Miranda, N.M.Z.; de Souza, A.C.; de Souza Costa Sobrinho, P.; Dias, D.R.; Schwan, R.F.; Ramos, C.L. Novel yeasts with potential probiotic characteristics isolated from the endogenous ferment of artisanal Minas cheese. Braz. J. Microbiol. 2023, 54, 1021–1033. [Google Scholar] [CrossRef] [PubMed]
  157. Gizachew, S.; Van Beeck, W.; Spacova, I.; Dekeukeleire, M.; Alemu, A.; Woldemedhin, W.M.; Mariam, S.H.; Lebeer, S.; Engidawork, E. Antibacterial and immunostimulatory activity of potential probiotic lactic acid bacteria isolated from Ethiopian fermented dairy products. Fermentation 2023, 9, 258. [Google Scholar] [CrossRef]
  158. Güley, Z.; Fallico, V.; Cabrera-Rubio, R.; O’Sullivan, D.; Marotta, M.; Pennone, V.; Smith, S.; Beresford, T. Diversity of the microbiota of traditional Izmir Tulum and Izmir brined Tulum cheeses and selection of potential probiotics. Foods 2023, 12, 3482. [Google Scholar] [CrossRef] [PubMed]
  159. Hadef, S.; Idoui, T.; Sifour, M.; Genay, M.; Dary-Mourot, A. Screening of wild lactic acid bacteria from algerian traditional cheeses and goat butter to develop a new probiotic starter culture. Probiotics Antimicrob. Proteins 2023, 2, 387–399. [Google Scholar] [CrossRef] [PubMed]
  160. Khaleghi, M.; Khorrami, S.; Jafari-Nasab, T. Pediococcus acidilactici isolated from traditional cheese as a potential probiotic with cytotoxic activity against doxorubicin-resistant MCF-7 cells. 3 Biotech 2023, 13, 170. [Google Scholar] [CrossRef] [PubMed]
  161. Nasr, N.M.; Abd-Alhalim, L.R. Characterization and identification of Lactobacillus rhamnosus and Enterococcus durans as probiotic potential isolated from selected dairy products in Egypt. J. Umm Al-Qura Univ. Appl. Sci. 2023. [Google Scholar] [CrossRef]
  162. Roldán-Pérez, S.; Gómez Rodríguez, S.L.; Sepúlveda-Valencia, J.U.; Ruiz Villadiego, O.S.; Márquez Fernández, M.E.; Montoya Campuzano, O.I.; Durango-Zuleta, M.M. Assessment of probiotic properties of lactic acid bacteria isolated from an artisanal Colombian cheese. Heliyon 2023, 9, e21558. [Google Scholar] [CrossRef] [PubMed]
  163. Zareie, Z.; Moayedi, A.; Garavand Garavand, F.; Tabar-Heydar, K.; Khomeiri, M.; Maghsoudlou, Y. Probiotic properties, safety assessment, and aroma-generating attributes of some lactic acid bacteria isolated from Iranian traditional cheese. Fermentation 2023, 9, 338. [Google Scholar] [CrossRef]
  164. Zommara, M.; El-Ghaish, S.; Haertle, T.; Chobert, J.M.; Ghanimah, M. Probiotic and technological characterization of selected Lactobacillus strains isolated from different Egyptian cheeses. BMC Microbiol. 2023, 23, 160. [Google Scholar] [CrossRef] [PubMed]
  165. Hanchi, H.; Mottawea, W.; Sebei, K.; Hammami, R. The genus Enterococcus: Between probiotic potential and safety concerns-an update. Front. Microbiol. 2018, 9, 1791. [Google Scholar] [CrossRef] [PubMed]
  166. Krawczyk, B.; Wityk, P.; Gałęcka, M.; Michalik, M. The many faces of Enterococcus spp.—Commensal, probiotic and opportunistic pathogen. Microorganisms 2021, 9, 1900. [Google Scholar] [CrossRef] [PubMed]
  167. EFSA BIOHAZ Panel (EFSA Panel on Biological Hazards); Koutsoumanis, K.; Allende, A.; Alvarez-Ordonez, A.; Bolton, D.; Bover-Cid, S.; Chemaly, M.; Davies, R.; De Cesare, A.; Hilbert, F.; et al. Statement on the update of the list of QPS-recommended biological agents intentionally added to food or feed as notified to EFSA 15: Suitability of taxonomic units notified to EFSA until September 2021. EFSA J. 2022, 20, 40. [Google Scholar] [CrossRef]
  168. Rasika, D.M.D.; Munasinghe, M.A.D.D.; Vidanarachchi, J.K.; da Cruz, A.G.; Ajlouni, S.; Ranadheera, C.S. Probiotics and prebiotics in non-bovine milk. Adv. Food Nutr. Res. 2020, 94, 339–384. [Google Scholar] [CrossRef] [PubMed]
  169. Ranadheera, C.S.; Evans, C.A.; Baines, S.K.; Balthazar, C.F.; Cruz, A.G.; Esmerino, E.A.; Freitas, M.Q.; Pimentel, T.C.; Wittwer, A.E.; Naumovski, N.; et al. Probiotics in goat milk products: Delivery capacity and ability to improve sensory attributes. Compr. Rev. Food Sci. Food Saf. 2019, 18, 867–882. [Google Scholar] [CrossRef] [PubMed]
  170. Ranadheera, C.S.; Naumovski, N.; Ajlouni, S. Non-bovine milk products as emerging probiotic carriers: Recent developments and innovations. Curr. Opin. Food Sci. 2018, 22, 109–114. [Google Scholar] [CrossRef]
  171. Balthazar, C.F.; Pimentel, T.C.; Ferrão, L.L.; Almada, C.N.; Santillo, A.; Albenzio, M.; Mollakhalili, N.; Mortazavian, A.M.; Nascimento, J.S.; Silva, M.C.; et al. Sheep milk: Physicochemical characteristics and relevance for functional food development. Compr. Rev. Food Sci. Food Saf. 2017, 16, 247–262. [Google Scholar] [CrossRef]
  172. McFarland, L.V. From yaks to yogurt: The history, development, and current use of probiotics. Clin. Infect. Dis. 2015, 60, S85–S90. [Google Scholar] [CrossRef]
  173. Shori, A.B. Camel milk and its fermented products as a source of potential probiotic strains and novel food cultures: A mini review. PharmaNutrition 2017, 5, 84–88. [Google Scholar] [CrossRef]
  174. Chieffi, D.; Fanelli, F.; Fusco, V. Legislation of probiotic foods and supplements. In Probiotics for Human Nutrition in Health and Disease; Leite de Souza, E., de Brito Alves, J.L., Fusco, V., Eds.; Academic Press: Cambridge, MA, USA, 2022; ISBN 9780323899086. [Google Scholar] [CrossRef]
  175. Di Lena, M.; Quero, G.M.; Santovito, E.; Verran, J.; De Angelis, M.; Fusco, V. A selective medium for isolation and accurate enumeration of Lactobacillus casei-group members in probiotic milks and dairy products. Int. Dairy J. 2015, 47, 27–36. [Google Scholar] [CrossRef]
  176. Fusco, V.; Fanelli, F.; Chieffi, D. Authenticity of probiotic foods and supplements: Up-to-date situation and methods to assess it. In Probiotics for Human Nutrition in Health and Disease; Leite de Souza, E., de Brito Alves, J.L., Fusco, V., Eds.; Academic Press: Cambridge, MA, USA, 2022; ISBN 9780323899086. [Google Scholar] [CrossRef]
  177. Fusco, V.; Fanelli, F.; Chieffi, D. Authenticity of probiotic foods and supplements: A pivotal issue to address. Crit. Rev. Food Sci. Nutr. 2021, 62, 6854–6871, Erratum in Crit. Rev. Food Sci. Nutr. 2021, 63, 4210–4215. [Google Scholar] [CrossRef]
  178. Leite de Souza, E.; de Brito Alves, J.L.; Fusco, V. (Eds.) Probiotics for Human Nutrition in Health and Disease; Academic Press: Cambridge, MA, USA, 2022; ISBN 978-0-323-89908-6. [Google Scholar] [CrossRef]
  179. Nazli, K.; Ceren, A.; Barbaros, O. Probiotic dairy-based beverages: A review. J. Funct. Foods 2019, 53, 62–75. [Google Scholar] [CrossRef]
  180. Pimentel, T.C.; Gomes de Oliveira, L.I.; de Souza, R.C.; Magnani, M. Probiotic ice cream: A literature overview of the technological and sensory aspects and health properties. Int. J. Dairy Technol. 2022, 75, 59–76. [Google Scholar] [CrossRef]
  181. Sakandar, H.A.; Zhang, H. Trends in probiotic(s)-fermented milks and their in vivo functionality: A review. Trends Food Sci. Technol. 2021, 110, 55–65. [Google Scholar] [CrossRef]
  182. Hadjimbei, E.; Botsaris, G.; Chrysostomou, S. Beneficial effects of yoghurts and probiotic fermented milks and their functional food potential. Foods 2022, 11, 2691. [Google Scholar] [CrossRef]
  183. Kaur, H.; Kaur, G.; Ali, S.A. Dairy-based probiotic-fermented functional foods: An update on their health-promoting properties. Fermentation 2022, 8, 425. [Google Scholar] [CrossRef]
  184. Vera-Santander, V.E.; Hernández-Figuero, R.H.; Jiménez-Munguía, M.T.; Mani-López, E.; López-Malo, A. Health benefits of consuming foods with bacterial probiotics, postbiotics, and their metabolites: A review. Molecules 2023, 28, 1230. [Google Scholar] [CrossRef]
  185. Quintieri, L.; Caputo, L.; Monaci, L.; Cavalluzzi, M.M.; Denora, N. Lactoferrin-derived peptides as a control strategy against skinborne staphylococcal biofilms. Biomedicines 2020, 8, 323. [Google Scholar] [CrossRef]
  186. Quintieri, L.; Fanelli, F.; Zühlke, D.; Caputo, L.; Logrieco, A.F.; Albrecht, D.; Riedel, K. Biofilm and pathogenesis-related proteins in the foodborne P. fluorescens ITEM 17298 with distinctive phenotypes during cold storage. Front. Microbiol. 2020, 11, 991. [Google Scholar] [CrossRef]
  187. Wang, Z.; Xu, J.; Zeng, X.; Du, Q.; Lan, H.; Zhang, J.; Pan, D.; Tu, M. Recent advances on antimicrobial peptides from milk: Molecular properties, mechanisms, and applications. J. Agric. Food Chem. 2023, 72, 80–93. [Google Scholar] [CrossRef] [PubMed]
  188. Sharma, P.; Kaur, H.; Kehinde, B.A.; Chhikara, N.; Sharma, D.; Panghal, A. Food-derived anticancer peptides: A review. Int. J. Pept. Res. Ther. 2021, 27, 55–70. [Google Scholar] [CrossRef]
  189. Tondo, A.R.; Caputo, L.; Mangiatordi, G.F.; Monaci, L.; Lentini, G.; Quintieri, L. Structure-based identification and design of angiotensin converting enzyme-inhibitory peptides from whey proteins. J. Agric. Food Chem. 2019, 68, 541–548. [Google Scholar] [CrossRef] [PubMed]
  190. 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] [PubMed]
  191. Shivanna, S.K.; Nataraj, B.H. Revisiting therapeutic and toxicological fingerprints of milk-derived bioactive peptides: An overview. Food Biosci. 2020, 38, 100771. [Google Scholar] [CrossRef]
  192. Gambacorta, N.; Caputo, L.; Quintieri, L.; Monaci, L.; Ciriaco, F.; Nicolotti, O. Rational discovery of antiviral whey protein-derived small peptides targeting the SARS-CoV-2 main protease. Biomedicines 2022, 10, 1067. [Google Scholar] [CrossRef]
  193. Sharma, K.; Sharma, K.K.; Sharma, A.; Jain, R. Peptide-based drug discovery: Current status and recent advances. Drug Discov. Today 2023, 28, 103464. [Google Scholar] [CrossRef]
  194. 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]
  195. Dullius, A.; Fassina, P.; Giroldi, M.; Goettert, M.I.; de Souza, C.F.V. A biotechnological approach for the production of branched chain amino acid containing bioactive peptides to improve human health: A review. Food Res. Int. 2020, 131, 109002. [Google Scholar] [CrossRef]
  196. Yuan, H.; Lv, J.; Gong, J.Y.; Xiao, G.N.; Zhu, R.Y.; Li, L.; Qiu, J.N. Secondary structures and their effects on antioxidant capacity of antioxidant peptides in yogurt. Int. J. Food Prop. 2018, 21, 2167–2180. [Google Scholar] [CrossRef]
  197. Tagliazucchi, D.; Martini, S.; Shamsia, S.; Helal, A.; Conte, A. Biological activities and peptidomic profile of in vitro-digested cow, camel, goat and sheep milk. Int. Dairy J. 2018, 81, 19–27. [Google Scholar] [CrossRef]
  198. Abd El-Salam, M.H.; El-Shibiny, S. Bioactive peptides of buffalo, camel, goat, sheep, mare, and yak milks and milk products. Food Rev. Int. 2013, 29, 1–23. [Google Scholar] [CrossRef]
  199. Boelsma, E.; Kloek, J. IPP-rich milk protein hydrolysate lowers blood pressure in subjects with stage 1 hypertension, a randomized controlled trial. Nutr. J. 2010, 9, 52. [Google Scholar] [CrossRef]
  200. Barati, M.; Jabbari, M.; Teymoori, F.; Farhadnejad, H.; Khalili-Moghadam, S.; Roshanravan, N.; Mosharkesh, E.; Kazemian, E.; Mirmiran, P.; Davoodi, S.H.; et al. Dairy-originated digestion-resistant and bioactive peptides increase the risk of hypertension: Tehran Lipid and Glucose Study. Hypertens. Res. 2021, 44, 1194–1204. [Google Scholar] [CrossRef]
  201. Jabbari, M.; Barati, M.; Shabani, M.; Kazemian, E.; Khalili-Moghadam, S.; Javanmardi, F.; Hatami, E.; Zeinalian, R.; Davoodi, S.H.; Rashidkhani, B.; et al. The association between consumption of dairy-originated digestion resistant and bioactive peptides and breast cancer risk: A case-control study. Nutr. Cancer 2022, 74, 2426–2435. [Google Scholar] [CrossRef]
  202. El-Sayed, M.I.; Awad, S.; Wahba, A.; El Attar, A.; Yousef, M.I.; Zedan, M. In vivo anti-diabetic and biological activities of milk protein and milk protein hydrolyaste. Adv. Dairy Res. 2016, 4, 1. [Google Scholar]
  203. Antony, P.; Vijayan, R. Bioactive peptides as potential nutraceuticals for diabetes therapy: A comprehensive review. Int. J. Mol. Sci. 2021, 22, 9059. [Google Scholar] [CrossRef] [PubMed]
  204. Abedin, M.M.; Chourasia, R.; Phukon, L.C.; Singh, S.P.; Rai, A.K. Characterization of ACE inhibitory and antioxidant peptides in yak and cow milk hard chhurpi cheese of the Sikkim Himalayan region. Food Chem. 2022, 13, 100231. [Google Scholar] [CrossRef] [PubMed]
  205. Redha, A.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]
  206. Khakhariya, R.; Sakure, A.A.; Maurya, R.; Bishnoi, M.; Kondepudi, K.K.; Padhi, S.; Rai, A.K.; Liu, Z.; Patil, G.; Mankad, M.; et al. A comparative study of fermented buffalo and camel milk with anti-inflammatory, ACE-inhibitory and anti-diabetic properties and release of bio active peptides with molecular interactions: In vitro, in silico and molecular study. Food Biosci. 2023, 52, 102373. [Google Scholar] [CrossRef]
  207. Ayoub, M.A.; Yap, P.-G.; Mudgil, P.; Khan, F.B.; Anwar, I.; Muhammad, K.; Gan, C.-Y.; Maqsood, S. Camel milk-derived bioactive peptides and diabetes: Molecular view and perspectives. J. Dairy Sci. 2023, 107, 649–668. [Google Scholar] [CrossRef]
  208. Abdel-Hamid, M.; Otte, J.; De Gobba, C.; Osman, A.; Hamad, E. Angiotensin I-converting enzyme inhibitory activity and antioxidant capacity of bioactive peptides derived from enzymatic hydrolysis of buffalo milk proteins. Int. Dairy J. 2016, 66, 91–98. [Google Scholar] [CrossRef]
  209. Li, Z.; Jiang, A.; Yue, T.; Wang, J.; Wang, Y.; Su, J. Purification and identification of five novel antioxidant peptides from goat milk casein hydrolysates. J. Dairy Sci. 2013, 96, 4242–4251. [Google Scholar] [CrossRef] [PubMed]
  210. Mudgil, P.; Kamal, H.; Yuen, G.C.; Maqsood, S. Characterization and identification of novel antidiabetic and anti-obesity peptides from camel milk protein hydrolysates. Food Chem. 2018, 259, 46–54. [Google Scholar] [CrossRef] [PubMed]
  211. Alavi, F.; Ciftci, O.N. Purification and fractionation of bioactive peptides through membrane filtration: A critical and application review. Trends Food Sci. Technol. 2023, 131, 118–128. [Google Scholar] [CrossRef]
  212. Mirzapour-Kouhdasht, A.; Garcia-Vaquero, M. Cardioprotective peptides from milk processing and dairy products: From bioactivity to final products including commercialization and legislation. Foods 2022, 11, 1270. [Google Scholar] [CrossRef] [PubMed]
  213. Korhonen, H. Milk-derived bioactive peptides: From science to applications. J. Funct. Foods 2009, 1, 177–187. [Google Scholar] [CrossRef]
  214. Estévez, N.; Fuciños, C.; Rodríguez-Sanz, A.; Rúa, M.L. Development and sensory test of a dairy product with ACE inhibitory and antioxidant peptides produced at a pilot plant scale. Food Chem. 2022, 394, 133459. [Google Scholar] [CrossRef]
  215. Ayyash, M.; Al-Dhaheri, A.S.; Al Mahadin, S.; Kizhakkayil, J.; Abushelaibi, A. In vitro investigation of anticancer, antihypertensive, antidiabetic, and antioxidant activities of camel milk fermented with camel milk probiotic: A comparative study with fermented bovine milk. J. Dairy Sci. 2018, 101, 900–911. [Google Scholar] [CrossRef]
  216. Hajian, N.; Salami, M.; Mohammadian, M.; Moghadam, M.; Emam-Djomeh, Z. Production of low-fat camel milk functional ice creams fortified with camel milk casein and its antioxidant hydrolysates. Appl. Food Biotechnol. 2020, 7, 95–102. [Google Scholar]
  217. Homayouni-Tabrizi, M.; Asoodeh, A.; Soltani, M. Cytotoxic and antioxidant capacity of camel milk peptides: Effects of isolated peptide on superoxide dismutase and catalase gene expression. J. Food Drug Anal. 2017, 25, 567–575. [Google Scholar] [CrossRef] [PubMed]
  218. Homayouni-Tabrizi, M.; Shabestarin, H.; Asoodeh, A.; Soltani, M. Identification of two novel antioxidant peptides from camel milk using digestive proteases: Impact on expression gene of superoxide dismutase (SOD) in hepatocellular carcinoma cell line. Int. J. Pept. Res. Ther. 2016, 22, 187–195. [Google Scholar] [CrossRef]
  219. Wali, A.; Yanhua, G.; Ishimov, U.; Yili, A.; Aisa, H.A.; Salikhov, S. Isolation and identification of three novel antioxidant peptides from the bactrian camel milk hydrolysates. Int. J. Pept. Res. Ther. 2020, 26, 641–650. [Google Scholar] [CrossRef]
  220. Nongonierma, A.B.; Paolella, S.; Mudgil, P.; Maqsood, S.; FitzGerald, R.J. Dipeptidyl peptidase IV (DPP-IV) inhibitory properties of camel milk protein hydrolysates generated with trypsin. J. Funct. Foods 2017, 34, 49–58. [Google Scholar] [CrossRef]
  221. Soleymanzadeh, N.; Mirdamadi, S.; Mirzaei, M.; Kianirad, M. Novel β-casein derived antioxidant and ACE-inhibitory active peptide from camel milk fermented by Leuconostoc lactis PTCC1899: Identification and molecular docking. Int. Dairy J. 2019, 97, 201–208. [Google Scholar] [CrossRef]
  222. Girardet, J.M.; Saulnier, F.; Gaillard, J.L.; Ramet, J.P.; Humbert, G. Camel (Camelus dromedarius) milk PP3: Evidence for an insertion in the amino-terminal sequence of the camel milk whey protein. Biochem. Cell Biol. 2000, 78, 19–26. [Google Scholar] [CrossRef] [PubMed]
  223. Quan, S.; Tsuda, H.; Miyamoto, T. Angiotensin i-converting enzyme inhibitory peptides in skim milk fermented with lactobacillus helveticus 130b4 from camel milk in inner mongolia, china. J. Sci. Food Agric. 2008, 88, 2688–2692. [Google Scholar] [CrossRef]
  224. Coscueta, E.R.; Batista, P.; Gomes, J.E.G.; da Silva, R.; Pintado, M.M. Screening of novel bioactive peptides from goat casein: In silico to in vitro validation. Int. J. Mol. Sci. 2022, 23, 2439. [Google Scholar] [CrossRef] [PubMed]
  225. Simsek, S.; Sánchez-Rivera, L.; El, S.N.; Karakaya, S.; Recio, I. Characterisation of in vitro gastrointestinal digests from low fat caprine kefir enriched with inulin. Int. Dairy J. 2017, 75, 68–74. [Google Scholar] [CrossRef]
  226. Hernández-Ledesma, B.; del Mar Contreras, M.; Recio, I. Antihypertensive peptides: Production, bioavailability and incorporation into foods. Adv. Colloid Interface Sci. 2011, 165, 23–35. [Google Scholar] [CrossRef]
  227. Puchalska, P.; Marina Alegre, M.L.; García López, M.C. Isolation and characterization of peptides with antihypertensive activity in foodstuffs. Crit. Rev. Food Sci. Nutr. 2015, 55, 521–551. [Google Scholar] [CrossRef]
  228. Ibrahim, H.R.; Ahmed, A.S.; Miyata, T. Novel angiotensin-converting enzyme inhibitory peptides from caseins and whey proteins of goat milk. J. Adv. Res. 2017, 8, 63–71. [Google Scholar] [CrossRef] [PubMed]
  229. Gong, H.; Gao, J.; Wang, Y.; Luo, Q.W.; Guo, K.R.; Ren, F.Z.; Mao, X.Y. Identification of novel peptides from goat milk casein that ameliorate high-glucose-induced insulin resistance in HepG2 cells. J. Dairy Sci. 2020, 103, 4907–4918. [Google Scholar] [CrossRef] [PubMed]
  230. Zhang, Y.; Chen, R.; Zuo, F.; Ma, H.; Zhang, Y.; Chen, S. Comparison of dipeptidyl peptidase IV-inhibitory activity of peptides from bovine and caprine milk casein by in silico and in vitro analyses. Int. Dairy J. 2016, 53, 37–44. [Google Scholar] [CrossRef]
  231. Chiozzi, R.Z.; Capriotti, A.L.; Cavaliere, C.; La Barbera, G.; Piovesana, S.; Samperi, R.; Laganà, A. Purification and identification of endogenous antioxidant and ACE-inhibitory peptides from donkey milk by multidimensional liquid chromatography and nanoHPLC-high resolution mass spectrometry. Anal. Bioanal. Chem. 2016, 408, 5657–5666. [Google Scholar] [CrossRef] [PubMed]
  232. Gu, H.; Liang, L.; Zhu, Z.; Mao, X. Preparation and identification of anti-breast cancer cells peptides released from yak milk casein. Front. Nutr. 2022, 9, 997514. [Google Scholar] [CrossRef] [PubMed]
  233. Pei, J.; Jiang, H.; Li, X.; Jin, W.; Tao, Y. Antimicrobial peptides sourced from post-butter processing waste yak milk protein hydrolysates. AMB Express 2017, 7, 217. [Google Scholar] [CrossRef] [PubMed]
  234. Yang, F.; He, X.; Chen, T.; Liu, J.; Luo, Z.; Sun, S.; Qin, D.; Huang, W.; Tang, Y.; Liu, C.; et al. Peptides isolated from yak milk residue exert antioxidant effects through Nrf2 signal pathway. Oxidative Med. Cell. Longev. 2021, 31, 9426314. [Google Scholar] [CrossRef]
  235. Wang, S.; Naumovski, N.; Ajlouni, S.; Ayyash, M.; Silva, R.; Balthazar, C.F.; Esmerino, E.A.; Freitas, M.Q.; da Silva, M.C.; Sant’Ana, A.S.; et al. Nonbovine milk and its products as sources of probiotics delivery: An overview of its viability, functionality and product quality characteristics. Int. J. Dairy. Technol. 2023, 76, 482–511. [Google Scholar] [CrossRef]
  236. Jia, W.; Du, A.; Fan, Z.; Shi, L. Novel insight into the transformation of peptides and potential benefits in brown fermented goat milk by mesoporous magnetic dispersive solid phase extraction-based peptidomics. Food Chem. 2022, 389, 133110. [Google Scholar] [CrossRef]
  237. Liu, Q.; Yang, M.; Zhao, B.; Yang, F. Isolation of antioxidant peptides from yak casein hydrolysate. RSC Adv. 2020, 10, 19844–19851. [Google Scholar] [CrossRef] [PubMed]
  238. Singh, T.P.; Arora, S.; Sarkar, M. Yak milk and milk products: Functional, bioactive constituents and therapeutic potential. Int. Dairy J. 2023, 142, 105637. [Google Scholar] [CrossRef]
  239. Lin, K.; Zhang, L.W.; Han, X.; Cheng, D.Y. Novel angiotensin I-converting enzyme inhibitory peptides from protease hydrolysates of Qula casein: Quantitative structure-activity relationship modeling and molecular docking study. J. Funct. Foods 2017, 32, 266–277. [Google Scholar] [CrossRef]
  240. Lin, K.; Zhang, L.; Han, X.; Meng, Z.; Zhang, J.; Wu, Y.; Cheng, D. Quantitative structure–activity relationship modeling coupled with molecular docking analysis in screening of angiotensin I-converting enzyme inhibitory peptides from Qula Casein hydrolysates obtained by two-enzyme combination hydrolysis. J. Agric. Food Chem. 2018, 66, 3221–3228. [Google Scholar] [CrossRef]
  241. Gómez-Ruiz, J.A.; Ramos, M.; Recio, I. Angiotensin-converting enzyme-inhibitory activity of peptides isolated from Manchego cheese. Stability under simulated gastrointestinal digestion. Int. Dairy J. 2004, 14, 1075–1080. [Google Scholar] [CrossRef]
  242. Quirós, A.; Hernández-Ledesma, B.; Ramos, M.; Amigo, L.; Recio, I. Angiotensin-converting enzyme inhibitory activity of peptides derived from caprine kefir. J. Dairy Sci. 2005, 88, 3480–3487. [Google Scholar] [CrossRef] [PubMed]
  243. Dalabasmaz, S.; de la Torre, E.P.; Gensberger-Reigl, S.; Pischetsrieder, M.; Rodríguez-Ortega, M.J. Identification of potential bioactive peptides in sheep milk kefir through peptidomic analysis at different fermentation times. Foods 2023, 12, 2974. [Google Scholar] [CrossRef]
  244. Rizzello, C.G.; Losito, I.; Gobbetti, M.; Carbonara, T.; De Bari, M.D.; Zambonin, P.G. Antibacterial activities of peptides from the water-soluble extracts of Italian cheese varieties. J. Dairy Sci. 2005, 88, 2348–2360. [Google Scholar] [CrossRef]
  245. Zhao, Q.; Zheng, W.; Yuan, Z.; Wang, X.; Huang, A. Anti-inflammatory effect of two novel peptides derived from Binglangjiang buffalo whey protein in lipopolysaccharide-stimulated RAW264. 7 macrophages. Food Chem. 2023, 429, 136804. [Google Scholar] [CrossRef]
  246. Reddi, S.; Shanmugam, V.P.; Kapila, S.; Kapila, R. Identification of buffalo casein-derived bioactive peptides with osteoblast proliferation activity. Eur. Food Res. Technol. 2016, 242, 2139–2146. [Google Scholar] [CrossRef]
  247. Kumar, N.; Devi, S.; Mada, S.B.; Reddi, S.; Kapila, R.; Kapila, S. Anti-apoptotic effect of buffalo milk casein derived bioactive peptide by directing Nrf2 regulation in starving fibroblasts. Food Biosci. 2020, 35, 100566. [Google Scholar] [CrossRef]
  248. Song, J.J.; Wang, Q.; Du, M.; Ji, X.M.; Mao, X.Y. Identification of dipeptidyl peptidase-IV inhibitory peptides from mare whey protein hydrolysates. J. Dairy Sci. 2017, 100, 6885–6894. [Google Scholar] [CrossRef] [PubMed]
  249. Waili, Y.; Gahafu, Y.; Aobulitalifu, A.; Chang, Z.; Xie, X.; Kawuli, G. Isolation, purification, and characterization of antioxidant peptides from fresh mare’s milk. Food Sci. Nutr. 2021, 9, 4018–4027. [Google Scholar] [CrossRef]
  250. Zaky, A.A.; Simal-Gandara, J.; Eun, J.-B.; Shim, J.-H.; Abd El-Aty, A.M. Bioactivities, applications, safety, and health benefits of bioactive peptides from food and by-products: A review. Front. Nutr. 2022, 8, 815640. [Google Scholar] [CrossRef]
  251. Regulation (EC) No. 1924/2006 of the European Parliament and of the council of 20 December 2006 on nutrition and health claims made on foods. Off. J. Eur. Union 2006, L404/9–L404/25.
  252. Chalamaiah, M.; Ulug, S.K.; Hong, H.; Wu, J. Regulatory requirements of bioactive peptides (protein hydrolysates) from food proteins. J. Funct. Foods 2019, 58, 123–129. [Google Scholar] [CrossRef]
  253. Ponstein-Simarro Doorten, A.Y.; vd Wiel, J.A.; Jonker, D. Safety evaluation of an IPP tripeptide-containing milk protein hydrolysate. Food Chem. Toxicol. 2009, 47, 55–61. [Google Scholar] [CrossRef]
  254. Anadón, A.; Martínez, M.A.; Ares, I.; Ramos, E.; Martínez-Larrañaga, M.R.; Contreras, M.M.; Ramos, M.; Recio, I. Acute and repeated dose (4 weeks) oral toxicity studies of two antihypertensive peptides, RYLGY and AYFYPEL, that correspond to fragments (90-94) and (143-149) from alpha(s1)-casein. Food Chem. Toxicol. 2010, 48, 1836–1845. [Google Scholar] [CrossRef]
  255. Dziuba, M.; Minkiewicz, P.; Dąbek, M. Peptides, specific proteolysis products, as molecular markers of allergenic proteins—In silico studies. Acta Sci. Pol. Technol. Aliment. 2013, 12, 101–112. [Google Scholar]
  256. Cifuentes, A. Foodomics: Principles and applications. In Foodomics: Advanced Mass Spectrometry in Modern Food Science and Nutrition; Cifuentes, A., Ed.; John Wiley & Sons: Hoboken, NJ, USA, 2013; pp. 1–13. [Google Scholar]
  257. Zheng, C.; Chen, A. System biological research on food quality for personalised nutrition and health using foodomics techniques: A review. J. Food Nutr. Res. 2014, 2, 608–616. [Google Scholar] [CrossRef]
  258. Dutta, B.; Lahiri, D.; Nag, M.; Abukhader, R.; Sarkar, T.; Pati, S.; Upadhye, V.; Pandit, S.; Amin, M.F.M.; Al Tawaha, A.R.M.S.; et al. Multi-omics approach in amelioration of food products. Front. Microbiol. 2022, 13, 955683. [Google Scholar] [CrossRef]
  259. Agregán, R.; Echegaray, N.; Nawaz, A.; Hano, C.; Gohari, G.; Pateiro, M.; Lorenzo, J.M. Foodomic-based approach for the control and quality improvement of dairy products. Metabolites 2021, 11, 818. [Google Scholar] [CrossRef]
  260. Kwoji, I.D.; Aiyegoro, O.A.; Okpeku, M.; Adeleke, M.A. ‘Multi-omics’ data integration: Applications in probiotics studies. NPJ Sci. Food 2023, 7, 25. [Google Scholar] [CrossRef]
  261. Nyholm, L.; Koziol, A.; Marcos, S.; Botnen, A.B.; Aizpurua, O.; Gopalakrishnan, S.; Limborg, M.T.; Gilbert, M.P.; Alberdi, A. Holo-omics: Integrated host-microbiota multi-omics for basic and applied biological research. iScience 2020, 23, 101414. [Google Scholar] [CrossRef]
  262. Uroš, A.; Djuro, J. Mass spectrometry based proteomics as foodomics tool in research and assurance of food quality and safety. Trends Food Sci. Technol. 2018, 77, 100–119. [Google Scholar] [CrossRef]
  263. Bolotin, A.; Wincker, P.; Mauger, S.; Jaillon, O.; Malarme, K.; Weissenbach, J.; Ehrlich, S.D.; Sorokin, A. The complete genome sequence of the lactic acid bacterium Lactococcus lactis ssp. lactis IL1403. Genome Res. 2001, 11, 731–753. [Google Scholar] [CrossRef] [PubMed]
  264. Johnson, B.R.; Klaenhammer, T.R. Impact of genomics on the field of probiotic research: Historical perspectives to modern paradigms. Antonie Van Leeuwenhoek 2014, 106, 141–156. [Google Scholar] [CrossRef]
  265. Ventura, M.; O’Flaherty, S.; Claesson, M.; Turroni, F.; Klaenhammer, T.; Van Sinderen, D.; O’Toole, P. Genome-scale analyses of health-promoting bacteria: Probiogenomics. Nat. Rev. Microbiol. 2009, 7, 61–71. [Google Scholar] [CrossRef]
  266. Xiao, Y.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Mining genome traits that determine the different gut colonization potential of Lactobacillus and Bifidobacterium species. Microb. Genom. 2021, 7, 000581. [Google Scholar] [CrossRef]
  267. Denou, E.; Pridmore, R.D.; Berger, B.; Panoff, J.-M.; Arigoni, F.; Brüssow, H. Identification of genes associated with the long-gut-persistence phenotype of the probiotic Lactobacillus johnsonii strain NCC533 using a combination of genomics and transcriptome analysis. J. Bacteriol. 2008, 190, 3161–3168. [Google Scholar] [CrossRef] [PubMed]
  268. Kankainen, M.; Paulin, L.; Tynkkynen, S.; von Ossowski, I.; Reunanen, J.; Partanen, P.; Satokari, R.; Vesterlund, S.; Hendrickx, A.P.A.; Lebeer, S.; et al. Comparative genomic analysis of Lactobacillus rhamnosus GG reveals pili containing a human- mucus binding protein. Proc. Natl. Acad. Sci. USA 2009, 106, 17193–17198. [Google Scholar] [CrossRef]
  269. Buron-Moles, G.; Chailyan, A.; Dolejs, I.; Forster, J.; Mikš, M.H. Uncovering carbohydrate metabolism through a genotype-phenotype association study of 56 lactic acid bacteria genomes. Appl. Microbiol. Biotechnol. 2019, 103, 3135–3152. [Google Scholar] [CrossRef]
  270. Ruiz, L.; Margolles, A.; Sánchez, B. Bile resistance mechanisms in Lactobacillus and Bifidobacterium. Front. Microbiol. 2013, 4, 396. [Google Scholar] [CrossRef]
  271. Ruiz, L.; Sánchez, B.; Ruas-Madiedo, P.; de los Reyes-Gavilán, C.G.; Margolles, A. Cell envelope changes in Bifidobacterium animalis ssp. lactis as a response to bile. FEMS Microbiol. Lett. 2007, 274, 316–322. [Google Scholar] [CrossRef] [PubMed]
  272. Shimizu, K.; Ito, M.; Katto, M.; Takada, T.; Oana, K.; Makino, H.; Okada, N.; Kurakawa, T.; Oishi, K. Identification of genes essential for bile acid resistance in the probiotic Lacticaseibacillus paracasei strain Shirota. Lett. Appl. Microbiol. 2023, 76, ovad062. [Google Scholar] [CrossRef] [PubMed]
  273. Reyes-Castillo, P.A.; González-Vázquez, R.; Torres-Maravilla, E.; Bautista-Hernández, J.I.; Zúñiga-León, E.; Leyte-Lugo, M.; Mateos-Sánchez, L.; Mendoza-Pérez, F.; Gutiérrez-Nava, M.A.; Reyes-Pavón, D.; et al. Bifidobacterium longum LBUX23 Isolated from feces of a newborn; potential probiotic properties and genomic characterization. Microorganisms 2023, 11, 1648. [Google Scholar] [CrossRef] [PubMed]
  274. Sundararaman, A.; Bansal, K.; Sidhic, J.; Patil, P.; Halami, P.M. Genome of Bifidobacterium longum NCIM 5672 provides insights into its acid-tolerance mechanism and probiotic properties. Arch. Microbiol. 2021, 203, 6109–6118. [Google Scholar] [CrossRef] [PubMed]
  275. Khaneghah, A.M.; Abhari, K.; Eş, I.; Soares, M.B.; Oliveira, R.B.A.; Hosseini, H.; Rezaei, M.; Balthazar, C.F.; Silva, R.; Cruz, A.G.; et al. Interactions between probiotics and pathogenic microorganisms in hosts and foods: A review. Trends Food Sci. Technol. 2020, 95, 205–218. [Google Scholar] [CrossRef]
  276. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [PubMed]
  277. Angelin, J.; Kavitha, M. Exopolysaccharides from probiotic bacteria and their health potential. Int. J. Biol. Macromol. 2020, 162, 853–865. [Google Scholar] [CrossRef]
  278. Kandasamy, S.; Yoo, J.; Yun, J.; Lee, K.H.; Kang, H.B.; Kim, J.E.; Oh, M.H.; Ham, J.S. Probiogenomic in-silico analysis and safety assessment of Lactiplantibacillus plantarum DJF10 strain isolated from Korean raw milk. Int. J. Mol. Sci. 2022, 23, 14494. [Google Scholar] [CrossRef] [PubMed]
  279. Felis, G.E.; Torriani, S.; Florez, A.B.; Mayo, B. Genomic characterisation of starter cultures and probiotic bacteria. In Probiotic Dairy Products, 2nd ed.; Tamine, A.Y., Thomas, L.V., Eds.; Wiley and Sons: Hoboken, NJ, USA; Oxford, UK, 2018; pp. 37–65. [Google Scholar]
  280. Abriouel, H.; Lerma, L.L.; Casado Muñoz Mdel, C.; Montoro, B.P.; Kabisch, J.; Pichner, R.; Cho, G.S.; Neve, H.; Fusco, V.; Franz, C.M.; et al. The controversial nature of the Weissella genus: Technological and functional aspects versus whole genome analysis-based pathogenic potential for their application in food and health. Front. Microbiol. 2015, 6, 1197. [Google Scholar] [CrossRef] [PubMed]
  281. Fanelli, F.; Montemurro, M.; Verni, M.; Garbetta, A.; Bavaro, A.R.; Chieffi, D.; Cho, G.S.; Franz, C.M.A.P.; Rizzello, C.G.; Fusco, V. Probiotic Potential and Safety Assessment of Type Strains of Weissella and Periweissella Species. Microbiol. Spectr. 2023, 11, e0304722. [Google Scholar] [CrossRef] [PubMed]
  282. EFSA (European Food Safety Authority). Scientific opinion on the maintenance of the list of QPS biological agents intentionally added to food and feed (2013. update). EFSA J. 2013, 11, 3449. [Google Scholar] [CrossRef]
  283. Papadimitriou, K.; Zoumpopoulou, G.; Foligne, B.; Alexandraki, V.; Kazou, M.; Pot, B.; Tsakalidou, E. Discovering probiotic microorganisms: In vitro, in vivo, genetic and omics approaches. Front. Microbiol. 2015, 6, 58. [Google Scholar] [CrossRef] [PubMed]
  284. Lugli, G.A.; Milani, C.; Turroni, F.; Duranti, S.; Ferrario, C.; Viappiani, A.; Mancabelli, L.; Mangifesta, M.; Taminiau, B.; Delcenserie, V.; et al. Investigation of the evolutionary development of the genus Bifidobacterium by comparative genomics. Appl. Environ. Microbiol. 2014, 80, 6383–6394. [Google Scholar] [CrossRef] [PubMed]
  285. Lugli, G.A.; Mangifesta, M.; Mancabelli, L.; Milani, C.; Turroni, F.; Viappiani, A.; van Sinderen, D.; Ventura, M. Compositional assessment of bacterial communities in probiotic supplements by means of metagenomic techniques. Int. J. Food Microbiol. 2019, 294, 1–9. [Google Scholar] [CrossRef] [PubMed]
  286. Mora, D.; Filardi, R.; Arioli, S.; Boeren, S.; Aalvink, S.; de Vos, W.M. Development of omics-based protocols for the microbiological characterization of multi-strain formulations marketed as probiotics: The case of VSL#3. Microb. Biotechnol. 2019, 12, 1371–1386. [Google Scholar] [CrossRef]
  287. Voth, E.; Khanna, S. The Integrative Human microbiome project: A mile stone in the understanding of the gut microbiome. Expert. Rev. Gastroenterol. Hepatol. 2020, 14, 639–642. [Google Scholar] [CrossRef]
  288. Liu, S.; Lu, S.Y.; Qureshi, N.; Enshasy, H.A.E.; Skory, C.D. Antibacterial property and metagenomic analysis of milk kefir. Probiotics Antimicrob. Proteins 2022, 14, 1170–1183. [Google Scholar] [CrossRef]
  289. Zeng, X.; Wang, Y.; Jia, H.; Wang, Z.; Gao, Z.; Luo, Y.; Sheng, Q.; Yuan, Y.; Yue, T. Metagenomic analysis of microflora structure and functional capacity in probiotic Tibetan kefir grains. Food Res. Int. 2022, 151, 110849. [Google Scholar] [CrossRef] [PubMed]
  290. Nalbantoglu, U.; Cakar, A.; Dogan, H.; Abaci, N.; Ustek, D.; Sayood, K.; Can, H. Metagenomic analysis of the microbial community in kefir grains. Food Microbiol. 2014, 41, 42–51. [Google Scholar] [CrossRef] [PubMed]
  291. Yasir, M.; Bibi, F.; Hashem, A.M.; Azhar, E.I. Comparative metagenomics and characterization of antimicrobial resistance genes in pasteurized and homemade fermented Arabian laban. Food Res. Int. 2020, 137, 109639. [Google Scholar] [CrossRef] [PubMed]
  292. Maoloni, A.; Blaiotta, G.; Ferrocino, I.; Mangia, N.P.; Osimani, A.; Milanović, V.; Cardinali, F.; Cesaro, C.; Garofalo, C.; Clementi, F.; et al. Microbiological characterization of Gioddu, an Italian fermented milk. Int. J. Food Microbiol. 2020, 323, 108610. [Google Scholar] [CrossRef]
  293. Shangpliang, H.N.J.; Tamang, J.P. Metagenome-assembled genomes for biomarkers of bio-functionalities in Laal dahi, an Indian ethnic fermented milk product. Int. J. Food Microbiol. 2023, 402, 110300. [Google Scholar] [CrossRef]
  294. Courant, F.; Antignac, J.P.; Dervilly-Pinel, G.; Le Bizec, B. Basics of mass spectrometry based metabolomics. Proteomics 2014, 14, 2369–2388. [Google Scholar] [CrossRef]
  295. Nicholson, J.K.; Holmes, E.; Lindon, J.C. Metabonomics and metabolomics techniques and their applications in mammalian systems. In The Handbook of Metabonomics and Metabolomics; Lindon, J.C., Nicholson, J.K., Holmes, E., Eds.; Elsevier: Amsterdam, The Netherlands, 2011. [Google Scholar]
  296. Sun, Y.; Guo, S.; Wu, T.J.; Zhang, L.-Y.; Kwok, Z.; Sun, H.; Zhang, J. Untargeted mass spectrometry-based metabolomics approach unveils biochemical changes in compound probiotic fermented milk during fermentation. NPJ Sci. Food 2023, 7, 21. [Google Scholar] [CrossRef] [PubMed]
  297. Zha, M.; Li, K.; Zhang, W.; Sun, Z.; Kwok, L.-Y.; Menghe, B.; Chen, Y. Untargeted mass spectrometry-based metabolomics approach unveils molecular changes in milk fermented by Lactobacillus plantarum P9. LWT-Food Sci. Technol. 2020, 140, 110759. [Google Scholar] [CrossRef]
  298. Ibáñez, C.; Simó, C.; García-Cañas, V.; Cifuentes, A.; Castro-Puyana, M. Metabolomics, peptidomics and proteomics applications of capillary electrophoresis-mass spectrometry in foodomics: A review. Anal. Chim. Acta 2013, 802, 1–13. [Google Scholar] [CrossRef]
  299. García-Cañas, V.; Simó, C.; Castro-Puyana, M.; Cifuentes, A. Recent advances in the application of capillary electromigration methods for food analysis and Foodomics. Electrophoresis 2014, 35, 147–169. [Google Scholar] [CrossRef]
  300. Agregán, R.; Echegaray, N.; López-Pedrouso, M.; Kharabsheh, R.; Franco, D.; Lorenzo, J.M. Proteomic advances in milk and dairy products. Molecules 2021, 26, 3832. [Google Scholar] [CrossRef]
  301. Monaci, L.; De Angelis, E.; Montemurro, N.; Pilolli, R. Comprehensive overview and recent advances in proteomics MS based methods for food allergens analysis. TrAC Trends Anal. Chem. 2018, 106, 21–36. [Google Scholar] [CrossRef]
  302. Toledo, R.; Bernal, M.D.; Marcilla, A. Proteomics of foodborne trematodes. J. Proteom. 2011, 74, 1485–1503. [Google Scholar] [CrossRef]
  303. Wilmes, P.; Heintz-Buschart, A.; Bond, P.L. A decade of metaproteomics: Where we stand and what the future holds. Proteomics 2015, 15, 3409–3417. [Google Scholar] [CrossRef] [PubMed]
  304. Almeida, A.; Bassols, A.; Bendixen, E.; Bhide, M.; Ceciliani, F.; Cristobal, S.; Eckersall, P.D.; Hollung, K.; Lisacek, F.; Mazzucchelli, G.; et al. Animal board invited review: Advances in proteomics for animal and food sciences. Animal 2015, 9, 1–17. [Google Scholar] [CrossRef] [PubMed]
  305. Siragusa, S.; De Angelis, M.; Calasso, M.; Campanella, D.; Minervini, F.; Di Cagno, R.; Gobbetti, M. Fermentation and proteome profiles of Lactobacillus plantarum strains during growth under food-like conditions. J. Proteom. 2014, 96, 366–380. [Google Scholar] [CrossRef] [PubMed]
  306. De Angelis, M.; Calasso, M.; Cavallo, N.; Di Cagno, R.; Gobbetti, M. Functional proteomics within the genus Lactobacillus. Proteomics 2016, 16, 946–962. [Google Scholar] [CrossRef] [PubMed]
  307. Ding, R.; Li, M.; Zou, Y.; Wang, Y.; Yan, C.; Zhang, H.; Wu, R.; Wu, J. Effect of normal and strict anaerobic fermentation on physicochemical quality and metabolomics of yogurt. Food Biosci. 2021, 46, 101368. [Google Scholar] [CrossRef]
  308. Zhang, S.; Li, D.; Zhang, X.; Sun, Y.; Xu, S.; Wang, X.M.; Zhang, N.; Wang, M.; Tian, H.; Li, C. Global transcriptomic analysis of Lactobacillus delbrueckii subsp. bulgaricus ATCC11842 reveals the role of LDB-RS05285 in the post-acidification of yogurt. Food Funct. 2021, 12, 9077–9086. [Google Scholar] [CrossRef] [PubMed]
  309. Pangallo, D.; Kraková, L.; Puškárová, A.; Šoltys, K.; Bučková, M.; Koreňová, J.; Budiš, J.; Kuchta, T. Transcription activity of lactic acid bacterial proteolysis-related genes during cheese maturation. Food Microbiol. 2019, 82, 416–425. [Google Scholar] [CrossRef]
  310. Mitra, S.; Mukhopadhyay, B.C.; Biswas, S.R. Potential application of the nisin Z preparation of Lactococcus lactis W8 in preservation of milk. Lett. Appl. Microbiol. 2011, 53, 98–105. [Google Scholar] [CrossRef] [PubMed]
  311. Suh, J.H. Critical review: Metabolomics in dairy science—Evaluation of milk and milk product quality. Food Res. Int. 2022, 154, 110984. [Google Scholar] [CrossRef] [PubMed]
  312. Pimentel, G.; Burton, K.J.; Vergères, G.; Dupont, D. The role of foodomics to understand the digestion/bioactivity relationship of food. Curr. Opin. Food Sci. 2018, 22, 67–73. [Google Scholar] [CrossRef]
  313. Madsen, C.T.; Refsgaard, J.C.; Teufel, F.G.; Kjærulff, S.K.; Wang, Z.; Meng, G.; Jessen, C.; Heljo, P.; Jiang, Q.; Zhao, X.; et al. Combining mass spectrometry and machine learning to discover bioactive peptides. Nat. Commun. 2022, 13, 6235. [Google Scholar] [CrossRef]
  314. Agyei, D.; Tsopmo, A.; Udenigwe, C.C. Bioinformatics and peptidomics approaches to the discovery and analysis of food-derived bioactive peptides. Anal. Bioanal. Chem. 2018, 410, 3463–3472. [Google Scholar] [CrossRef]
  315. Daroit, D.J.; Brandelli, A. In vivo bioactivities of food protein-derived peptides—A current review. Curr. Opin. Food Sci. 2021, 39, 120–129. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the exploitation of omics technologies for the identification and characterization of probiotics and bioactive peptides from milk and dairy products.
Figure 1. Schematic representation of the exploitation of omics technologies for the identification and characterization of probiotics and bioactive peptides from milk and dairy products.
Foods 13 00601 g001
Table 2. Potential probiotic microorganisms isolated from dairy products (updated from Fusco et al., 2022 [34]).
Table 2. Potential probiotic microorganisms isolated from dairy products (updated from Fusco et al., 2022 [34]).
StrainSpecies aSource bTests Reference
832Saccharomyces (S.) cerevisiaeFeta cheese (sheep milk)In vitro cholesterol assimilation.[96]
56
D4
D7
H 78
SR 82
SR41
SR246
S. cerevisiaeDanish blue veined cheese
Gorgonzola, Italy
Gorgonzola, Italy
Cheese, Europe
Spontaneous fermented cow milk, Sudan
In vitro probiotic characterization: acid and bile tolerance, adhesion to non-tumorigenic porcine jejunal epithelial cell line, cytokine assay.[41]
BFE6058
BFE6059
Lb. acidophilus
Lb. acidophilus
Kule naoto, Maasai fermented milk In vitro probiotic and safety characterization: tolerance to acid and simulated GIT, antigenotoxic properties (ability to protect cells from DNA damage), hydrophobicity cholesterol assimilation, adhesion, bile salts deconjugation, auto-aggregation, biogenic amine production, DNAse activity, hemolysis, mucin degradation, antibiotic susceptibility.[48]
F6Lb. fermentumTraditional dairy product in Inner MongoliaIn vitro probiotic characterization: tolerance to acid and simulated GIT, auto- and co-aggregation. [97]
SM-B
SM-1
Lb. brevis
Lb. plantarum
Brazilian regional ovine cheeseIn vitro probiotic and safety characterization: antioxidant activity, resistance to acid and bile salts, auto-aggregation, bile salt deconjugation, hydrophobicity, antagonistic activity, beta galactosidase activity, hemolysis.[98]
AQ71E. faeciumAzerbaijani Motal cheese (goat or sheep milk)In vitro probiotic and safety characterization: antimicrobial activity, partial purification of bacteriocin, kinetics of bacteriocin production, detection of bacteriocin genes, acid and bile salt tolerance, antibiotic susceptibility, virulence genes detection, auto- and co-aggregation, enzymatic activity.[99]
ZS07
K21
Lb. plantarum
Lb. plantarum
Slovak Bryndza cheese (ewe milk)In vitro probiotic and safety characterization: biogenic amine production, bile salt deconjugation, β-galactosidase activity, resistance to GIT, antibiotic susceptibility.[100]
L10
L11
L13
K. marxianus
S. cerevisiae
K. lactis
Autochthon cheese startersIn vitro probiotic characterization: cell surface hydrophobicity, survival in simulated GIT, auto and co-aggregation with pathogens, antimicrobial activity.[101]
Mb26Lb. plantarumFarmhouse goat’s milk cheeseIn vitro probiotic and safety characterization: biofilm formation, bile salt hydrolase activity, resistance to HCl and oxgall, detection of plantaricin genes, production of hydrogen peroxide and bacteriocines, adhesion and inhibition of L. monocytogenes CECT 4032, production of biogenic amines, functional enzymatic activities, DNase activity, antibiotic susceptibility.[102]
Three halotolerant lactobacilli (name of the strains not provided)Lb. plantarum,
Lb. pentosus,
Lb. acidipiscis
Ripened Mexican tropical cheese (double cream Chiapas cheese)In vitro functional characterization: bile salt deconjugation, antimicrobial activity, β-galactosidase activity, tolerance to simulated orogastrointestinal transit, adhesion to mucin, hydrophobicity.[103]
FT259Lb. paraplantarumBrazilian semi-hard artisanal cheeseIn vitro probiotic and safety characterization: tolerance to low pH and bile salts, antimicrobial activity, adhesion to Caco-2 cells, protease susceptibility and antilisterial activity of the antimicrobial compound produced by FT259; survival in simulated gastric juice, antibiotic susceptibility.[104]
SJRP57Lb. delbruiekii subsp. bulgaricusWater-buffalo mozzarella cheeseIn vitro probiotic and safety characterization: β-galactosidase activity, hydrophobicity, auto- and co-aggregation, tolerance, and effect of matrices on the survival under simulated GIT, adhesion and virulence genes detection, antibiotic and medicine susceptibility.[105]
PRA205
PRA211
PRA172
Lb. casei
Lb. rhamnosus
Lb. rhamnosus
Ripened Parmigiano Reggiano cheese In vitro probiotic and safety characterization: acid, bile salt and lysozyme tolerance, bile salts deconjugation, tolerance to simulated gastric and pancreatic juices, cell surface hydrophobicity, auto-aggregation, antibiotic susceptibility.[106]
LP2Lb. fermentumTulum cheeseIn vitro probiotic and safety characterization: enzymatic activity, adhesion, survival in simulated GIT, antimicrobial activity, cholesterol assimilation, antibiotic susceptibility.[107]
ZGPR3-18
BGGO5-47
Lb. plantarum
Lb. rhamnosus
Croatian fresh soft cheese and Serbian white pickled cheeseIn vitro probiotic and safety characterization: bile salt hydrolase activity, adhesion, cell surface hydrophobicity, survival in simulated GIT, assimilation of prebiotic substrates, cholesterol assimilation, capacity to modulate the immune response on GALT (gut associated lymphoid tissues) from rats, antibiotic susceptibility.[108]
FS10
PM8
Lb. rhamnosus
Lb. paracasei
Ragusano (cow milk) and Pecorino (sheep milk) Siciliano cheesesIn vitro probiotic and safety characterization: adhesion, hemolytic and bile salt hydrolase activities, tolerance to simulated GIT, hydrophobicity, antimicrobial activity, co- and auto-aggregation, antibiotic susceptibility.[109]
SJRP55Leuc. mesenteroidesBrazilian water buffalo mozzarella cheeseIn vitro probiotic and safety characterization: auto and co-aggregation, adhesion, cell surface hydrophobicity, low pH, bile and NaCl tolerance, medication and antibiotic susceptibility, bile salts deconjugation, enzymatic activity.[110]
ZIM 2408Kluyveromyces (K.). lactisTraditional cheese from Serbia and CroatiaIn vitro probiotic characterization: adhesion, tolerance to simulated GIT, proliferation of gut-associated lymphoid tissue cells in the presence of non-viable yeast strain. [111]
SJRP17E. duranscheeseIn vitro probiotic and safety characterization: tolerance to NaCl, hydrophobicity, biogenic amine production, tolerance to simulated GIT, bile salt hydrolase activity, auto and co-aggregation, mucin degradation, virulence gene detection, medication and antibiotic susceptibility.[112]
KJ722784Lb. plantarumChhurpi (Indian dried cottage cheese)In vitro probiotic characterization: cholesterol removal, exopolysaccharide production, tolerance to acid, auto-aggregation, lysozyme and bile salt, antimicrobial activity, cell surface hydrophobicity, β-galactosidase activity, hemolytic activity, antibiotic susceptibility.[113]
KLDS 1.8701Lb. helveticusChinese traditional cheeseIn vitro probiotic characterization: antimicrobial activity, tolerance to simulated GIT.[114]
1133
1086-1
1089
1138
1059
1141
1197
Lb casei
Lb. plantarum
Lb. casei
Lb. casei
Lb. buchneri
Lb. plantarum
Lb. plantarum
Tibetan qula (raw yak milk cheese)In vitro probiotic and safety characterization: antimicrobial activity, acid and bile salt tolerance, antibiotic susceptibility, tolerance to simulated GIT, cell surface hydrophobicity.[115]
B7Lb. plantarumMinas artisanal cheeseProtective effects of milk fermented by B7 on Salmonella enterica serovar Typhimurium infection in BALB/c mice (in vivo).[116]
26
27
55
56
Lb. plantarumPortuguese raw ewe milk semisoft cheeseIn vitro antihemolytic, antimutagenic, anti-inflammatory, antioxidant and antimicrobial activities of bioactive peptides produced in the probiotic fermented milk.[117]
SJRP30
SJRP145
SJRP146
Lb. fermentum
Lb. casei
Lb. casei
Water buffalo mozzarella cheeseIn vitro probiotic and safety characterization: β-galactosidase activity, adhesion, aggregation and colonization factors, tolerance to simulated GIT, antimicrobial activity, antibiotic susceptibility, mucin degradation, detection of genes encoding adhesion, virulence, antibiotic resistance, and biogenic amine production.[118]
LP4Lb. plantarumMinas artisanal cheeseIn vitro probiotic and safety characterization: bile salt and artificial gastric juice tolerance, hydrogen peroxide production, antimicrobial susceptibility, antagonistic activity. In vivo protection against Salmonella Typhimurium infection in mice.[119]
17bKL1
17bKlL2
NS2KL9
NS1KM2
14KM1
6688 KM
K. lactis
K. lactis
K. marxianus
K. marxianus
K. marxianus
K. marxianus
Fiore sardo cheese (raw ewe milk)In vitro probiotic and safety characterization: tolerance to GIT, bile salt hydrolase activity, hydrophobicity, auto-aggregation, adhesion to Caco-2 cells, hemolytic activity, susceptibility to antifungal agents, antimicrobial activity.[120]
K5Lb. paracaseiFeta-type cheese (sheep milk)In vitro probiotic and safety characterization: antibiotic susceptibility and tolerance to simulated GIT.[121]
DP3
DP21
Lb. plantarum
Lb. casei
Iranian artisanal cheesesIn vitro technological, probiotic and safety characterization: bile salt and acid tolerance, auto-aggregation, cholesterol assimilation, adhesion to human intestinal cells, bile salt hydrolase activity, cell surface hydrophobicity, exopolysaccharide production, antagonistic activity, hemolytic activity, antibiotic susceptibility, monitoring of the pH and microbial survival of the probiotic fermented milk with added DP3 and DP21.[122]
L3C1E8Lb. plantarumPico cheeseIn vitro probiotic characterization: low pH, bile salts and pancreatin tolerance, biofilm formation, adhesion, auto-aggregation, ability to inhibit the adhesion of E. coli ATCC 25922 to HT-29 cells, hydrophobicity. Production of conjugated linoleic acid from free linoleic acid.[123]
J20
J23
J24
J25
J27
J28
J32
J37
Lb. fermentum
Lb. fermentum
Lb. pentosus
Lb. plantarum
Lb. pentosus
Lb. fermentum
Lb. fermentum
Lb. pentosus
Cocido cheeseIn vitro technological, probiotic and safety characterization: survival at pasteurization temperatures, survival under cold, lyophilizing, and freezing conditions, low pH and bile salt tolerance, hydrophobicity, auto-aggregation, mucin degradation, β-galactosidase activity, antibiotic susceptibility, hemolytic activity. In vivo study of the immunomodulatory effect of the probiotic fermented milk.[124]
ACA-DC 2640
ACA-DC 4039
ACA-DC 264
ACA-DC 170
Lb. plantarum
Lb. plantarum
Streptococcus (St.) thermophilus
St. thermophilus
Traditional Greek dairy products (Kasseri and Feta sheep milk cheeses)In vitro adherence ability, antibacterial activity, and anti-inflammatory properties.[125]
OB15E. faecalisTunisian Rigouta cheeseIn vitro probiotic and safety characterization: acid and bile salt tolerance, hydrophobicity, auto-aggregation, biofilm formation, adhesion to Caco-2 cells, gelatinase activity, hemolysis and cytotoxicity in Caco-2 cells, antibiotic susceptibility, virulence in the Galleria mellonella model, virulence gene detection.[126]
SJRP38
SJRP43
Lb. casei
Lb. fermentum
Water buffalo mozzarella cheeseIn vitro probiotic characterization: survival of the probiotic strains and of milk fermented with the probiotic strains to simulated GIT, auto- and co-aggregation, hydrophobicity, β-galactosidase production, virulence genes detection.[127]
DSM 32386Lb. brevisAlpine cheeseIn vitro probiotic and safety characterization: γ-aminobutyric acid (GABA) production, Tolerance to pH, bile and pancreatic fluid, antibiotic susceptibility, antibiotic resistance gene detection.[128]
FS103Lb. paracaseiSheep cheeseIn vitro technological, probiotic and characterization: survival in the probiotic sheep and cow fermented milk during cold storage, tolerance to simulated GIT, acid and bile salt tolerance, antibiotic susceptibility, antagonistic activity.[129]
PE24
PE25
PE44
PE61
PE85
PE86
Lb. paracasei
Lb. rhamnosus
Lb. rhamnosus
Lb. rhamnosus
Lb. paracasei
Lb. paracasei
Piacentino Ennese PDO cheeseIn vitro probiotic and safety characterization: survival in simulated GIT, acid, lysozyme and bile salt tolerance, hydrophobicity, antimicrobial activity, anti-inflammatory activity, adhesion, auto- and co-aggregation, antioxidant activity, preliminary identification of metabolites responsible for antagonistic activity against pathogens, virulence and antibiotic resistance genes detection, biogenic amine production, DNAse, gelatinase, hemolytic and mucin degradation, antibiotic susceptibility and MIC determination.[130]
C1Lb21
GiLb5
G4Lb7
Lb. brevis
Lb. plantarum
Lb. pentosus
Serpa Cheese (soft cheese)In vitro probiotic and safety characterization: aggregation activity, tolerance to simulated GIT, cell surface hydrophobicity, growth on prebiotic, short-chain fatty acid (SCFA) production, biogenic amine production, antibiotic susceptibility. [131]
E297E. faeciumMinas Frescal cheeseIn vitro probiotic and safety characterization: survival in simulated GIT, gelatinase, antimicrobial resistance, virulence and antibiotic resistance genes detection, lipase and DNase production, hemolysis.[132]
A6Lb. brevisMinas artisanal cheeseIn vitro probiotic and safety characterization: bile salts and artificial gastric juice tolerance, hydrogen peroxide production, antimicrobial activity, antibiotic susceptibility.[133]
PMD74E. lactisEzine cheese (made from ewe, cow and goat milk)In vitro functional and safety characterization: bile salt hydrolysis (BSH) and mucin degradation activity, survival in simulated GIT, antimicrobial activity, antibiotic susceptibility, hemolytic and gelatinase activity, amino acid decarboxylase detection, detection of virulence genes.[134]
B7
D1
Lb. plantarum
Lb. rhamnosus
Minas artisanal cheeseIn vivo (in mice) and in vitro functional characterization: acid and bile salt tolerance, antimicrobial susceptibility.[135]
SMVDUDB2Pediococcus acidilacticiKalarei (traditional cheese)In vitro probiotic and safety characterization: exopolysaccharide production, auto and co-aggregation, tolerance to simulated GIT, hydrophobicity, antagonistic activity, antibiotic susceptibility.[136]
L3C21M6Lb. paracaseiArtisanal Pico cheeseIn vitro probiotic and safety characterization: acid, bile salts and pancreatin tolerance, bile salts deconjugation, adhesion, virulence genes detection, antibiotic susceptibility. In vitro cholesterol-lowering activity and ability to degrade histamine.[137]
OSY-EGY
E. duransEgyptian artisanal cheeseIn vitro probiotic and safety characterization: resistance to low pH and bile salts, auto-aggregation, adhesion, hydrophobicity, cholesterol-lowering activity, antioxidant effect, antimicrobial activity, heamolysis, gelatinase production, cytotoxicity in human colorectal adenocarcinoma cell line Caco-2 cells, antibiotic susceptibility, virulence genes detection.[138]
KMJC4
KMJC1
Lb.plantarum
Lb. brevis
Iranian Jug cheese (made from cow milk or a mixture of sheep and cow milks)In vitro functional and safety characterization: acid and bile salt tolearance, survival in simulated GIT, adhesion capacity, antimicrobial activity, heamolysis, antibiotic susceptibility.[139]
Not specifiedPediococcus acidilacticiWara, Nigerian unripened soft cheeseIn vitro probiotic and safety characterization: acid and bile salt tolerance, survival in simulated GIT, antimicrobial activity, auto-aggregation, adhesion to hydrocarbon assay, exopolysaccharide production, heamolytic activity.[140]
Os4
Kor14
Lb. plantarum
Lb. plantarum
Polish regional cheesesIn vitro technological and probiotic characterization: enzymatic profile, adhesion, tolerance to simulated GIT, anti-staphylococcal activity, antibiotic resistance. Assessment of the anti-staphylococcal activity in the probiotic skim milk.[141]
B9, B13 and B38Lb. brevisAlgerian homemade cheeses made from either cow, sheep or goat milkIn vitro probiotic and safety characterization: tolerance to simulated GIT, hydrophobicity, adhesion capacity, auto and co-aggregation, antimicrobial activity, bile salt deconjugation, in vitro cholesterol, lowering ability, antioxidant activity, antibiotic susceptibility.[142]
B14L. acidophilusIranian cheeseIn vitro functional and safety characterization: acid and bile salt tolerance, resistance to simulated GIT, cell surface hydrophobicity, auto- and co-aggregation, adhesion capacity, antibiotic susceptibility, hemolytic activity, biogenic amine production.[143]
RI53
RI42
E. faeciumAhvaz semi-skimmed natural cheese
Ardabil full-fat natural cheese
In vitro probiotic and safety characterization: aggregation properties, acid and bile salt tolerance, survival in simulated GIT, production of exopolysaccharides, antimicrobial activity, antibiotic susceptibility.[144]
5C
1D
9 KE
3 TB
7 KB
3B
E. faecalis
E. hiriae
E. durum
E. faecium
E. faecalis
Motal cheese (traditional raw milk Iranian cheese)In vitro functional and safety characterization: survival under simulated GIT, adhesion to Caco-2 cells, antibacterial activity, auto- and co-aggregation, cholesterol assimilation, hemolysis, antibiotic susceptibility, virulence gene detection.[145]
1QB77Lb. plantarumBrazilian artisanal cheeseIn vitro probiotic and safety characterization: auto-aggregation, low pH and bile salt tolerance, hydrophobicity, tolerance to simulated GIT, adhesion, biogenic amine genes detection, antagonistic activity, antimicrobial properties of the potential probiotic strain in microscale cheeses.[146]
S1113
S104
S1121
S202
E. faecium
E. durans
E. durans
E. durans
Artisanal white cheeses from TurkeyIn vitro probiotic and safety characterization: antibiotic susceptibility bile salts and simulated gastric juice tolerance, antimicrobial activity.[147]
894E. faeciumTurkish Tulum cheeseIn vitro probiotic and safety characterization: auto-aggregation, antibacterial activity, bile and simulated gastric juice tolerance, cell surface hydrophobicity, cholesterol removal, β-galactosidase activity, exopolysaccharide production, antibiotic susceptibility, hemolytic activity, biogenic amine production.[148]
SIM12
SIS16
1734
Lh43
Lb. helveticusItalian hard cheesesIn vitro assessment of the immunomodulatory properties (cytokine production in culture media and in cheese). In vitro probiotic characterization: proteolytic, peptidase, antioxidant and β-galactosidase activities, ability to produce folate. [149]
MT1-MT6Lactip. plantarumTen different traditional Iranian cheeses produced either from cow, ewe or goat milkIn vitro probiotic and safety characterization: resistance to acid, bile salts and simulated GIT, antibiotic susceptibility, antibacterial activity.[150]
T40L. paracaseiTenate cheeseIn vitro probiotic characterization: resistance to acid and bile salt, antimicrobial activity.[151]
SJ14Lactp. plantarumAlgerian
traditional cheese “Jben”, made from with raw cow, goat, or sheep milk
In vitro probiotic and safety characterization: acid and bile salt tolerance, antibacterial and antifungal activities, exopolysaccharides production, adhesion to Caco-2 cells, antibiotic susceptibility.[152]
12 not typed isolatesIsolates of the Enterococcus genus by phenotypic testsLabneh Anbaris (goat milk cheese)In vitro functional characterization: acid and bile tolerance, antibacterial activity, ACE inhibitory activity. [153]
AD73L. plantarumMilk kefirIn vitro probiotic characterization: adhesion to Caco-2 cells, antibacterial activity, acid and bile salt tolerance, cytotoxic activity in Caco-2 cells, survival in lab-scale production of probiotic Chevre cheese.[154]
14
15
16
47
60
70
Not typed isolates that were identified by biochemical methods as Lactobacillus spp.Ethiopian spiced cottage cheeseIn vitro probiotic and safety characterization: hydrophobicity, acid and bile salt tolerance, antimicrobial activity, hemolysis, antibiotic susceptibility.[155]
9 strains isolated from rala
3 strains isolated from pingo
Yarrowia lipolytica
Kodamaea ohmeri
Endogenous ferment (pingo-whey or rala-grated ripened cheese) of artisanal Minas cheeseIn vitro probiotic and safety characterization: acid and bile tolerance, auto-aggregation, hydrophobicity, antibiotic susceptibility, co-aggregation with pathogens, antimicrobial activity.[156]
54B,
54C,
55A,
55B,
95E
Lb. plantarum
Lb. plantarum
Lb. plantarum
Lb. pentosus
P. pentosaceus
Ethiopian spontaneously fermented cheeseIn vitro probiotic and safety characterization: survival in simulated GIT, antimicrobial activity, in vitro immunostimulatory activity, antibiotic susceptibility.[157]
4R15
9N2
3R2
Lb. paracaseiTulum cheese (Turkish goat milk cheese)In vitro probiotic and safety characterization: bile salt hydrolase activity, tolerance to simulated GIT, antimicrobial activity, adhesion to HT-29 cells, biogenic amine production, antibiotic susceptibility.[158]
B15
BM10
C30
Lb. plantarum,
Lb. casei,
E. durans
Bouhezza cheese (made from raw goat or sheep milk)
Bouchezza cheese
Goat’s butter
In vitro probiotic and safety characterization: phenol, acid and bile salt tolerance, hydrophobicity, auto- and co-aggregation, antimicrobial activity, exopolysaccharide production, hemolysis, antibiotic susceptibility.[159]
CFSP. acidilacticiIranian cheeseIn vitro probiotic and safety characterization: NaCl, acid and bile salt tolerance, antimicrobial activity, in vitro anticancer assessment, antibiotic susceptibility.[160]
LAB-2
LAB-3
LAB-6
Lb. rhamnosus
Lb. rhamnosus
E. durans
Egyptian fermented dairy samplesIn vitro probiotic and safety characterization: acid and bile salt tolerance, antimicrobial activity, antibiotic susceptibility.[161]
16
26
28
29
21
P. pentosaceus
Leucc. mesenteroides
Lb. casei
Lb. fermentum
E. faecium
Colombian double creame cheeseIn vitro probiotic and safety characterization: acid and bile salt tolerance, auto-aggregation, hydrophobicity, exopolysaccharide production, antimicrobial activity, hemolysis, antibiotic susceptibility.[162]
L11
L13
L33
Lb. pentosus
E. faecium
Lb. plantarum
Iranian cheeseIn vitro probiotic and safety characterization: hydrophobicity, auto- and co-aggregation, antimicrobial activity, hemolysis, antibiotic susceptibility, biogenic amine production.[163]
BD3
BR4
MR2
Lb. paracasei
Lb. plantarum
Lb. fermentum
Egyptian cheesesIn vitro probiotic and safety characterization: resistance to gastric acidity and bile salts, exopolysaccharide production, auto-aggregation, hydrophobicity, bile salt hydrolysis, hemolysis, antibiotic susceptibility.[164]
a The genus Lactobacillus has been reclassified into 25 genera [94]. In this table, the old and the new nomenclature are provided. b Where not specified, the milk used for the production of dairy products is cow’s milk.
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

Quintieri, L.; Fanelli, F.; Monaci, L.; Fusco, V. Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides. Foods 2024, 13, 601. https://doi.org/10.3390/foods13040601

AMA Style

Quintieri L, Fanelli F, Monaci L, Fusco V. Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides. Foods. 2024; 13(4):601. https://doi.org/10.3390/foods13040601

Chicago/Turabian Style

Quintieri, Laura, Francesca Fanelli, Linda Monaci, and Vincenzina Fusco. 2024. "Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides" Foods 13, no. 4: 601. https://doi.org/10.3390/foods13040601

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