Next Article in Journal
Incorporation of Blue Honeysuckle Juice into Fermented Goat Milk: Physicochemical, Sensory and Antioxidant Characteristics and In Vitro Gastrointestinal Digestion
Next Article in Special Issue
Microbial Properties of Raw Milk throughout the Year and Their Relationships to Quality Parameters
Previous Article in Journal
Protective Effects of Four Natural Antioxidants on Hydroxyl-Radical-Induced Lipid and Protein Oxidation in Yak Meat
Previous Article in Special Issue
Effect of Adding Bifidobacterium animalis BZ25 on the Flavor, Functional Components and Biogenic Amines of Natto by Bacillus subtilis GUTU09
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Characterization, High-Density Fermentation, and the Production of a Directed Vat Set Starter of Lactobacilli Used in the Food Industry: A Review

1
Key Laboratory of Agricultural and Animal Products Storage & Processing of Guizhou Province, Guizhou University, Guiyang 550025, China
2
Department of Brewing Engineering, Moutai University, Renhuai 564507, China
3
College of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
4
School of Chemistry and Chemical Engineering, Guizhou University, Guiyang 550025, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Foods 2022, 11(19), 3063; https://doi.org/10.3390/foods11193063
Submission received: 16 August 2022 / Revised: 15 September 2022 / Accepted: 29 September 2022 / Published: 2 October 2022
(This article belongs to the Special Issue Molecular Methods in Food Quality and Microbiological Safety)

Abstract

:
Lactobacilli have been widely concerned for decades. Bacteria of the genus Lactobacillus have been commonly employed in fermented food to improve the appearance, smell, and taste of food or prolong its shelf-life. They comprise 261 species (by March 2020) that are highly diverse at the phenotypic, ecological, and genotypic levels. Some Lactobacilli strains have been documented to be essential probiotics, which are defined as a group of living microorganisms that are beneficial to the health of the host when ingested in sufficiency. However, the characterization, high-density fermentation, and the production of a directed vat set (DVS) starter of Lactobacilli strains used in the food industry have not been systematically reported. This paper mainly focuses on reviewing Lactobacilli as functional starter cultures in the food industry, including different molecular techniques for identification at the species and strain levels, methods for evaluating Lactobacilli properties, enhancing their performance and improving the cell density of Lactobacilli, and the production techniques of DVS starter of Lactobacilli strains. Moreover, this review further discussed the existing problems and future development prospects of Lactobacilli in the food industry. The viability and stability of Lactobacilli in the food industry and gastrointestinal environment are critical challenges at the industrial scale. The new production equipment and technology of DVS starter of Lactobacilli strains will have the potential for large-scale application, for example, developing low-temperature spray drying, freezing granulation drying, and spray freeze-drying.

Graphical Abstract

1. Introduction

Lactobacilli are Gram-positive rod, non-spore-forming, catalase-negative bacteria that commonly colonize the human intestine and have essential physiological functions in the human body [1,2]. Microscopically, these bacteria represent non-motile, thin rods that differ from long to short. Sometimes, they are present in coryneform, bent morphology, or chains. Lactobacilli comprise 261 species (by March 2020) that are highly diverse at the phenotypic, ecological, and genotypic levels [3]. Moreover, Lactobacilli have been documented to be important probiotics, defined as a group of living microorganisms that are beneficial to the host’s health when ingested in sufficiency [4].
Lactobacilli may be added as starters, which are used to ferment and produce specific changes in the chemical composition and sensory properties of foods [5]. It can produce amylase, protease, dehydrogenase, decarboxylase, β-glucosidase, and peptidase during the fermentation, thereby can be widely used in the food industry for the production of yogurt [6], cheese [7], sourdough [8], sausages [9], cucumber pickles [10], olives [11], sauerkraut [12], and so on. However, there is some diversity in the Lactobacilli used in fermented food, depending on the food matrix. One example is Lactobacillus plantarum, which is used as a starter culture in meat and wine (malolactic) fermentation [5], whereas L. bulgaricus can be found as the primary starter culture in yogurt fermentation [13].
The nutritional quality may increase during fermentation by Lactobacilli strains because the hydrolytic enzymes produced by the bacteria hydrolyze the complex macromolecule into simpler forms [14]. The characteristics of Lactobacilli strains in fermented food mainly include antimicrobial activity [15], acid and bile tolerance [16], gastrointestinal transit tolerance [17], cell surface properties (such as auto-aggregation, co-aggregation, and bacterial hydrophobicity) [18], metabolic products (such as bacteriocin, organic acids, fatty acids, hydrogen peroxide, bioactive peptides, and cell wall components) [19], and the evaluation of safety such as antibiotic resistance [20]. Additionally, Lactobacilli strains with high activity and stability can meet the rapid development of industrial food production [16]. The most common methods for long-term storage of Lactobacilli strains are freezing, spraying, and fluidized bed drying [21,22]. However, the latest progress, existing problems, and prospects on characterization, high-density fermentation, and directed vat set starter production of Lactobacilli used in the food industry still lacks systematic excavation. This paper mainly focuses on reviewing Lactobacilli as functional starter cultures in the food industry, including different molecular techniques for identification at the species and strain levels, methods for evaluating Lactobacilli properties, improving their performance and improving the cell density of Lactobacilli, the production techniques of DVS starters of Lactobacilli strains and future perspectives to be overcome in this area. Moreover, this review further discusses the existing problems and future development prospects of Lactobacilli in the food industry.

2. Characterization of Lactobacilli Strains

2.1. Screening out Lactobacilli Strains

Lactobacilli used in the fermented food industry are diverse and many (Table 1). Due to their excellent fermentation performance, they are extensively used to ferment food based on various raw materials, including milk, meat, cereals, fruits, vegetables, and seafood. Their commercial products, including probiotics, have ample market space [23]. So, screening out one or several new strains with excellent fermentation performance and potential probiotic properties is very meaningful work. High-throughput screening technology is a method for the quick selection of certain strains of Lactobacilli species with outstanding performance (such as extracellular polysaccharides, bacteriocin, gamma amino acid, butyric acid, short-chain fatty acid, etc.) [24]. These Lactobacilli strains can be traditionally isolated from a wide range of sources, such as human and animal mucosal membranes, plants or material of plant origin, and fermented food.
We constructed a phylogenetic tree containing 64 Lactobacilli species based on the related Lactobacilli 16S rRNA gene sequence derived from National Center for Biotechnology Information (https://www.ncbi.nlm.nih.gov/) (Figure 1). It shows that the Lactobacilli species used in the food industry are not clustered in a particular branch but are almost evenly distributed in all branches of the Lactobacilli phylogenetic tree. Based on this, the other species in this genus may also have potential use value in the field of food, livestock breeding, and medicine. Given that Lactobacilli have strain specificity, the traditionally safe Lactobacilli strains cannot ensure the safety of all strains in the species. Thus, the identification and detection of Lactobacilli are of significance.
Generally, microorganisms always contain desirable and undesirable characteristics. A qualified candidate for use in the food industry initially requires a desirable characterization, especially safety. For Lactobacilli strains, we expect to select strains with probiotic properties. These characteristics of new Lactobacilli strains need to be evaluated by in vitro and in vivo experiments to understand their potential probiotic properties and industrial uses. Table 2 summarizes in vitro evaluation assays of Lactobacilli strains used in screening.

2.2. Identification and Safety Assessment of Lactobacilli Strains

The safety status of Lactobacilli used in food production has become the focus of the attention of manufacturers and government departments. Although European Qualified Presumption of Safety regulations have identified microorganisms that can be safely used in food [91], some safety aspects must be evaluated before the newly screened cultures are applied to fermented food. For a newly screened Lactobacilli strain, its inherent characteristics need to be tested in vitro. First, we need to identify the isolate to make sure it is what we want and safe, and it can preliminarily predict whether the strains of this genus can be fully put into production [44]. Figure 2 describes some essential characterization contents of the new Lactobacilli strain.
The identification methods of Lactobacilli use phenotypic methods and molecular identification methods. In contrast to phenotypic approaches, molecular identification and characterization tools can distinguish even between closely related groups of species, which are indistinguishable based on phenotype, which is far more consistent, quick, trustworthy, and reproducible [46,47]. The most commonly employed molecular techniques for the identification of Lactobacilli can be divided into two groups: species-specific identification techniques (including amplified ribosomal DNA restriction analysis (ARDRA) and 16S and 23S rRNA sequencing) and strain-specific identification techniques (including ribotyping, restriction enzyme analysis (REA) with pulsed-field gel electrophoresis (PFGE), genetic probes/DNA dot blot, multiplex PCR using specific primers, randomly amplified polymorphic DNA (RAPD), amplified fragment length polymorphism (AFLP), PCR-denaturing gradient gel electrophoresis (DGGE), and fluorescent in situ hybridization (FISH)) [92,93,94,95,96,97,98,99,100,101,102,103,104,105,106]. As an overview, these methods and evaluations are listed in Table 3. Taxonomy and phylogeny of the genus Lactobacillus have been recognized as rather complicated, because of a great number of species with a diverse group of species [3]. From the Table 3, it is clear that for reliable species determination within this genus, a polyphasic approach based primarily on one or more molecular methods is required. Additionally, the International Committee on Systematic Bacteriology has acknowledged polyphasic taxonomy as a trustworthy method for describing species and revising the current nomenclature of specific bacterial groupings. In a recent study, Zheng et al. [3] proposed reclassification of the genus Lactobacillus into 25 genera including the emended genus Lactobacillus, which includes host-adapted organisms that have been referred to as the Lactobacillus delbrueckii group, Paralactobacillus and 23 novel genera including Holzapfelia, Amylolactobacillus, Bombilactobacillus, Companilactobacillus, Lapidilactobacillus, Agrilactobacillus, Schleiferilactobacillus, Loigolactobacilus, Lacticaseibacillus, Latilactobacillus, Dellaglioa, Liquorilactobacillus, Ligilactobacillus, Lactiplantibacillus, Furfurilactobacillus, Paucilactobacillus, Limosilactobacillus, Fructilactobacillus, Acetilactobacillus, Apilactobacillus, Levilactobacillus, Secundilactobacillus, and Lentilactobacillus. This reclassification reflects the phylogenetic position of the micro-organisms and groups Lactobacilli into robust clades with shared ecological and metabolic properties that can anticipate the addition of new species shortly.
Then, the relevant antibiotic susceptibility is usually determined and evaluated according to the protocol provided by the European Food Safety Agency (EFSA) [107]. Microdilution broth tests on test tubes, disk diffusion [108], and commercial ready-to-use kits [109] have been used to determine the physical sensitivity of known antibiotics to newly screened strains. Hemolytic activity was also investigated [49]. The production of various enzymes should also be evaluated. Maybe they are the cause of pathogenicity. Strains should be tested for known human toxins (e.g., cytolysin) by appropriate in vitro analysis. Detecting the toxicity of pathogenic genes and metabolites is also conducted; several Lactobacilli can decarboxylate and reduce amino acids in food to produce biogenic amines, which can cause poisoning symptoms if they accumulate in excess amounts in the body [110]. These in vitro experimental analyses are simple and rapid in determining the safety of a newly screened strain and avoid the use of harmful strains. For example, a hemolytic and toxin-producing strain can easily be excluded from further analysis [49]. The false negative strains created by in vitro experimental research are concerning. Therefore, further in vivo experiments are needed, including animal models and clinical applications [11,82].

2.3. Potential Probiotic Functionalities of Lactobacilli Strains

Some Lactobacilli have been reported as strains with high probiotic potential and support efforts to improve probiotic quality, such as L. salivarius strains BCRC14759 and BCRC 12574, with the highest exopolysaccharide production [111], L. johnsonii ZLJ010, with better adaptation to the gut environment and its probiotic functionalities [112], and L. helveticus D75 and D76 that can inhibit the growth of pathogens and pathobionts [20]. However, Lactobacilli strains in the probiotic market are still limited, and Lactobacilli strains with potential probiotic properties should be explored. An important aspect is to evaluate the selected Lactobacilli in vitro and find their probiotic potential. Some in vitro probiotic performance evaluations of the strains include survival under stress (low pH, high bile salt, high osmotic pressure, high oxygen, oxidation, starvation, etc.), adhesion ability, and antibacterial, antioxidation, cholesterol-lowering, and anticancer activities (Table 2).
As probiotics, Lactobacilli colonizing the intestine to reach 1 × 106 CFU is necessary for its probiotic effect [113]. Lactobacilli can survive in the robust acid environment in the gastric juice and high bile salt concentration in the small intestine, which are two criteria for screening good probiotic Lactobacilli strains. The acid and bile salt tolerance of Lactobacilli strains use the rate of viable bacteria incubated in various acid pH environments as an indicator in in vitro assays. Additionally, many studies conducted artificially simulated gastric juice tolerance and animal model tests of probiotic Lactobacilli [23,72,76]. The survival rate was used as an index to evaluate probiotic Lactobacilli’s acid and bile salt tolerance.
Adhesion is another of the essential characteristics of probiotic bacteria that contributes to the colonization of probiotics in the gastrointestinal tract [67]. The ability of the bacteria to stick with hydrocarbons determines the extent of adhesion to the epithelial cells in the gastrointestinal tract, known as cell surface hydrophobicity [63]. The direct method of cell surface hydrophobicity of bacteria is to determine the change of absorbance of the supernatant of bacterial cell solution at 600 nm after treatment with hydrocarbons such as n-hexadecane and toluene. More precisely, the adhesion of Lactobacilli strains to mucin has also been determined [65]. Moreover, commercial kits for determining these mucins have been reported and can be used for high-throughput screening [66].
Intestinal epithelial cell (IEC) lines are often presumed to better represent conditions in the tissues of the GIT. Several studies have been conducted using human epithelial cell lines (such as HT-29, HT-29MTX, and Caco-2) to screen the adhesion of probiotic strains [67]. Additionally, other studies have focused on the self-aggregation of probiotics [18], which is also related to adhesion.
Lactobacilli strains can secrete lactic acid and other organic acids, lowering the environment’s pH and thereby inhibiting other microorganisms’ growth [70]. Additionally, Lactobacilli strains produce medicinal probiotic metabolites and bacteriocin BACs, often used as biological preservatives in the food industry, arousing people’s attention [114]. These metabolites have antagonistic activity against bacteria genetically similar to producing bacteria, which are immune to their own BACs. BACs have also been considered biologically active molecules with potential activities for human health, such as use as antiviral and anticancer drugs. BACs are extracellular antimicrobial peptides synthesized by ribosomes. They have extensive antibacterial activity and are a safe alternative to antibiotics. As a result, the shelf life of naturally fermented foods has increased. Therefore, screening high-yield BAC probiotic Lactobacilli strains from naturally fermented food should be an option. Researchers have also studied the production and characterization of BACs by different probiotics [12]. Additionally, the combined culture of different probiotics may produce new antibacterial products [115,116].
Some reports show that Lactobacilli strains have antioxidant activity and can be used as antioxidants in food, stabilizing food’s color, flavor, and taste [80]. Additionally, Lactobacilli strains can reduce the oxidative stress injury of Caco-2 cells and improve the antioxidant capacity under oxidative stress. Firstly, the tolerance of Lactobacilli strains to hydrogen peroxide was studied [79]. The antioxidant capacity of Lactobacilli strains was evaluated by measuring the hydroxyl radical scavenging capacity of cell-free extracts of these strains. These strains can produce metabolites such as superoxide dismutase, glutathione, and extracellular polysaccharide to inhibit oxidation.

2.4. Fermentation Performance of Lactobacilli Strains

As a lactic acid starter, Lactobacilli strains should be tolerant to harsh conditions, such as temperature changes, osmotic pressure (high fat and protein concentration in milk and meat and high salt in kimchi), and lactic acid accumulation. These characteristics can ensure the rapid adaptation and growth of microorganisms to bring good physical properties and taste to the products. Due to different food components, some of them are used for specific food manufacturing, such as yogurt and cheese (L. delbrueckii), fermented vegetables (L. plantarum and L. pentosus), and fermented meat (L. pentosus).
The diversity of lactic acid food produced by Lactobacilli strains requires that the fermentation characteristics of these strains are different [117]. For example, Lactobacilli strains used in meat processing should be able to improve the flavor of end products without producing biogenic amines, because these compounds are produced by the deacidification of free amino acids and have toxic effects on human intestines. Studies have revealed that Lactobacilli strains with protein hydrolytic activity [73], which belong to homogeneous fermentation, can significantly reduce the biogenic amines of fermented sausage. The production of bacteriocin by Lactobacilli strains, for example, is another feature of evaluating the development of meat products by Lactobacilli strains. It can inhibit the growth of pathogenic bacteria and increase the shelf life of products. As mentioned above, the antibacterial activity of Lactobacilli strains was screened to resist various pathogens evaluating the production of nisin against Listeria monocytogenes, Clostridium perfringens, Bacillus cereus, and Staphylococcus aureus [51,69,70,71].
Lactobacilli strains produce large amounts of lactic acid, a non-volatile, odorless compound that contributes to the aroma of the product [118]. Therefore, the production of another fermentation performance flavor molecule was evaluated by gas chromatography–mass spectrometry. The main aroma components include aldehydes, organic acids, higher alcohols, esters, carboxylic acids, and ketones [119,120]. Lactobacilli strains convert precursor molecules into aromatic compounds by secreting various extracellular enzymes [73,74,75]. In a protein-rich environment, proteolytic enzymes play a major role in forming aromatic molecules from the amino acids released by complex proteins. For example, milk is rich in casein, and Lactobacilli strains used in yogurt and cheese convert these precursor molecules into flavor substances. Lipid degradation also plays a vital role in the aroma formation of fermented meat and dairy products [121].

2.5. Health Functions of Lactobacilli Strains

From the functional characteristics, the existence of Lactobacilli strains in the intestinal microbiota is related to the host’s health status [122,123]. Clinical research showed that the reduction or increase in the proportion of this genus in the human body would produce health problems, such as irritable bowel syndrome [124], human immunodeficiency virus [125], obesity [126], type 2 diabetes [127], etc. Several strains with probiotic properties of specific metabolites have been successfully included in various functional foods (Figure 3). Although the mechanism of their role is uncertain, the health-promoting effect of these strains is related to the strains themselves and their active metabolic substances (such as extracellular polysaccharide, bacteriocin, polypeptide, short-chain fatty acid, etc.). Several meta-analyses and systematic reviews [128,129,130,131,132,133] support the health effects of probiotic Lactobacilli strains and specific metabolites produced by Lactobacilli strains in treatment cases, including acute rotavirus diarrhea in children, antibiotic-related diarrhea in children, Helicobacter pylori infection, allergic rhinitis, high blood pressure, hyperlipidemia, and other diseases. Additionally, based on its excellent physiological characteristics and probiotic function, Lactobacillus has become an important direction in the field of probiotic function. Given the specificity of the Lactobacilli strains, the new strains may have potential unique functional characteristics. Table 4 summarizes several vital functions of Lactobacilli strains.

2.6. Performance Development and Improvement of Lactobacilli Strains

As mentioned above, numerous species of Lactobacilli are used in food production (Table 1), including improving traditional food and developing new products. On the one hand, Lactobacilli strains can enhance the quality of fermented food and, on the other hand, prolong the storage period of food as a preservative. Therefore, the excellent characteristics of Lactobacilli strains are the key to their application in the food industry. However, Lactobacilli strains have specificity themselves. Different strains of the same species show significant differences; therefore, new characteristics can be found [138]. Thus, scholars are committed to screening new Lactobacilli strains.
On the one hand, the growth of naturally screened Lactobacilli strains is limited by physical and chemical factors, such as pH [139,140], oxygen [141,142], osmotic stress [143], temperature [140], carbohydrate substrates[144], and other factors [145,146]. On the other hand, the yields of beneficial metabolites of naturally screened Lactobacilli strains, such as lactic acid [147], γ-aminobutyric acid [148,149], extracellular polysaccharide [149], and bacteriocin [19] are relatively low and cannot meet the requirements of industrial production. Therefore, reasonable breeding strategies are used to improve the performance of Lactobacilli strains with potential application in the food industry.
One method is mutagenesis breeding. Mutation breeding of Lactobacilli strains can change the genetic structure and function of Lactobacilli strains, and then screen mutants to obtain the required high-yield and high-quality strains [150]. It is the most basic modern breeding method. The breeding speed is fast, the cost is low, the time is short, and the method is simple, mainly including physical, chemical, and biological mutagens. Chemical mutagenesis primarily uses nitrosoguanidine, diethyl sulfate, and other chemicals. These chemicals are harmful to the human body. Thus, they are not widely used in the food industry. A limited number of studies focused on the biological mutagenesis of Lactobacilli strains, mainly involving transposon mutations [151,152]. Physical mutagenesis of Lactobacilli strains commonly uses ultraviolet [153] or microwave radiation [154]. Given the possible tolerance of traditional radiation technology of Lactobacilli strains, new mutation technologies, such as heavy ion beam irradiation and plasma mutation breeding, have recently appeared [155,156,157]. The operation of traditional mutation breeding is simple, and the experimental conditions are not high; the mutation is random, and the workload is enormous despite the introduction of high-throughput screening technology in the mutation process [155,158,159].
Another method is metabolic engineering, a continuation, and upgrade of gene engineering technology. This method can directionally change the functional characteristics of Lactobacilli strains and compensate for the shortcomings of classical mutagenesis screening [160,161,162,163,164]. The metabolic strategies of Lactobacilli mainly focus on the changes in pyruvate metabolism to produce essential fermentation end products, such as sweeteners, spices, aromatic compounds, and complex biosynthetic pathways, leading to the production of extracellular polysaccharides and vitamins [165]. Currently, the most commonly used methods for metabolic engineering of Lactobacilli include whole-genome amplification [166], genome shuffling [167,168], and genome editing (plasmid-based homologous recombination, Red/RecET-mediated double-stranded DNA recombination, and single-stranded DNA recombination) [169,170]. However, the safety of these methods for metabolic engineering to change the metabolic characteristics of Lactobacilli is worth considering and is not accepted by the European Union [171].

2.7. Role of Lactobacilli Strains in Food Production

The primary role of Lactobacilli strains in dairy processing (such as yogurt, dahi, kefir, koumiss, and cheese) is not only to improve the nutritional value but also to produce lactic acid, butyric acid, a variety of amino acids, and vitamins and other metabolites, resulting in a unique food flavor. Additionally, these strains use dairy products as a carrier to promote human health due to their probiotic effect [172]. The application of Lactobacilli strains to meat products can improve the appearance of meat products, promote the improvement of taste, inhibit the growth of spoilage bacteria, reduce the generation of nitrite and greatly improve the overall quality of meat products [27].
In turn, fermented foods as a carrier play a role in transporting and storing these excellent strains. On the one hand, these strains were found in traditional fermented foods, which characterized their excellent properties. On the other hand, these strains were intensively inoculated into conventional fermented food to improve product control. Fermented fruits can be produced by natural fermentation of the surface flora spontaneously formed (such as Lactobacilli and Pediococcus spp.) or inoculated with fermentation starter (such as L. plantarum, L. rhamnosus, and L. acidophilus). Food nutritionists are developing a new generation of fermented fruit products with special biological and unique sensory characteristics [5,173,174]. Fermented vegetable products can positively impact human health because they are rich in substances beneficial to human beings (such as dietary fiber, minerals, antioxidants, and vitamins). The principle is to use Lactobacilli strains attached to vegetables and several artificially selected excellent strains to carry out a series of microbial fermentations and finally obtain the finished pickle. The Lactobacilli contained in kimchi can promote human gastrointestinal peristalsis, reduce fat, and enhance immunity [10,120].
The fermentation of probiotic strains with excellent performance has attracted people’s attention. The screened new strains are often used in the development of new products. In recent years, several Lactobacilli strains have been widely used in various functional foods due to their unique physiological efficacy and flavor, such as active Lactobacilli drinks and solid drinks [119,175]. With the deepening of relevant research, Lactobacilli will be used in human health conditioning treatments as a probiotic functional food to a greater extent, and the application direction will be more extensive.
Lactobacilli can also be applied to preserving food, such as meat, fruit, vegetables, seafood, etc. These Lactobacilli strains are used as biological preservatives due to the following manifestations: (1) produce organic acids, such as lactic acid and acetic acid, to inhibit the growth and reproduction of most spoilage bacteria; (2) H2O2 production activates the catalase thiocyanate system in milk; (3) produce small proteins or peptides similar to bacteriocin, etc., [176,177].

3. High-Density Fermentation

High-density cell culture of Lactobacilli strains is a critical step in producing direct vast set starters and a key challenge at the industrial scale. The application of various emerging culture technologies with equipment to culture the bacteria can significantly increase the density of the bacteria compared with traditional culture, thereby increasing the specific productivity of the target product and offering a fermentation process that obtains more bacteria at a lower cost.
The high-density fermentation methods of the Lactobacilli strain mainly involve optimizing medium composition and culture conditions [178]. The current high-density fermentation methods for increasing the concentration of bacteria have certain advantages and disadvantages (Table 5).
Table 5. The current high-density-culture methods showing advantages and disadvantages.
Table 5. The current high-density-culture methods showing advantages and disadvantages.
Current High-Density-Culture Methods AdvantageDisadvantages
Buffer salt culture [179]Add a buffer salt that has no effect on the strain or has a growth-promoting impact on the culture medium to improve the buffering capacity of the fermentation broth and control the stability of pH within a specific range, easy to operate.The buffering capacity of the buffer salt is limited and can only play a role within a specific range.
Chemical neutralization culture [180]Add lye (such as NaOH, ammonia, and CaCO3) to the culture system to control the pH value of the fermentation system, easy to operate.With the continuous addition of lye and the accumulation of metabolites, too high salt concentration will inhibit the growth of bacteria.
Dialysis culture [181]Remove part of the small molecular metabolites produced by the bacteria while providing fresh nutrients to the culture solution.A small processing volume, a long dialysis process, and large equipment investment are also not conducive to industrialization.
Fed-batch culture (non-feedback mode and feedback mode) [182,183,184,185]Effectively eliminates substrate inhibition and acid inhibition and is simple to operateInadequate utilization of nutrients; Limited by container volume
Cross-flow culture [186]Due to cross-flow filtration, the high viscosity produced by cells is reduced, which is conducive to cell recovery and high concentrationHigh equipment cost; Requires more professional operators; It is easy to block the membrane module.
Circulating culture (sedimentation, centrifugation, and membrane filtration) [187,188]Through technologies such as sedimentation, centrifugation, and membrane filtration, the cells are intercepted, the culture medium flows out, and then a certain amount of fresh culture medium is added to obtain high-density cells. It shortens the production time and saves a lot of power, workforce, water, and steamIn the circulation process, the strains quickly degenerate and are polluted, resulting in economic losses; The utilization rate of nutrients was lower than that of batch culture.
Recent advances in high-density fermentation methods showed that the fed-batch culture strategies, including non-feedback modes, such as intermittent feeding, constant feeding, exponential feeding, and feedback mode, are the most widely used for the high-density growth of Lactobacilli strains (Table 5). The combination of high-density cell culture methods is the current and future development trend for studying Lactobacillus.

4. Production of DVS Starter of Lactobacilli

Fermented food has experienced the methods of natural fermentation, inoculation fermentation, and pure culture fermentation, that is, a directed vat set (DVS) starter developed today [189]. A DVS starter has a stronger fermentation activity and higher viable bacteria (>1 × 1011 CFU/g). This starter can be directly inoculated during the production of lactic acid fermented food, has no intermediate subculture process, is easy to use, and is a new commercial production strain with a stable product quality [189]. The DVS starters of Lactobacilli are dry powders made from the selection of excellent strains, proliferation, and culture in a liquid medium, concentration, and separation, combined with a biological protective agent and drying (Figure 4). Fermented foods often require more than one strain. For example, yogurt usually includes Streptococcus thermophilus and L. bulgaricus. Additionally, adding other strains enhances fermentation performance and potential probiotic properties. Therefore, many researchers have studied the compound DVS starters for manufacturing various foods (e.g., yogurt, cheese, fermented meats, and vegetables) and probiotic products.
There are still deficiencies in production technologies for DVS starters, manifested in four aspects: strain breeding, optimization of culture medium, high-density culture, and cell drying. Drying is the last and most critical step in preparing DVS starters with high activity and stability. The most common methods for cell drying of Lactobacilli strains are freeze and spray drying [190]. Freeze-drying technology is the best choice for producing bacterial powder because microorganisms are sensitive to heat. The process utilizes the phase change of water when it is lower than the triple point, freezing the free water in the culture solution and sublimating the ice crystals into water vapor under a vacuum [190]. After the freeze-dried cells reach low temperature, dryness, vacuum, and other conditions, their metabolic activities stop, and they are in a dormant state and can be stored for a long time. During the freeze-drying process, the bacterial cells will be damaged due to freezing and drying. Therefore, the appropriate Lactobacilli species must be selected. The study showed that the freeze-dried survival rate of Lactobacilli is lower than that of Lactococcus, and this can be observed at the end of logarithmic growth and at the same time [190]. Additionally, the culture medium, freezing temperature, and cell membrane composition affect the freeze drying of bacteria [191]. Even if low product temperatures are applied, varying degrees of viability recovery have been reported for freeze-dried bacteria.
Another drying method is spray drying. Spray drying is high-speed centrifugation or high-pressure method, spraying the starter containing the bacteria into extremely fine droplets in a dry heat medium, allowing the water to evaporate quickly, thereby obtaining a dry bacterial sample [192]. Compared with freeze-drying and freezing methods, spray-drying technology has the advantages of simple equipment, low cost, and suitability for large-scale production, transportation, and storage of probiotics. However, this method will cause specific damage to bacteria and affect their survival rate due to their exposure to dry heat and rapid dehydration. Some effective heat protection agents such as lactose, trehalose, whey protein isolate, and skimmed milk [8,193,194] are selected to protect Lactobacilli cells during the spray drying process.
Recently, some emerging drying technologies for the production of DVS starters of Lactobacilli, such as low-temperature spray-drying technology [195], freezing granulation drying technology [196], and spray freeze-drying [197], have been developed, which contribute to solving the problem of low survival rate.
Producing probiotics with high activity is the key to ensuring product quality and enhancing market competitiveness. In the traditional batch process, each operation unit is discontinuous, increasing the risk of bacteria exposure. Therefore, the development trend of the probiotic industry involves the development of production equipment for automatic continuous cell culture, separation, drying, and other operating units to realize continuous production. Combined with the advantages of freeze and spray drying, the equipment and technology for developing low-temperature spray drying, freezing granulation drying, and spray freeze-drying have potential for large-scale application [189,190,196,197]. The preparation process of cryogenic bacteria is also one of the essential directions for preparing highly active probiotics [198]. The development of packaging machines and related supporting equipment under low temperatures is a problem that needs further solving in producing cryogenic bacteria in China.

5. Conclusions and Outlook

A large number of published studies showed that bacteria belonging to Lactobacilli could spontaneously form a microbial group in fermented food, such as in food processing micro-factories, continuously transporting substances beneficial to human health. With the advancement of modern molecular biotechnology, various Lactobacilli species in fermented food have been identified, several complete genomes have been obtained, and the probiotic mechanism of Lactobacilli species has been revealed. The separation, identification, and characterization of Lactobacilli species from various foods showed differences in fermentation performance, acid production, acid tolerance, bile resistance, antimicrobial activity, cholesterol-lowering ability, and antibiotic resistance. Highly active Lactobacilli species preparations can be obtained by high-density cultivation and used as DVS starters to realize the industrialization of fermented foods.
Screening out excellent Lactobacilli strains from various food resources has always been an important research topic. At present, diverse Lactobacilli species are used in the food industry. Foods fermented by Lactobacilli species not only improve the original flavor of the food but also increase the food’s nutritional value, which has a good health effect on the human body. In the future, we can use the advantages and characteristics of Lactobacilli species fermentation, conduct an in-depth study of the health functions of Lactobacilli species and protection technology of high-efficiency live bacteria protection technology, and develop new products with health-care functions. In addition, with the increase in the maturity of multi-omics technology, the application of multi-omics technology in Lactobacilli species is increasingly being favored and valued. The comprehensive applications of genomics, transcriptomics, proteomics, and metabonomics can reveal the genetic information of Lactobacilli species, analyze physiological and metabolic mechanisms of Lactobacilli, penetrate the adaptation mechanism of Lactobacilli species to physiological and environmental changes, and explore the molecular mechanism of beneficial functions of Lactobacilli species. All of these will promote the rapid development of the Lactobacillus fermenting food industry.

Author Contributions

Y.L., S.X., and Y.D.: conceptualization, original draft, writing—review, and editing; X.W.: review; X.Z.: writing—review and editing; C.L. and L.H.: conceptualization, original draft, writing—review and editing, supervision and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Key Agricultural Project of Guizhou Province (QKHZC-[2021]YB278, QKHZC-[2021]YB184, QKHZC-[2021]YB142, QKHZC-[2019]2382, and QKHZC-[2016]2580), the High-Level Innovative Talents Training Project of Guizhou Province (QKHPTRC-GCC[2022]026-1), and the Natural Science Foundation of China (31870002 and 31660010), Qiankehe talents project ([2018]5781 and [2017]5788-11), and Guizhou Province Science and Technology Project, grant number [2019]1078.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors have no competing interests to disclose.

References

  1. Iseppi, R.; Zurlini, C.; Cigognini, I.M.; Cannavacciuolo, M.; Sabia, C.; Messi, P. Eco-friendly edible packaging systems based on live-Lactobacillus kefiri MM5 for the control of Listeria monocytogenes in fresh vegetables. Foods 2022, 11, 2632. [Google Scholar] [CrossRef] [PubMed]
  2. Xie, Y.; Wang, Y.; Han, Y.; Zhang, J.; Wang, S.; Lu, S.; Wang, H.; Lu, F.; Jia, L. Complete genome sequence of a novel Lactobacillus paracasei TK1501 and its application in the biosynthesis of isoflavone aglycones. Foods 2022, 11, 2807. [Google Scholar] [CrossRef] [PubMed]
  3. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; Toole, P.W.O.; 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. Micr. 2020, 70, 2782–2858. [Google Scholar] [CrossRef] [PubMed]
  4. Reid, G. Probiotics: Definition, scope and mechanisms of action. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 17–25. [Google Scholar] [CrossRef]
  5. Brizuela, N.; Tymczyszyn, E.E.; Semorile, L.C.; Valdes La Hens, D.; Delfederico, L.; Hollmann, A.; Bravo-Ferrada, B. Lactobacillus plantarum as a malolactic starter culture in winemaking: A new (old) player? Electron. J. Biotechn. 2019, 38, 10–18. [Google Scholar] [CrossRef]
  6. Shori, A.B.; Aljohani, G.S.; Al-zahrani, A.J.; Al-sulbi, O.S.; Baba, A.S. Viability of probiotics and antioxidant activity of cashew milk-based yogurt fermented with selected strains of probiotic Lactobacillus spp. LWT 2022, 153, 112482. [Google Scholar] [CrossRef]
  7. Yang, W.; Hao, X.; Zhang, X.; Zhang, G.; Li, X.; Liu, L.; Sun, Y.; Pan, Y. Identification of antioxidant peptides from cheddar cheese made with Lactobacillus helveticus. LWT 2021, 141, 110866. [Google Scholar] [CrossRef]
  8. Ilha, E.C.; Da Silva, T.; Lorenz, J.G.; de Oliveira Rocha, G.; Sant Anna, E.S. Lactobacillus paracasei isolated from grape sourdough: Acid, bile, salt, and heat tolerance after spray drying with skim milk and cheese whey. Eur. Food Res. Technol. 2015, 240, 977–984. [Google Scholar] [CrossRef]
  9. Sun, F.; Kong, B.; Chen, Q.; Han, Q.; Diao, X. N-nitrosoamine inhibition and quality preservation of Harbin dry sausages by inoculated with Lactobacillus pentosus, Lactobacillus curvatus and Lactobacillus sake. Food Control 2017, 73, 1514–1521. [Google Scholar] [CrossRef]
  10. Zhou, M.; Zheng, X.; Zhu, H.; Li, L.; Zhang, L.; Liu, M.; Liu, Z.; Peng, M.; Wang, C.; Li, Q.; et al. Effect of Lactobacillus plantarum enriched with organic/inorganic selenium on the quality and microbial communities of fermented pickles. Food Chem. 2021, 365, 130495. [Google Scholar] [CrossRef]
  11. Guantario, B.; Zinno, P.; Schifano, E.; Roselli, M.; Perozz, G.; Uccelletti, C.P.D.; Devirgiliis, C. In vitro and in vivo selection of potentially probiotic Lactobacilli from Nocellara del Belice table olives. Front. Microbiol. 2018, 9, 595. [Google Scholar] [CrossRef] [Green Version]
  12. Song, J.; Peng, S.; Yang, J.; Zhou, F.; Suo, H. Isolation and identification of novel antibacterial peptides produced by Lactobacillus fermentum SHY10 in Chinese pickles. Food Chem. 2021, 348, 129097. [Google Scholar] [CrossRef] [PubMed]
  13. He, Z.; Zheng, J.; He, L.; Li, C.; Hu, P.; Tao, H.; Wang, X. Evaluation of the effect of essential oil addition on the quality parameters and predicted shelf life of potato yogurt. J. Food Protect. 2021, 84, 1069–1079. [Google Scholar] [CrossRef] [PubMed]
  14. Shori, A.B.; Rashid, F.; Baba, A.S. Effect of the addition of phytomix-3+ mangosteen on antioxidant activity, viability of lactic acid bacteria, type 2 diabetes key-enzymes, and sensory evaluation of yogurt. LWT 2018, 94, 33–39. [Google Scholar] [CrossRef]
  15. Divyashree, S.; Anjali, P.G.; Somashekaraiah, R.; Sreenivasa, M.Y. Probiotic properties of Lactobacillus casei—MYSRD 108 and Lactobacillus plantarum-MYSRD 71 with potential antimicrobial activity against Salmonella paratyphi. Biotechnol. Rep. 2021, 32, e00672. [Google Scholar] [CrossRef]
  16. Xu, J.; Guo, L.; Zhao, N.; Meng, X.; Zhang, J.; Wang, T.; Wei, X.; Fan, M. Response mechanisms to acid stress of acid-resistant bacteria and biotechnological applications in the food industry. Crit. Rev. Biotechnol. 2022, 42, 1–17. [Google Scholar] [CrossRef]
  17. García-Gamboa, R.G.M.O. Assessment of intermediate and long chains agave fructan fermentation on the growth of intestinal bacteria cultured in a gastrointestinal tract simulator. Rev. Mex. Ing. Quim. 2019, 19, 827–838. [Google Scholar] [CrossRef] [Green Version]
  18. Tuo, Y.; Yu, H.; Ai, L.; Wu, Z.; Guo, B.; Chen, W. Aggregation and adhesion properties of 22 Lactobacillus strains. J. Dairy Sci. 2013, 96, 4252–4257. [Google Scholar] [CrossRef] [Green Version]
  19. Gaspar, C.; Donders, G.G.; de Oliveira, R.P.; Queiroz, J.A.; Tomaz, C.; de Oliveira, J.M.; de Oliveira, A.P. Bacteriocin production of the probiotic Lactobacillus acidophilus KS400. AMB Express 2018, 8, 153. [Google Scholar] [CrossRef]
  20. Toropov, V.; Demyanova, E.; Shalaeva, O.; Sitkin, S.; Vakhitov, T. Whole-genome sequencing of Lactobacillus helveticus D75 and D76 confirms safety and probiotic potential. Microorganisms 2020, 8, 329. [Google Scholar] [CrossRef]
  21. Meireles Mafaldo, Í.; de Medeiros, V.P.B.; Da Costa, W.K.A.; Da Costa Sassi, C.F.; Da Costa Lima, M.; de Souza, E.L.; Eduardo Barão, C.; Colombo Pimentel, T.; Magnani, M. Survival during long-term storage, membrane integrity, and ultrastructural aspects of Lactobacillus acidophilus 05 and Lacticaseibacillus casei 01 freeze-dried with freshwater microalgae biomasses. Food Res. Int. 2022, 159, 111620. [Google Scholar] [CrossRef] [PubMed]
  22. Whittington, H.D.; Dagher, S.F.; Bruno-Bárcena, J.M. Production and conservation of starter cultures: From “backslopping” to controlled fermentations. In How Fermented Foods Feed a Healthy Gut Microbiota: A Nutrition Continuum; Azcarate-Peril, M.A., Arnold, R.R., Bruno-Bárcena, J.M., Eds.; Springer International Publishing: Cham, Switzerland, 2019; pp. 125–138. [Google Scholar] [CrossRef]
  23. Reque, P.M.; Brandelli, A. Encapsulation of probiotics and nutraceuticals: Applications in functional food industry. Trends Food Sci. Tech. 2021, 114, 1–10. [Google Scholar] [CrossRef]
  24. Zeng, W.; Guo, L.; Xu, S.; Chen, J.; Zhou, J. High-throughput screening technology in industrial biotechnology. Trends Biotechnol. 2020, 38, 888–906. [Google Scholar] [CrossRef] [PubMed]
  25. Tanasupawat, S.; Shida, O.; Okada, S.; Komagata, K. Lactobacillus acidipiscis sp. Nov. and Weissella thailandensis sp. Nov., isolated from fermented fish in Thailand. Int. J. Syst. Evol. Micr. 2000, 50 Pt 4, 1479. [Google Scholar] [CrossRef]
  26. Aarti, C.; Khusro, A.; Varghese, R.; Arasu, M.V.; Agastian, P.; Al-Dhabi, N.A.; Ilavenil, S.; Choi, K.C. In vitro studies on probiotic and antioxidant properties of Lactobacillus brevis strain LAP2 isolated from Hentak, a fermented fish product of North-East India. LWT 2017, 86, 438–446. [Google Scholar] [CrossRef]
  27. Liu, J.; Lin, C.; Zhang, W.; Yang, Q.; Meng, J.; He, L.; Deng, L.; Zeng, X. Exploring the bacterial community for starters in traditional high-salt fermented Chinese fish (Suanyu). Food Chem. 2021, 358, 129863. [Google Scholar] [CrossRef] [PubMed]
  28. Kittibunchakul, S.; Yuthaworawit, N.; Whanmek, K.; Suttisansanee, U.; Santivarangkna, C. Health beneficial properties of a novel plant-based probiotic drink produced by fermentation of brown rice milk with GABA-producing Lactobacillus pentosus isolated from Thai pickled weed. J. Funct. Foods 2021, 86, 104710. [Google Scholar] [CrossRef]
  29. Luz, C.; Calpe, J.; Manuel Quiles, J.; Torrijos, R.; Vento, M.; Gormaz, M.; Mañes, J.; Meca, G. Probiotic characterization of Lactobacillus strains isolated from breast milk and employment for the elaboration of a fermented milk product. J. Funct. Foods 2021, 84, 104599. [Google Scholar] [CrossRef]
  30. Hameed, A.M.; Elkhtab, E.; Mostafa, M.S.; Refaey, M.M.M.; Hassan, M.A.A.; Abo El-Naga, M.Y.; Hegazy, A.A.; Rabie, M.M.; Alrefaei, A.F.; Abdulaziz Alfi, A.; et al. Amino acids, solubility, bulk density and water holding capacity of novel freeze-dried cow’s skimmed milk fermented with potential probiotic Lactobacillus plantarum Bu-Eg5 and Lactobacillus rhamnosus Bu-Eg6. Arab J. Chem. 2021, 14, 103291. [Google Scholar] [CrossRef]
  31. Ong, L.; Shah, N.P. Release and identification of angiotensin-converting enzyme-inhibitory peptides as influenced by ripening temperatures and probiotic adjuncts in Cheddar cheeses. LWT 2008, 41, 1555–1566. [Google Scholar] [CrossRef]
  32. Tang, W.; Li, C.; He, Z.; Pan, F.; Pan, S.; Wang, Y. Probiotic properties and cellular antioxidant activity of Lactobacillus plantarum MA2 isolated from Tibetan kefir grains. Probiotics Antimicrob. Proteins 2018, 10, 523–533. [Google Scholar] [CrossRef] [PubMed]
  33. Jeong, D.; Kim, D.; Kang, I.; Kim, H.; Song, K.; Kim, H.; Seo, K. Characterization and antibacterial activity of a novel exopolysaccharide produced by Lactobacillus kefiranofaciens DN1 isolated from kefir. Food Control 2017, 78, 436–442. [Google Scholar] [CrossRef]
  34. Talib, N.; Mohamad, N.E.; Yeap, S.K.; Hussin, Y.; Aziz, M.N.M.; Masarudin, M.J.; Sharifuddin, S.A.; Hui, Y.W.; Ho, C.L.; Alitheen, N.B. Isolation and characterization of Lactobacillus spp. from kefir samples in Malaysi. Molecules 2019, 24, 2606. [Google Scholar] [CrossRef] [Green Version]
  35. Fei, Y.; Liu, L.; Liu, D.; Chen, L.; Tan, B.; Fu, L.; Li, L. Investigation on the safety of Lactobacillus amylolyticus L6 and its fermentation properties of tofu whey. LWT 2017, 84, 314–322. [Google Scholar] [CrossRef]
  36. He, Y. The Isolation and Identification of Lactic Acid Bacteria from Naturally Fermented Tofu Whey and the Application of the Strains in the Acidic Whey Tofu. Master’s Thesis, Jiangnan University, Wuxi, China, 2019. [Google Scholar]
  37. Aljuobori, A.; Abdullah, N.; Zulkifli, I.; Soleimani, A.F.; Liang, J.B.; Oskoueian, E. Lactobacillus salivarius fermentation reduced glucosinolate and fibre in canola meal. J. Food Res. 2014, 3, 95–102. [Google Scholar] [CrossRef]
  38. Li, M.; Xu, Y.; Zhang, J. Effect of lactic acid bacteria fermentation on the quality of starchy foods. China Brew. 2020, 39, 13–18. [Google Scholar] [CrossRef]
  39. Han, M.; Wang, X.; Zhang, M.; Ren, Y.; Yue, T.; Gao, Z. Effect of mixed Lactobacillus on the physicochemical properties of cloudy apple juice with the addition of polyphenols-concentrated solution. Food Biosci. 2021, 41, 101049. [Google Scholar] [CrossRef]
  40. Yang, X.; Zhou, J.; Fan, L.; Qin, Z.; Chen, Q.; Zhao, L. Antioxidant properties of a vegetable–fruit beverage fermented with two Lactobacillus plantarum strains. Food Sci. Biotechnol. 2018, 27, 1719–1726. [Google Scholar] [CrossRef]
  41. Jans, C.; Lagler, S.; Lacroix, C.; Meile, L.; Stevens, M.J.A. Complete genome sequences of Lactobacillus curvatus KG6, L. Curvatus MRS6, and Lactobacillus sakei FAM18311, isolated from fermented meat products. Genome Announc. 2017, 5, e00915-17. [Google Scholar] [CrossRef]
  42. Chaillou, S.P.; Monique, M.C.S.; Dudez, A.; Martin, V.R.; Beaufils, S.; Re, E.D.; Bossy, R.; Loux, V.; Zagorec, M. The complete genome sequence of the meat-borne lactic acid bacterium Lactobacillus sakei 23K. Nat. Biotechnol. 2005, 23, 1527–1533. [Google Scholar] [CrossRef] [Green Version]
  43. Letunic, I.; Bork, P. Interactive Tree of Life (iTOL) v5: An online tool for phylogenetic tree display and annotation. Nucleic Acids Res. 2021, 49, W293–W296. [Google Scholar] [CrossRef]
  44. Briges, M. The classification of Lactobacilli by means of physiological tests. J. Gen. Microbiol. 1953, 9, 234–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Garcia, E.F.; Luciano, W.A.; Xavier, D.E.; Da Costa, W.C.A.; de Sousa Oliveira, K.; Franco, O.L.; de Morais Júnior, M.A.; Lucena, B.T.L.; Picão, R.C.; Magnani, M.; et al. Identification of lactic acid bacteria in fruit pulp processing byproducts and potential probiotic properties of selected Lactobacillus strains. Front. Microbiol. 2016, 7, 1371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Riaz Rajoka, M.S.; Mehwish, H.M.; Siddiq, M.; Haobin, Z.; Zhu, J.; Yan, L.; Shao, D.; Xu, X.; Shi, J. Identification, characterization, and probiotic potential of Lactobacillus rhamnosus isolated from human milk. LWT 2017, 84, 271–280. [Google Scholar] [CrossRef]
  47. Karami, S.; Roayaei, M.; Hamzavi, H.; Bahmani, M.; Hassanzad-Azar, H.; Leila, M.; Rafieian-Kopaei, M. Isolation and identification of probiotic from local dairy and evaluating their antagonistic effect on pathogens. Int. J. Pharm. Investig. 2017, 7, 137–141. [Google Scholar] [CrossRef]
  48. Anisimova, E.A.; Yarullina, D.R. Antibiotic resistance of Lactobacillus strains. Curr. Microbiol. 2019, 76, 1407–1416. [Google Scholar] [CrossRef]
  49. Saroj, S.D.; Maudsdotter, L.; Tavares, R.; Jonsson, A.-B. Lactobacilli interfere with Streptococcus pyogenes hemolytic activity and adherence to host epithelial cells. Front. Microbiol. 2016, 7, 1176. [Google Scholar] [CrossRef] [Green Version]
  50. Alamdary, S.Z.; Bakhshi, B. Lactobacillus acidophilus attenuates toxin production by Vibrio cholerae and shigella dysenteriae following intestinal epithelial cells infection. Microb. Pathog. 2020, 149, 104543. [Google Scholar] [CrossRef]
  51. Boricha, A.A.; Shekh, S.L.; Pithv, S.P.; Ambalam, P.S.; Vyas, B.R.M. In vitro evaluation of probiotic properties of Lactobacillus species of food and human origin. LWT 2019, 106, 201–208. [Google Scholar] [CrossRef]
  52. Kieliszek, M.; Pobiega, K.; Piwowarek, K.; Kot, A.M. Characteristics of the proteolytic enzymes produced by lactic acid bacteria. Molecules 2021, 26, 1858. [Google Scholar] [CrossRef]
  53. Larsen, N.; de Souza, C.B.; Krych, L.; Kot, W.; Leser, T.D.; Sørensen, O.B.; Blennow, A.; Venema, K.; Jespersen, L. Effect of potato fiber on survival of Lactobacillus species at simulated gastric conditions and composition of the gut microbiota in vitro. Food Res. Int. 2019, 125, 108644. [Google Scholar] [CrossRef] [PubMed]
  54. Larsen, N.; Cahú, T.B.; Isay Saad, S.M.; Blennow, A.; Jespersen, L. The effect of pectins on survival of probiotic Lactobacillus spp. in gastrointestinal juices is related to their structure and physical properties. Food Microbiol. 2018, 74, 11–20. [Google Scholar] [CrossRef]
  55. Andrade, R.; Santos, E.; Azoubel, P.; Ribeiro, E. Increased survival of Lactobacillus rhamnosus ATCC 7469 in guava juices with simulated gastrointestinal conditions during refrigerated storage. Food Biosci. 2019, 32, 100470. [Google Scholar] [CrossRef]
  56. Liao, N.; Luo, B.; Gao, J.; Li, X.; Zhao, Z.; Zhang, Y.; Ni, Y.; Tian, F. Oligosaccharides as co-encapsulating agents: Effect on oral Lactobacillus fermentum survival in a simulated gastrointestinal tract. Biotechnol. Lett. 2019, 41, 263–272. [Google Scholar] [CrossRef] [PubMed]
  57. Nguyen, T.H.; Kim, Y.; Kim, J.; Jeong, Y.; Park, H.M.; Kim, J.W.; Kim, J.; Kim, H.; Paek, N.; Kang, C. Evaluating the cryoprotective encapsulation of the lactic acid bacteria in simulated gastrointestinal conditions. Biotechnol. Bioprocess Eng. 2020, 25, 287–292. [Google Scholar] [CrossRef]
  58. Fijałkowski, K.; Peitler, D.; Rakoczy, R.; Żywicka, A. Survival of probiotic lactic acid bacteria immobilized in different forms of bacterial cellulose in simulated gastric juices and bile salt solution. LWT 2016, 68, 322–328. [Google Scholar] [CrossRef]
  59. Xu, M.; Zhong, F.; Zhu, J. Evaluating metabolic response to light exposure in Lactobacillus species via targeted metabolic profiling. J. Microbiol. Methods 2017, 133, 14–19. [Google Scholar] [CrossRef]
  60. Parlindungan, E.; May, B.; Jones, O. Metabolic insights into the effects of nutrient stress on Lactobacillus plantarum B21. Front. Mol. Biosci. 2019, 6, 75. [Google Scholar] [CrossRef] [Green Version]
  61. Hosseini Nezhad, M.; Hussain, M.A.; Britz, M.L. Stress responses in probiotic Lactobacillus casei. Crit. Rev. Food Sci. 2015, 55, 740–749. [Google Scholar] [CrossRef]
  62. Palomino, M.M.; Waehner, P.M.; Fina Martin, J.; Ojeda, P.; Malone, L.; Sánchez Rivas, C.; Prado Acosta, M.; Allievi, M.C.; Ruzal, S.M. Influence of osmotic stress on the profile and gene expression of surface layer proteins in Lactobacillus acidophilus ATCC 4356. Appl. Microbiol. Biot. 2016, 100, 8475–8484. [Google Scholar] [CrossRef]
  63. Samak, G.; Rao, R.; Rao, R. Lactobacillus casei and epidermal growth factor prevent osmotic stress-induced tight junction disruption in caco-2 cell monolayers. Cells 2021, 10, 3578. [Google Scholar] [CrossRef] [PubMed]
  64. Carasi, P.; Ambrosis, N.; De Antoni, G.; Bressollier, P.; Urdaci, M.; Serradell, M. Adhesion properties of potentially probiotic Lactobacillus kefiri to gastrointestinal mucus. J. Dairy Res. 2014, 81, 16–23. [Google Scholar] [CrossRef] [PubMed]
  65. Klopper, K.B.; Deane, S.M.; Dicks, L.M.T. Aciduric strains of Lactobacillus reuteri and Lactobacillus rhamnosus, isolated from Human Feces, have strong adhesion and aggregation properties. Probiotics Antimicrob. Proteins 2018, 10, 89–97. [Google Scholar] [CrossRef] [PubMed]
  66. Sophatha, B.; Piwat, S.; Teanpaisan, R. Adhesion, anti-adhesion and aggregation properties relating to surface charges of selected Lactobacillus strains: Study in Caco-2 and H357 cells. Arch. Microbiol. 2020, 202, 1349–1357. [Google Scholar] [CrossRef]
  67. Rocha-Mendoza, D.; Kosmerl, E.; Miyagusuku-Cruzado, G.; Giusti, M.M.; Jiménez-Flores, R.; García-Cano, I. Growth of lactic acid bacteria in milk phospholipids enhances their adhesion to Caco-2 cells. J. Dairy Sci. 2020, 103, 7707–7718. [Google Scholar] [CrossRef]
  68. Zavala, L.; Golowczyc, M.A.; Vandamme, P.; Abraham, A.G. Selected Lactobacillus strains isolated from sugary and milk kefir reduce Salmonella infection of epithelial cells in vitro. Benef. Microbes 2016, 7, 585–595. [Google Scholar] [CrossRef]
  69. Chen, C.; Lai, C.; Huang, H.; Huang, W.; Toh, H.; Weng, T.; Chuang, Y.; Lu, Y.; Tang, H.-J. Antimicrobial activity of Lactobacillus species against carbapenem-resistant Enterobacteriaceae. Front. Microbiol. 2019, 10, 789. [Google Scholar] [CrossRef]
  70. de Souza Rodrigues, J.Z.; Passos, M.R.; de Macêdo Neres, N.S.; Almeida, R.S.; Pita, L.S.; Santos, I.A.; Silveira, P.H.S.; Reis, M.M.; Santos, I.P.; de Oliveira Negrão Ricardo, L.; et al. Antimicrobial activity of Lactobacillus fermentum TcUESC01 against Streptococcus mutans UA159. Microb. Pathog. 2020, 142, 104063. [Google Scholar] [CrossRef]
  71. Dubourg, G.; Elsawi, Z.; Raoult, D. Assessment of the in vitro antimicrobial activity of Lactobacillus species for identifying new potential antibiotics. Int. J. Antimicrob. Agents 2015, 46, 590–593. [Google Scholar] [CrossRef]
  72. Sornsenee, P.; Singkhamanan, K.; Sangkhathat, S.; Saengsuwan, P.; Romyasamit, C. Probiotic properties of Lactobacillus species isolated from fermented palm sap in Thailand. Probiotics Antimicrob. Proteins 2021, 13, 957–969. [Google Scholar] [CrossRef]
  73. Aguirre, L.; Hebert, E.M.; Garro, M.S.; de Giori, G.S. Proteolytic activity of Lactobacillus strains on soybean proteins. LWT 2014, 59, 780–785. [Google Scholar] [CrossRef]
  74. Esteban-Torres, M.; Mancheño, J.M.; de Las Rivas, B.; Muñoz, R. Characterization of a halotolerant lipase from the lactic acid bacteria Lactobacillus plantarum useful in food fermentations. LWT 2015, 60, 246–252. [Google Scholar] [CrossRef] [Green Version]
  75. Guo, J.; Xie, Y.; Yu, Z.; Meng, G.; Wu, Z. Effect of Lactobacillus plantarum expressing multifunctional glycoside hydrolases on the characteristics of alfalfa silage. Appl. Microbiol. Biotechnol. 2019, 103, 7983–7995. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, G.; Yu, H.; Feng, X.; Tang, H.; Xiong, Z.; Xia, Y.; Ai, L.; Song, X. Specific bile salt hydrolase genes in Lactobacillus plantarum AR113 and relationship with bile salt resistance. LWT 2021, 145, 111208. [Google Scholar] [CrossRef]
  77. Rai, A.K.; Sanjukta, S.; Jeyaram, K. Production of angiotensin I converting enzyme inhibitory (ACE-I) peptides during milk fermentation and their role in reducing hypertension. Crit. Rev. Food Sci. 2017, 57, 2789–2800. [Google Scholar] [CrossRef]
  78. Moslehishad, M.; Ehsani, M.R.; Salami, M.; Mirdamadi, S.; Ezzatpanah, H.; Naslaji, A.N.; Moosavi-Movahedi, A.A. The comparative assessment of ACE-inhibitory and antioxidant activities of peptide fractions obtained from fermented camel and bovine milk by Lactobacillus rhamnosus PTCC 1637. Int. Dairy J. 2013, 29, 82–87. [Google Scholar] [CrossRef]
  79. Vanessa Moraes Ramalho Castro, M.D.M.S.; Guerra, A.F.; Riger, C.J.; Laureano-Melo, R.; Luchese, R.H. Role of milk and honey in the tolerance of Lactobacilli to oxidative stress. Braz. J. Microbiol. 2021, 52, 883–893. [Google Scholar] [CrossRef]
  80. Zhang, L.; Liu, C.; Li, D.; Zhao, Y.; Zhang, X.; Zeng, X.; Yang, Z.; Li, S. Antioxidant activity of an exopolysaccharide isolated from Lactobacillus plantarum C88. Int. J. Biol. Macromol. 2013, 54, 270–275. [Google Scholar] [CrossRef]
  81. Ahmadi, M.A.; Ebrahimi, M.T.; Mehrabian, S.; Tafvizi, F.; Bahrami, H.; Dameshghian, M. Antimutagenic and anticancer effects of lactic acid bacteria isolated from Tarhana through Ames test and phylogenetic analysis by 16S rDNA. Nutr. Cancer 2014, 66, 1406–1413. [Google Scholar] [CrossRef]
  82. Pepoyan, A.Z.; Balayan, M.H.; Malkhasyan, L.; Manvelyan, A.; Bezhanyan, T.; Paronikyan, R.; Tsaturyan, V.V.; Tatikyan, S.; Kamiya, S.; Chikindas, M.L. Effects of probiotic Lactobacillus acidophilus strain INMIA 9602 Er 317/402 and putative probiotic Lactobacilli on DNA damages in the small intestine of Wistar Rats in vivo. Probiotics Antimicrob. Proteins 2019, 11, 905–909. [Google Scholar] [CrossRef]
  83. Tukenmez, U.; Aktas, B.; Aslim, B.; Yavuz, S. The relationship between the structural characteristics of Lactobacilli-EPS and its ability to induce apoptosis in colon cancer cells in vitro. Sci. Rep. 2019, 9, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Amiri, S.; Rezaei Mokarram, R.; Sowti Khiabani, M.; Rezazadeh Bari, M.; Alizadeh Khaledabad, M. In situ production of conjugated linoleic acid by Bifidobacterium lactis BB12 and Lactobacillus acidophilus LA5 in milk model medium. LWT 2020, 132, 109933. [Google Scholar] [CrossRef]
  85. Ameen, F.A.; Hamdan, A.M.; El-Naggar, M.Y. Assessment of the heavy metal bioremediation efficiency of the novel marine lactic acid bacterium, Lactobacillus plantarum MF042018. Sci. Rep. 2020, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Son, S.; Jeon, H.; Jeon, E.B.; Lee, N.; Park, Y.; Kang, D.; Paik, H. Potential probiotic Lactobacillus plantarum Ln4 from kimchi: Evaluation of β-galactosidase and antioxidant activities. LWT 2017, 85, 181–186. [Google Scholar] [CrossRef]
  87. Chamberlain, C.A.; Hatch, M.; Garrett, T.J. Metabolomic profiling of oxalate-degrading probiotic Lactobacillus acidophilus and Lactobacillus gasseri. PLoS ONE 2019, 14, e0222393. [Google Scholar] [CrossRef] [PubMed]
  88. Li, P.; Gu, Q.; Yang, L.; Yu, Y.; Wang, Y. Characterization of extracellular vitamin B12 producing Lactobacillus plantarum strains and assessment of the probiotic potentials. Food Chem. 2017, 234, 494–501. [Google Scholar] [CrossRef] [PubMed]
  89. Markowiak-Kopeć, P.; Śliżewska, K. The effect of probiotics on the production of short-chain fatty acids by human intestinal microbiome. Nutrients 2020, 12, 1107. [Google Scholar] [CrossRef] [Green Version]
  90. Hamzehlou, P.; Sepahy, A.A.; Mehrabian, S.; Hosseini, F. Production of vitamins B3, B6 and B9 by Lactobacillus Isolated from traditional yogurt samples from 3 cities in Iran, Winter 2016. Appl. Food Biotechnol. 2018, 5, 107–120. [Google Scholar] [CrossRef]
  91. Bourdichon, F.; Laulund, S.; Tenning, P. Inventory of microbial species with a rationale: A comparison of the IDF/EFFCA inventory of microbial food cultures with the EFSA Biohazard Panel qualified presumption of safety. FEMS Microbiol. Lett. 2019, 366, fnz048. [Google Scholar] [CrossRef]
  92. Tsai, C.; Lai, C.; Yu, B.; Tsen, H. Use of PCR primers and probes based on the 23S rRNA and internal transcription spacer (its) gene sequence for the detection and enumerization of Lactobacillus acidophilus and Lactobacillus plantarum in feed supplements. Anaerobe 2010, 16, 270–277. [Google Scholar] [CrossRef]
  93. Singh, H.; Kongo, J.M.; Borges, A.; Ponte DJ, B.; Griffiths, M.W. Lactic acid bacteria isolated from raw milk cheeses: Ribotyping, antimicrobial activity against selected food pathogens and resistance to common antibiotics. J. Food Process. Technol. 2015, 6, 485. [Google Scholar] [CrossRef]
  94. Galanis, A.; Kourkoutas, Y.; Tassou, C.C.; Chorianopoulos, N. Detection and identification of probiotic Lactobacillus plantarum strains by multiplex PCR using RAPD-derived primers. Int. J. Mol. Sci. 2015, 16, 25141–25153. [Google Scholar] [CrossRef] [PubMed]
  95. Khemariya, P.; Singh, S.; Jaiswal, N.; Chaurasia, S.N.S. Isolation and ddentification of Lactobacillus plantarum from vegetable samples. Food Biotechnol. 2016, 30, 49–62. [Google Scholar] [CrossRef]
  96. Kim, E.; Yang, S.; Lim, B.; Park, S.H.; Rackerby, B.; Kim, H. Design of PCR assays to specifically detect and identify 37 Lactobacillus species in a single 96 well plate. BMC Microbiol. 2020, 20, 1–14. [Google Scholar] [CrossRef] [Green Version]
  97. Jarocki, P.; Komoń-Janczara, E.; Glibowska, A.; Dworniczak, M.; Pytka, M.; Korzeniowska-Kowal, A.; Wzorek, A.; Kordowska-Wiater, M. Molecular routes to specific identification of the Lactobacillus casei group at the species, subspecies and strain level. Int. J. Mol. Sci. 2020, 21, 2694. [Google Scholar] [CrossRef]
  98. Felis, G.E.; Dellaglio, F.; Mizzi, L.; Torriani, S. Comparative sequence analysis of a recA gene fragment brings new evidence for a change in the taxonomy of the Lactobacillus casei group. Int. J. Syst. Evol. Micr. 2001, 51, 2113–2117. [Google Scholar] [CrossRef] [Green Version]
  99. Rodas, A.M.; Ferrer, S.; Pardo, I.Y. Polyphasic study of wine Lactobacillus strains: Taxonomic implications. Int. J. Syst. Evol. Microbiol. 2005, 55, 197–207. [Google Scholar] [CrossRef]
  100. Adesulu-Dahunsi, A.T.; Sanni, A.I.; Jeyaram, K.; Banwo, K. Genetic diversity of Lactobacillus plantarum strains from some indigenous fermented foods in Nigeria. LWT 2017, 82, 199–206. [Google Scholar] [CrossRef]
  101. Lu, W.; Kong, W.; Yang, P.; Kong, J. A one-step PCR-based method for specific identification of 10 common lactic acid bacteria and Bifidobacterium in fermented milk. Int. Dairy J. 2015, 41, 7–12. [Google Scholar] [CrossRef]
  102. Maleki Kakelar, H.; Barzegari, A.; Hanifian, S.; Barar, J.; Omidi, Y. Isolation and molecular identification of Lactobacillus with probiotic potential from abomasums driven rennet. Food Chem. 2019, 272, 709–714. [Google Scholar] [CrossRef]
  103. Kaur, J.; Sharma, A.; Lee, S.; Park, Y. Molecular typing of Lactobacillus brevis isolates from Korean food using repetitive element-polymerase chain reaction. Food Sci. Technol. Int. 2018, 24, 341–350. [Google Scholar] [CrossRef] [PubMed]
  104. Yang, H.; Liu, T.; Zhang, G.; Chen, J.; Gu, J.; Yuan, L.; He, G. Genotyping of Lactobacillus sanfranciscensis isolates from Chinese traditional sourdoughs by multilocus sequence typing and multiplex RAPD-PCR. Int. J. Food Microbiol. 2017, 258, 50–57. [Google Scholar] [CrossRef] [PubMed]
  105. Garofalo, C.; Bancalari, E.; Milanović, V.; Cardinali, F.; Osimani, A.; Sardaro, M.L.S.; Bottari, B.; Bernini, V.; Aquilanti, L.; Clementi, F.; et al. Study of the bacterial diversity of foods: PCR-DGGE versus LH-PCR. Int. J. Food Microbiol. 2017, 242, 24–36. [Google Scholar] [CrossRef] [PubMed]
  106. Pasulka, A.L.; Howes, A.L.; Kallet, J.G.; VanderKelen, J.; Villars, C. Visualization of probiotics via epifluorescence microscopy and fluorescence in situ hybridization (FISH). J. Microbiol. Methods 2021, 182, 106151. [Google Scholar] [CrossRef]
  107. Chatzopoulou, S.; Eriksson, N.L.; Eriksson, D. Improving risk assessment in the European Food Safety Authority: Lessons from the European Medicines Agency. Front. Plant Sci. 2020, 11, 349. [Google Scholar] [CrossRef] [Green Version]
  108. Morris, C.P.; Bergman, Y.; Tekle, T.; Fissel, J.A.; Tamma, P.D.; Simner, P.J.; Burnham, C.D. Cefiderocol antimicrobial susceptibility testing against multidrug-resistant Gram-negative bacilli: A comparison of disk diffusion to broth microdilution. J. Clin. Microbiol. 2020, 59, e01649-20. [Google Scholar] [CrossRef] [PubMed]
  109. Yusuf, E.; van Westreenen, M.; Goessens, W.; Croughs, P. The accuracy of four commercial broth microdilution tests in the determination of the minimum inhibitory concentration of colistin. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 1–8. [Google Scholar] [CrossRef]
  110. Kumar, S.; Pattanaik, A.K.; Jadhav, S.E. Potent health-promoting effects of a synbiotic formulation prepared from Lactobacillus acidophilus NCDC15 fermented milk and Cichorium intybus root powder in Labrador dogs. Curr. Res. Biotechnol. 2021, 3, 209–214. [Google Scholar] [CrossRef]
  111. Chiu, S.; Chen, C.; Wang, L.; Huang, L. Whole-genome sequencing of Lactobacillus salivarius strains BCRC 14759 and BCRC 12574. Genome Announc. 2017, 5, e01336-17. [Google Scholar] [CrossRef]
  112. Zhang, W.; Wang, J.; Zhang, D.; Liu, H.; Wang, S.; Wang, Y.; Ji, H. Complete genome sequencing and comparative genome characterization of Lactobacillus johnsonii ZLJ010, a potential probiotic with health-promoting properties. Front. Genet. 2019, 10, 812. [Google Scholar] [CrossRef] [Green Version]
  113. Pasolli, E.; De Filippis, F.; Mauriello, I.E.; Cumbo, F.; Walsh, A.M.; Leech, J.; Cotter, P.D.; Segata, N.; Ercolini, D. Large-scale genome-wide analysis links lactic acid bacteria from food with the gut microbiome. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  114. Rodríguez-Sánchez, S.; Fernández-Pacheco, P.; Seseña, S.; Pintado, C.; Palop, M.L. Selection of probiotic Lactobacillus strains with antimicrobial activity to be used as biocontrol agents in food industry. LWT 2021, 143, 111142. [Google Scholar] [CrossRef]
  115. Geng, T.; He, F.; Su, S.; Sun, K.; Zhao, L.; Zhao, Y.; Bao, N.; Pan, L.; Sun, H. Probiotics Lactobacillus rhamnosus GG ATCC53103 and Lactobacillus plantarum JL01 induce cytokine alterations by the production of TCDA, DHA, and succinic and palmitic acids, and enhance immunity of weaned piglets. Res. Vet. Sci. 2021, 137, 56–67. [Google Scholar] [CrossRef] [PubMed]
  116. Meng, L.; Zhu, X.; Tuo, Y.; Zhang, H.; Li, Y.; Xu, C.; Mu, G.; Jiang, S. Reducing antigenicity of β-lactoglobulin, probiotic properties and safety evaluation of Lactobacillus plantarum AHQ-14 and Lactobacillus bulgaricus BD0390. Food Biosci. 2021, 42, 101137. [Google Scholar] [CrossRef]
  117. Melini, F.; Melini, V.; Luziatelli, F.; Ficca, A.G.; Ruzzi, M. Health-promoting components in fermented foods: An up-to-date systematic review. Nutrients 2019, 11, 1189. [Google Scholar] [CrossRef] [Green Version]
  118. de Oliveira, P.M.; Santos, L.P.; Coelho, L.F.; Avila Neto, P.M.; Sass, D.C.; Contiero, J. Production of L (+) Lactic Acid by Lactobacillus casei Ke11: Fed batch fermentation strategies. Fermentation 2021, 7, 151. [Google Scholar] [CrossRef]
  119. Allgeyer, L.C.; Miller, M.J.; Lee, S.Y. Sensory and microbiological quality of yogurt drinks with prebiotics and probiotics. J. Dairy Sci. 2010, 93, 4471–4479. [Google Scholar] [CrossRef]
  120. Tang, S.; Cheng, Y.; Wu, T.; Hu, F.; Pan, S.; Xu, X. Effect of Lactobacillus plantarum-fermented mulberry pomace on antioxidant properties and fecal microbial community. LWT 2021, 147, 111651. [Google Scholar] [CrossRef]
  121. Lv, X.; Chen, M.; Huang, Z.; Guo, W.; Ai, L.; Bai, W.; Yu, X.; Liu, Y.; Rao, P.; Ni, L. Potential mechanisms underlying the ameliorative effect of Lactobacillus paracasei FZU103 on the lipid metabolism in hyperlipidemic mice fed a high-fat diet. Food Res. Int. 2021, 139, 109956. [Google Scholar] [CrossRef]
  122. Stearns, J.C.; Lynch, M.D.J.; Senadheera, D.B.; Tenenbaum, H.C.; Goldberg, M.B.; Cvitkovitch, D.G.; Croitoru, K.; Moreno-Hagelsieb, G.; Neufeld, J.D. Bacterial biogeography of the human digestive tract. Sci. Rep. 2011, 1, 170. [Google Scholar] [CrossRef] [Green Version]
  123. Xiao, L.; Feng, Q.; Liang, S.; Sonne, S.B.; Xia, Z.; Qiu, X.; Li, X.; Long, H.; Zhang, J.; Zhang, D.; et al. A catalog of the mouse gut metagenome. Nat. Biotechnol. 2015, 33, 1103–1108. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, H.-N.C.; Wu, H.A.; DE Srjhfb Chen, Y.-Z.; Chen, Y.-J.; Shen, X.-Z.; Liu, T.-T. Altered molecular signature of intestinal microbiota in irritable bowel syndrome patients compared with healthy controls: A systematic review and meta-analysis. Digest. Liver Dis. 2017, 49, 331–337. [Google Scholar] [CrossRef] [PubMed]
  125. Yang, L.; Poles, M.; Fisch, G.; Ma, Y.; Nossa, C.; Phelan, J.; Pei, Z. HIV induced immunosuppression is associated with colonization of the proximal gut by environmental bacteria. AIDS 2016, 30, 19–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef] [PubMed]
  127. Forslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Krogh Pedersen, H. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 2015, 528, 262–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Khalesi, S.; Sun, J.; Buys, N.; Jayasinghe, R. Effect of probiotics on blood pressure. Hypertension 2014, 64, 897–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Ahmadi, E.; Alizadeh-Navaei, R.; Rezai, M.S. Efficacy of probiotic use in acute rotavirus diarrhea in children: A systematic review and meta-analysis. Caspian J. Intern. Med. 2015, 6, 187–195. [Google Scholar]
  130. Peng, Y.; Li, A.; Yu, L.; Qin, G. The role of probiotics in prevention and treatment for patients with allergic rhinitis: A systematic review. Am. J. Rhinol. Allergy 2015, 29, 292–298. [Google Scholar] [CrossRef]
  131. Szajewska, H.; Ruszczyński, M.; Radzikowski, A. Probiotics in the prevention of antibiotic-associated diarrhea in children: A meta-analysis of randomized controlled trials. J. Pediatr. 2006, 149, 367–372.e1. [Google Scholar] [CrossRef]
  132. Zhang, M.; Qian, W.; Qin, Y.; He, J.; Zhou, Y. Probiotics in helicobacter pylori eradication therapy: A systematic review and meta-analysis. World J. Gastroenterol. 2015, 21, 4345–4357. [Google Scholar] [CrossRef]
  133. Jang, Y.J.; Kim, W.; Han, D.H.; Lee, K.; Ko, G. Lactobacillus fermentum species ameliorate dextran sulfate sodium-induced colitis by regulating the immune response and altering gut microbiota. Gut Microbes 2019, 10, 696–711. [Google Scholar] [CrossRef]
  134. Li, X.; Wang, N.; Yin, B.; Fang, D.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Lactobacillus plantarum X1 with α-glucosidase inhibitory activity ameliorates type 2 diabetes in mice. Rsc. Adv. 2016, 6, 63536–63547. [Google Scholar] [CrossRef]
  135. Lim, E. Evaluation of the anti-Helicobacter pylori and cytotoxic properties of the antimicrobial substances from Lactobacillus acidophilus BK13 and Lactobacillus paracasei BK57. Korean J. Microbiol. 2015, 51, 156–168. [Google Scholar] [CrossRef] [Green Version]
  136. Rajoka, M.R.R.; Zhao, H.; Lu, Y.; Lian, Z.; Li, N.; Hussain, N.; Shao, D.; Jin, M.; Li, Q.; Shi, J. Anticancerpotential against cervix cancer (HeLa) cell line of probiotic Lactobacillus casei and Lactobacillus paracasei strains isolated from human breast milk. Food Funct. 2018, 9, 2705–2715. [Google Scholar] [CrossRef] [PubMed]
  137. Suo, H.; Liu, S.; Li, J.; Ding, Y.; Wang, H.; Zhang, Y.; Zhao, X.; Song, J. Lactobacillus paracasei ssp. Paracasei YBJ01 reduced d-galactose-induced oxidation in male Kuming mice. J. Dairy Sci. 2018, 101, 10664–10674. [Google Scholar] [CrossRef] [Green Version]
  138. Lee, I.; Caggianiello, G.; van Swam, I.I.; Taverne, N.; Meijerink, M.; Bron, P.A.; Spano, G.; Kleerebezem, M. Strain-specific features of extracellular polysaccharides and their impact on Lactobacillus plantarum-host interactions. Appl. Environ. Microbiol. 2016, 82, 3959–3970. [Google Scholar] [CrossRef] [Green Version]
  139. Sanhueza, E.; Paredes-Osses, E.; González, C.L.; García, A. Effect of pH in the survival of Lactobacillus salivarius strain UCO_979C wild type and the pH acid acclimated variant. Electron. J. Biotechnol. 2015, 18, 343–346. [Google Scholar] [CrossRef] [Green Version]
  140. Zhu, M.; Xie, R.; Chen, L.; You, M.; Gou, W.; Chen, C.; Li, P.; Cai, Y. Milk production and quality of lactating yak fed oat silage prepared with a low-temperature-tolerant lactic acid bacteria inoculant. Foods 2021, 10, 2437. [Google Scholar] [CrossRef]
  141. Zotta, T.; Parente, E.; Ricciardi, A. Aerobic metabolism in the genus Lactobacillus: Impact on stress response and potential applications in the food industry. J. Appl. Microbiol. 2017, 122, 857–869. [Google Scholar] [CrossRef] [Green Version]
  142. Vázquez, J.A.; Mirón, J.; González, M.P.; Murado, M.A. Effects of aeration on growth and on production of bacteriocins and other metabolites in cultures of eight strains of lactic acid bacteria. Appl. Biochem. Biotechnol. 2005, 127, 111–124. [Google Scholar] [CrossRef]
  143. Tian, X.; Wang, Y.; Chu, J.; Mohsin, A.; Zhuang, Y. Exploring cellular fatty acid composition and intracellular metabolites of osmotic-tolerant mutant Lactobacillus paracasei NCBIO-M2 for highly efficient lactic acid production with high initial glucose concentration. J. Biotechnol. 2018, 286, 27–35. [Google Scholar] [CrossRef] [PubMed]
  144. Panwar, D.; Kapoor, M. Transcriptional analysis of galactomannooligosaccharides utilization by Lactobacillus plantarum WCFS1. Food Microbiol. 2020, 86, 103336. [Google Scholar] [CrossRef] [PubMed]
  145. Lee, J.H.; Hwang, C.E.; Cho, E.J.; Song, Y.H.; Kim, S.C.; Cho, K.M. Improvement of nutritional components and in vitro antioxidative properties of soy-powder yogurts using Lactobacillus plantarum. J. Food Drug Anal. 2018, 26, 1054–1065. [Google Scholar] [CrossRef] [PubMed]
  146. Glušac, J.; Stijepić, M.; Đurđević-Milošević, D.; Milanović, S.; Kanurić, K.; Vukić, V. Growth and viability of Lactobacillus delbrueckii subsp. bulgaricus and Streptococcus thermophilus in traditional yoghurt enriched by honey and whey protein concentrate. Iran J. Vet. Res. 2015, 16, 249–254. [Google Scholar] [PubMed]
  147. Guimarães, A.; Santiago, A.; Teixeira, J.A.; Venâncio, A.; Abrunhosa, L. Anti-aflatoxigenic effect of organic acids produced by Lactobacillus plantarum. Int. J. Food Microbiol. 2018, 264, 31–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Villegas, J.M.; Brown, L.; Savoy De Giori, G.; Hebert, E.M. Optimization of batch culture conditions for GABA production by Lactobacillus brevis CRL 1942, isolated from quinoa sourdough. LWT 2016, 67, 22–26. [Google Scholar] [CrossRef]
  149. Jiang, Y.; Yang, Z. A functional and genetic overview of exopolysaccharides produced by Lactobacillus plantarum. J. Funct. Foods 2018, 47, 229–240. [Google Scholar] [CrossRef]
  150. Bachmann, H.; Pronk, J.T.; Kleerebezem, M.; Teusink, B. Evolutionary engineering to enhance starter culture performance in food fermentations. Curr. Opin. Biotech. 2015, 32, 1–7. [Google Scholar] [CrossRef]
  151. Ito, M.; Kim, Y.; Tsuji, H.; Takahashi, T.; Kiwaki, M.; Nomoto, K.; Danbara, H.; Okada, N. Transposon mutagenesis of probiotic Lactobacillus casei identifies asnH, an asparagine synthetase gene involved in its immune-activating capacity. PLoS ONE 2014, 9, e83876. [Google Scholar] [CrossRef] [Green Version]
  152. Perpetuini, G.; Scornec, H.; Tofalo, R.; Serror, P.; Schirone, M.; Suzzi, G.; Corsetti, A.; Cavin, J.F.; Licandro-Seraut, H. Identification of critical genes for growth in olive brine by transposon mutagenesis of Lactobacillus pentosus C11. Appl. Environ. Microbiol. 2013, 79, 4568–4575. [Google Scholar] [CrossRef]
  153. Joshi, D.S.; Singhvi, M.S.; Khire, J.M.; Gokhale, D.V. Strain improvement of Lactobacillus lactis for D-lactic acid production. Biotechnol. Lett. 2010, 32, 517–520. [Google Scholar] [CrossRef] [PubMed]
  154. Lin, H.; Chen, X.; Yu, L.; Xu, W.; Wang, P.; Zhang, X.; Li, W.; Li, C.; Ren, N. Screening of Lactobacillus rhamnosus strains mutated by microwave irradiation for increased lactic acid production. Afr. J. Microbiol. Res. 2012, 6, 6055–6065. [Google Scholar] [CrossRef]
  155. Iang, A.L.; Hu, W.; Li, W.J.; Liu, L.; Tian, X.J.; Liu, J.; Wang, S.Y.; Lu, D.; Chen, J.H. Enhanced production of L-lactic acid by Lactobacillus thermophiles SRZ50 mutant generated by high-linear energy transfer heavy ion mutagenesis. Eng. Life Sci. 2018, 18, 626–634. [Google Scholar] [CrossRef] [Green Version]
  156. Xu, F.; Li, Q.; Wang, S.; Bai, J.; Dong, M.; Xiao, G.; Wang, J. Lactobacillus casei JY300-8 generated by 12C6+ beams mutagenesis inhibits tumor progression by modulating the gut microbiota in mice. J. Funct. Foods 2021, 87, 104779. [Google Scholar] [CrossRef]
  157. Gao, C.; Yang, F.; Liu, Y. Plasma mutation breeding of high yield γ-aminobutyric acid lactic acid bacteria. Gene Sci. Eng. 2017, 1, 8–18. [Google Scholar] [CrossRef]
  158. Chen, Y.; Tian, X.W.; Li, Q.H.; Li, Y.; Zhuang, Y.P. Target-site directed rational high-throughput screening system for high sophorolipids production by Candida bombicola. Bioresour. Technol. 2020, 315, 123856–123864. [Google Scholar] [CrossRef]
  159. Lv, X.; Song, J.; Yu, B.; Liu, H.; Li, C.; Zhuang, Y.; Wang, Y. High-throughput system for screening of high L-lactic acid productivity strains in deep-well microtiter plates. Bioprocess Biosyst. Eng. 2016, 39, 1737–1747. [Google Scholar] [CrossRef]
  160. Hugenholtz, J.; Sybesma, W.; Groot, M.N.; Wisselink, W.; Ladero, V.; Burgess, K.; van Sinderen, D.; Piard, J.; Eggink, G.; Smid, E.J.; et al. Metabolic engineering of lactic acid bacteria for the production of nutraceuticals. In Lactic Acid Bacteria: Genetics, Metabolism and Applications, 2nd ed.; Siezen, R.J., Kok, J., Abee, T., Schaafsma, G., Eds.; Springer: Berlin, Germany, 2002; Volume 82, pp. 217–235. [Google Scholar]
  161. Pan, H.; Zhan, J.; Yang, H.; Wang, C.; Liu, H.; Zhou, H.; Zhou, H.; Lu, X.; Su, X.; Tian, Y. Improving the acid resistance of Tannase TanBLp (AB379685) from Lactobacillus plantarum ATCC14917T by site-specific mutagenesis. Indian J. Microbiol. 2022, 62, 96–102. [Google Scholar] [CrossRef]
  162. Tian, X.; Liu, X.; Zhang, Y.; Chen, Y.; Hang, H.; Chu, J.; Zhuang, Y. Metabolic engineering coupled with adaptive evolution strategies for the efficient production of high-quality L-lactic acid by Lactobacillus paracasei. Bioresour. Technol. 2021, 323, 124549. [Google Scholar] [CrossRef]
  163. Upadhyaya, B.P.; DeVeaux, L.C.; Christopher, L.P. Metabolic engineering as a tool for enhanced lactic acid production. Trends Biotechnol. 2014, 32, 637–644. [Google Scholar] [CrossRef]
  164. Tian, X.; Chen, H.; Liu, H.; Chen, J. Recent advances in lactic acid production by lactic acid bacteria. Appl. Biochem. Biotech. 2021, 193, 4151–4171. [Google Scholar] [CrossRef] [PubMed]
  165. Papagianni, M. Metabolic engineering of lactic acid bacteria for the production of industrially important compounds. Comput. Struct. Biotec. 2012, 3, e201210003. [Google Scholar] [CrossRef] [Green Version]
  166. Ye, L.; Zhao, H.; Li, Z.; Wu, J.C. Improved acid tolerance of Lactobacillus pentosus by error-prone whole genome amplification. Bioresour. Technol. 2013, 135, 459–463. [Google Scholar] [CrossRef] [PubMed]
  167. Hospet, R.; Thangadurai, D.; Cruz-Martins, N.; Sangeetha, J.; Appaiah, K.A.A.; Chowdhury, Z.Z.; Bedi, N.; Soytong, K.; Al Tawahaj, A.R.M.; Jabeen, S.; et al. Genome shuffling for phenotypic improvement of industrial strains through recursive protoplast fusion technology. Crit. Rev. Food Sci. 2021, 1, 1–10. [Google Scholar] [CrossRef]
  168. Sun, J.; Liu, H.; Dang, L.; Liu, J.; Wang, J.; Lu, Z.; Lu, Y. Genome shuffling of Lactobacillus plantarum 163 enhanced antibacterial activity and usefulness in preserving orange juice. Lett. Appl. Microbiol. 2021, 73, 741–749. [Google Scholar] [CrossRef] [PubMed]
  169. van Pijkeren, J.P.; Britton, R.A. Precision genome engineering in lactic acid bacteria. Microb. Cell Factories 2014, 13, S10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Börner, R.A.; Kandasamy, V.; Axelsen, A.M.; Nielsen, A.T.; Bosma, E.F. Genome editing of lactic acid bacteria: Opportunitie for food, feed, pharma and biotech. FEMS Microbiol. Lett. 2019, 366, fny291. [Google Scholar] [CrossRef]
  171. Plavec, T.V.; Berlec, A. Safety aspects of genetically modified lactic acid bacteria. Microorganisms 2020, 8, 297. [Google Scholar] [CrossRef] [Green Version]
  172. Panesar, P.S. Fermented dairy products: Starter cultures and potential nutritional benefits. Food Nutr. Sci. 2011, 2, 47–51. [Google Scholar] [CrossRef] [Green Version]
  173. Goveas, L.C.; Ashwath, K.S.; Nazerath, B.R.; Dsouza, O.; Ullekh; Umesh, A.; Muddappa, V.S. Development of coconut water-based exopolysaccharide rich functional beverage by fermentation with probiotic Lactobacillus plantarum SVP2. Biocatal. Agric. Biotechnol. 2021, 34, 102030. [Google Scholar] [CrossRef]
  174. Szutowska, J. Functional properties of lactic acid bacteria in fermented fruit and vegetable juices: A systematic literature review. Eur. Food Res. Technol. 2020, 246, 357–372. [Google Scholar] [CrossRef]
  175. Crispín-Isidro, G.; Lobato-Calleros, C.; Espinosa-Andrews, H.; Alvarez-Ramirez, J.; Vernon-Carter, E.J. Effect of inulin and agave fructans addition on the rheological, microstructural and sensory properties of reduced-fat stirred yogurt. LWT 2016, 62, 438–444. [Google Scholar] [CrossRef]
  176. Ozogul, F.; Ozcelik, S.; Ozogul, Y.; Yilmaz, M.T. Seafood infusion broths as novel sources to produce organic acids using selected lactic acid bacteria strains. Food Biosci 2021, 43, 101227. [Google Scholar] [CrossRef]
  177. Siedler, S.; Balti, R.; Neves, A.R. Bioprotective mechanisms of lactic acid bacteria against fungal spoilage of food. Curr. Opin. Biotech. 2019, 56, 138–146. [Google Scholar] [CrossRef]
  178. Papizadeh, M.; Rohani, M.; Nahrevanian, H.; Hosseini, S.N.; Shojaosadati, S.A.; Pourshafie, M.R. Using various approaches of design of experiments for high cell density production of the functionally probiotic Lactobacillus plantarum strain RPR42 in a cane molasses-based medium. Curr. Microbiol. 2020, 77, 1756–1766. [Google Scholar] [CrossRef] [PubMed]
  179. E, J.; Ma, L.; Chen, Z.; Ma, R.; Zhang, Q.; Sun, R.; He, Z.; Wang, J. Effects of buffer salts on the freeze-drying survival rate of Lactobacillus plantarum LIP-1 based on transcriptome and proteome analyses. Food Chem. 2020, 326, 126849. [Google Scholar] [CrossRef]
  180. Singhvi, M.; Zendo, T.; Sonomoto, K. Free lactic acid production under acidic conditions by lactic acid bacteria strains: Challenges and future prospects. Appl. Microbiol. Biot. 2018, 102, 5911–5924. [Google Scholar] [CrossRef]
  181. Bähr, C.; Leuchtle, B.; Lehmann, C.; Becker, J.; Jeude, M.; Peinemann, F.; Arbter, R.; Büchs, J. Dialysis shake flask for effective screening in fed-batch mode. Biochem. Eng. J. 2012, 69, 182–195. [Google Scholar] [CrossRef]
  182. Costas Malvido, M.; Alonso González, E.; Pérez Guerra, N. Nisin production in realkalized fed-batch cultures in whey with feeding with lactose- or glucose-containing substrates. Appl. Microbiol. Biotechnol. 2016, 100, 7899–7908. [Google Scholar] [CrossRef]
  183. Kawai, M.; Tsuchiya, A.; Ishida, J.; Yoda, N.; Yashiki-Yamasaki, S.; Katakura, Y. Suppression of lactate production in fed-batch culture of some lactic acid bacteria with sucrose as the carbon source. J. Biosci. Bioeng. 2020, 129, 535–540. [Google Scholar] [CrossRef]
  184. Hu, J.; Lin, Y.; Zhang, Z.; Xiang, T.; Mei, Y.; Zhao, S.; Liang, Y.; Peng, N. High-titer lactic acid production by Lactobacillus pentosus FL0421 from corn stover using fed-batch simultaneous saccharification and fermentation. Bioresour. Technol. 2016, 214, 74–80. [Google Scholar] [CrossRef] [PubMed]
  185. Cui, S.; Sadiq, F.A.; Xu, Z.; Liu, Z.; Zhao, J.; Zhang, H.; Chen, W. High-density cultivation of Lactobacillus and Bifidobacterium using an automatic feedback feeding method. LWT 2019, 112, 108232. [Google Scholar] [CrossRef]
  186. Zhao, L.; Tang, Z.; Gu, Y.; Shan, Y.; Tang, T. Investigate the cross-flow flat-plate photobioreactor for high-density culture of microalgae. Asia-Pac. J. Chem. Eng. 2018, 13, e2247. [Google Scholar] [CrossRef]
  187. Chen, P.; Hong, Z.; Cheng, C.; Ng, I.; Lo, Y.; Nagarajan, D.; Chang, J. Exploring fermentation strategies for enhanced lactic acid production with polyvinyl alcohol-immobilized Lactobacillus plantarum 23 using microalgae as feedstock. Bioresour. Technol. 2020, 308, 123266. [Google Scholar] [CrossRef]
  188. Cha, K.H.; Lee, E.H.; Yoon, H.S.; Lee, J.H.; Kim, J.Y.; Kang, K.; Park, J.; Jin, J.B.; Ko, G.; Pan, C. Effects of fermented milk treatment on microbial population and metabolomic outcomes in a three-stage semi-continuous culture system. Food Chem. 2018, 263, 216–224. [Google Scholar] [CrossRef]
  189. Gao, M.J.; Nie, Y.F.; Zhang, H.X.; Xie, Y.H.; Hao, H.W.; Liu, H. Application of directed vat set yogurt starters by space Lactobacillus reuteri. Nat. Prod. Res. Dev. 2019, 31, 136–142. [Google Scholar] [CrossRef]
  190. Fonseca, F.; Cenard, S.; Passot, S. Freeze-drying of lactic acid bacteria. In Cryopreservation and Freeze-Drying Protocols, 1st ed.; Wolkers, W., Oldenhof, H., Eds.; Springer: New York, NY, USA, 2015; Volume 1257, pp. 477–488. [Google Scholar] [CrossRef]
  191. Gallardo-Rivera, C.; Báez-González, J.G.; García-Alanís, K.G.; Torres-Alvarez, C.; Dares-Sánchez, K.; Szymanski, A.; Amaya-Guerra, C.A.; Castillo, S. Effect of three types of drying on the viability of lactic acid bacteria in Foam-Mat dried yogurt. Processes 2021, 9, 2123. [Google Scholar] [CrossRef]
  192. Moreira, M.T.C.; Martins, E.; Perrone, Í.T.; de Freitas, R.; Queiroz, L.S.; de Carvalho, A.F. Challenges associated with spray drying of lactic acid bacteria: Understanding cell viability loss. Compr. Rev. Food Sci. Food Saf. 2021, 20, 3267–3283. [Google Scholar] [CrossRef]
  193. Hao, F.; Fu, N.; Ndiaye, H.; Woo, M.W.; Jeantet, R.; Chen, X.D. Thermotolerance, survival, and stability of lactic acid bacteria after spray drying as affected by the increase of growth temperature. Food Bioprocess Tech. 2021, 14, 120–132. [Google Scholar] [CrossRef]
  194. Lu, W.; Fu, N.; Woo, M.W.; Chen, X.D. Exploring the interactions between Lactobacillus rhamnosus GG and whey protein isolate for preservation of the viability of bacteria through spray drying. Food Funct. 2021, 12, 2995–3008. [Google Scholar] [CrossRef]
  195. Mamun, A.A.; Payap, M.; Jaruwan, M. Viability of Lactobacillus plantarum TISTR 2083 in protectant during low-temperature drying and storage. Sains Malays. 2021, 50, 2227–2238. [Google Scholar] [CrossRef]
  196. Shanmugam, S. Granulation techniques and technologies: Recent progresses. Bioimpacts 2015, 5, 55–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Vishali, D.A.; Monisha, J.; Sivakamasundari, S.K.; Moses, J.A.; Anandharamakrishnan, C. Spray freeze drying: Emerging applications in drug delivery. J. Control. Release 2019, 300, 93–101. [Google Scholar] [CrossRef] [PubMed]
  198. Choi, I.S.; Ko, S.H.; Kim, H.M.; Chun, H.H.; Lee, K.H.; Yang, J.E.; Jeong, S.; Park, H.W. Shelf-life extension of freeze-dried Lactobacillus brevis WiKim0069 using supercooling pretreatment. LWT 2019, 112, 108230. [Google Scholar] [CrossRef]
Figure 1. Neighbor-joining phylogenetic tree based on 16S rRNA gene sequences showing the relationship between known species in Lactobacillus, produced and rendered by an online tool—Interactive Tree of Life (https://itol.embl.de/, accessed on 21 March 2022) [43]. The lilac background color represents Lactobacillus species commonly used in the food industry (including fermented fish products, fermented dairy products, fermented soy products, fermented starch foods, fermented fruit and vegetable, fermented meat products, etc.), and the pink background color represents others (including human gastrointestinal tract, vagina, oral cavity, etc.).
Figure 1. Neighbor-joining phylogenetic tree based on 16S rRNA gene sequences showing the relationship between known species in Lactobacillus, produced and rendered by an online tool—Interactive Tree of Life (https://itol.embl.de/, accessed on 21 March 2022) [43]. The lilac background color represents Lactobacillus species commonly used in the food industry (including fermented fish products, fermented dairy products, fermented soy products, fermented starch foods, fermented fruit and vegetable, fermented meat products, etc.), and the pink background color represents others (including human gastrointestinal tract, vagina, oral cavity, etc.).
Foods 11 03063 g001
Figure 2. Characterization and evaluation procedures of Lactobacilli strains.
Figure 2. Characterization and evaluation procedures of Lactobacilli strains.
Foods 11 03063 g002
Figure 3. Probiotic properties of Lactobacillus spp.
Figure 3. Probiotic properties of Lactobacillus spp.
Foods 11 03063 g003
Figure 4. Production process of DVS starters.
Figure 4. Production process of DVS starters.
Foods 11 03063 g004
Table 1. Lactobacilli species in fermented foods.
Table 1. Lactobacilli species in fermented foods.
Fermented ProductsLactobacilli Species PresentReferences
Fermented fish productsL. acidipiscis, L. brevis, L. delbrueckii, L. fermentum, L. pentosus, L. plantarum, L. versmoldensis[25,26,27]
Fermented dairy productsL. delbrueckii, L. bulgaricus, L. fermentum, L. plantarum, L. kefiri, L. paracasei subsp. Paracasei, L. rhamnosus, L. curvatus[6,28,29,30,31,32,33,34]
Fermented soy productsL. amylophilus, L. buchneri, L. delbrueckii, L. fermentum, L. paracasei, L. plantarum, L. salivarius[35,36,37]
Fermented starch foodsL. acidophilus, L. brevis, L. casei, L. bulgaricus, L. oryzae, L. pentosus, L. reuteri, L. rhamnosus, L. rossiae, L. sakei, L. curvatus, L. panis, L. sanfranciscensis[38]
Fermented fruit and vegetableL. acidophilus, L. brevis, L. casei, L. fermentum, L. pentosus, L. plantarum[28,39,40]
Fermented meat productsL. sakei, L. curvatus[41,42]
Table 2. Characteristics of Lactobacilli strains.
Table 2. Characteristics of Lactobacilli strains.
CharacteristicAssaysRepresentative References
SafetyStrain identification (including physiological and biochemical tests, molecular level)[44,45,46,47]
Antibiotic resistance[48]
Hemolytic activity[49]
Determination of potential metabolites (enzyme production, toxin production, production of biogenic amines)[50,51,52]
Tolerance to stressLow pH and bile (for example, artificial gastric and pancreatic juices and GIT simulators)[17,53,54,55,56,57,58]
Growth environment (for example, nutrition substrate, osmotic pressure, light, temperature, oxygen)[59,60,61,62]
Adhesion abilityCell surface hydrophobicity[63]
Adhesion to mucus (for example, adhesion to mucin)[64,65,66]
Adhesion to Caco-2/TC7 cells[67,68]
Antimicrobial activityProduction of antimicrobial metabolites such as lactic acid and bacteriocin against pathogenic bacteria (e.g., streak methods, disk diffusion methods, turbidimetric assays, biofluorescence analysis)[69,70,71]
Autoaggregation, Coaggregation[18,72]
Technological propertiesProteolytic activity (e.g., production of various proteases)[73]
Lipolytic activity (e.g., production of lipases)[74]
Carbohydrate degradation activity (e.g., production of various glycosidases, amylases, cellulases)[75]
Reduce cardiovascular diseaseCholesterol degradation tests (e.g., Bile salt hydrolase activity)[76]
Metabolites such as peptides inhibit the ACE activity[77,78]
AntioxidantTolerance to hydrogen peroxide[79]
Metabolites such as the antioxidant activity of extracellular polysaccharides, peptides[78,80]
AnticancerAmes test[81]
Comet assay[82]
Nitrosamine degradation assay[9]
Inducing apoptosis of cancer cells test[83]
Additional characteristicsConjugated linoleic acid test[84]
The removal of heavy metals[85]
β-Galactosidase activity analysis[86]
Determination of oxalic acid degradation[87]
Determination of production of short-chain unsaturated fatty acids and vitamins[88,89,90]
Table 3. Molecular approaches used in discrimination among the genus Lactobacillus.
Table 3. Molecular approaches used in discrimination among the genus Lactobacillus.
Methods UsedCommentsSpecies Identified and SourceReference
23S rDNA probeProbes unequivocally differentiated L. acidophilus and L. plantarum isolates.L. acidophilus, L. pentosus, L. plantarum species isolates from feed supplements or starter products[92]
RibotypingGood discrimination at strains level based upon differences in rRNA.Some L. paracasei ss. paracasei strains as the dominant ones from raw milk cheeses[93]
RAPDGood discrimination at strains level.L. plantarum 2035 and L. plantarum ACA-DC 2640 isolated from Feta cheese[94]
Species-specific PCR (plantaricin biosynthesis protein gene)Rapid and preliminary screening of L. plantarum from large vegetable samples before performing a battery of phenotypic and molecular methods.L. plantarum from vegetable samples[95]
Species-specific PCR using 16S rRNA or unique genes primersSuccessful in the species detected in 17 products matched those indicated on their labels, whereas the remaining products contained species other than those appearing on the label.Some Lactobacillus spp., 19 probiotics and 12 dairy products[96]
Genus- and species-specific PCR, multiplex PCR,
real-time HRM analysis, RFLP-PCR, rep-PCR, RAPD-PCR, AFLP-PCR, and proteomic methods such as MALDI-TOF MS typing and SDS-PAGE fingerprinting
Multiplex PCR and MALDI-TOF MS were the most valuable methods to identify the tested bacteria at the species level. At the strain level, the AFLP-PCR method showed the highest discriminatory power.L. casei group, two international collections of microorganisms—the Japan Collection of Microorganisms (JCM) and Belgian Coordinated Collections of Microorganisms (BCCM)[97]
Comparative sequence analysis, stretches of rec A geneSuccessful in a clear separation of all type strains in distinct branches; identification of L. casei ATCC 393 and L. casei ATCC 334 as L. zeae and L. paracasei, respectively.L. casei, L. paracasei (both subspecies), L. rhamnosus, L. zeae, strains from a commercial probiotic product.[98]
16S ARDRA, RAPD, Eco RI ribotyping13 wine strains typed as L. paracasei/casei, based on similar band pattern as L. paracasei type strain and L. casei ATCC 334.L. casei/L. paracasei from wine[99]
PFGEGood discrimination at strain level based upon different bacterial strains.The strains of L. plantarum isolated from the different fermented foods[100]
One-step PCR-based, using 16S rRNA genes primersSuccessful differentiation among 10 common lactic acid bacteria at the species level.L. delbrueckii and others from fermented milk[101]
16S ARDRA, ribotyping, RAPDOnly RAPD and ribotyping could discriminate between the type strains of both species.L. plantarum, L. pentosus, Wine isolates[99]
PCR-ARDRA (Taq I), RAPDARDRA and RAPD approaches may demonstrate a robust efficiency in the discrimination of unknown isolates.L. acidophilus, L. planetarum, and L. fermentum from abomasums driven rennet[102]
Repetitive-element PCRCould rapidly and easily differentiate L. brevis species at strains level.The closely related strains of L. brevis species[103]
Multi-locus sequence typing (MLST) and multiplex RAPD-PCRTargeting different genetic variations under the combination of MLST and multiplex-RAPD analysisL. sanfranciscensis, Chinese traditional sourdoughs[104]
PCR-DGGE, length-heterogeneity PCR (LH-PCR)Good discrimination at strains level.Type and reference strains of L. brevis DSMZ 20556 and L. plantarum DSMZ 2601[105]
FISHRapid and accurate way to identify and quantify bacterial species.L. plantarum (Probiotic products)[106]
Table 4. Some crucial functions of Lactobacilli strains.
Table 4. Some crucial functions of Lactobacilli strains.
Functional PropertiesExampleReference
Regulating immune systemJang et al. evaluated immunometabolic functions of L. fermentum strains (KBL374 and KBL375) isolated from the feces of healthy Koreans.[133]
Regulating the balance of blood glucose, blood lipid, and blood pressureLi et al. found that L. plantarum X1 can alleviate the symptoms of diabetes by improving the level of short-chain fatty acids in type 2 diabetic mice.[134]
Antimicrobial activityLim et al. found that L. paracasei BK 57 has antagonistic effect on Helicobacter pylori and can be used as potential antibiotics.[135]
Lower blood pressureOng et al. found that L. paracasei can isolate and purify ACE inhibitory peptides from cheddar cheese.[31]
AntitumorRajoka et al. found that the antiproliferative activity of the fermentation supernatant of L. paracasei SR 4 on cervical cancer cells was up to 89%. L. paracasei showed high anti-cancer activity by promoting the up-regulation of BAX, BAD, caspase3, caspase8, and caspase9 genes and down-regulating the expression of the Bcl-2 gene.[136]
AntioxidantSuo et al. found that L. paracasei ybJ 01 can significantly improve D-galactose, induced the ability of serum superoxide dismutase (SOD), glutathione peroxidase and total antioxidant in mice, and inhibited the production of malondialdehyde.[137]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lu, Y.; Xing, S.; He, L.; Li, C.; Wang, X.; Zeng, X.; Dai, Y. Characterization, High-Density Fermentation, and the Production of a Directed Vat Set Starter of Lactobacilli Used in the Food Industry: A Review. Foods 2022, 11, 3063. https://doi.org/10.3390/foods11193063

AMA Style

Lu Y, Xing S, He L, Li C, Wang X, Zeng X, Dai Y. Characterization, High-Density Fermentation, and the Production of a Directed Vat Set Starter of Lactobacilli Used in the Food Industry: A Review. Foods. 2022; 11(19):3063. https://doi.org/10.3390/foods11193063

Chicago/Turabian Style

Lu, Yun, Shuqi Xing, Laping He, Cuiqin Li, Xiao Wang, Xuefeng Zeng, and Yifeng Dai. 2022. "Characterization, High-Density Fermentation, and the Production of a Directed Vat Set Starter of Lactobacilli Used in the Food Industry: A Review" Foods 11, no. 19: 3063. https://doi.org/10.3390/foods11193063

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