Next Article in Journal
Effect of Surface Roughness of Deciduous and Permanent Tooth Enamel on Bacterial Adhesion
Next Article in Special Issue
Impact of Pomegranate on Probiotic Growth, Viability, Transcriptome and Metabolism
Previous Article in Journal
Antibiotic Resistance in Neisseria gonorrhoeae: Challenges in Research and Treatment
Previous Article in Special Issue
Probiotics: Protecting Our Health from the Gut
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Applications of Probiotic-Based Multi-Components to Human, Animal and Ecosystem Health: Concepts, Methodologies, and Action Mechanisms

by
Sonagnon Kouhounde
1,
Kifouli Adéoti
2,
Majid Mounir
3,
Alessandro Giusti
4,
Paulo Refinetti
5,
Akaninyene Otu
6,7,8,
Emmanuel Effa
7,8,
Bassey Ebenso
9,
Victor O. Adetimirin
10,
Josep Mercader Barceló
11,
Ousmane Thiare
12,
Holy N. Rabetafika
13 and
Hary L. Razafindralambo
13,14,*
1
Laboratoire des Sciences Biologiques Appliquées, Université Aube Nouvelle, Bobo-Dioulasso 01 BP 234, Burkina Faso
2
Laboratoire de Microbiologie et de Technologie Alimentaire (LAMITA), Faculté des Sciences et Techniques, Université d’Abomey-Calavi, Cotonou 01BP 526, Benin
3
Department of Food Science and Nutrition, Biotransformations Laboratory, Hassan II Institute of Agronomy and Veterinary Medicine, Rabat Instituts, Rabat 10112, Morocco
4
CyRIC-Cyprus Research and Innovation Center Ltd., Nicosia 2414, Cyprus
5
REM Analytics SA, 1870 Monthey, Switzerland
6
Leeds Teaching Hospitals NHS Trust, Leeds LS1 3EX, UK
7
Foundation for Healthcare Innovation and Development (FHIND), Calabar 540271, Nigeria
8
Department of Internal Medicine, University of Calabar, Calabar 540271, Nigeria
9
Leeds Institute of Health Sciences, University of Leeds, Leeds LS2 9NL, UK
10
Department of Crop and Horticultural Sciences, University of Ibadan, Ibadan 200284, Nigeria
11
Molecular Biology and One Health Research Group (MolONE), University of the Balearic Islands, 07120 Palma, Spain
12
Department of Computer Science, Faculty of Applied Sciences and Technology, Université Gaston Berger de Saint Louis, Saint-Louis BP 234, Senegal
13
ProBioLab, Campus Universitaire de la Faculté de Gembloux Agro-Bio Tech/Université de Liège, B-5030 Gembloux, Belgium
14
BioEcoAgro Joint Research Unit, TERRA Teaching and Research Centre, Microbial Processes and Interactions, Gembloux Agro-Bio Tech/Université de Liège, B-5030 Gembloux, Belgium
*
Author to whom correspondence should be addressed.
Microorganisms 2022, 10(9), 1700; https://doi.org/10.3390/microorganisms10091700
Submission received: 30 June 2022 / Revised: 12 August 2022 / Accepted: 20 August 2022 / Published: 24 August 2022

Abstract

:
Probiotics and related preparations, including synbiotics and postbiotics, are living and non-living microbial-based multi-components, which are now among the most popular bioactive agents. Such interests mainly arise from the wide range and numerous beneficial effects of their use for various hosts. The current minireview article attempts to provide an overview and discuss in a holistic way the concepts, methodologies, action mechanisms, and applications of probiotic-based multi-components in human, animal, plant, soil, and environment health. Probiotic-based multi-component preparations refer to a mixture of bioactive agents, containing probiotics or postbiotics as main functional ingredients, and prebiotics, protectants, stabilizers, encapsulating agents, and other compounds as additional constituents. Analyzing, characterizing, and monitoring over time the traceability, performance, and stability of such multi-component ingredients require relevant and sensitive analytical tools and methodologies. Two innovative profiling and monitoring methods, the thermophysical fingerprinting thermogravimetry–differential scanning calorimetry technique (TGA-DSC) of the whole multi-component powder preparations, and the Advanced Testing for Genetic Composition (ATGC) strain analysis up to the subspecies level, are presented, illustrated, and discussed in this review to respond to those requirements. Finally, the paper deals with some selected applications of probiotic-based multi-components to human, animal, plant, soil and environment health, while mentioning their possible action mechanisms.

1. Introduction

During the 20th century, probiotics have been recognized and used as beneficial live microorganisms only for human and animal health [1]. A few years later, the concept of probiotics has been extended and applied to plant growth and protection, soil fertilizing [2], and depolluting [3]. Now, probiotics are considered to be a biotool par excellence that offers multiple potential solutions in improving human life for food and nutrition security [4], disease prevention [5,6,7,8], and environment protection [9,10]. Beside high functional interests, probiotics also benefit the material status of natural [11], safe or “generally recognized as safe” (GRAS) and “qualified presumption of safety” (QPS) [12], and renewable, i.e., cultivable and inexhaustible, biomass sources [13].
Probiotics are live microorganisms, mainly belonging to lactic acid bacteria (Lactobacilli), Bifidobacteria, soil-based bacteria (Bacillus sp.), and yeast (Saccharomyces sp.) groups [14] with different species and strains from food and non-food sources [15]. Their multiple functionalities in promoting human and ecosystem health result from their capacity to control pathogens, reduce toxin and polluting substances, and increase nutrient bioavailability through three main action mechanisms [16]. These include, among others: (1) surface and nutrient competition ability against pathogens through cell wall macromolecular structures (e.g., S-proteins and exopolysaccharides) and secreted amphiphilic compounds (e.g., biosurfactants); (2) antimicrobial production (e.g., bacteriocins, antiviral agents, enzymes, antioxidant compounds); and (3) immunomodulation activity to immune cells. In fact, probiotics act as immunomodulators by increasing the growth of healthy components and restoring the normal gut ecological niche [17]. Probiotics can stimulate phagocytic activity, balance pro- and anti-inflammatory cytokines, enhance the production of immunoglobulin (IgA) by plasma cells, and generate bioactive peptides.
From a technological viewpoint, probiotics are commonly produced by culture in fermenters, and used as functional ingredients in formulated food and non-food products, including fermented foods and beverages, diet supplements, drugs, and biological and cosmetic products [18]. Rarely used in pure forms, probiotics are mostly prepared and formulated with a wide range of other functional compounds for preserving, and even enhancing, cell performance, viability, and stability [19,20]. These components include thermo- and cryo-protectors [21], prebiotics [22], and encapsulating agents [23,24], or another probiotic species for preparing multi-strain products. When probiotics are combined with prebiotics, represented mainly by oligosaccharides, phenolic compounds, or polyunsaturated fatty acids, which serve as selective nutrients for probiotics [25], the multi-component preparations are called synbiotics.
When microbial cells are inactivated by thermal processing (e.g., pasteurization, tyndallization, autoclaving), and no longer contain viable probiotic cells, or the preparation consists of probiotic fragments or their metabolites, the term postbiotics is used [26].
The concept of “probiotic-based multi-components” is therefore more appropriate to design the preparations or formulations of probiotic multi-strains, synbiotics, or postbiotics, which are all beneficial for human, animal, and ecosystem health [27,28] when used under adequate conditions. In terms of analysis and characterization, the use only of gold-standard methods (e.g., genotype and phenotype profiling) is insufficient for identifying and fingerprinting all components, particularly the “additives” included in these probiotic-based multi-components. Other analytical and physical chemical tools are needed for identifying and quantifying, for instance, prebiotics and protectants in such formulations [29].
The objective of this review paper is to (1) define the concept of probiotic-based multi-components; (2) describe two emerging analytical techniques for the global profiling of multi-component and multi-strain products, and monitoring the probiotic strain interactions over time; and (3) address relevant applications in human, animal, and ecosystem health, including plant, soil and environment.

2. Probiotic-Based Multi-Components

2.1. Probiotics and Synbiotics

Probiotic-based multi-components are products that contain either one strain or a mixture of strains [30] and additional functional compounds. These are mainly thermo- or cryoprotectants [20,21], mainly carbohydrates (e.g., lactose, maltose, trehalose, maltodextrins), proteins (e.g., skim milk powder, whey protein), minerals (e.g., Ca++), and other compounds (e.g., glycerol), but also antioxidants such as ascorbic acid, tocopherol, and flavonoids [31]. Such additional compounds are essentially used to protect microbial cells against the changes in stress parameters such as temperature, pressure, oxygen exposure, and relative humidity, which cause losses in cell viability during the drying process of the probiotic culture and the storage of the resulting dry material [32,33]. Other functional compounds found in probiotic preparations are encapsulating agents when coating/encapsulation techniques are used for ensuring sufficient survival rates of microbial cells, until they reach the human or animal gut [34]. Among the current components used in such processes are hydrocolloid biopolymers, mainly constituted by proteins and polysaccharides, either for bulk or single-cell encapsulation systems [24]. According to the physical state of the product (dry or wet form), other extra additives and functional components used in probiotic formulations, for instance, anticaking agents, minerals, buffers, etc., can influence the performance of probiotic cells in terms of metabolic activity and survivability [35].
When prebiotics are combined with mono- or multi-strain probiotic preparations, the multi-component systems are called synbiotics [36]. Prebiotics are selective nutrients for beneficial microorganisms harbored by the host, mainly carbohydrate compounds such as inulin and fructo- and galacto-oligosaccharides (FOSs and GOSs), which are considered safe food ingredients in the European Union [25]. Moreover, as renewable and sustainable resources with a relatively low cost, these biocomponents appear to be eco-friendly and economically advantageous for use in agro-food sectors. Some foods and plant-based materials such as soybean extracts, kojiglycosylceramides, grape extracts, tea polyphenols, and seaweed extracts can also stimulate the proliferation of beneficial microorganisms in the intestine, and are considered as prebiotics [37]. Although all current prebiotics are carbohydrates, some polyphenols compounds have emerged and mediate beneficial physiological effects by modulating the gut microbiota [38].
Synbiotics as mixtures of probiotics and prebiotics can be designed in complement to independently target the host microorganisms, or in synergism, for which the prebiotic is selectively utilized by the co-administrated microorganisms to achieve one or more health benefits [36]. Consequently, such probiotic-based multi-components can be designed using a multitude of combinations with a wide range of properties and functions. The use of synbiotics with regards to the synergistic aspect confers them economic and environmental assets. Numerous benefits of synbiotics to human health have been shown [36] in comparison with those of animal [39] and plant cases in nutrition and health [40]. Synbiotic-based multi-components also appear to be relevant for promoting both food nutritional security and sustainable agriculture, due to their roles as biofertilizers and biopesticides [2]. Table 1 lists some common synbiotics with their components in probiotics and prebiotics, in addition to their commercial names.

2.2. Postbiotics

The term “postbiotics” has been used for several years in different contexts, and the definition varies from one author to another, sometimes leading to confusion. Substances released by or produced through the metabolic activity of a microorganism that exerts a beneficial effect on the host, directly or indirectly, or substances of microorganism origin that confer beneficial effects to the host and differ from substances of a prebiotic nature, or non-viable probiotics, inanimate microorganisms and/or their components, paraprobiotics, and ghostbiotics, are among the terms currently used to name postbiotics [41,42]. A panel of scientist experts has declared that postbiotics are preparations of inanimate microorganisms and/or their components that confers a health benefit on the host [43]. The most recent concept of postbiotics uses the term “substances derived after the microorganisms are no longer alive, inanimate, dead or inactivated”, including intact cells or structural fragments of microbes such as the cell wall [26]. From a chemical viewpoint, postbiotics are heterogeneous multi-components of microbial metabolites from cell-free supernatants (e.g., supernatants of L. acidophilus, L. casei, L. rhamnosus GG, S. cerevisae, and S. boulardii cultures), or microbial fragments and lysates prepared by chemical and mechanical techniques such as sonication and heat treatments [42]. Metabolite-based postbiotics include exopolysaccharides (EPSs), e.g., β-glucan, antioxidant enzymes, short-chain fatty acids (SCFAs) such as acetic, propionic, and butyric acids, and vitamins; those from cell wall components are lipoteichoic acid, teichoic acids, peptidoglycan, cell surface proteins, and polysaccharides [44]. Some information on the potential applications of postbiotics for human health and their mechanisms of action related to antibacterial, antiviral, antioxidant, and anticancer activities have extensively been reviewed by different authors [45,46,47].

3. Innovative Profiling and Monitoring Methodologies

3.1. Thermophysical Profiling

3.1.1. Principle

A thermal profiling and fingerprinting method combines both thermogravimetry and differential scanning calorimetry techniques (TGA–DSC). This is one of the most convenient techniques for characterizing and analyzing powder-based products [48,49,50]. This calorimetric-coupling technique has recently been used for the first time to fingerprint probiotic-based powder products [51]. Such an original approach provides unique qualitative and quantitative data related to the decomposition and transition phases for each probiotic powder sample, regardless of its complexity, by monitoring the changes in the material mass and energetic content under a constant temperature rise. Its potential validity has been shown by a comparative analysis using proteomics and intestinal permeability in vivo tests [52].

3.1.2. Practice of TGA-DSC Coupling Method

The TGA-DSC thermal analysis consists in heating a sample while simultaneously monitoring its mass and energy content by gravimetric analysis and heat flow measurements, respectively. The TGA-DSC instrument includes a calorimeter, which is a special furnace for controlling and measuring temperature changes of the material, and a microbalance for mass measurements. A powder sample is weighted with high precision (±0.01 mg), and then deposited onto an aluminum crucible. Runs are performed by simultaneously monitoring the changes in the sample mass (m) and heat flow (HF) as a function of the linearly increasing temperature under defined conditions. The workflow, a synthetic example of thermal profiling, and a fingerprinting analysis of probiotic powder samples are provided in Figure 1. Such a method provides TGA (sample mass vs. time/temperature), DTG (TGA first derivative), and DSC (heat flux vs. time/temperature) curves from which a series of unique thermophysical data of each sample can be extracted (Table 2).

3.1.3. Examples of Mono- and Multi-Strain Thermophysical Profiling

Illustrative examples of thermal profiling (Figure 2) and fingerprint data (Table 3) of representative mono-strains and various lots of multi-strain samples are unique. The reproducibility of the method has been demonstrated by an interlaboratory TGA profiles of a same multi-strain formulation [53].
In these examples, probiotic strains can be classified by their thermostability, that is, their main decomposition (Tmax) or transition (Tm) temperatures, extracted from TGA or DSC curves, respectively. For instance, the probiotic sample of B. subtilis appears the most thermoresistant with the highest Tmax = 351 °C and Tm = 353 °C, due to the bacterial cell capacity to sporulate [54], whereas L. bulgaricus is the least thermoresistant, depending on the structure and composition of the strain cell wall. Moreover, the residual material at 600 °C is an indicator of the purity and dose of the probiotic cells, which include in their components a variety of macromolecules, e.g., proteins and polysaccharides (higher R600), compared to the minor additional ingredients, often constituted by smaller compounds (lower R600) [55]. In addition, a variability in the same brand of multi-strain samples prepared in different countries (e.g., VSL#3) can be detected by TGA-DSC analysis [52] due to the difference in the carbohydrate-based protectant used (e.g., maltose, maltodextrin, starch). Figure 3a demonstrates the protectant impact (e.g., maltose) on the multi-strain formulation profile and fingerprint. Both major and minor ingredients in probiotic products are detectable through the present thermal profiling and fingerprinting method. Figure 3b illustrates the difference in thermal profiles between a formulated strain (L. plantarum ATCC 8014) containing lyoprotectants, compared to the same one in pure form.

3.1.4. Advantages of the TGA-DSC Approach

The TGA-DSC profiling and fingerprinting method has many advantages compared to the gold standard approaches (e.g., phenotyping and genotyping) in quality control and authentication of probiotic powder-based products. It instantly reveals information on both probiotic strain and additional functional ingredients (e.g., cryoprotectants and antioxidants) in the formulation. In fact, these additional ingredients can also contribute to the functionality and performance of the product, and deserve careful attention. Moreover, the method is rapid, highly reproducible, sensitive, and adaptable to a high throughput analysis while requiring only a small amount of sample without pre-treatment. Besides the pure analytical aspects, this method can also provide some relevant fundamental information on the thermostability of the probiotic strains and the product formulation itself.

3.2. Advanced Testing for Genetic Composition (ATGC)

Advanced Testing for Genetic Composition (ATGC) can be considered as a “next generation genotyping” technique. It was built upon the use of Cycling Temperature Capillary Electrophoresis (CTCE) developed first in the Thilly Laboratory at MIT [56] and then at the Norwegian Radium Hospital.
ATGC is a targeted measurement technique that provides greater precision, specificity, and versatility than rtPCR, while being just as cost-effective and fast. It has been developed to translate discovery data from untargeted analysis (16S, shotgun metagenomics, etc.) into tests for routine use. Due to its speed and cost, ATGC is ideally suited for probiotic quality control (QC), microbiome monitoring in humans for personalized treatments, and soil microbiome analysis for precision agriculture.

3.2.1. Origins of ATGC

Initially known as “Constant Denaturing Capillary electrophoresis” (CDCE), ATGC was first developed as a technique to detect and quantify genetic variations. In the 1990s it was successfully used to measure the mutational spectrum of γ-DNA polymerase [57], Pfu [58], and β-DNA polymerase [59]. Later, Cycling Temperature during the electrophoresis has been introduced as a means to improve the flexibility of the technique [60,61]. With this addition, the fragment selection process could be automated, and bio-informatic techniques were developed to enable the automatic identification of suitable fragments for separation using CTCE [62]. A first automated algorithm was then developed to design fragments that enable the complete analysis of human mitochondrial DNA [63]. This assay, once established, was successfully used in several lineage tracing applications [64,65,66,67,68]. Transitioning from CTCE to ATGC was achieved through the further development of algorithms and software for the design of assays in more complex genetic mixes (such as microbiomes).

3.2.2. Principles of ATGC

The ATGC workflow has four steps, namely: (1) DNA extraction: the protocol depends on the samples being analyzed; (2) PCR: a classical PCR (not rtPCR) to amplify the target fragments; (3) CTCE: measuring the relative abundance of the different fragment variants in a sample; and (4) data analysis: combining the results of several fragments to produce an accurate profile of a sample’s composition.
CTCE enables an accurate and fast analysis of single nucleotide polymorphisms (SNPs) [61]. Because it relies on the separation of fragments after PCR, where fragments are almost identical (i.e., only one SNP difference), CTCE is not affected by PCR amplification biases, which is a significant drawback of rtPCR [69,70].
A single SNP changes the melting profile of a double-stranded DNA fragment. As the temperature is cycled throughout the electrophoresis, the two variants spend a different amount of time in open and closed configuration, which results in the separations shown on Figure 4.
The area under the peak is a direct estimate of the relative abundance of one variant relative to the other. Leveraging the ability to accurately quantify the relative abundance between two fragments differing by a single SNP, it is possible to design assays in which one primer specifically amplifies two species of micro-organisms. Combining several such primers, one can effectively “cover” a genus, for quantitative and precise profiling. Selecting such primers is not trivial and requires extensive bio-informatics. Using a property platform made available by REM Analytics (Monthey, Switzerland), it is possible to “map” a large number of genetic sequences. The results can be displayed in an interactive 3D map. From Figure 5, it is clear from this map that there are some comparisons that are more appropriate than others. It is easy to compare L. gasseri to L. johnsonii. However, a direct comparison between L. gasseri and L. helveticus would be impractical. It also appears that a grouping of L. gasseri with L. johnsonii to be compared with a group including L. crispatus and L. helveticus would be possible. However, the genetic map of Lactobacillus species and subspecies is different compared to that of Bifidobacterium (Figure 6). B. breve and B. longum form a clique, whereas B. animalis behaves almost like a separate genus, rather than a species within a genus. From this map, it is inferred that Bifidobacterium must be analyzed at the subspecies level, and that the species level analysis provides insufficient information.
From Figure 5 and Figure 6, it is possible to intelligently select primers of interest. Looking at the map in Figure 6, we see that individual Bifidobacterium species behave almost like genera, which have several sequences clustered together. Thus, further detailed maps are required for each individual species. Figure 7 shows a map of subspecies of B. longum. It can be seen that B. longum subspecies longum and infantis form distinct clusters, making the design of a subspecies primer effective and meaningful.

3.2.3. Probiotic Quality Control Application

Guarantying the composition of multi-strain probiotics is a challenge in quality control [71]. Classical microbiology techniques based on selective culturing cannot distinguish between species and/or subspecies of the same strain. Furthermore, these techniques take several days to produce a result, days in which a batch cannot be released from production. Recently the use of antibody binding followed by flow cytometry [72] was proposed. However, polyclonal antibodies remain difficult to use in routine QC applications, and the transition from polyclonal to monoclonal antibodies is very expensive.
ATGC, with a fast assay design process, specificity, sensitivity, and precision, provides a valid alternative. Assays can be designed for species-level discrimination, and subspecies or strains. Several mixtures of species are first produced and measured by ATGC. These mixtures are then added together in equal volume, and the relative abundance measured in the original mix is used to predict the relative abundance in this second mix. The results of four samples run in triplicate show an average discrepancy between observed and predicted values of 2% with a 95% confidence interval of ±4% (Figure 8).

3.2.4. Human Microbiome Application

ATGC is an ideal tool to bring human microbiome measurements from a “discovery phase”, in which the whole spectrum of possible micro-organisms is investigated with NGS, to a validation and diagnostic phase, in which specific assays are used to analyze subsets of the microbiome.
For example, in the vaginal microbiome, large-scale studies using sequencing techniques [73] have demonstrated that only a few species of Lactobacillus are present in the healthy vagina. Relying on these data, REM Analytics has developed a vaginal microbiome assay that produces quantitative and reliable results on the main micro-organisms species of the woman’s reproductive tract: L. iners, L. crisptus, L. helveticus, L. jensenii, L. gasseri, and G. vaginalis. Yoni Solutions (Monthey, Switzerland) is currently commercializing this assay, to be used in personalizing microbiome treatments for women suffering from recurrent vaginal infections or implantation failure.
Similarly, for baby gut microbiomes, existing evidence demonstrates that the ability of babies to metabolize different human milk oligosaccharides (HMOs) is determined by their population of Bifidobacterium [74]. In this case, subspecies of B. longum and B. animalis must be distinguished in order to better predict the HMO metabolism, which is in turn important for the recommendation of specific formula, or of probiotics to infants with digestive troubles. REM Analytics is currently commercializing a subspecies level Bifidobacterium assay for research uses only, and testing it as part of the NUTRISHIELD project (H2020 Grant agreement number 818110).
As the examples above illustrate, ATGC has great potential to be used in specific diagnostics when the target list of micro-organisms has been identified. It can also be used with data from metabolic studies, enabling the identification of key micro-organisms involved in a function and deemed important. Furthermore, it can be used in combination with probiotics to monitor specific changes induced by the probiotics.

3.2.5. Soil Microbiome Application

Soil is a complex ecosystem in which several genera of bacteria interact with fungi and other organisms (not always micro). Obtaining a general profile of the soil microbiome is difficult, but can be achieved using NGS with a very large number of reads. However, such general profiles are not useful for precision agriculture. Chemical soil analyses measure specific micro-nutrients and elements of known relevance to crops. Similarly, microbiological analyses must develop specialized assays focusing on known functions for which a clear corrective strategy exists when deficiencies are identified.
Leveraging the significant advances in soil microbiome of recent years, it is possible to identify specific families of micro-organisms with known effects on soil. For example, the Bacillus, and Bacillus-related genera, are known beneficial microbes for soil, with several having well-characterized roles in crop growth. Furthermore, the number of such micro-organisms available on the market to be used as biofertilizers or biopesticides is growing fast. A list of species within the Bacillus family targeted by an existing ATGC assay is shown in Table 4. Each element has well-known functions, and is available for purchase on the market while being approved by regulatory authorities for use in agriculture. The same approach can also be developed to study mycorrhizal fungi for improving their use in agriculture.

3.2.6. Limitations, Challenges, and Future Developments

ATGC, like most other microbiome profiling techniques, requires accurate DNA databases to design assays, in addition to high-quality reference material to calibrate and validate such assays. In the microbiome field, such material can be difficult to find. This is especially true in niche fields, such as women’s health, in which very few reference genomes are available for most bacteria. Furthermore, bacterial libraries such as ATCC or DSMZ have a limited number of strains isolated from women’s genital track that can be used as reference material.
ATGC is less susceptible to poor sequence databases than NGS or 16S, since assays can be calibrated once designed, and their specificity, precision, and detection profiles are well established once they are deployed. This means that having reliable reference material remains critical. In the agricultural field, there are several laboratories with extensive libraries of agricultural micro-organisms. This makes the acquisition of high-quality reference material straightforward.
In the area of probiotic quality control, reference material is available and has high quality. The precision, sensitivity, speed, and cost of ATGC makes it therefore a perfect tool for monitoring the relative abundance of strains in mixed probiotics, ensuring consistency across batches. Demonstrative results from internal research (not yet published) exist with the following types of probiotics:
  • Lactobacillus and Bifidobacterium
  • Spore-forming (e.g., Bacillus)
  • Gram-negative (e.g., Hafnia alvei, E. coli Nissle)

4. Applications and Action Mechanisms

4.1. Human Health

Humans are a reservoir of diverse group of microbes, which together constitute the human microbiome. This microbiome plays a key role in modulating the host internal environment, defending the body against infectious organisms and maintaining the health of humans [84]. The emergence of superbugs resistant to commonly used antibiotics suggests that the development of simple, low-cost, and intrinsic approaches to maintaining health are crucial. Probiotics have been shown to supplement the host microflora and protect against various pathogens by improving gut barrier function and activate specific genes in host cells, thereby stimulating the host’s immune response [85]. The gut microbiota in humans exert systemic effects on host health, metabolism, nutrition, and the immune system, which accounts for their designation as a “hidden metabolic organ” [86]. The evolution of the gut microflora from birth through adulthood is influenced by diet, genetic make-up, lifestyle and age of the host, and use of antibiotics [84]. Imbalances in the composition and function of intestinal microbes, referred to as gut dysbiosis, are associated with various human diseases [87]. Consequently, manipulation of intestinal microbiota, through diets that stimulate beneficial bacteria colonization of the GIT [88] and the administration of probiotics [89], holds promise for maintaining health and treatment of diseases. A shift from the healthy symbiosis between the microbiota and the host to persistent dysbiosis has also been identified as a factor in obesity [90]. Probiotics supplement host microflora and provide protection against various enteric pathogens, with demonstrated remarkable functional attributes for meeting most of the basic human nutritional and clinical supplementation requirements [84]. Although probiotics are essentially beneficial gut microorganisms, some species of probiotics are not part of the normal human gut flora, and the beneficial effects observed are not the same for different strains [91]. The majority of probiotics are species from three genera, viz., Lactobacillus, Bifidobacterium, and Saccharomyces [92,93,94]. The most-used vehicles for prebiotic administration have been pharmaceutical formulas and dairy products [95].
Probiotics have antipathogenic, antidiabetic, anti-obesity, anti-inflammatory, anticancer, anti-allergic, anti-anxiety, and angiogenic properties in humans [84]. These properties have been successfully harnessed to induce remission in ulcerative colitis [96] and reduce both weight and blood pressure [97]. Probiotics have also been shown to ameliorate infection and antibiotic-associated diarrhea, Clostridium difficile–associated diarrhea, and conditions such as allergic rhinitis and atopic dermatitis (eczema) [97]. Further research is required into the long-term utility and safety of probiotics in various disease conditions. Probiotics have been used to treat gastrointestinal (GI) and non-GI conditions that include traveler’s diarrhea, acute infectious diarrhea in infants and children, antibiotic-associated diarrhea, irritable bowel syndrome, and ulcer and atopic dermatitis [98,99], with effects also exercised on the brain and central nervous system [84,100] and cancer cells [101]. The advantages of probiotics are, however, more clearly demonstrated for GI-related diseases [98]. Probiotics significantly reduce the risk for diarrhea [102,103], with greater effectiveness obtained in children than adults [104]. Their effectiveness at reducing the frequency of antibiotic-associated diarrhea has also been demonstrated [103,105,106]. Probiotic strains L. fermentum NCMB 52221 and 8829 have shown considerable potency for suppressing colorectal cancer cells in vitro [101]. Probiotics and their fermented metabolites (postbiotics) have shown activities that counter oxidative stress, a factor in ageing, in middle-aged mice [95].
Probiotics have been trialed as a therapy for necrotizing enterocolitis (NEC). NEC is a serious inflammatory gastrointestinal disease that primarily affects premature infants and has a mortality rate as high as 50%. A Cochrane review with a meta-analysis of twenty-four eligible trials involving preterm infants <37 weeks gestational age or <2500 g birth weight showed that enteral probiotic supplementation significantly reduced the incidence of severe NEC (≥stage 2) and no systemic infection with the probiotic organism was reported in the trials [107]. Putative mechanisms for probiotic action in the gut include: (1) upregulation of cytoprotective genes; (2) competition with other microbes; (3) downregulation of pro-inflammatory gene expression; (4) production of butyrate and other short chain fatty acids that nourish colonocytes; (5) support of barrier maturation and function; and (6) regulation of cellular immunity and Th1:Th2 balance [97]. Outstanding issues to address include determining which probiotic to use, whether infants <1000 g benefit, and how to mitigate the risk of probiotic sepsis.
Research in animal models has shown that important components in mammalian milk, such as sialylated galacto-oligosaccharides (GOSs), reduce the occurrence of NEC in neonatal rats [108]. This could account for the 6–10-fold lower NEC risk in breast-fed infants compared to formula-fed infants. Indeed, GOSs appear to shape the components of the intestinal microbiome. Complex polysaccharides such as β-glucan (BGL) with anti-inflammatory properties have also shown promise in boosting growth performance and intestinal epithelium functions in weaned pigs, and hens [109,110]. More research is needed into the applicability of BGL in managing gastrointestinal inflammatory conditions such as NEC in humans.
Probiotics also play an important role in dentistry, since oral infections are considered prime among other infections affecting humans. Effects of probiotics on oral health are both direct and indirect. Some probiotics produce digestive enzymes for metabolizing proteins and carbohydrates. Several randomized clinical trials have shown the possible benefits of probiotic dairy products for oral health in children, adolescents, adults, and the elderly [95]. These studies indicate a role for probiotics in caries prophylaxis. The incorporation of probiotics into dairy products is due to their ability to neutralize acidic conditions that promote dental caries, the irreversible microbial disease of the calcified tissues of the teeth [111], and suppression of the caries pathogen. Given consumers’ concerns about allergens and lactose intolerance in respect of traditional dairy food matrices, there have been concerted efforts towards the development of cereals, soy, fruits, vegetables, and chocolate as innovative food matrices [112,113]. Although most probiotics are safe, they may sometimes come with side effects that include constipation, flatulence, hiccups, nausea, infection, and rashes [98]. In recent years, probiotic strains have been considered a powerful ally in fighting and preventing respiratory tract infections [99]. Reduction in upper and lower respiratory tract infections from the administration of probiotics bacteria has been reported [114]. The increasing evidence between gut and lung function, resulting from gut–lung cross-talk, suggests a possible role for probiotics in the management of COVID-19, caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) that assumed a pandemic status in February 2020 [115].
In terms of action mechanisms, probiotics are involved in the maintenance of health through diverse and interconnected mechanisms. Probiotics produce vitamins, enhance nutrient absorption, and possess enzymatic activities, such as β-glucurodinase, β-galactosidase, and bile salt hydrolase, among other, that are essential for the host metabolism [97,116]. Probiotics modify microbiota populations through the production of short chain fatty acids (SCFAs), which alter luminal pH, and antimicrobial compounds, such as bacteriocin [117]. Probiotics stimulate the production of mucin glycoproteins and secretory immunoglobulin A (sIgA) by globet and B cells, respectively [117]. Mucin is necessary for probiotic adhesion to the intestinal mucosa, while impairing the adhesion of pathogen bacteria. sIgA serves as the first line of defense in protecting the gut from pathogens. Probiotics further modulate the immune system by interacting with toll-like receptors, thereby leading to the activation of the innate immune response; activating T-regulatory cells; and increasing the production of anti-inflammatory cytokines and reducing proinflammatory cytokines [97,118]. The production of SCFAs also plays an important role in the immune and inflammatory responses [119]. Moreover, SCFAs activate insulin sensitivity and fatty acid oxidation in muscle, decrease lipolysis and increase adipogenesis in adipose tissue, and enhance satiety through the stimulation of intestinal glucagon-like peptide 1 secretion [97,120]. The relevance of the gut microbiome on distal organs has led to defining the terms gut–brain, gut–lung, and gut–skin axes, among others. In nervous system disorders, the production of neuroactive compounds plays a significant role [121]. Finally, the interplay between the gut microbiome and other host microbiomes (lung, skin) is thought to contribute to the development of respiratory and skin diseases, in addition to the mitigation of symptoms [122,123,124,125]. Figure 9 illustrates the action mechanisms of probiotics for promoting human health.
When probiotics are mixed with prebiotics, the resulting synbiotic preparation can develop either complementary or synergistic actions for human health [36]. Synbiotics help to manage several disease pathologies by targeting host gut microbiota, which play a crucial role in metabolism and protection against pathogens [126,127,128]. Synbiotics can act in balancing the gut microbiota by adjusting the Firmicutes-to-Bacteroidetes ratio, inhibiting harmful bacteria through direct antagonism (such as Klebsiella, Escherichia coli, and C. difficile) or excluding the latter by competitive adhesion, and accelerating the recovery to a healthy gut microbiome, e.g., by maintaining intestinal pH, producing important metabolites, and improving the gut mucosal barrier [129]. Health claims from clinical studies on synbiotics are linked to the gut health in order to treat inflammatory bowel syndrome (IBS) and inflammatory bowel disease (IBD), metabolic disease, and colorectal cancers. Other health claims relate to the treatment of systemic diseases such as allergies, hypocholesterolemia, osteoporosis, hepatic encelphalopathy; gut–brain axis diseases such as autism, depression, and anxiety [130]; and gut–lung axis respiratory diseases such as SARS-CoV-2 pathogenesis [131,132]. Some selected examples of synbiotic health benefits claimed from clinical studies are listed in Table 5. For further clinical trial results from studies conducted around the world, some databases are available online (https://www.clinicaltrials.gov/, accessed on 8 June 2021; https://www.clinicaltrialsregister.eu/, accessed on 1 January 2021).
An alternative strategy to achieve the human health benefits of probiotics is the administration of bioactive compound-based preparations derived from probiotics, i.e., postbiotics [43]. They have several advantages over probiotics in terms of safety and production costs. Postbiotic health benefits rely on their antimicrobial, antioxidant, anticancer, and immunomodulatory potentials [44]. The postbiotic compounds with antimicrobial activities include bacteriocins and other peptides, SCFAs, organic acids, and hydrogen peroxide. The probiotic antioxidant enzymes catalase, superoxide dismutase, and glutathione peroxidase reduce the concentration of reactive oxygen species. Bacteriocins, and, in particular, enterocin, have cytostatic and apoptotic effects against cancer cells. The health-promoting effects of postbiotics include favoring mineral absorption, relieving constipation, preventing intestinal inflammation, controlling glycaemia, and reducing food allergies. Recent clinical trials have demonstrated the impact of postbiotics in a wide age range of individuals. In infants, the inclusion of postbiotics in an infant formula modifies the fecal microbiome and metabolome towards a profile closer to that observed in breast-fed infants [142]. In middle-aged individuals, the intake of urolithin A, a postbiotic metabolite of ellagitannins, improves muscle performance [143]. Figure 10 summarizes the potential applications of postbiotics in promoting human health.

4.2. Animal Health

The main use of probiotics, prebiotics, postbiotics, and synbiotics in animal feeding is associated with their verified efficacy in modulation of the intestinal microbiota. Administration of probiotic strains, both individual and combined, may have a significant effect on absorption and utilization of feed, resulting in a daily increase in body weight and an increase in total body weight of various animals, including turkeys, chicken, piglets, sheep, goats, cattle, and horses. Probiotic microorganisms mostly intended for animals include Lactobacillus (e.g., L. brevis, L. casei, L. crispatus, L. farciminisa, L. fermentum), Bifidobacterium (e.g., B. animalis, B. longum, B. pseudolongum, B. thermophilum), other lactic acid bacteria (e.g., Enterococcus faecalis and faecium, Lactococcus lactis, Leuconostoc citreum, Pediococcus acidilactici) and some species of Bacillus, Saccharomyces, Kluyveromyces, and Aspergillus [144].

4.2.1. Poultry

In recent decades, antibiotics have been widely added to poultry diets to maintain animal health and to prevent enteric diseases, which would impair productivity, increase mortality, and contaminate poultry products for human consumption [145]. Increased bacterial resistance to antibiotics in humans has caused an increase in public and governmental interest in eliminating sub-therapeutic use of antibiotics in livestock. Prebiotics and probiotics are two alternative approaches that can alter the intestinal microbiota and immune system to inhibit colonization by pathogens and therefore have the potential to prevent enteric diseases in poultry production [146]. The application of probiotics in poultry is strictly associated with the concept of competitive exclusion (CE) [147], which protect chicks against C. jejuni, Listeria monocytogenes, pathogenic E. coli, Yersinia enterocolitica, and C. perfrigens [148]. Furthermore, Lactobacillus-based probiotic cultures significantly reduced Salmonella enteritidis recovery in challenged neonatal broiler chicks [147].
The supplementation of synbiotics increased average daily gain but reduced the feed/gain ratio in broilers from 1 to 42 days of age. Moreover, dietary synbiotic inclusion increased breast yield and decreased abdominal fat in broilers [146]. By contrast, synbiotic supplementation lowered the cooking loss during heat treatment in a water bath, malondialdehyde (MDA) content, and total Cr content in the thigh muscle in broilers [146]. Regarding meat quality, lipid peroxidation is one of the most common causes of meat quality degradation in chicken, and can reduce nutritious value, produce taste and texture issues, and change the look of the meat [146]. By reducing MDA accumulation in the thigh muscle, synbiotic supplementation reduced meat lipid peroxidation, which may be favorable to meat quality and shelf life [146]. Supplementation of synbiotic to broilers’ diet at the dosage of 1.5 g·kg−1, composed of probiotics (B. subtilis, Bacillus licheniformis, and Clostridium butyricum) and prebiotics (yeast cell wall and xylooligosaccharide), can promote growth performance (increased weight gain and feed utilization efficiency), improve carcass characteristics (elevated breast muscle yield and reduced abdominal fat yield) and meat quality (increased pH24 h value in the breast muscle and decreased cooking loss in the thigh muscle), and reduce the product of lipid peroxidation (MDA) and Cr accumulations in the thigh muscle in broilers [146]. A probiotic strain, L. plantarum UY, inhibited the proliferation of influenza A (IFV) virus in the animal lung in a dose-dependent manner [149] and also stimulated the Thelper cells type 1 (Th1) immune response, resulting in higher synthesis of secretary IgA, leading to the removal of IFV from the lung.

4.2.2. Pigs and Piglets

Pigs have specific immune and intestinal functions and weaning is a difficult period that can lead to a reduction in growth performance. Consequently, during this period, pigs are highly susceptible to pathogenic microorganisms, such as enterotoxigenic Escherichia coli (ETEC), causing enteric diseases [114,150]. Typical plant-based feeds contain 2.3–3.8% of xylans, which can reduce nutrient digestibility and induce a propitious environment for the growing of harmful bacteria, changing the gut associate microbiota in newly weaned pigs [151]. The application of LAB probiotics has been linked by several authors with beneficial effects in models of gastrointestinal infection using small animals. However, reports of the efficacy of probiotic treatment in alleviating intestinal infection in large animals remain scarce [152]. Synbiotics enhanced growth performance by reducing diarrhea, immune response, and oxidative stress in the jejunum [153]. In fact, exogenous enzymes (e.g., xylanase) have been successfully used to hydrolyze the β-1,4 backbone of xylan, releasing xylan oligosaccharides (XOSs) and, consequently, reducing the non-starch polysaccharide (NSP) content and the viscosity of digesta, increasing the digestibility of nutrients [154,155]. The use of the synbiotic mixture associated with 0.05% of herbal mixture showed an average weight gain [151].
The use of synbiotics promoted a smaller villus:depth crypt ratio, except when associated with 0.1% of herbal mixture, which was similar to the diet without additives [151]. Supplementation with a combination of a probiotic originating from anaerobic microbiota (bacteria—109 CFU/mL, yeast—105 CFU/mL, molds—103 CFU/mL) and a prebiotic (malto-oligosacharides, sodium acetate, ammonia citrate) results in improved digestion of nutrients and reduced emission of harmful gases, and prevents bacterial infections during the weaning period [156].

4.2.3. Ruminants

In recent decades, there have been considerable improvements in ruminant production, and these advances must continue in order to meet growing demands. Currently available data regarding effects of synbiotics on animal health are insufficient and require further studies. However, they clearly indicate the effective synergistic action of probiotics and prebiotics in the reduction of populations of bacterial gastrointestinal pathogens [157]. A method to manipulate the microbiota of the rumen during its growth period is to directly provide activators and/or probiotics, to establish a balance in the microbiota, which is more efficient during growth than in adults [158]. Recent studies suggest that integrating pre- and probiotics into ruminant feeds may improve various aspects of ruminant performance, mitigate disease, promote overall animal health and well-being, and reduce the environmental impacts of ruminant production. LAB and yeasts (S. cerevisiae) are used as ruminal activators/probiotics for their ability to affect the dynamics of the microbiota in the rumen and the way in which nutrients are decomposed [159]. It has been confirmed that individual or combined supplementation with Saccharomyces cerevisiae and L. acidophilus improved the growth performance of growing goats. Synbiotics provide these benefits by favorably modulating the microbial environment within the gastrointestinal tract of ruminant animals [160]. Although the mechanisms of action exerted by probiotics on ruminants are not well elucidated, dietary probiotic dosage to ruminants enhances development and maturation, growth and performance, milk production and composition, nutrient digestibility, feed efficiency, pathogen reduction, and mitigation of gastrointestinal diseases [161]. The addition of mix feed additives can affect the kinetics of gas and methane production, and not the level of pH or dry matter and organic matter digestibility. In dairy cows, probiotics containing live yeast boosted food intake, improved feed efficiency, average daily gain, and total weight, and increased milk yield and quality [162,163]. Probiotics and prebiotics, alone or in combination, in the diet of lambs finished under subtropical climate conditions may assist in reducing the unfavorable effects of high ambient heat load on dietary energy utilization [164]. Lambs supplied with probiotics and/or prebiotics showed higher gain efficiency and a lower ratio of observed-to-expected diet net energy compared to controls, with little influence on carcass features, whole cuts, or visceral mass. Table 6 lists the effects of some species of microbial pre-/pro-/synbiotics administrated under defined mode and dose conditions on various ruminant hosts.

4.2.4. Fish

Application of biofriendly feed additives such as probiotics, prebiotics, and synbiotics are becoming popular dietary supplements with the potential to not only improve growth performance, but, in some cases, to also enhance immune competence and the overall well-being of fish and crustaceans [176]. Probiotics not only improve the health status of cultured animals but also help to ensure the safety of consumers [177]. The most commonly used are bacterial probiotics strains (e.g., Bacillus sp., Lactobacillus sp., Bifidobacterium sp., Pseudomonas sp., Streptococcus sp., Arthrobacters sp., Microbacterium sp., Phaeobacter sp., Streptomyces sp., Enterococcus sp., Lactococcus sp., Micrococcus sp., etc.), yeast probiotics (e.g., S. cerevisiae, Debrayomyces hansenii), micro-algal probiotics (e.g., Tetrasehnis suecica, Spirulina platensis), and bacteriophages probiotics (e.g., Bacteriophages sp.) [176]. The main roles played by probiotics in fish are: (1) improvement in growth and feed utilization of aquaculture species; (2) assist with the provision of essential nutrients and micronutrients such as vitamins and essential fatty and amino acids to the host species; (3) improvement in hemato-biochemical parameters as they allow a significant increase in the abundance of erythrocytes, but also elevate the number of white blood cells (WBCs), the latter enhancing non-specific immunity associated with neutrophils and macrophages; (4) improvement in fish culture systems through enhanced disease resistance, in addition to general health benefits to fish [176,178,179,180]. Moreover, under stressful situations, fish experience oxidative stress, causing lipid peroxidation and excessive malondialdehyde production (MDA) [181], which threaten the functionality of body tissues and cells and pose a risk of DNA damage [182]. A recent study showed that dietary Pediococcus acidilactici (PA) and pistachio hull-derived polysaccharide (PHDP) + PA used as a synbiotic resulted in low MDA levels in Nile tilapia, thus improving the antioxidative capacity [183]. Furthermore, synbiotics can improve the quality of water with beneficial influences on fish production. Gram-positive bacteria (e.g., Bacillus sp.) efficiently convert organic substances into carbon dioxide, whereas Gram-negative bacteria convert relatively more organic matter into biomass or bacterial slime [184]. For example, Lactobacillus sp. used as a probiotic simultaneously eliminated nitrogen and pathogens from polluted shrimp farms and then decreased fish mortality [185].

4.3. Plant and Soil Health

In the past decade, probiotics have been much applied to a wide range of industries such as aquaculture, food industries, human medicine, and agriculture. Some studies have been focused on successful practices, mechanisms of probiotics activities, and methods for optimizing the successful use of strains [186,187]. According to research results published in agriculture fields, the microbiome community known as probiotics can offer benefits to plant growth promotion, nutrient use efficiency (Figure 11), and pest and phytopathogen control [188,189]. Although many authors have demonstrated the interactions of probiotics with plants, a very little knowledge is available in the literature on the action mechanisms of prebiotics in the ecosystem. Results from research on forest ecosystems showed that fungal and bacterium communities can respond to environmental changes in accordance with host trees [190]. Vassilev et al. [191] demonstrated that Piriformospora indica, a beneficial microorganism for plants, can be used to produce a phosphate-enriched fermentation liquid through a repeated-batch fermentation process for improving soil fertility and plant productivity. In other work, it was proved that Bacillus amyloliquefaciens BChi1 and Paraburkholderia fungorum BRRh-4 can also increase growth and fruit yield of strawberries, and enhance their functional properties, such as the content of total antioxidants, carotenoids, flavonoids, phenolics, and anthocyanins [192]. In addition, other work demonstrated that microbial and biochemical indicators of soil health can be used to assess the ecological risk of soil. These results confirmed that soil respiration can be used for estimations of the soil ecological conditions and microbiological activity [193].
Basically, the ecosystem has been defined as a system of two components, constituting living organisms and inanimate or physical factors, respectively called biotic and abiotic components [195]. Biotic components comprise animals, microbial organisms, and plants, which are fed by nutrients, among which prebiotics and postbiotics constitute important elements. Prebiotics are molecules capable of stimulating both the intestinal microflora and other bacterial populations, including those growing in agricultural soils, by improving plant and soil health. Diverse sources of plant prebiotics such as fructo-oligosaccharides (FOSs), inulin, and galacto-oligosaccharides (GOS) are commonly cited, but xylans, pectins, and fructans are also among substrates used as carbohydrate-based prebiotics [196]. Information on postbiotics is very limited and associated research is quite recent. However, their role has been tested recently on animal, human, and plant health. It has been reported that postbiotics contribute to promoting plant growth by enhancing proliferation of shoots and rooting, and also having biocontrol effects on plants [194,197]. Limited studies on the effect of postbiotics on plants are available. Indeed, these derivate molecules from the plant probiotic microorganisms’ metabolism play mediating roles between probiotics and plants, acting as plant growth activators or in the defense of plants against certain stresses. Postbiotics interact via biochemical mechanisms with plant cellular membrane receptors through transduction of systemic signals, which leads to changes in plant gene expression at the plant level [198]. A large number of molecules obtained from probiotics activities act on plants, and significantly contribute to enhancing plant health performance, such as in terms of growth, yield, and resistance to stresses (biotic and abiotic).

4.4. Environmental Health

Probiotics play an important role as remediation agents, helping the host in responding to environmental changes. Certain genera also act as bioremediation or decomposing agents of hazardous substances [10], such as the case of a bacterial consortium (Xanthomonadaceae, Brachybacterium sp., Bhargavaea sp., Gordonia sp., Thalassospira sp., Pseudomonas sp., Dietzia sp., Mesorhizobium sp., Cytophaga sp., Martelella sp.), providing an innovative bioremediation approach. In this work, chitosan used as an encapsulated agent can stimulate the bacterial community of mangrove sediments [199]. Bioremediation in this case is based on the use of probiotics to degrade, reduce, or remove pollutants in the environment. The working mechanism of bioremediation involves several technical aspects such as biotransformation, biodegradation, mineralization, phytohydraulics, bioaccumulation, and biovolatilization, where the degrading microbes remove, transform, modify, and/or convert a complex compound of pollutants into simpler and less-toxic compounds. This bioremediation system has been successfully applied in cleaning contaminated sites [200], agricultural land [201], ground water [202], surface water [203], and sea water [204].
Conventional remediation strategies for most types of environmental contamination are not only expensive but also ineffective, especially in low contaminant concentrations [9]. Probiotics-assisted remediation has come forward as a cheap and easy alternative. Probiotics can act through four main action mechanisms divided into two categories, the binding and enzymatic degrading activities of toxins and pollutants, as summarized in Figure 12. LABs, yeasts, and soil probiotic bacteria are able to bind both organic toxins (e.g., mycotoxins and pesticides) and nonorganic pollutants (e.g., heavy metals) [3] through chelation, adsorption, and precipitation mechanisms. The nature and structure of the cell wall, surface macromolecules such as S-layer proteins and polysaccharides, and the environment conditions (e.g., pH and temperature), are among the factors that control the binding capacity of probiotics, which in turn depends on the surface hydrophobicity and electrical charge [3]. The binding mechanisms of toxins may also result from physical degradation of petroleum hydrocarbons [205]. Another mechanism is the production of enzyme-degrading toxins, such as organophosphorus-based pesticides [206], or proteolytic activity [207].
By combining probiotics with prebiotics, the resulting synbiotics are expected to develop a higher detoxifying capacity since prebiotics support the viability and functionalities of probiotics, which can improve the binding capacity of the mixture. The rare research work conducted on the synbiotic beneficial effects on bioremediation involved the combination of Lactobacilli and Bifidobacteria with inulin (prebiotic) for removing various substances, especially Pb [208].
Concerning postbiotics, it has been reported that both viable and nonviable LABs were able to bind toxic secondary metabolites such as fumonisin [209]. As the removal of mycotoxins involves an adhesion-type mechanism to cell wall components, rather than a covalent binding or binding by the metabolism, dead cells retain their binding ability [210]. Another case of postbiotic activity demonstrated in vivo was the removal of ochratoxin A from a liquid medium of foods using a mixture of sterilized yeast and a fermentation residue of beer (40:60). The binding action for toxin removal implied physical interactions with the cell wall since the changes in pH affected the degree of the activity [211].

5. Conclusions

As natural microbial-based and multifunctional materials, the probiotic-based multi-components described in this review article refer to bioactive agent mixtures derived from alive (probiotics and synbiotics) and nonalive (postbiotics) probiotic cells. In such a preparation, the microbial-derived components constitute the main functional ingredients, whereas prebiotics, protectors, stabilizers, and encapsulating agents are among the additional ingredients. Analyzing, characterizing, and monitoring the traceability, performance, and stability of such multi-component ingredients over time require convenient and sensitive measuring methodologies such as the TGA-DSC calorimetric technique and ATGC genetic strain analysis up to subspecies. Such methodologies are able to provide qualitative and quantitative profiling of both microbial- and non-microbial components in the preparation, as overviewed and discussed in this article. The applications of probiotic-based multi-components are not limited to human and animal health, but also extend to the promotion of the health of plants, the soil, and the environment, that is, our ecosystem health. Now, they can be used as biosupplements in food and feed, biopesticides and biofertilizers for promoting plant and soil health, and bioremediation and depolluting agents for cleaning up and protecting the environment. A large amount of effort is still needed to obtain insight into their multiple and complex mechanisms of action through unimaginable interactions among microbial and non-microbial components. The use of an efficacious combination of living and non-living entities from natural resources is among the promising “one health” approaches for tackling the disruption of human, animal, plant, and environmental health arising from climate change, urbanization, ocean acidification, and other calamities at the possible origin of emerging infectious diseases and epidemics worldwide.

Author Contributions

Conceptualization, review and editing, H.L.R.; drawing original pictures, H.N.R. and J.M.B., S.K., K.A., M.M., A.G., P.R., A.O., E.E., B.E., V.O.A., J.M.B., O.T., H.N.R. and H.L.R. contributed to the writing of various sections of the article. All authors have read and agreed to the published version of the manuscript.

Funding

This work has received funding by the European Union’s Horizon 2020 Research and Innovation Program through NUTRISHIELD project (https://nutrishield-project.eu/) under Grant Agreement No. 818110, European Union’s Horizon Europe Research, innovation program through URBANE under the grant Agreement 101059232 and by the Swiss State Secretariat for Education (SERI) under the contract number 22.00083. This paper reflects only the authors views; the European Union is not liable for any use that may be made of the information contained therein.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Fuller, R. Probiotics in Man and Animals. J. Appl. Bacteriol. 1989, 66, 365–378. [Google Scholar] [CrossRef] [PubMed]
  2. Song, D.; Ibrahim, S.; Hayek, S. Recent Application of Probiotics in Food and Agricultural Science. In Probiotics; Rigobelo, E., Ed.; InTech: London, UK, 2012; ISBN 978-953-51-0776-7. [Google Scholar]
  3. Zoghi, A.; Khosravi-Darani, K.; Sohrabvandi, S. Surface Binding of Toxins and Heavy Metals by Probiotics. Mini Rev. Med. Chem. 2014, 14, 84–98. [Google Scholar] [CrossRef] [PubMed]
  4. Ebenso, B.; Otu, A.; Alessandro, G.; Philipe, C.; Victor, A.; Razafindralambo, H. Nature-Based One Health Approaches to Urban Agriculture Can Deliver Food and Nutrition Security. Front. Nutr. 2022, 9, 1–9. [Google Scholar] [CrossRef] [PubMed]
  5. Lee, N.-K.; Paik, H.-D. Prophylactic Effects of Probiotics on Respiratory Viruses Including COVID-19: A Review. Food Sci. Biotechnol. 2021, 30, 773–781. [Google Scholar] [CrossRef]
  6. Razafindralambo, H. Trends in Probiotic Applications; Studium Press LLC: Houston, TX, USA, 2018; ISBN 1-62699-110-3. [Google Scholar]
  7. Singh, K.; Rao, A. Probiotics: A Potential Immunomodulator in COVID-19 Infection Management. Nutr. Res. 2021, 87, 1–12. [Google Scholar] [CrossRef]
  8. Soleymani, S.; Ebrahimi, F.; Rezaeizadeh, H.; Rahimi, R. Probiotics and Cancer. In Nutraceuticals and Cancer Signaling: Clinical Aspects and Mode of Action; Jafari, S.M., Nabavi, S.M., Silva, A.S., Eds.; Food Bioactive Ingredients; Springer International Publishing: Cham, Switzerland, 2021; pp. 467–527. ISBN 978-3-030-74035-1. [Google Scholar]
  9. Goyal, P.; Belapurkar, P.; Kar, A. A Review on in Vitro and in Vivo Bioremediation Potential of Environmental and Probiotic Species of Bacillus and Other Probiotic Microorganisms for Two Heavy Metals, Cadmium and Nickel. Biosci. Biotechnol. Res. Asia 2019, 16, 01–13. [Google Scholar] [CrossRef]
  10. Helmy, Q.; Kardena, E.; Gustiani, S. Probiotics and Bioremediation. In Microorganisms; IntechOpen: London, UK, 2019. [Google Scholar]
  11. Yadav, M.K.; Kumari, I.; Singh, B.; Sharma, K.K.; Tiwari, S.K. Probiotics, Prebiotics and Synbiotics: Safe Options for next-Generation Therapeutics. Appl. Microbiol. Biotechnol. 2022, 106, 505–521. [Google Scholar] [CrossRef]
  12. Cavattoni, F. Probiotic Products: Technological and Regulatory Aspects. In Trends in Probiotic Applications; Hary Razafindralambo: Houxton, TX, USA, 2018; pp. 3–12. [Google Scholar]
  13. Cottet, C.; Ramírez Tapias, Y.A.; Delgado, J.F.; de la Osa, O.; Salvay, A.G.; Peltzer, M.A. Biobased Materials from Microbial Biomass and Its Derivatives. Materials 2020, 13, 1263. [Google Scholar] [CrossRef]
  14. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S. Expert Consensus Document: The International Scientific Association for Probiotics and Prebiotics Consensus Statement on the Scope and Appropriate Use of the Term Probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
  15. Rabetafika, H.-N.; Razafindralambo, A.; Razafindralambo, H. Probiotics from Food and Non-Food Sources. Trends Probiotic Appl. 2018, 53, 33–49. [Google Scholar]
  16. Oelschlaeger, T.A. Mechanisms of Probiotic Actions—A Review. Int. J. Med. Microbiol. 2010, 300, 57–62. [Google Scholar] [CrossRef]
  17. Zambori, C.; Cumpănăşoiu, C.; Moţ, D.; Huţu, I.; Gurban, C.; Tîrziu, E. The Antimicrobial Role of Probiotics in the Oral Cavity in Humans and Dogs. Anim. Sci. Biotechnol. 2014, 47, 126–130. [Google Scholar]
  18. Muller, J.A.; Ross, R.P.; Fitzgerald, G.F.; Stanton, C. Manufacture of Probiotic Bacteria. In Prebiotics and Probiotics Science and Technology; Springer: New York, NY, USA, 2009; Volume 1, pp. 725–759. [Google Scholar]
  19. Baral, K.C.; Bajracharya, R.; Lee, S.H.; Han, H.-K. Advancements in the Pharmaceutical Applications of Probiotics: Dosage Forms and Formulation Technology. Int. J. Nanomed. 2021, 16, 7535. [Google Scholar] [CrossRef]
  20. Huang, S.; Vignolles, M.-L.; Chen, X.D.; Le Loir, Y.; Jan, G.; Schuck, P.; Jeantet, R. Spray Drying of Probiotics and Other Food-Grade Bacteria: A Review. Trends Food Sci. Technol. 2017, 63, 1–17. [Google Scholar] [CrossRef]
  21. Su, Y.; Zheng, X.; Zhao, Q.; Fu, N.; Xiong, H.; Wu, W.D.; Chen, X.D. Spray Drying of Lactobacillus Rhamnosus GG with Calcium-Containing Protectant for Enhanced Viability. Powder Technol. 2019, 358, 87–94. [Google Scholar] [CrossRef]
  22. Charalampopoulos, D.; Rastall, R.A. Prebiotics and Probiotics Science and Technology; Springer: New York, NY, USA, 2009; Volume 1. [Google Scholar]
  23. Burgain, J.; Gaiani, C.; Linder, M.; Scher, J. Encapsulation of Probiotic Living Cells: From Laboratory Scale to Industrial Applications. J. Food Eng. 2011, 104, 467–483. [Google Scholar] [CrossRef]
  24. Centurion, F.; Basit, A.W.; Liu, J.; Gaisford, S.; Rahim, M.A.; Kalantar-Zadeh, K. Nanoencapsulation for Probiotic Delivery. ACS Nano 2021, 15, 18653–18660. [Google Scholar] [CrossRef]
  25. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D. The International Scientific Association for Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
  26. Vinderola, G.; Sanders, M.E.; Salminen, S. The Concept of Postbiotics. Foods 2022, 11, 1077. [Google Scholar] [CrossRef]
  27. Elko, N.; Foster, D.; Kleinheinz, G.; Raubenheimer, B.; Brander, S.; Kinzelman, J.; Kritzer, J.P.; Munroe, D.; Storlazzi, C.; Sutula, M.; et al. Human and Ecosystem Health in Coastal Systems. Shore Beach 2022, 90, 28. [Google Scholar]
  28. Vignieri, S.; Fahrenkamp-Uppenbrink, J. Ecosystem Earth. Science 2017, 356, 258–259. [Google Scholar] [CrossRef]
  29. Razafindralambo, H. Advances in Physical Chemistry Tools for Probiotic Characterization. In Trends Probiotic Applications; Razafindralambo, H., Ed.; Studium Press LLC: Houston, TX, USA, 2018; pp. 50–81. [Google Scholar]
  30. Trush, E.A.; Poluektova, E.A.; Beniashvilli, A.G.; Shifrin, O.S.; Poluektov, Y.M.; Ivashkin, V.T. The Evolution of Human Probiotics: Challenges and Prospects. Probiotics Antimicrob. Proteins 2020, 12, 1291–1299. [Google Scholar] [CrossRef]
  31. Niki, E. Free Radicals in the 1900’s: From in Vitro to in Vivo. Free Radic. Res. 2000, 33, 693–704. [Google Scholar] [CrossRef]
  32. Barbosa, J.; Borges, S.; Teixeira, P. Influence of Sub-Lethal Stresses on the Survival of Lactic Acid Bacteria after Spray-Drying in Orange Juice. Food Microbiol. 2015, 52, 77–83. [Google Scholar] [CrossRef]
  33. Iaconelli, C.; Lemetais, G.; Kechaou, N.; Chain, F.; Bermúdez-Humarán, L.G.; Langella, P.; Gervais, P.; Beney, L. Drying Process Strongly Affects Probiotics Viability and Functionalities. J. Biotechnol. 2015, 214, 17–26. [Google Scholar] [CrossRef]
  34. Rajam, R.; Subramanian, P. Encapsulation of Probiotics: Past, Present and Future. Beni-Suef Univ. J. Basic Appl. Sci. 2022, 11, 46. [Google Scholar] [CrossRef]
  35. Grumet, L.; Tromp, Y.; Stiegelbauer, V. The Development of High-Quality Multispecies Probiotic Formulations: From Bench to Market. Nutrients 2020, 12, 2453. [Google Scholar] [CrossRef]
  36. Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M. The International Scientific Association for Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
  37. Cheng, C.; Yu, X.; Geng, F.; Wang, L.; Yang, J.; Huang, F.; Deng, Q. Review on the Regulation of Plant Polyphenols on the Stability of Polyunsaturated-Fatty-Acid-Enriched Emulsions: Partitioning Kinetic and Interfacial Engineering. J. Agric. Food Chem. 2022, 70, 3569–3584. [Google Scholar] [CrossRef]
  38. Bindels, L.B.; Delzenne, N.M.; Cani, P.D.; Walter, J. Towards a More Comprehensive Concept for Prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 303–310. [Google Scholar] [CrossRef]
  39. Malik, J.K.; Prakash, A.; Srivastava, A.K.; Gupta, R.C. Synbiotics in Animal Health and Production. In Nutraceuticals in Veterinary Medicine; Springer: Cham, Switzerland, 2019; pp. 287–301. [Google Scholar]
  40. Tabacchioni, S.; Passato, S.; Ambrosino, P.; Huang, L.; Caldara, M.; Cantale, C.; Hett, J.; Del Fiore, A.; Fiore, A.; Schlüter, A. Identification of Beneficial Microbial Consortia and Bioactive Compounds with Potential as Plant Biostimulants for a Sustainable Agriculture. Microorganisms 2021, 9, 426. [Google Scholar] [CrossRef] [PubMed]
  41. Tsilingiri, K.; Rescigno, M. Postbiotics: What Else? Benef. Microbes 2013, 4, 101–107. [Google Scholar] [CrossRef] [PubMed]
  42. Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics—A Step beyond Pre-and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef] [PubMed]
  43. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  44. Rad, A.H.; Hosseini, S.; Pourjafar, H. Postbiotics as Dynamic Biological Molecules for Antimicrobial Activity: A Mini-Review. Biointerface Res. Appl. Chem. 2022, 12, 6543–6556. [Google Scholar]
  45. Abbasi, A.; Sheykhsaran, E.; Kafil, H.S. Postbiotics: Science, Technology and Applications; Bentham Science Publishers: Sharjah, United Arab Emirates, 2021; ISBN 1-68108-839-8. [Google Scholar]
  46. Aghebati-Maleki, L.; Hasannezhad, P.; Abbasi, A.; Khani, N. Antibacterial, Antiviral, Antioxidant, and Anticancer Activities of Postbiotics: A Review of Mechanisms and Therapeutic Perspectives. Biointerface Res. Appl. Chem. 2021, 12, 2629–2645. [Google Scholar]
  47. Wegh, C.A.; Geerlings, S.Y.; Knol, J.; Roeselers, G.; Belzer, C. Postbiotics and Their Potential Applications in Early Life Nutrition and Beyond. Int. J. Mol. Sci. 2019, 20, 4673. [Google Scholar] [CrossRef]
  48. Asachi, M.; Nourafkan, E.; Hassanpour, A. A Review of Current Techniques for the Evaluation of Powder Mixing. Adv. Powder Technol. 2018, 29, 1525–1549. [Google Scholar] [CrossRef]
  49. Loskyll, J.; Maier, W.F.; Stoewe, K. Application of a Simultaneous TGA-DSC Thermal Analysis System for High-Throughput Screening of Catalytic Activity. ACS Comb. Sci. 2012, 14, 600–604. [Google Scholar] [CrossRef]
  50. Yu, L. Amorphous Pharmaceutical Solids: Preparation, Characterization and Stabilization. Adv. Drug Deliv. Rev. 2001, 48, 27–42. [Google Scholar] [CrossRef]
  51. Razafindralambo, H.; Razafindralambo, A.; Blecker, C. Thermophysical Fingerprinting of Probiotic-Based Products. Sci. Rep. 2019, 9, 1–8. [Google Scholar]
  52. Razafindralambo, H.; Correani, V.; Fiorucci, S.; Mattei, B. Variability in Probiotic Formulations Revealed by Proteomics and Physico-Chemistry Approach in Relation to the Gut Permeability. Probiotics Antimicrob. Proteins 2020, 12, 1193–1202. [Google Scholar] [CrossRef]
  53. Razafindralambo, H. Application of a Rapid Micro-Calorimetry Coupling Method in Quality and Integrity Control of Probiotic-Based Products. In New Visions in Science and Technology; Book Publisher International: West Bengal, India, 2021; Volume 2, pp. 6–11. ISBN 978-93-91882-98-3. [Google Scholar]
  54. Higgins, D.; Dworkin, J. Recent Progress in Bacillus Subtilis Sporulation. FEMS Microbiol. Rev. 2012, 36, 131–148. [Google Scholar] [CrossRef]
  55. Zhao, F.; Jin, H.; Shen, X.; Li, Q.; Liu, X.; Zhang, L.; Sun, Z.; Yu, J. Effect of the Administration of Probiotics on the Fecal Microbiota of Adult Individuals. Food Sci. Nutr. 2021, 9, 6471–6479. [Google Scholar] [CrossRef]
  56. Khrapko, K.; Hanekamp, J.S.; Thilly, W.G.; Belenkii, A.; Foret, F.; Karger, B.L. Constant Denaturant Capillary Electrophoresis (CDCE): A High Resolution Approach to Mutational Anaylsis. Nucleic Acids Res. 1994, 22, 364–369. [Google Scholar] [CrossRef]
  57. Khrapko, K.; Coller, H.A.; André, P.C.; Li, X.-C.; Hanekamp, J.S.; Thilly, W.G. Mitochondrial Mutational Spectra in Human Cells and Tissues. Proc. Natl. Acad. Sci. USA 1997, 94, 13798–13803. [Google Scholar] [CrossRef] [Green Version]
  58. Andre, P.; Kim, A.; Khrapko, K.; Thilly, W.G. Fidelity and Mutational Spectrum of Pfu DNA Polymerase on a Human Mitochondrial DNA Sequence. Genome Res. 1997, 7, 843–852. [Google Scholar] [CrossRef]
  59. Sudo, H.; Li-Sucholeiki, X.-C.; Marcelino, L.A.; Gruhl, A.N.; Herrero-Jimenez, P.; Zarbl, H.; Willey, J.C.; Furth, E.E.; Morgenthaler, S.; Coller, H.A. Fetal–Juvenile Origins of Point Mutations in the Adult Human Tracheal–Bronchial Epithelium: Absence of Detectable Effects of Age, Gender or Smoking Status. Mutat. Res./Fundam. Mol. Mech. Mutagenesis 2008, 646, 25–40. [Google Scholar] [CrossRef]
  60. Ekstrøm, P.O.; Warren, D.J.; Thilly, W.G. Separation Principles of Cycling Temperature Capillary Electrophoresis. Electrophoresis 2012, 33, 1162–1168. [Google Scholar] [CrossRef]
  61. Ekstrøm, P.O.; Khrapko, K.; Li-Sucholeiki, X.-C.; Hunter, I.W.; Thilly, W.G. Analysis of Mutational Spectra by Denaturing Capillary Electrophoresis. Nat. Protoc. 2008, 3, 1153–1166. [Google Scholar] [CrossRef]
  62. Ekstrøm, P.O.; Nakken, S.; Johansen, M.; Hovig, E. Automated Amplicon Design Suitable for Analysis of DNA Variants by Melting Techniques. BMC Res. Notes 2015, 8, 667. [Google Scholar] [CrossRef] [PubMed]
  63. Refinetti, P.; Morgenthaler, S.; Ekstrøm, P.O. Cycling Temperature Capillary Electrophoresis: A Quantitative, Fast and Inexpensive Method to Detect Mutations in Mixed Populations of Human Mitochondrial DNA. Mitochondrion 2016, 29, 65–74. [Google Scholar] [CrossRef] [PubMed]
  64. Arstad, C.; Refinetti, P.; Kristensen, A.T.; Giercksky, K.-E.; Ekstrøm, P.O. Is Detection of Intraperitoneal Exfoliated Tumor Cells after Surgical Resection of Rectal Cancer a Prognostic Factor of Survival? BMC Cancer 2017, 17, 406. [Google Scholar] [CrossRef] [PubMed]
  65. Refinetti, P.; Arstad, C.; Thilly, W.G.; Morgenthaler, S.; Ekstrøm, P.O. Mapping Mitochondrial Heteroplasmy in a Leydig Tumor by Laser Capture Micro-Dissection and Cycling Temperature Capillary Electrophoresis. BMC Clin. Pathol. 2017, 17, 6. [Google Scholar] [CrossRef] [PubMed]
  66. Arstad, C.; Refinetti, P.; Warren, D.; Giercksky, K.-E.; Ekstrøm, P.O. Scanning the Mitochondrial Genome for Mutations by Cycling Temperature Capillary Electrophoresis. Mitochondrial DNA Part A 2018, 29, 19–30. [Google Scholar] [CrossRef] [PubMed]
  67. Arstad, C.; Taskén, K.; Refinetti, P.; Axcrona, U.; Giercksky, K.-E.; Ekstrøm, P.O. Somatic Mitochondrial DNA Point Mutations Used as Biomarkers to Demonstrate Genomic Heterogeneity in Primary Prostate Cancer. Prostate Cancer 2020, 2020, 7673684. [Google Scholar] [CrossRef] [PubMed]
  68. Refinetti, P.; Morgenthaler, S.; Thilly, W.G.; Arstad, C.; Ekstrøm, P.O. Tracing of Human Tumor Cell Lineages by Mitochondrial Mutations. Front. Oncol. 2020, 10, 523860. [Google Scholar] [CrossRef]
  69. Karlen, Y.; McNair, A.; Perseguers, S.; Mazza, C.; Mermod, N. Statistical Significance of Quantitative PCR. BMC Bioinform. 2007, 8, 131. [Google Scholar] [CrossRef]
  70. Ruijter, J.M.; Ramakers, C.; Hoogaars, W.M.H.; Karlen, Y.; Bakker, O.; Van den Hoff, M.J.B.; Moorman, A. Amplification Efficiency: Linking Baseline and Bias in the Analysis of Quantitative PCR Data. Nucleic Acids Res. 2009, 37, e45. [Google Scholar] [CrossRef]
  71. Jackson, S.A.; Schoeni, J.L.; Vegge, C.; Pane, M.; Stahl, B.; Bradley, M.; Goldman, V.S.; Burguière, P.; Atwater, J.B.; Sanders, M.E. Improving End-User Trust in the Quality of Commercial Probiotic Products. Front. Microbiol. 2019, 10, 739. [Google Scholar] [CrossRef]
  72. Chiron, C.; Tompkins, T.A.; Burguière, P. Flow Cytometry: A Versatile Technology for Specific Quantification and Viability Assessment of Micro-Organisms in Multistrain Probiotic Products. J. Appl. Microbiol. 2018, 124, 572–584. [Google Scholar] [CrossRef]
  73. Petrova, M.I.; Lievens, E.; Malik, S.; Imholz, N.; Lebeer, S. Lactobacillus Species as Biomarkers and Agents That Can Promote Various Aspects of Vaginal Health. Front. Physiol. 2015, 6, 81. [Google Scholar] [CrossRef]
  74. Sela, D.A.; Mills, D.A. Nursing Our Microbiota: Molecular Linkages between Bifidobacteria and Milk Oligosaccharides. Trends Microbiol. 2010, 18, 298–307. [Google Scholar] [CrossRef]
  75. Hashem, A.; Tabassum, B.; Abd_Allah, E.F. Bacillus Subtilis: A Plant-Growth Promoting Rhizobacterium That Also Impacts Biotic Stress. Saudi J. Biol. Sci. 2019, 26, 1291–1297. [Google Scholar] [CrossRef]
  76. Ghahremani, Z.; Escudero, N.; Beltrán-Anadón, D.; Saus, E.; Cunquero, M.; Andilla, J.; Loza-Alvarez, P.; Gabaldón, T.; Sorribas, F.J. Bacillus Firmus Strain I-1582, a Nematode Antagonist by Itself and through the Plant. Front. Plant Sci. 2020, 11, 796. [Google Scholar] [CrossRef]
  77. Roh, J.-Y.; Choi, J.-Y.; Li, M.-S.; Jin, B.-R.; Je, Y.-H. Bacillus Thuringiensis as a Specific, Safe, and Effective Tool for Insect Pest Control. J. Microbiol. Biotechnol. 2007, 17, 547–559. [Google Scholar]
  78. Basak, B.B.; Biswas, D.R. Influence of Potassium Solubilizing Microorganism (Bacillus Mucilaginosus) and Waste Mica on Potassium Uptake Dynamics by Sudan Grass (Sorghum Vulgare Pers.) Grown under Two Alfisols. Plant Soil 2009, 317, 235–255. [Google Scholar] [CrossRef]
  79. Puri, A.; Padda, K.P.; Chanway, C.P. Seedling Growth Promotion and Nitrogen Fixation by a Bacterial Endophyte Paenibacillus Polymyxa P2b-2R and Its GFP Derivative in Corn in a Long-Term Trial. Symbiosis 2016, 69, 123–129. [Google Scholar] [CrossRef]
  80. Azcón, R.; del Carmen Perálvarez, M.; Roldán, A.; Barea, J.-M. Arbuscular Mycorrhizal Fungi, Bacillus Cereus, and Candida Parapsilosis from a Multicontaminated Soil Alleviate Metal Toxicity in Plants. Microb. Ecol. 2010, 59, 668–677. [Google Scholar] [CrossRef]
  81. Shen, Z.; Wang, B.; Lv, N.; Sun, Y.; Jiang, X.; Li, R.; Ruan, Y.; Shen, Q. Effect of the Combination of Bio-Organic Fertiliser with Bacillus Amyloliquefaciens NJN-6 on the Control of Banana Fusarium Wilt Disease, Crop Production and Banana Rhizosphere Culturable Microflora. Biocontrol Sci. Technol. 2015, 25, 716–731. [Google Scholar] [CrossRef]
  82. Masood, S.; Zhao, X.Q.; Shen, R.F. Bacillus Pumilus Promotes the Growth and Nitrogen Uptake of Tomato Plants under Nitrogen Fertilization. Sci. Hortic. 2020, 272, 109581. [Google Scholar] [CrossRef]
  83. Kang, S.-M.; Radhakrishnan, R.; You, Y.-H.; Joo, G.-J.; Lee, I.-J.; Lee, K.-E.; Kim, J.-H. Phosphate Solubilizing Bacillus Megaterium Mj1212 Regulates Endogenous Plant Carbohydrates and Amino Acids Contents to Promote Mustard Plant Growth. Indian J. Microbiol. 2014, 54, 427–433. [Google Scholar] [CrossRef]
  84. Kerry, R.G.; Patra, J.K.; Gouda, S.; Park, Y.; Shin, H.-S.; Das, G. Benefaction of Probiotics for Human Health: A Review. J. Food Drug Anal. 2018, 26, 927–939. [Google Scholar] [CrossRef]
  85. Rao, S.C.; Athalye-Jape, G.K.; Deshpande, G.C.; Simmer, K.N.; Patole, S.K. Probiotic Supplementation and Late-Onset Sepsis in Preterm Infants: A Meta-Analysis. Pediatrics 2016, 137, e20153684. [Google Scholar] [CrossRef]
  86. Shehata, A.A.; Yalçın, S.; Latorre, J.D.; Basiouni, S.; Attia, Y.A.; Abd El-Wahab, A.; Visscher, C.; El-Seedi, H.R.; Huber, C.; Hafez, H.M. Probiotics, Prebiotics, and Phytogenic Substances for Optimizing Gut Health in Poultry. Microorganisms 2022, 10, 395. [Google Scholar] [CrossRef]
  87. Illiano, P.; Brambilla, R.; Parolini, C. The Mutual Interplay of Gut Microbiota, Diet and Human Disease. FEBS J. 2020, 287, 833–855. [Google Scholar] [CrossRef] [PubMed]
  88. Bottari, B.; Quartieri, A.; Prandi, B.; Raimondi, S.; Leonardi, A.; Rossi, M.; Ulrici, A.; Gatti, M.; Sforza, S.; Nocetti, M. Characterization of the Peptide Fraction from Digested Parmigiano Reggiano Cheese and Its Effect on Growth of Lactobacilli and Bifidobacteria. Int. J. Food Microbiol. 2017, 255, 32–41. [Google Scholar] [CrossRef] [PubMed]
  89. Yan, F.; Polk, D.B. Probiotics and Immune Health. Curr. Opin. Gastroenterol. 2011, 27, 496. [Google Scholar] [CrossRef] [PubMed]
  90. Breton, J.; Galmiche, M.; Déchelotte, P. Dysbiotic Gut Bacteria in Obesity: An Overview of the Metabolic Mechanisms and Therapeutic Perspectives of next-Generation Probiotics. Microorganisms 2022, 10, 452. [Google Scholar] [CrossRef]
  91. Senok, A.C.; Ismaeel, A.Y.; Botta, G.A. Probiotics: Facts and Myths. Clin. Microbiol. Infect. 2005, 11, 958–966. [Google Scholar] [CrossRef]
  92. Mizock, B.A. Probiotics. Dis. -A-Mon. DM 2015, 61, 259–290. [Google Scholar] [CrossRef]
  93. Pandey, K.R.; Naik, S.R.; Vakil, B.V. Probiotics, Prebiotics and Synbiotics-a Review. J. Food Sci. Technol. 2015, 52, 7577–7587. [Google Scholar] [CrossRef]
  94. Suez, J.; Zmora, N.; Segal, E.; Elinav, E. The Pros, Cons, and Many Unknowns of Probiotics. Nat. Med. 2019, 25, 716–729. [Google Scholar] [CrossRef]
  95. Da Cruz, M.F.; Magno, M.B.; Jural, L.A.; Pimentel, T.C.; Masterson, D.; Esmerino, E.A.; Ramos, G.L.P.A.; Gomila, J.V.; Silva, M.C.; Cruz, A.G. Probiotics and Dairy Products in Dentistry: A Bibliometric and Critical Review of Randomized Clinical Trials. Food Res. Int. 2022, 157, 111228. [Google Scholar] [CrossRef]
  96. Ganji-Arjenaki, M.; Rafieian-Kopaei, M. Probiotics Are a Good Choice in Remission of Inflammatory Bowel Diseases: A Meta Analysis and Systematic Review. J. Cell. Physiol. 2018, 233, 2091–2103. [Google Scholar] [CrossRef]
  97. Plaza-Diaz, J.; Ruiz-Ojeda, F.J.; Gil-Campos, M.; Gil, A. Mechanisms of Action of Probiotics. Adv. Nutr. 2019, 10, S49–S66. [Google Scholar] [CrossRef]
  98. Islam, S.U. Clinical Uses of Probiotics. Medicine 2016, 95, e2658. [Google Scholar] [CrossRef]
  99. Liu, Y.; Tran, D.Q.; Rhoads, J.M. Probiotics in Disease Prevention and Treatment. J. Clin. Pharmacol. 2018, 58, S164–S179. [Google Scholar] [CrossRef]
  100. Tillisch, K. The Effects of Gut Microbiota on CNS Function in Humans. Gut Microbes 2014, 5, 404–410. [Google Scholar] [CrossRef]
  101. Kahouli, I.; Malhotra, M.; Alaoui-Jamali, M.; Prakash, S. In-Vitro Characterization of the Anti-Cancer Activity of the Probiotic Bacterium Lactobacillus Fermentum NCIMB 5221 and Potential against Colorectal Cancer. J. Cancer Sci. 2015, 7, 224–235. [Google Scholar]
  102. Hilton, E.; Kolakowski, P.; Singer, C.; Smith, M. Efficacy of Lactobacillus GG as a Diarrheal Preventive in Travelers. J. Travel Med. 1997, 4, 41–43. [Google Scholar] [CrossRef]
  103. McFarland, L.V. Meta-Analysis of Probiotics for the Prevention of Traveler’s Diarrhea. Travel Med. Infect. Dis. 2007, 5, 97–105. [Google Scholar] [CrossRef]
  104. Sazawal, S.; Hiremath, G.; Dhingra, U.; Malik, P.; Deb, S.; Black, R.E. Efficacy of Probiotics in Prevention of Acute Diarrhoea: A Meta-Analysis of Masked, Randomised, Placebo-Controlled Trials. Lancet Infect. Dis. 2006, 6, 374–382. [Google Scholar] [CrossRef]
  105. Cremonini, F.; Di Caro, S.; Nista, E.C.; Bartolozzi, F.; Capelli, G.; Gasbarrini, G.; Gasbarrini, A. Meta-Analysis: The Effect of Probiotic Administration on Antibiotic-Associated Diarrhoea. Aliment. Pharmacol. Ther. 2002, 16, 1461–1467. [Google Scholar] [CrossRef]
  106. Naidoo, K.; Gordon, M.; Fagbemi, A.O.; Thomas, A.G.; Akobeng, A.K. Probiotics for Maintenance of Remission in Ulcerative Colitis. Cochrane Database Syst. Rev. 2011, CD007443. [Google Scholar] [CrossRef]
  107. AlFaleh, K.; Anabrees, J. Probiotics for Prevention of Necrotizing Enterocolitis in Preterm Infants. Evid.-Based Child Health A Cochrane Rev. J. 2014, 9, 584–671. [Google Scholar] [CrossRef]
  108. Autran, C.A.; Schoterman, M.H.; Jantscher-Krenn, E.; Kamerling, J.P.; Bode, L. Sialylated Galacto-Oligosaccharides and 2′-Fucosyllactose Reduce Necrotising Enterocolitis in Neonatal Rats. Br. J. Nutr. 2016, 116, 294–299. [Google Scholar] [CrossRef]
  109. Vetvicka, V.; Oliveira, C. β(1-3)(1-6)-D-Glucans Modulate Immune Status in Pigs: Potential Importance for Efficiency of Commercial Farming. Ann. Transl. Med. 2014, 2, 16. [Google Scholar]
  110. Jacob, J.P.; Pescatore, A.J. Barley β-Glucan in Poultry Diets. Ann. Transl. Med. 2014, 2, 20. [Google Scholar]
  111. Alok, A.; Singh, I.D.; Singh, S.; Kishore, M.; Jha, P.C.; Iqubal, M.A. Probiotics: A New Era of Biotherapy. Adv. Biomed. Res. 2017, 6, 31. [Google Scholar] [CrossRef]
  112. Fiocco, D.; Longo, A.; Arena, M.P.; Russo, P.; Spano, G.; Capozzi, V. How Probiotics Face Food Stress: They Get by with a Little Help. Crit. Rev. Food Sci. Nutr. 2020, 60, 1552–1580. [Google Scholar] [CrossRef] [PubMed]
  113. Pimentel, T.C.; da Costa, W.K.A.; Barão, C.E.; Rosset, M.; Magnani, M. Vegan Probiotic Products: A Modern Tendency or the Newest Challenge in Functional Foods. Food Res. Int. 2021, 140, 110033. [Google Scholar] [CrossRef] [PubMed]
  114. Campbell, K. How Some Probiotic Scientists Are Working to Address COVID-19. 2020. Available online: https://isappscience.org/how-some-probiotic-and-prebiotic-scientists-are-working-to-address-covid-19/ (accessed on 29 June 2022).
  115. Bottari, B.; Castellone, V.; Neviani, E. Probiotics and COVID-19. Int. J. Food Sci. Nutr. 2021, 72, 293–299. [Google Scholar] [CrossRef] [PubMed]
  116. Markowiak, P.; Śliżewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef]
  117. Van Zyl, W.F.; Deane, S.M.; Dicks, L.M.T. Molecular Insights into Probiotic Mechanisms of Action Employed against Intestinal Pathogenic Bacteria. Gut Microbes 2020, 12, 1831339. [Google Scholar] [CrossRef]
  118. Burgueño, J.F.; Abreu, M.T. Epithelial Toll-like Receptors and Their Role in Gut Homeostasis and Disease. Nat Rev Gastroenterol Hepatol 2020, 17, 263–278. [Google Scholar] [CrossRef]
  119. Mirzaei, R.; Dehkhodaie, E.; Bouzari, B.; Rahimi, M.; Gholestani, A.; Hosseini-Fard, S.R.; Keyvani, H.; Teimoori, A.; Karampoor, S. Dual Role of Microbiota-Derived Short-Chain Fatty Acids on Host and Pathogen. Biomed. Pharmacother. 2022, 145, 112352. [Google Scholar] [CrossRef]
  120. Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; van Harsselaar, J.; et al. Short Chain Fatty Acids in Human Gut and Metabolic Health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef]
  121. Lai, Y.; Dhingra, R.; Zhang, Z.; Ball, L.M.; Zylka, M.J.; Lu, K. Toward Elucidating the Human Gut Microbiota–Brain Axis: Molecules, Biochemistry, and Implications for Health and Diseases. Biochemistry 2021. [Google Scholar] [CrossRef]
  122. Fabbrizzi, A.; Amedei, A.; Lavorini, F.; Renda, T.; Fontana, G. The Lung Microbiome: Clinical and Therapeutic Implications. Intern. Emerg. Med. 2019, 14, 1241–1250. [Google Scholar] [CrossRef] [Green Version]
  123. Gutiérrez-Castrellón, P.; Gandara-Martí, T.; Abreu, Y.; Abreu, A.T.; Nieto-Rufino, C.D.; López-Orduña, E.; Jiménez-Escobar, I.; Jiménez-Gutiérrez, C.; López-Velazquez, G.; Espadaler-Mazo, J. Probiotic Improves Symptomatic and Viral Clearance in Covid19 Outpatients: A Randomized, Quadruple-Blinded, Placebo-Controlled Trial. Gut Microbes 2022, 14, 2018899. [Google Scholar] [CrossRef]
  124. Mercader-Barceló, J.; Truyols-Vives, J.; Río, C.; López-Safont, N.; Sala-Llinàs, E.; Chaplin, A. Insights into the Role of Bioactive Food Ingredients and the Microbiome in Idiopathic Pulmonary Fibrosis. Int. J. Mol. Sci. 2020, 21, 6051. [Google Scholar] [CrossRef]
  125. Sinha, S.; Lin, G.; Ferenczi, K. The Skin Microbiome and the Gut-Skin Axis. Clin. Dermatol. 2021, 39, 829–839. [Google Scholar] [CrossRef]
  126. Khan, I.; Ullah, N.; Zha, L.; Bai, Y.; Khan, A.; Zhao, T.; Che, T.; Zhang, C. Alteration of Gut Microbiota in Inflammatory Bowel Disease (IBD): Cause or Consequence? IBD Treatment Targeting the Gut Microbiome. Pathogens 2019, 8, 126. [Google Scholar] [CrossRef]
  127. Li, H.-Y.; Zhou, D.-D.; Gan, R.-Y.; Huang, S.-Y.; Zhao, C.-N.; Shang, A.; Xu, X.-Y.; Li, H.-B. Effects and Mechanisms of Probiotics, Prebiotics, Synbiotics, and Postbiotics on Metabolic Diseases Targeting Gut Microbiota: A Narrative Review. Nutrients 2021, 13, 3211. [Google Scholar] [CrossRef]
  128. Sergeev, I.N.; Aljutaily, T.; Walton, G.; Huarte, E. Effects of Synbiotic Supplement on Human Gut Microbiota, Body Composition and Weight Loss in Obesity. Nutrients 2020, 12, 222. [Google Scholar] [CrossRef]
  129. Kvakova, M.; Bertkova, I.; Stofilova, J.; Savidge, T.C. Co-Encapsulated Synbiotics and Immobilized Probiotics in Human Health and Gut Microbiota Modulation. Foods 2021, 10, 1297. [Google Scholar] [CrossRef]
  130. Krumbeck, J.A.; Walter, J.; Hutkins, R.W. Synbiotics for Improved Human Health: Recent Developments, Challenges, and Opportunities. Annu. Rev. Food Sci. Technol. 2018, 9, 451–479. [Google Scholar] [CrossRef]
  131. Ahmadi Badi, S.; Tarashi, S.; Fateh, A.; Rohani, P.; Masotti, A.; Siadat, S.D. From the Role of Microbiota in Gut-Lung Axis to SARS-CoV-2 Pathogenesis. Mediat. Inflamm. 2021, 2021, 6611222. [Google Scholar] [CrossRef]
  132. Antunes, A.E.; Vinderola, G.; Xavier-Santos, D.; Sivieri, K. Potential Contribution of Beneficial Microbes to Face the COVID-19 Pandemic. Food Res. Int. 2020, 136, 109577. [Google Scholar] [CrossRef]
  133. Eslamparast, T.; Zamani, F.; Hekmatdoost, A.; Sharafkhah, M.; Eghtesad, S.; Malekzadeh, R.; Poustchi, H. Effects of Synbiotic Supplementation on Insulin Resistance in Subjects with the Metabolic Syndrome: A Randomised, Double-Blind, Placebo-Controlled Pilot Study. Br. J. Nutr. 2014, 112, 438–445. [Google Scholar] [CrossRef]
  134. Rogha, M.; Esfahani, M.Z.; Zargarzadeh, A.H. The Efficacy of a Synbiotic Containing Bacillus Coagulans in Treatment of Irritable Bowel Syndrome: A Randomized Placebo-Controlled Trial. Gastroenterol. Hepatol. Bed Bench 2014, 7, 156. [Google Scholar]
  135. Dinleyici, E.C.; Dalgic, N.; Guven, S.; Ozen, M.; Kara, A.; Arica, V.; Metin-Timur, O.; Sancar, M.; Kurugol, Z.; Tanir, G. The Effect of a Multispecies Synbiotic Mixture on the Duration of Diarrhea and Length of Hospital Stay in Children with Acute Diarrhea in Turkey: Single Blinded Randomized Study. Eur. J. Pediatrics 2013, 172, 459–464. [Google Scholar] [CrossRef]
  136. Worthley, D.L.; Le Leu, R.K.; Whitehall, V.L.; Conlon, M.; Christophersen, C.; Belobrajdic, D.; Mallitt, K.-A.; Hu, Y.; Irahara, N.; Ogino, S. A Human, Double-Blind, Placebo-Controlled, Crossover Trial of Prebiotic, Probiotic, and Synbiotic Supplementation: Effects on Luminal, Inflammatory, Epigenetic, and Epithelial Biomarkers of Colorectal Cancer. Am. J. Clin. Nutr. 2009, 90, 578–586. [Google Scholar] [CrossRef]
  137. Dehghani, H.; Heidari, F.; Mozaffari-Khosravi, H.; Nouri-Majelan, N.; Dehghani, A. Synbiotic Supplementations for Azotemia in Patients with Chronic Kidney Disease: A Randomized Controlled Trial. Iran. J. Kidney Dis. 2016, 10, 351–357. [Google Scholar]
  138. Javadi, L.; Khoshbaten, M.; Safaiyan, A.; Ghavami, M.; Abbasi, M.M.; Gargari, B.P. Pro-and Prebiotic Effects on Oxidative Stress and Inflammatory Markers in Non-Alcoholic Fatty Liver Disease. Asia Pac. J. Clin. Nutr. 2018, 27, 1031–1039. [Google Scholar]
  139. Ahanchian, H.; Jafari, S.A.; Ansari, E.; Ganji, T.; Kiani, M.A.; Khalesi, M.; Momen, T.; Kianifar, H. A Multi-Strain Synbiotic May Reduce Viral Respiratory Infections in Asthmatic Children: A Randomized Controlled Trial. Electron. Physician 2016, 8, 2833. [Google Scholar] [CrossRef]
  140. Wu, K.-G.; Li, T.-H.; Peng, H.-J. Lactobacillus Salivarius plus Fructo-Oligosaccharide Is Superior to Fructo-Oligosaccharide Alone for Treating Children with Moderate to Severe Atopic Dermatitis: A Double-Blind, Randomized, Clinical Trial of Efficacy and Safety. Br. J. Dermatol. 2012, 166, 129–136. [Google Scholar] [CrossRef]
  141. Haghighat, N.; Mohammadshahi, M.; Shayanpour, S.; Haghighizadeh, M.H.; Rahmdel, S.; Rajaei, M. The Effect of Synbiotic and Probiotic Supplementation on Mental Health Parameters in Patients Undergoing Hemodialysis: A Double-Blind, Randomized, Placebo-Controlled Trial. Indian J. Nephrol. 2021, 31, 149. [Google Scholar]
  142. Rodriguez-Herrera, A.; Tims, S.; Polman, J.; Porcel Rubio, R.; Muñoz Hoyos, A.; Agosti, M.; Lista, G.; Corvaglia, L.T.; Knol, J.; Roeselers, G. Early-Life Fecal Microbiome and Metabolome Dynamics in Response to an Intervention with Infant Formula Containing Specific Prebiotics and Postbiotics. Am. J. Physiol.-Gastrointest. Liver Physiol. 2022, 322, G571–G582. [Google Scholar] [CrossRef]
  143. Singh, A.; D’Amico, D.; Andreux, P.A.; Fouassier, A.M.; Blanco-Bose, W.; Evans, M.; Aebischer, P.; Auwerx, J.; Rinsch, C. Urolithin A Improves Muscle Strength, Exercise Performance, and Biomarkers of Mitochondrial Health in a Randomized Trial in Middle-Aged Adults. Cell Rep. Med. 2022, 3, 100633. [Google Scholar] [CrossRef] [PubMed]
  144. Ricci, A.; Allende, A.; Bolton, D.; Chemaly, M.; Davies, R.; Girones, R.; Herman, L.; Koutsoumanis, K.; Lindqvist, R.; Nørrung, B.; et al. Scientific Opinion on the Update of the List of QPS-recommended Biological Agents Intentionally Added to Food or Feed as Notified to EFSA. EFSA J. 2017, 15, e04664. [Google Scholar] [CrossRef] [PubMed]
  145. Patterson, J.A.; Burkholder, K.M. Application of Prebiotics and Probiotics in Poultry Production. Poult. Sci. 2003, 82, 627–631. [Google Scholar] [CrossRef] [PubMed]
  146. Cheng, Y.; Chen, Y.; Li, X.; Yang, W.; Wen, C.; Kang, Y.; Wang, A.; Zhou, Y. Effects of Synbiotic Supplementation on Growth Performance, Carcass Characteristics, Meat Quality and Muscular Antioxidant Capacity and Mineral Contents in Broilers: Effects of Synbiotic Supplementation. J. Sci. Food Agric. 2017, 97, 3699–3705. [Google Scholar] [CrossRef]
  147. Gaggìa, F.; Mattarelli, P.; Biavati, B. Probiotics and Prebiotics in Animal Feeding for Safe Food Production. Int. J. Food Microbiol. 2010, 141, S15–S28. [Google Scholar] [CrossRef]
  148. Stanczuk, J.; Zdunczyk, Z.; Juskiewicz, J.; Jankowski, J. Indices of Response of Young Turkeys to Diets Containing Mannan- Oligosaccharide or Inulin Raciono, Turin Č Io Monooligosacharid Ų Ir Inulino, Poveikis. Veterinarija Ir Zootechnika 2005, 31, 98–101. [Google Scholar]
  149. Kawashima, T.; Hayashi, K.; Kosaka, A.; Kawashima, M.; Igarashi, T.; Tsutsui, H.; Tsuji, N.M.; Nishimura, I.; Hayashi, T.; Obata, A. Lactobacillus Plantarum Strain YU from Fermented Foods Activates Th1 and Protective Immune Responses. Int. Immunopharmacol. 2011, 11, 2017–2024. [Google Scholar] [CrossRef]
  150. Guevarra, R.B.; Lee, J.H.; Lee, S.H.; Seok, M.J.; Kim, D.W.; Kang, B.N.; Johnson, T.J.; Isaacson, R.E.; Kim, H.B. Piglet Gut Microbial Shifts Early in Life: Causes and Effects. J. Anim. Sci. Biotechnol. 2019, 10, 1–10. [Google Scholar] [CrossRef]
  151. Zangeronimo, M.G.; Cantarelli, V.d.S.; Fialho, E.T.; Amaral, N.d.O.; Silveira, H.; Pereira, L.d.M.; Pereira, L.J. Herbal Extracts and Symbiotic Mixture Replacing Antibiotics in Piglets at the Initial Phase. Rev. Bras. De Zootec. 2011, 40, 1045–1051. [Google Scholar] [CrossRef]
  152. Casey, P.G.; Gardiner, G.E.; Casey, G.; Bradshaw, B.; Lawlor, P.G.; Lynch, P.B.; Leonard, F.C.; Stanton, C.; Ross, R.P.; Fitzgerald, G.F.; et al. A Five-Strain Probiotic Combination Reduces Pathogen Shedding and Alleviates Disease Signs in Pigs Challenged with Salmonella Enterica Serovar Typhimurium. Appl. Environ. Microbiol. 2007, 73, 1858–1863. [Google Scholar] [CrossRef]
  153. Duarte, M.E.; Tyus, J.; Kim, S.W. Synbiotic Effects of Enzyme and Probiotics on Intestinal Health and Growth of Newly Weaned Pigs Challenged with Enterotoxigenic F18+ Escherichia coli. Front. Vet. Sci. 2020, 7, 573. [Google Scholar] [CrossRef]
  154. Cadogan, D.J.; Choct, M. Pattern of Non-Starch Polysaccharide Digestion along the Gut of the Pig: Contribution to Available Energy. Anim. Nutr. 2015, 1, 160–165. [Google Scholar] [CrossRef]
  155. Passos, A.A.; Park, I.; Ferket, P.; von Heimendahl, E.; Kim, S.W. Effect of Dietary Supplementation of Xylanase on Apparent Ileal Digestibility of Nutrients, Viscosity of Digesta, and Intestinal Morphology of Growing Pigs Fed Corn and Soybean Meal Based Diet. Anim. Nutr. 2015, 1, 19–23. [Google Scholar] [CrossRef]
  156. Lee, Y.K. Selection and Maintenance of Probiotic Microorganisms. In Handbook of Probiotics and Prebiotics, 2nd ed.; John Wiley & Sons, Ltd: Hoboken, NJ, USA, 2008; pp. 177–187. ISBN 978-0-470-13544-0. [Google Scholar]
  157. Markowiak, P.; Śliżewska, K. The Role of Probiotics, Prebiotics and Synbiotics in Animal Nutrition. Gut Pathog. 2018, 10, 21. [Google Scholar] [CrossRef]
  158. Weimer, P.J. Redundancy, Resilience, and Host Specificity of the Ruminal Microbiota: Implications for Engineering Improved Ruminal Fermentations. Front. Microbiol. 2015, 6, 296. [Google Scholar] [CrossRef] [Green Version]
  159. Melara, E.G.; Avellaneda, M.C.; Valdivié, M.; García-Hernández, Y.; Aroche, R.; Martínez, Y. Probiotics: Symbiotic Relationship with the Animal Host. Animals 2022, 12, 719. [Google Scholar] [CrossRef]
  160. Stover, M.G.; Watson, R.R.; Collier, R.J. Pre- and Probiotic Supplementation in Ruminant Livestock Production. In Probiotics, Prebiotics, and Synbiotics: Bioactive Foods in Health Promotion; Academic Press: Cambridge, MA, USA, 2016; pp. 25–36. ISBN 978-0-12-802189-7. [Google Scholar]
  161. Reuben, R.C.; Elghandour, M.M.M.Y.; Alqaisi, O.; Cone, J.W.; Márquez, O.; Salem, A.Z.M. Influence of Microbial Probiotics on Ruminant Health and Nutrition: Sources, Mode of Action and Implications. J. Sci. Food Agric. 2022, 102, 1319–1340. [Google Scholar] [CrossRef]
  162. Stein, D.R.; Allen, D.T.; Perry, E.B.; Bruner, J.C.; Gates, K.W.; Rehberger, T.G.; Mertz, K.; Jones, D.; Spicer, L.J. Effects of Feeding Propionibacteria to Dairy Cows on Milk Yield, Milk Components, and Reproduction. J. Dairy Sci. 2006, 89, 111–125. [Google Scholar] [CrossRef]
  163. Poppy, G.D.; Rabiee, A.R.; Lean, I.J.; Sanchez, W.K.; Dorton, K.L.; Morley, P.S. A Meta-Analysis of the Effects of Feeding Yeast Culture Produced by Anaerobic Fermentation of Saccharomyces Cerevisiae on Milk Production of Lactating Dairy Cows. J. Dairy Sci. 2012, 95, 6027–6041. [Google Scholar] [CrossRef]
  164. Estrada-Angulo, A.; Zapata-Ramírez, O.; Castro-Pérez, B.I.; Urías-Estrada, J.D.; Gaxiola-Camacho, S.; Angulo-Montoya, C.; Ríos-Rincón, F.G.; Barreras, A.; Zinn, R.A.; Leyva-Morales, J.B.; et al. The Effects of Single or Combined Supplementation of Probiotics and Prebiotics on Growth Performance, Dietary Energetics, Carcass Traits, and Visceral Mass in Lambs Finished under Subtropical Climate Conditions. Biology 2021, 10, 1137. [Google Scholar] [CrossRef]
  165. Uyeno, Y.; Shigemori, S.; Shimosato, T. Effect of Probiotics/Prebiotics on Cattle Health and Productivity. Microbes Environ. 2015, 30, 126–132. [Google Scholar] [CrossRef]
  166. Xu, H.; Huang, W.; Hou, Q.; Kwok, L.; Sun, Z.; Ma, H.; Zhao, F.; Lee, Y.-K.; Zhang, H. The Effects of Probiotics Administration on the Milk Production, Milk Components and Fecal Bacteria Microbiota of Dairy Cows. Sci. Bull. 2017, 62, 767–774. [Google Scholar] [CrossRef]
  167. Stella, A.V.; Paratte, R.; Valnegri, L.; Cigalino, G.; Soncini, G.; Chevaux, E.; Dell’Orto, V.; Savoini, G. Effect of Administration of Live Saccharomyces Cerevisiae on Milk Production, Milk Composition, Blood Metabolites, and Faecal Flora in Early Lactating Dairy Goats. Small Rumin. Res. 2007, 67, 7–13. [Google Scholar] [CrossRef]
  168. Callaway, T.R.; Edrington, T.S.; Anderson, R.C.; Nisbet, D.J. Efficiency of the Use of Pre- and Probiotics for Dairy Cows. In Proceedings of the 3rd International Livestock Farming Systems Symposium, Aberdeen, Scotland, 12 September 1996; pp. 33–54. [Google Scholar]
  169. Fleige, S.; Preißinger, W.; Meyer, H.H.D.; Pfaffl, M.W. Effect of Lactulose on Growth Performance and Intestinal Morphology of Pre-Ruminant Calves Using a Milk Replacer Containing Enterococcus Faecium. Animal 2007, 1, 367–373. [Google Scholar] [CrossRef]
  170. Morrison, S.J.; Dawson, S.; Carson, A.F. The Effects of Mannan Oligosaccharide and Streptococcus Faecium Addition to Milk Replacer on Calf Health and Performance. Livest. Sci. 2010, 131, 292–296. [Google Scholar] [CrossRef]
  171. Jonova, S.; Ilgaza, A.; Zolovs, M. The Impact of Inulin and a Novel Synbiotic (Yeast Saccharomyces Cerevisiae Strain 1026 and Inulin) on the Development and Functional State of the Gastrointestinal Canal of Calves. Vet. Med. Int. 2021, 2021, 1–9. [Google Scholar] [CrossRef]
  172. Guowei, S.; He, Y.; Wan, H.; Hui, Y.; Li, H. Effects of Prebiotics on Antioxidant Activity of Goat Milk Fermented by Lactobacillus Plantarum L60. Acta Univ. Cibiniensis. Ser. E Food Technol. 2017, 21, 11–18. [Google Scholar] [CrossRef]
  173. Han, G.; Gao, X.; Duan, J.; Zhang, H.; Zheng, Y.; He, J.; Huo, N.; Pei, C.; Li, H.; Gu, S. Effects of Yeasts on Rumen Bacterial Flora, Abnormal Metabolites, and Blood Gas in Sheep with Induced Subacute Ruminal Acidosis. Anim. Feed Sci. Technol. 2021, 280, 115042. [Google Scholar] [CrossRef]
  174. Lettat, A.; Nozière, P.; Silberberg, M.; Morgavi, D.P.; Berger, C.; Martin, C. Rumen Microbial and Fermentation Characteristics Are Affected Differently by Bacterial Probiotic Supplementation during Induced Lactic and Subacute Acidosis in Sheep. BMC Microbiol. 2012, 12, 142. [Google Scholar] [CrossRef] [PubMed]
  175. Zapata, O.; Cervantes, A.; Barreras, A.; Monge-Navarro, F.; González-Vizcarra, V.M.; Estrada-Angulo, A.; Urías-Estrada, J.D.; Corona, L.; Zinn, R.A.; Martínez-Alvarez, I.G.; et al. Effects of Single or Combined Supplementation of Probiotics and Prebiotics on Ruminal Fermentation, Ruminal Bacteria and Total Tract Digestion in Lambs. Small Rumin. Res. 2021, 204, 106538. [Google Scholar] [CrossRef]
  176. Rohani, M.F.; Islam, S.M.; Hossain, M.K.; Ferdous, Z.; Siddik, M.A.B.; Nuruzzaman, M.; Padeniya, U.; Brown, C.; Shahjahan, M. Probiotics, Prebiotics and Synbiotics Improved the Functionality of Aquafeed: Upgrading Growth, Reproduction, Immunity and Disease Resistance in Fish. Fish Shellfish Immunol. 2022, 120, 569–589. [Google Scholar] [CrossRef] [PubMed]
  177. Tanwar, J.; Das, S.; Fatima, Z.; Hameed, S. Multidrug Resistance: An Emerging Crisis. Interdiscip. Perspect. Infect. Dis. 2014, 2014, 541340. [Google Scholar] [CrossRef] [PubMed]
  178. Irianto, A.; Austin, B. Probiotics in Aquaculture. J. Fish Dis. 2002, 25, 633–642. [Google Scholar] [CrossRef]
  179. Prasad, L.; Baghel, D.; Kumar, V.; Laxmi, P.; Dharmendra Singh, B.; Vikas, K. Role and Prospects of Probiotics Use in Aquaculture. Aquacult 2003, 4, 247–251. [Google Scholar]
  180. Verschuere, L.; Rombaut, G.; Sorgeloos, P.; Verstraete, W. Probiotic Bacteria as Biological Control Agents in Aquaculture. Microbiol. Mol. Biol. Rev. 2000, 64, 655. [Google Scholar] [CrossRef]
  181. Dawood, M.A.; Koshio, S. Recent Advances in the Role of Probiotics and Prebiotics in Carp Aquaculture: A Review. Aquaculture 2016, 454, 243–251. [Google Scholar] [CrossRef]
  182. Hoseinifar, S.H.; Yousefi, S.; Van Doan, H.; Ashouri, G.; Gioacchini, G.; Maradonna, F.; Carnevali, O. Oxidative Stress and Antioxidant Defense in Fish: The Implications of Probiotic, Prebiotic, and Synbiotics. Rev. Fish. Sci. Aquac. 2020, 29, 198–217. [Google Scholar] [CrossRef]
  183. Mohammadi, G.; Hafezieh, M.; Karimi, A.; Azra, M.N.; Van Doan, H.; Tapingkae, W.; Abdelrahman, H.A.; Dawood, M.A.O. The Synergistic Effects of Plant Polysaccharide and Pediococcus Acidilactici as a Synbiotic Additive on Growth, Antioxidant Status, Immune Response, and Resistance of Nile Tilapia (Oreochromis Niloticus) against Aeromonas Hydrophila. Fish Shellfish. Immunol. 2021, 120, 304–313. [Google Scholar] [CrossRef]
  184. El-Haroun, E.R.; Goda, A.M.A.S.; Kabir Chowdhury, M.A. Effect of Dietary Probiotic Biogen® Supplementation as a Growth Promoter on Growth Performance and Feed Utilization of Nile Tilapia Oreochromis Niloticus (L.). Aquac. Res. 2006, 37, 1473–1480. [Google Scholar] [CrossRef]
  185. Michael, E.T.; Amos, S.O.; Hussaini, L.T. A Review on Probiotics Application in Aquaculture. Fish. Aquac. J. 2014, 5, 1. [Google Scholar] [CrossRef]
  186. Batista, K.S.; de Albuquerque, J.G.; de Vasconcelos, M.H.A.; Bezerra, M.L.R.; da Silva Barbalho, M.B.; Oliveira, R.P.; de Souza Aquino, J. Probiotics and Prebiotics: Potential Prevention and Therapeutic Target for Nutritional Management of COVID-19? Nutr. Res. Rev. 2021, 1–42. [Google Scholar] [CrossRef]
  187. McKenzie, V.J.; Kueneman, J.G.; Harris, R.N. Probiotics as a Tool for Disease Mitigation in Wildlife: Insights from Food Production and Medicine. Ann. N. Y. Acad. Sci. 2018, 1429, 18–30. [Google Scholar] [CrossRef]
  188. Ray, P.; Lakshmanan, V.; Labbé, J.L.; Craven, K.D. Microbe to Microbiome: A Paradigm Shift in the Application of Microorganisms for Sustainable Agriculture. Front. Microbiol. 2020, 11, 622926. [Google Scholar] [CrossRef]
  189. Haque, M.M.; Mosharaf, M.K.; Khatun, M.; Haque, M.A.; Biswas, M.S.; Islam, M.S.; Islam, M.M.; Shozib, H.B.; Miah, M.M.U.; Molla, A.H. Biofilm Producing Rhizobacteria with Multiple Plant Growth-Promoting Traits Promote Growth of Tomato under Water-Deficit Stress. Front. Microbiol. 2020, 11, 542053. [Google Scholar] [CrossRef]
  190. Park, K.H.; Oh, S.-Y.; Yoo, S.; Fong, J.J.; Kim, C.S.; Jo, J.W.; Lim, Y.W. Influence of Season and Soil Properties on Fungal Communities of Neighboring Climax Forests (Carpinus cordata and Fraxinus rhynchophylla). Front. Microbiol. 2020, 11, 2643. [Google Scholar] [CrossRef]
  191. Vassilev, N.; Eichler-Löbermann, B.; Flor-Peregrin, E.; Martos, V.; Reyes, A.; Vassileva, M. Production of a Potential Liquid Plant Bio-Stimulant by Immobilized Piriformospora Indica in Repeated-Batch Fermentation Process. AMB Express 2017, 7, 1–7. [Google Scholar] [CrossRef]
  192. Rahman, M.; Sabir, A.A.; Mukta, J.A.; Khan, M.; Alam, M.; Mohi-Ud-Din, M.; Miah, M.; Rahman, M.; Islam, M.T. Plant Probiotic Bacteria Bacillus and Paraburkholderia Improve Growth, Yield and Content of Antioxidants in Strawberry Fruit. Sci. Rep. 2018, 8, 1–11. [Google Scholar]
  193. Niemeyer, J.C.; Lolata, G.B.; de Carvalho, G.M.; Da Silva, E.M.; Sousa, J.P.; Nogueira, M.A. Microbial Indicators of Soil Health as Tools for Ecological Risk Assessment of a Metal Contaminated Site in Brazil. Appl. Soil Ecol. 2012, 59, 96–105. [Google Scholar] [CrossRef]
  194. Vassilev, N.; Vassileva, M.; Martos, V.; Garcia del Moral, L.F.; Kowalska, J.; Tylkowski, B.; Malusá, E. Formulation of Microbial Inoculants by Encapsulation in Natural Polysaccharides: Focus on Beneficial Properties of Carrier Additives and Derivatives. Front. Plant Sci. 2020, 11, 270. [Google Scholar]
  195. González-Salazar, C.; Stephens, C.R.; Marquet, P.A. Comparing the Relative Contributions of Biotic and Abiotic Factors as Mediators of Species’ Distributions. Ecol. Model. 2013, 248, 57–70. [Google Scholar] [CrossRef]
  196. Dwivedi, S.; Sahrawat, K.; Puppala, N.; Ortiz, R. Plant Prebiotics and Human Health: Biotechnology to Breed Prebiotic-Rich Nutritious Food Crops. Electron. J. Biotechnol. 2014, 17, 238–245. [Google Scholar] [CrossRef]
  197. Moradipour, M.; Saberi-Riseh, R.; Mohammadinejad, R.; Hosseini, A. Nano-Encapsulation of Plant Growth-Promoting Rhizobacteria and Their Metabolites Using Alginate-Silica Nanoparticles and Carbon Nanotube Improves Ucb1 Pistachio Micropropagation. J. Microbiol. Biotechnol. 2019, 29, 1096–1103. [Google Scholar] [CrossRef]
  198. Harman, G.; Khadka, R.; Doni, F.; Uphoff, N. Benefits to Plant Health and Productivity from Enhancing Plant Microbial Symbionts. Front. Plant Sci. 2021, 11, 610065. [Google Scholar] [CrossRef]
  199. Angelim, A.L.; Costa, S.P.; Farias, B.C.S.; Aquino, L.F.; Melo, V.M.M. An Innovative Bioremediation Strategy Using a Bacterial Consortium Entrapped in Chitosan Beads. J. Environ. Manag. 2013, 127, 10–17. [Google Scholar] [CrossRef]
  200. Yadav, K.K.; Gupta, N.; Kumar, V.; Singh, J.K. Bioremediation of Heavy Metals from Contaminated Sites Using Potential Species: A Review. Indian J. Environ. Prot. 2017, 37, 65. [Google Scholar]
  201. Baez-Rogelio, A.; Morales-García, Y.E.; Quintero-Hernández, V.; Muñoz-Rojas, J. Next Generation of Microbial Inoculants for Agriculture and Bioremediation. Microb. Biotechnol. 2017, 10, 19–21. [Google Scholar] [CrossRef]
  202. Farhadian, M.; Vachelard, C.; Duchez, D.; Larroche, C. In Situ Bioremediation of Monoaromatic Pollutants in Groundwater: A Review. Bioresour. Technol. 2008, 99, 5296–5308. [Google Scholar] [CrossRef]
  203. Cao, W.; Zhang, H.; Wang, Y.; Pan, J. Bioremediation of Polluted Surface Water by Using Biofilms on Filamentous Bamboo. Ecol. Eng. 2012, 42, 146–149. [Google Scholar] [CrossRef]
  204. Voskoboinikov, G.M.; Matishov, G.G.; Metelkova, L.O.; Zhakovskaya, Z.A.; Lopushanskaya, E.M. Participation of the Green Algae Ulvaria Obscura in Bioremediation of Sea Water from Oil Products. In Proceedings of the Doklady Biological Sciences; Pleiades Publishing: New York, NY, USA, 2018; Volume 481, pp. 139–141. [Google Scholar]
  205. Fragoso ados Santos, H.; Duarte, G.A.S.; Rachid, C.T.d.C.; Chaloub, R.M.; Calderon, E.N.; Marangoni, L.F.d.B.; Bianchini, A.; Nudi, A.H.; Do Carmo, F.L.; van Elsas, J.D. Impact of Oil Spills on Coral Reefs Can Be Reduced by Bioremediation Using Probiotic Microbiota. Sci. Rep. 2015, 5, 1–11. [Google Scholar] [CrossRef] [Green Version]
  206. Sarlak, Z.; Khosravi-Darani, K.; Rouhi, M.; Garavand, F.; Mohammadi, R.; Sobhiyeh, M.R. Bioremediation of Organophosphorus Pesticides in Contaminated Foodstuffs Using Probiotics. Food Control 2021, 126, 108006. [Google Scholar] [CrossRef]
  207. Nybom, S.M.K.; Dziga, D.; Heikkilä, J.E.; Kull, T.P.J.; Salminen, S.J.; Meriluoto, J.A.O. Characterization of Microcystin-LR Removal Process in the Presence of Probiotic Bacteria. Toxicon 2012, 59, 171–181. [Google Scholar] [CrossRef] [PubMed]
  208. Mirza Alizadeh, A.; Hosseini, H.; Mohseni, M.; Eskandari, S.; Sohrabvandi, S.; Hosseini, M.-J.; Tajabadi-Ebrahimi, M.; Mohammadi-Kamrood, M.; Nahavandi, S. Analytic and Chemometric Assessments of the Native Probiotic Bacteria and Inulin Effects on Bioremediation of Lead Salts. J. Sci. Food Agric. 2021, 101, 5142–5153. [Google Scholar] [CrossRef] [PubMed]
  209. Niderkorn, V.; Boudra, H.; Morgavi, D.P. Binding of Fusarium Mycotoxins by Fermentative Bacteria In Vitro. J. Appl. Microbiol. 2006, 101, 849–856. [Google Scholar] [CrossRef] [PubMed]
  210. Sampaio Baptista, A.; Horii, J.; Antonia Calori-Domingues, M.; Micotti da Glória, E.; Mastrodi Salgado, J.; Roberto Vizioli, M. The Capacity of Manno-Oligosaccharides, Thermolysed Yeast and Active Yeast to Attenuate Aflatoxicosis. World J. Microbiol. Biotechnol. 2004, 20, 475–481. [Google Scholar] [CrossRef]
  211. Shetty, P.H.; Hald, B.; Jespersen, L. Surface Binding of Aflatoxin B1 by Saccharomyces Cerevisiae Strains with Potential Decontaminating Abilities in Indigenous Fermented Foods. Int. J. Food Microbiol. 2007, 113, 41–46. [Google Scholar] [CrossRef]
Figure 1. Main steps of the thermal probiotic powder profiling and fingerprinting experimentation: (a) from powder weighing to TDA-DSC analysis, and (b) output data.
Figure 1. Main steps of the thermal probiotic powder profiling and fingerprinting experimentation: (a) from powder weighing to TDA-DSC analysis, and (b) output data.
Microorganisms 10 01700 g001aMicroorganisms 10 01700 g001b
Figure 2. TGA curves of (a) mono-strains and (b) multi-strains.
Figure 2. TGA curves of (a) mono-strains and (b) multi-strains.
Microorganisms 10 01700 g002
Figure 3. Thermal curves of (a) maltose-containing formulation vs. maltose and (b) pure vs. formulated L. plantarum ATCC8014.
Figure 3. Thermal curves of (a) maltose-containing formulation vs. maltose and (b) pure vs. formulated L. plantarum ATCC8014.
Microorganisms 10 01700 g003
Figure 4. Basic principles of separation by CTCE “Reprinted with permission from Ref. [60]. 2022, John Wiley & Sons”.
Figure 4. Basic principles of separation by CTCE “Reprinted with permission from Ref. [60]. 2022, John Wiley & Sons”.
Microorganisms 10 01700 g004
Figure 5. Example of genetic map of a subset of Lactobacillus species.
Figure 5. Example of genetic map of a subset of Lactobacillus species.
Microorganisms 10 01700 g005
Figure 6. Example of a Bifidobacterium map.
Figure 6. Example of a Bifidobacterium map.
Microorganisms 10 01700 g006
Figure 7. A genetic map of B. longum subspecies.
Figure 7. A genetic map of B. longum subspecies.
Microorganisms 10 01700 g007
Figure 8. ATGC analysis of Lactobacilli mixtures.
Figure 8. ATGC analysis of Lactobacilli mixtures.
Microorganisms 10 01700 g008
Figure 9. Illustration of action mechanisms of probiotics for promoting human health. GLP1: glucagon-like peptide 1, SCFAs: short-chain fatty acids, sIgA: soluble immunoglobulin A, TLR: toll-like receptor.
Figure 9. Illustration of action mechanisms of probiotics for promoting human health. GLP1: glucagon-like peptide 1, SCFAs: short-chain fatty acids, sIgA: soluble immunoglobulin A, TLR: toll-like receptor.
Microorganisms 10 01700 g009
Figure 10. Potential applications of postbiotics in human health.
Figure 10. Potential applications of postbiotics in human health.
Microorganisms 10 01700 g010
Figure 11. Diagram showing the three strategies for microbial soil–plant management based on prebiotics, probiotics, and postbiotics approaches. Legend: Full lines show the direct effect, dashed lines show the interactions, dotted lines show the formulation/production processes [194].
Figure 11. Diagram showing the three strategies for microbial soil–plant management based on prebiotics, probiotics, and postbiotics approaches. Legend: Full lines show the direct effect, dashed lines show the interactions, dotted lines show the formulation/production processes [194].
Microorganisms 10 01700 g011
Figure 12. Main mechanisms of action of probiotics in remediation.
Figure 12. Main mechanisms of action of probiotics in remediation.
Microorganisms 10 01700 g012
Table 1. Some examples of synbiotics, their probiotic and prebiotic components, and trade names.
Table 1. Some examples of synbiotics, their probiotic and prebiotic components, and trade names.
Synbiotics
ProbioticsPrebioticsProducts
Lacticaseibacillus paracasei YIT 9029
(strain Shirota: LcS)
Bifidobacterium breve YIT 12272 (BbrY)
GOSSuper Synbiotics LBG-P
(Yakult Honsha Co., Ltd., Tokyo, Japan)
Streptococcus thermophilus
Lacticaseibacillus rhamnosus
Lactobacillus acidophilus
B. infantis
B. lactis
FOS + Ascorbic acidProbiotical
(Laboratoires Phacobel Belgium, Soheit-Tinlot, Belgium)
B. breveShort chain scGOS
Long chain lcFOS
Danone Nutricia Research, Utrecht, The Netherlands
Bacillus coagulans MTCC 5856Cranberry fiberLactocran (Sabinsa Corporation, Piscataway, NJ, USA)
B. lactis, B. breve, B. infantis, B. longumXOSPrebioMed™ XOS (designs for health, Suffield, CT, USA)
Mix of Bifidobacteria and LactobacilliWhole fruit Indian Pomegranate
(Punica granatum)
(>40% Polyphenolic + Phenolic Bioactives)
DS-01 (Seed Health, Los Angeles, CA, USA)
L. acidophilus
Lacticaseibacillus casei, L. rhamnosus
Lactiplantibacillus plantarum,
S. thermophilus
B. longum
Oat Bran (10% β-Glucan fiber)
Organic Red Beetroot
Inulin from Organic Chicory Root
Beta Glucan Synbiotic (BioImmersion Inc, Bellevue, WA, USA)
B. animalis,
Enterococcus faecium,
Limosilactobacillus reuteri
Ligilactobacillus salivarius
Pediococcus acidilactici
InulinPoultryStar® (ME BIOMIN GmbH, Niederösterreich, Austria)
Enterococcus faeciumFOSBiomin® IMBO (ME BIOMIN GmbH, Niederösterreich, Austria)
L. acidophilus
L. casei
L. salivarius
L. plantarum, L. rhamnosus
Levilactobacillus brevis
B. bifidum
B. lactis
S. thermophilus
InulinSynbiotic poultry (Vetafarm, Wagga Wagga, Australia)
Table 2. Main thermophysical quantities from the thermal profiling of probiotic-based products.
Table 2. Main thermophysical quantities from the thermal profiling of probiotic-based products.
Thermophysical QuantitiesMeaningSource
Tmax [°C]Maximum temperature of main decomposition
(first derivative or DTG main peak)
TGA
Vmax [Kmin−1]Maximum rate of main decomposition
(first derivative or DTG main peak)
TGA
Tm [°C]Temperature of phase transition of main decompositionDSC
ΔHm [J/g]Enthalpy of phase transition of main decomposition
(area of the curve)
DSC
R600 [%]Black carbon and mineral compounds at the end of the
temperature scan (600 °C)
TGA
Table 3. Thermophysical data of mono-strain and multi-strain (VSL#3) samples of probiotic formulations.
Table 3. Thermophysical data of mono-strain and multi-strain (VSL#3) samples of probiotic formulations.
Probiotic FormulationsTmaxVmaxTmΔHmR600
[°C]100 × [min−1][°C][J/g][%]
Mono-strains
B. subtilis350.7 ± 0.73.93 ± 0.11352.9 ± 0.9−23.0 ± 3.024.0 ± 1.1
B. longum314.3 ± 0.17.04 ± 0.07310.9 ± 0.4−5.1 ± 1.712.8 ± 0.3
S. boulardii275.4 ± 0.32.60 ± 0.03279.5 ± 0.433.3 ± 2.033.4 ± 0.3
S. thermophilus264.6 ± 0.14.01 ± 0.03273.2 ± 1.012.4 ± 1.232.7 ± 0.2
L. rhamnosus GG228.2 ± 0.56.44 ± 0.06228.1 ± 1.026.1 ± 4.024.5 ± 0.3
L. bulgaricus165.2 ± 0.03.21 ± 0.04165.6 ± 0.0−97.9 ± 1.332.2 ± 0.1
Multi-strains VSL#3
Lot 606035301.2 ± 0.22.97 ± 0.02305.8 ± 1.731.8 ± 4.530.1 ± 0.0
Lot 45752294.1 ± 0.94.24 ± 0.03290.0 ± 3.070.0 ± 8.328.4 ± 0.6
Lot 10151198206.9 ± 2.13.00 ± 0.01196.4 ± 4.5−33.5 ± 2.832.9 ± 0.9
Lot 3302E10204.3 ± 3.03.13 ± 0.11193.7 ± 4.6−41.2 ± 11.533.4 ± 0.2
Table 4. A list of Bacillus family species and their application in agriculture for ATGC assay.
Table 4. A list of Bacillus family species and their application in agriculture for ATGC assay.
SpeciesKnown FunctionReference
Bacillus subtilisSolubilize soil (phosphorus), enhance nitrogen fixation, and produce siderophores that promote its growth and suppresses the growth of pathogens[75]
Cytobacillus firmusNematode antagonist[76]
Bacillus thuringiensisKnown pesticide activities[77]
Bacillus mucilaginosusSolubilize potassium from minerals in soil so that plants, such as food crops, are able to use it[78]
Paenibacillus polymyxaFix nitrogen, making it available to plants[79]
Bacillus cereusRegenerate contaminated soils and promote mycorrhizae growth[80]
Bacillus amyloliquefaciensPrevent a series of bacterial pathogens in crops[81]
Bacillus pumilusPromote plant growth, fix nitrogen, and prevent the germination of several fungal pathogens on crop roots[82]
Bacillus megateriumSolubilizes phosphates, and promotes plant growth through signaling[83]
Table 5. Example of human health benefits of synbiotics claimed from clinical studies.
Table 5. Example of human health benefits of synbiotics claimed from clinical studies.
Health OutcomesProbiotic StrainsPrebioticsReference
Gut Intestinal tractTreatment of overweight and metabolic syndromeL. casei PXN 37, L. rhamnosus PXN 54,
S. thermophilus PXN 66, B. breve PXN 25,
L. acidophilus PXN 35, B. longum PXN 30,
L. bulgaricus PXN 39
FOS[133]
Treatment of IBSBacillus coagulansFOS[134]
Acute diarrheaL. acidophilus, L. rhamnosus, B. bifidum,
B. longum, Enterococcus faecium
FOS[135]
Colorectal cancerB. lactisResistant starch[136]
KidneyTreatment of chronic kidney
disease
L. casei, L. acidophilus, L. bulgaricus, L. rhamnosus,
B. breve, B. longum, S. thermophilus
FOS[137]
LiverTreatment of non-alcoholic fatty liver disease
Prevention of infections after liver transplant
B. longum
L. acidophilus
Inulin HP[138]
LungReduction of viral respiratory infections in asthmatic childrenL. casei, L. rhamnosus, S. thermophilus, B. breve,
L. acidophilus, B. infantis, L. bulgaricus
FOS[139]
Skin/dermTreatment of atopic dermatitisL. salivarius PM-A0006FOS[140]
BrainImprovement in mental health in hemodialysis patientsL. acidophilus strain T16, B. bifidum strain BIA-6,
B. lactis strain BIA-7, B. longum strain BIA-8
Equal mix of FOS, GOS and inulin[141]
Table 6. Effect of some microbial pre/pro/synbiotics on ruminant production.
Table 6. Effect of some microbial pre/pro/synbiotics on ruminant production.
Ruminant HostPre-/Pro-/SynbioticsMode of Administration/DoseEffectReference
Dairy cowsL. casei and L. plantarumCombination of both in the feed
(50 g/day)
Increases the milk production and the contents of milk immunoglobulin G, lactoferrin, lysozyme and lactoperoxidase[165,166]
Propionibacterium spp. and
S. cerevisiae
Oral administration, mixed in feedImproves the feed conversion rate, milk production and dry matter intake[162]
S. cerevisiaeOral administration, mixed in feed (0.2 g/day)Improves the feed conversion rate, milk production and dry matter intake[167]
Fructo-oligosacchrides (FOSs) and Mannan-oligosaccharides (MOSs)Oral administrationProvides specific bacteria with a competitive advantage in the gut.[168]
Enterococcus faecium + lactuloseOral administration 109 colony forming units (probiotics) + 1–3% dry matter (prebiotics)Decreases the ileal villus height, the depth of the crypts in the cecum, and the surface area of lymph follicles from Peyer’s patches[169]
Streptococcus faecium + Mannan-oligosaccharide (MOS)Oral administration (0.6 kg/day)Improves fecal consistency and reduced the fecal score of calves[170]
S. cerevisiae strain 1026 + InulinOral administration
(probiotic 5 g + prebiotic 6 g)
Impacts positively the development of morphological structures of digestive systems[171]
GoatsL. reuteri, L. alimentarius,
Enterococcus faecium and
B. bifidum
Oral administration, resuspended in milk (1 mL/two feeds per day)Improves the microbial environment and intestinal health, in addition to the acid profile of milk, with an increase in unsaturated fatty acids, mainly linoleic, linolenic and conjugated linoleic acids, and a decrease in the atherogenic index[159]
Inulin, fructo-oligosaccharide, galacto-oligosaccharide and
xylo-oligosaccharide
0.4 to 0.6% in milkAntioxidant activity and promotes the development of functional goat milk[172]
SheepS. cerevisiae and two strains of rumen-derived Diutina rugosaOral administration (100 mL)Stabilizes the ruminal pH, improves the richness of rumen microflora, relieves acidosis and inflammation, and prevents subacute ruminal acidosis[173,174]
Propionibacterium P63, L. plantarum and L. rhamnosusIntraruminal cannula (2 g/day)Stabilizes the pH of the rumen and prevents acidosis[173,174]
Mannan-oligosaccharide and
b-glucans
Intraruminal cannulaAdditive effects on digestion and fermentation[175]
CattleEnterococcus faecium strain 26 and Clostridium butyricum strain MiyariOral administrationReduces the ruminal pH and the concentration of lactic acid in the ruminal fluid, thus preventing acidosis[159]
Cellooligosaccharide (CE), Mannan-oligosaccharides (MOSs) and fructo-oligosaccharides (FOSs) such as Galactosyl-lactose in combination with spray-dried bovine serumOral administration of supplemented milk replacerReduces the incidence and severity of enteric disease and modulate the intestinal bacterial community in calves[165]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kouhounde, S.; Adéoti, K.; Mounir, M.; Giusti, A.; Refinetti, P.; Otu, A.; Effa, E.; Ebenso, B.; Adetimirin, V.O.; Barceló, J.M.; et al. Applications of Probiotic-Based Multi-Components to Human, Animal and Ecosystem Health: Concepts, Methodologies, and Action Mechanisms. Microorganisms 2022, 10, 1700. https://doi.org/10.3390/microorganisms10091700

AMA Style

Kouhounde S, Adéoti K, Mounir M, Giusti A, Refinetti P, Otu A, Effa E, Ebenso B, Adetimirin VO, Barceló JM, et al. Applications of Probiotic-Based Multi-Components to Human, Animal and Ecosystem Health: Concepts, Methodologies, and Action Mechanisms. Microorganisms. 2022; 10(9):1700. https://doi.org/10.3390/microorganisms10091700

Chicago/Turabian Style

Kouhounde, Sonagnon, Kifouli Adéoti, Majid Mounir, Alessandro Giusti, Paulo Refinetti, Akaninyene Otu, Emmanuel Effa, Bassey Ebenso, Victor O. Adetimirin, Josep Mercader Barceló, and et al. 2022. "Applications of Probiotic-Based Multi-Components to Human, Animal and Ecosystem Health: Concepts, Methodologies, and Action Mechanisms" Microorganisms 10, no. 9: 1700. https://doi.org/10.3390/microorganisms10091700

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