Next Article in Journal
Exposure of Wild Ruminants to Toxoplasma gondii in Alpine Ecosystems, NE Spain
Previous Article in Journal
Evaluating the Role of Social Media in Veterinary Anatomy and Clinical Education: A Student-Based Study
Previous Article in Special Issue
Nano-Encapsulated Cumin Oil and Bacillus subtilis Enhance Growth Performance, Immunity, Oxidative Stability, and Intestinal Integrity in Growing Rabbits Under High Ambient Temperature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Swine-Derived Probiotics and Their Metabolites as an Alternative to Veterinary Antibiotics

1
Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
2
Quanzhou Animal Husbandry Station, Quanzhou 362000, China
3
Fujian Provincial Animal Husbandry Station, Fuzhou 350001, China
*
Author to whom correspondence should be addressed.
Vet. Sci. 2025, 12(11), 1100; https://doi.org/10.3390/vetsci12111100
Submission received: 21 August 2025 / Revised: 7 October 2025 / Accepted: 12 November 2025 / Published: 18 November 2025

Simple Summary

Prompted by the need to combat antibiotic overuse-induced bacterial resistance in livestock farming—a serious threat to food safety and public health—probiotics and their metabolites have emerged as vital alternatives, proving effective in enhancing animal growth, immunity, and gut microecological balance when administered via feed, water, or fermentation. Swine-derived strains show particular promise due to their host compatibility, pathogen-inhibiting potential, and cost-effectiveness. This review systematically analyzes their isolation, metabolite functions, mechanisms of action, and application efficacy while evaluating innovations and future prospects to advance sustainable, antibiotic-free animal farming.

Abstract

The intensive development of livestock and poultry farming has heavily relied on antibiotics, leading to widespread antimicrobial resistance and posing serious threats to food safety and public health. As the industry transitions towards reduced antibiotic use and sustainable animal production, probiotics and their metabolites have garnered attention as functional alternatives. Probiotics are typically administered in the form of microecological preparations by mixing them into feed or water, offering advantages in cost-effectiveness and ease of use, with demonstrated efficacy in promoting animal health. Swine-derived probiotics, in particular, demonstrate host-specific advantages due to their natural adaptation to the porcine gastrointestinal environment, which improves intestinal colonization, pathogen inhibition, and immune modulation. Their metabolites, including short-chain fatty acids, bacteriocins, and exopolysaccharides, further contribute to these benefits through antimicrobial, anti-inflammatory, and barrier-strengthening effects. Recent studies have demonstrated improvements in average daily gain (18–22%) and feed conversion ratio (12–15%), along with a reduction in diarrhea incidence (up to 40–45%) in weaned piglets supplemented with certain probiotic consortia. It should be noted, however, that part of the supporting evidence is derived from in vitro or non-porcine models, and practical outcomes in swine may vary depending on husbandry conditions, probiotic strain, and husbandry conditions. This review systematically summarizes the isolation and identification of swine-derived probiotics, the active components and functions of their metabolites, and the mechanisms of action and application effects of these metabolites as antibiotic-alternative feed additives. It primarily focuses on innovative research advances in probiotic metabolites for enhancing antibacterial activity and improving pig growth performance. Furthermore, the review discusses the prospects for commercial applications and future research directions, aiming to provide theoretical foundations and technical references for green and healthy farming practices.

1. Probiotics and Their Beneficial Effects

Antibiotics, as crucial agents in the prevention and treatment of bacterial infections, are wildely utilized in animal husbandry [1]. However, their extensive application has resulted in significant threats, including the dissemination of antibiotic resistance genes, disruption of intestinal microbiota, and environmental pollution [2]. In global swine production, the prolonged and excessive use of antibiotics has facilitated the spread of resistant bacteria and increased drug residues, threatening both product safety and public health [3]. Consequently, many regions are implementing stricter regulations. For instance, the European Union has long enforced restrictions on antibiotic growth promoters, while market pressures in the United States and Brazil are also prompting a shift toward reduced usage. In China, the “Feed Antibiotic Ban” (Announcement No. 194, 2019) officially prohibited the use of antibiotic feed additives as of July 2020, accelerating the need for safe and effective antibiotic alternatives (AAs) [4].
Various AAs candidates, including enzymes, plant extracts, acidifiers, and probiotics, as well as probiotics and their metabolites, have received increasing attention as functional feed additives [5]. Probiotics are live microorganisms conferring health benefits to the host and are commonly applied in animal husbandry as feed additives [6,7]. Major genera include Bifidobacterium, Lactobacillus, and Bacillus (Table 1) [8]. Their beneficial effects are mediated through multiple mechanisms, including nutrient digestion, immune modulation, intestinal development, and microecological maintenance (Figure 1). Under experimental conditions, certain probiotic supplements have been shown to reduce diarrhea incidence by 30–40% and improve the feed conversion ratio by 5–10% in weaned piglets [9,10].
In swine production, swine-derived probiotics, as indigenous commensals, offer distinct advantages over exogenous strains. Beneficial microorganisms isolated from healthy pigs, such as lactic acid bacteria, bacilli, and yeasts, typically exhibit superior gut colonization capacity [25,26]. This enhanced colonization not only competitively excludes opportunistic pathogens but also strengthens the host’s resistance to infections, a mechanism referred to as colonization resistance. For example, certain swine-derived Lactobacillus strains have been demonstrated to reduce E. coli colonization by 2–3 log units in challenged piglets [27]. Furthermore, swine-derived probiotics produce various bioactive substances through fermentation metabolism, including short-chain fatty acids (SCFAs), antimicrobial peptides, and exopolysaccharides [28]. Owing to their unique value in maintaining intestinal microecological homeostasis, inhibiting pathogen colonization and growth, and enhancing host immune defense functions, swine-derived probiotics and their metabolites have become crucial targets for developing novel AAs strategies [29,30].
Metabolites derived from single strains often exhibit limitations such as a narrow activity spectrum or insufficient potency, and their industrial-scale production is constrained by fermentation process parameters. In recent years, consortia-based synergistic fermentation technology has garnered significant attention due to the diversity of metabolic pathways and the functional synergy and complementarity among strains [31]. This article systematically elucidates the isolation and identification of swine-derived probiotics, the active components and functions of their metabolites, the mechanisms of action, and application efficacy of AAs feed additives based on swine-derived probiotics and their metabolites, and reviews cutting-edge research advances in this field. The goal is to provide a robust theoretical foundation for the development of novel and highly efficient AAs feed additives.

2. Isolation, Identification and Selection of Swine-Derived Beneficial Strains

The isolation and identification of swine-derived beneficial strains form the foundation for developing novel probiotic agents and AAs. This approach combines conventional microbiological methods with advanced omics technologies, offering both substantial scientific challenges and application value.
The standard isolation procedure typically begins with the aseptic collection of gastrointestinal contents, fecal samples, or colostrum from healthy pigs (Figure 2). Strains isolated from colostrum (lactobacilli and bifidobacteria) typically exhibit superior capabilities in gut colonization and immune regulation. In contrast, fecal-derived strains (butyrate-producing clostridia) are generally more adept at dietary fiber decomposition, competitive exclusion of pathogens, and tolerance to digestive stresses [32]. Following collection, samples undergo serial dilution in sterile phosphate-buffered saline (PBS) before being evenly spread onto selective solid media. These plates are subsequently incubated in an anaerobic workstation (oxygen concentration < 0.1%) at 37 °C for 48–72 h to accurately simulate the intestinal anaerobic microenvironment. This approach significantly enhances target strain enrichment efficiency, yielding a 3- to 5-fold increase compared to conventional methods [33,34]. Following incubation, individual colonies are selected and purified through three consecutive streak-plating generations on fresh media to ensure genetic homogeneity.
While these classical culture-based techniques remain foundational for probiotic isolation, recent methodological advances have greatly expanded the resolution and scope of strain characterization through multi-omics integration. Current identification technologies demonstrate a trend toward multidimensional integration. The combination of traditional physiological-biochemical characterization with 16S rRNA gene sequencing enables preliminary taxonomic classification at the phylum/genus level [35,36]. However, its resolution is limited for distinguishing closely related species due to the high conservation of the 16S rRNA gene. In contrast, whole-genome sequencing (WGS) coupled with average nucleotide identity analysis achieves precise discrimination of closely related strains exhibiting >99% homology, substantially enhancing identification resolution and efficiency [37]. Nevertheless, the cost and bioinformatic demands of WGS can limit its routine use. For functional characterization, metabolomics techniques such as ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) facilitate systematic identification of strain-specific metabolites such as short-chain fatty acids and vitamins [38]. Similarly, transcriptomics (RNA-seq) elucidates the molecular regulatory networks that underlie stress responses including acid and bile salt tolerance [39]. These multi-omics technologies provide a critical technological underpinning for screening high-performance probiotic strains. By integrating genomic data with phenotypic metabolomic profiles, researchers can select strains based not only on their genetic potential but also on their documented functional output. This approach enables the identification of candidates with enhanced survival, colonization, and metabolic efficacy in the swine gastrointestinal environment.
Following genomic and functional screening, it is imperative to validate the viability and robustness of candidate strains under simulated practical conditions. This step bridges the gap between genetic potential and practical application.
In vitro viability assessments serve as a primary, cost-effective screening tool. Key tests include evaluating tolerance to simulated gastric acid (pH 2.0–3.0) and intestinal bile salts, with survival quantified by plate counts or flow cytometry; assessing thermal stability at feed pelleting temperatures (60–80 °C); and determining storage stability in various carrier matrices. Data from these studies provide critical go/no-go decisions for advancing candidates to in vivo testing.
In vivo studies are indispensable, as in vitro systems cannot fully replicate the complex gastrointestinal environment. These trials involve administering the probiotic and tracking viable cells through fecal samples to confirm their survival and gut transit. Post-euthanasia analysis of digestive contents and mucosal tissues further assesses the strain’s ability to colonize specific niches (ileum, colon) [40]. Crucially, in vivo viability data must be correlated with health and performance outcomes—including gut morphology, immune markers, and growth performance—to establish a direct link between the presence of the viable probiotic and its functional benefits.

3. Metabolites Derived from Beneficial Microorganisms of Swine Origin

3.1. Short-Chain Fatty Acids (SCFAs)

SCFAs, the principal end-products of dietary fiber fermentation by gut microbiota, play a pivotal regulatory role in maintaining intestinal homeostasis and pro-/anti-inflammatory balance in swine [41,42]. Dominant components include acetate, propionate, and butyrate, collectively accounting for approximately 80% of total SCFAs (Table 2). Butyrate plays a particularly significant physiological role as the preferred energy substrate for colonic epithelial cells, being directly absorbed and utilized in mucosal barrier fortification [43,44]. Schilderink [31] demonstrated in cell cultures and rodent models that butyrate significantly upregulates tight junction proteins (Claudin-1 and ZO-1) through histone deacetylase (HDAC) inhibition, thereby enhancing intestinal barrier integrity and reducing pathogen and endotoxin translocation risks (Figure 3).
At the metabolic regulatory level, SCFAs suppress key lipogenesis gene expression via the peroxisome proliferator-activated receptor gamma (PPAR-γ) pathway while synergistically enhancing mineral bioavailability through organic acid chelation, ultimately optimizing growth performance [45,46]. Their immunomodulatory functions include: Promoting regulatory T cell (Treg) differentiation via histone acetylation-mediated epigenetic modification; Polarizing macrophages toward an anti-inflammatory phenotype through G protein-coupled receptor 43 (GPR43) signaling, effectively mitigating stress-induced inflammation in weaned piglets [47]. These mechanisms, while well-documented in murine systems, require further validation in swine.
Furthermore, SCFAs selectively inhibit pathogenic bacteria (Escherichia coli) proliferation by reducing intestinal pH, while simultaneously facilitating metabolic cross-feeding with commensals such as Clostridium butyricum to restore microbial equilibrium [48,49].
Table 2. Mechanisms of action and effects of short-chain fatty acids produced by probiotics.
Table 2. Mechanisms of action and effects of short-chain fatty acids produced by probiotics.
CompoundMechanismEffects
Acetic acidAcetate serves as an energy source for colonic cells, mucosal epithelial cells, and muscle tissue. It also participates in hepatic metabolism and crosses the blood–brain barrier to regulate appetite through central mechanisms.The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health [50]
Propionic acidPropionate, absorbed in the colon, is primarily used for hepatic gluconeogenesis. It enhances intestinal epithelial activity and suppresses the synthesis of cholesterol and lipids.Propionate alleviates palmitic acid-induced endoplasmic reticulum stress by enhancing autophagy in calf hepatic cells [51]
Butyric acidButyrate acts as the main energy substrate for the colon and cecum. It strengthens the intestinal barrier and exerts anti-inflammatory effects via HDAC inhibition, while also modulating immune cell differentiation.Short-chain fatty acids and their producing organisms: An overlooked therapy for IBD? [52]
Pentoic acidValerate inhibits Th17 cell proliferation and IL-17A production, downregulates key Th17-related genes, and promotes IL-10 expression along with metabolic reprogramming, thereby reducing pathogenicity.The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes [53]

3.2. Antimicrobial Peptides

Naturally produced antimicrobial compounds derived from swine-derived beneficial strains—such as bacteriocins, organic acids, and defensins—exhibit potent broad-spectrum antimicrobial activity (Table 3) [54,55]. These compounds are proposed to help maintain intestinal microecological homeostasis and defend against pathogen invasion [56,57,58,59]. Among them, bacteriocins, as ribosomally synthesized antimicrobial peptides, demonstrate high target specificity and biosafety [60,61]. Within porcine microbiota, bacteriocins produced by Lactobacillus spp. have been most extensively studied. In vitro studies, such as that by Du Kun [62] demonstrated that plantaricin secreted by Lactobacillus plantarum disrupts cell membrane integrity and induces transmembrane pore formation, leading to collapse of ion gradients and leakage of macromolecules, thereby effectively inhibiting the proliferation of Gram-negative pathogens such as Escherichia coli and Salmonella spp. (Figure 3).
Notably, specific bacteriocins can inhibit pathogen biofilm formation by interfering with quorum-sensing systems, potentially blocking their adhesion to intestinal epithelial cells and reducing infection risks [63,64]. As reported by Grande Burgos [65], the cyclic bacteriocin enterocin AS-48 from Enterococcus not only effectively inhibits Staphylococcus aureus biofilm development but also suppresses the growth of Listeria monocytogenes, Salmonella, and enterotoxigenic Escherichia coli.
Compared to conventional antibiotics, bacteriocins may exhibit a lower potential for inducing resistance due to their specific targets. They can be enzymatically degraded by proteases (trypsin) in the gastrointestinal tract without residue accumulation in animal products, establishing them as promising antibiotic alternatives [66,67]. However, their efficacy and stability under commercial swine production conditions require further investigation.
Table 3. Common Bioactive Peptides produced by probiotics and their mechanisms of action.
Table 3. Common Bioactive Peptides produced by probiotics and their mechanisms of action.
CompoundMechanismEffect
BacteriocinsProteinaceous toxins that inhibit the growth of closely related bacterial strains and foodborne pathogens by forming pores in the cell membrane, disrupting cell wall synthesis, or inhibiting enzyme activity.Bacteriocins: Properties and potential use as antimicrobials [68]
Bioactive PeptidesPeptides released during the fermentation of milk, legumes, or other proteins by probiotics. Exhibits antihypertensive (ACE-inhibition), antioxidant, antimicrobial, immunomodulatory, and opioid-like activities.Bioactive peptides from meat: Current status on production, biological activity, safety, and regulatory framework [69]
Surface Layer ProteinsCrystalline protein structures coating the cell surface. Contribute to adhesion to the intestinal epithelium, competitive exclusion of pathogens, modulation of immune responses, and maintenance of epithelial barrier function.Lactobacillus surface layer proteins: structure, function and applications [70]

3.3. Exopolysaccharides (EPS)

EPS from swine-derived probiotics are proposed to regulate gastrointestinal health and immune function through multiple mechanisms (Figure 3). Their bioactivities encompass antioxidant effects, immunomodulation, intestinal mucosal barrier construction, and microbial homeostasis maintenance(Table 4) [71,72,73]. Recent in vitro studies highlight EPS’s potent in vitro antioxidant activity: it enhances cellular self-defense capacity while effectively scavenging free radicals and mitigating reactive oxygen species (ROS)-induced oxidative damage, thereby preserving cellular structural integrity [74,75].
Regarding immunomodulation, in vitro evidence suggests EPS enhances host–pathogen resistance by activating immune cells including macrophages, B lymphocytes, and T lymphocytes [76,77,78]. This process involves binding to immune cell surface receptors, triggering downstream signaling transduction pathways that promote immune cell activation and functional enhancement, ultimately elevating systemic immune responses [79].
For intestinal mucosal barrier protection, EPS operates through dual mechanisms: interaction with mucins in the intestinal mucus layer to form a physical barrier that inhibits pathogen epithelial adhesion [80], while simultaneously stimulating goblet cells to secrete MUC2 mucin, increasing mucus layer thickness by 2- to 3-fold to block pathogen invasion [81]. It is important to note that “increasing mucus layer thickness by 2- to 3-fold,” are primarily supported by rodent or in vitro studies and require direct validation in swine. In microbial homeostasis regulation, EPS may function as a prebiotic to enhance microbial diversity and promote the proliferation of beneficial bacteria while suppressing the colonization of pathogens such as Clostridium perfringens, although data from swine are limited [82,83].
Table 4. Common Polysaccharides produced by probiotics and their mechanisms of action.
Table 4. Common Polysaccharides produced by probiotics and their mechanisms of action.
CompoundMechanismEffects
Exopolysaccharides (EPS)High-molecular-weight sugar polymers secreted into the environment. Form a protective biofilm, aid in adhesion to surfaces, protect the producing bacterium from harsh conditions (acid, bile), and modulate the host immune system towards an anti-inflammatory state.Exopolysaccharides from probiotic bacteria and their health potential [84]
Lipopolysaccharides (LPS) (from specific probiotic strains)Component of the outer membrane of Gram-negative bacteria. Probiotic-derived LPS often has a less inflammatory structure (penta-acylated lipid A) that can act as an antagonist to more inflammatory LPS from pathogens, thereby training the immune system and reducing excessive inflammation.The role of lipopolysaccharide in the development of atopy in humans [85]
Capsular Polysaccharides (CPS)Tightly bound polysaccharide layer surrounding the cell. Provides physical protection against phagocytosis and antimicrobial peptides, facilitates biofilm formation, and can directly interact with host pattern-recognition receptors to elicit immunomodulatory effects.The Intestinal Commensal, Bacteroides fragilis, Modulates Host Responses to Viral Infection and Therapy: Lessons for Exploration during Mycobacterium tuberculosis Infection [86]

3.4. Vitamins and Coenzymes

Vitamins (including B-complex vitamins and vitamin K) and coenzymes (coenzyme A, NAD+) derived from porcine beneficial strain metabolism are thought to enhance host nutritional metabolic efficiency and physiological functions by engaging core metabolic pathways and signaling networks [87,88,89]. Lactobacillus reuteri-synthesized 5-methyltetrahydrofolate (5-MTHF), a highly bioavailable active folate, exhibits 42% higher absorption efficiency mediated by the reduced folate carrier (RFC) compared to synthetic folate, a finding demonstrated in human studies. Its potential to prevent megaloblastic anemia in swine remains to be evaluated [90,91]. Riboflavin (vitamin B2), as a precursor of flavin adenine dinucleotide (FAD), increases mitochondrial oxidative phosphorylation efficiency by 18% through activation of the succinate dehydrogenase complex, while simultaneously modulating Th17/Treg cell balance via the aryl hydrocarbon receptor (AHR) signaling axis to exert immunoregulatory functions; however, its efficacy and optimal dosage in swine require further investigation [92]. Menaquinone-7 (MK-7), a vitamin K2 component serving as an essential cofactor for γ-glutamyl carboxylase (GGCX), promotes γ-carboxylation of osteocalcin, increasing bone ash content by 8–10% and significantly improving limb-hoof structural integrity in breeding swine [93,94]. Furthermore, the coenzyme nicotinamide adenine dinucleotide (NAD+) has been demonstrated in cellular and animal models to enhance intestinal epithelial cell autophagy through SIRT1 pathway activation, thereby mitigating oxidative stress damage; its protective role and mechanism in swine intestinal health warrant further validation [95].

4. Mechanisms and Efficacy of Swine-Derived Probiotic Metabolites

Swine-derived probiotic metabolites represent a promising class of natural bioactive compounds that contribute to intestinal health through multidimensional mechanisms including the promotion of morphological development, immune modulation, enhancement of nutrient absorption, metabolic optimization, and maintenance of microbial homeostasis. These integrated functions suggest considerable potential as part of antibiotic-reduction strategies, although their application requires further validation under commercial conditions (Figure 3).

4.1. Enhancement of Intestinal Morphological Development and Physical Barrier Function

Probiotic metabolites contribute to the reinforcement of intestinal barrier integrity by promoting the proliferation and differentiation of epithelial cells. Short-chain fatty acids (SCFAs), produced by swine-derived Lactobacillus, have been shown to improve jejunal villus development and increase the villus height-to-crypt depth (V/C) ratio, which may enhance nutrient absorption capacity [96,97,98]. For instance, Hao Ding [99] reported increased ileal villus height and V/C ratio in weaned piglets, suggesting a potential expansion of the absorptive surface area.
In terms of mucus barrier enhancement, propionic and butyric acids appear to operate through synergistic mechanisms involving stimulation of goblet cell proliferation and activation of the MAPK pathway, leading to upregulation of MUC2 mucin secretion. Some studies report statistically significant thickening of the mucus layer (p < 0.05), which could help inhibit pathogen adhesion [100,101,102]. Additionally, Yue Yingxue [103] observed that butyrate from Clostridium butyricum increased occludin expression by 1.8-fold (p < 0.01), indicating a strengthening of barrier function.
Furthermore, SCFAs may also modulate intestinal motility via binding to G protein-coupled receptors (GPR41/43), with some swine studies indicating increased peristalsis frequency and reduced chyme transit time, thereby potentially limiting exposure to luminal pathogens [104,105,106,107]. It should be noted, however, that such effects may vary with metabolite concentration and host physiological state.

4.2. Immunomodulation and Antimicrobial Substance Production

Swine-derived probiotics and their metabolites exhibit bidirectional immunomodulatory properties. Some studies report elevated serum immunoglobulin (IgM, IgG) and interleukin-2 (IL-2) [108,109], along with increased secretory IgA in intestinal tissues, suggesting enhanced systemic and mucosal immunity [110,111]. For example, Wang [112] observed increased serum immunoglobulins in piglets supplemented with Clostridium butyricum and Enterococcus faecalis, accompanied by reduced diarrhea incidence. Mizumachi [113] reported that fermented feed from Lactobacillus plantarum LQ80 reduced tumor necrosis factor-alpha (TNF-α) levels and increased total serum IgM/IgG concentrations in weaned piglets compared to unfermented diets (Figure 3).
On the other hand, antimicrobial compounds such as bacteriocins EPS may inhibit pathogen adhesion and biofilm formation through membrane perforation (disrupting pathogen membrane integrity to cause ion imbalance and content leakage) and competitive exclusion (occupying intestinal binding sites) [114,115]. Ashlesha Kaushik [116] confirmed that nisin from Lactococcus lactis induces membrane perforation in methicillin-resistant Staphylococcus aureus, significantly impairing virulence factor expression. Bifidobacterial EPS specifically blocks surface adhesins (fimbriae, flagella) of pathogens, reducing adherence of enteropathogenic Escherichia coli to Caco-2 enterocytes by 37–52% [117,118,119]. Beyond organic acids and bacteriocins, hydrogen peroxide produced by lactobacilli and commensal microbiota constitutes a critical antimicrobial mechanism [120,121]. It diminishes pathogen virulence, compromises epithelial invasiveness, or penetrates cells to disrupt transcriptional regulation and signal transduction pathways, ultimately inducing pathogen death [122], though its practical significance in swine gut environments remains investigational.
Furthermore, metabolites from beneficial strains have been shown in some models to alleviate inflammatory signaling, such as the TLR4/NF-kappaB pathway, which is implicated in barrier dysfunction [123]. Yin [124] discovered that metabolites from porcine beneficial strains alleviate dextran sulfate sodium (DSS)-induced colitis and intestinal barrier damage in mice by suppressing TLR4/NF-kappaB inflammatory signaling. Zhang [125] further demonstrated that Lactobacillus plantarum ameliorates Clostridium perfringens-induced barrier dysfunction and inflammatory cascades through coordinated regulation of TLR4/NF-kappaB signaling and mitophagy.

4.3. Enhanced Nutrient Absorption Efficiency and Metabolic Pathway Optimization

Probiotic metabolites may improve nutrient digestion and absorption efficiency by enhancing digestive enzyme activity, optimizing the intestinal microenvironment, and regulating host metabolic gene expression [126]. Key enzymes such as amylases and proteases produced through their metabolism effectively degrade macromolecular nutrients (starch and proteins) in feed into absorbable micromolecules (glucose and amino acids), potentially increasing the digestibility of crude protein and other nutrients in nursery pigs [127]. Zhou Yuanli [128] demonstrated that feeding Lactobacillus plantarum-fermented feed significantly enhanced protease, lipase, and α/β-amylase activities (p < 0.05) in intestinal contents of nursery pigs, thereby improving nutrient absorption efficiency and increasing beneficial bacterial abundance (Figure 3).
SCFAs also contribute to metabolic regulation: acetate and propionate participate in lipogenesis and gluconeogenesis, while butyrate serves as an energy source for colonocytes and may activate PPAR-γ signaling, potentially improving metabolic efficiency [129,130]. Tang [131] observed improvements in backfat thickness and lean meat percentage in finishing pigs supplemented with Clostridium butyricum.
Furthermore, B vitamins synthesized by probiotics synergize with host metabolic enzymes: vitamin B12 mediates methylmalonyl-CoA mutase activity to enhance carbohydrate and lipid metabolism, resulting in a 40% increase (p < 0.01) in serum vitamin B12 levels in piglets and significant improvements in growth performance indicators such as average daily gain [132].

4.4. Modulation of Gastrointestinal Microbial Balance and Intestinal Health Maintenance

Swine-derived beneficial strains and their metabolites establish a stable microecological equilibrium system by targeted regulation of gut microbial community structure and function, suppressing pathogen proliferation while promoting colonization of beneficial bacteria [133,134]. This mechanism constitutes the core foundation for antibiotic-alternative additives to achieve “bacteriotherapy” functionality [135,136], primarily manifested through: SCFAs produced by beneficial bacterial metabolism significantly lowering intestinal pH to create an acidic environment inhibitory to pathogen growth. Such selective pressure directly suppresses replication of pathogenic bacteria including Escherichia coli and Salmonella spp., while simultaneously stimulating proliferation of beneficial bacteria such as Bifidobacterium and Lactobacillus, thereby optimizing microbial community structure [137,138].
Bai Peidian [139] demonstrated that feeding compound probiotic preparations significantly increased the relative abundance of Faecalibacterium in piglet cecum (+28.6%, p < 0.05) compared to controls, whereas abundances of Campylobacter and Lachnospira decreased by 19.3% and 14.7%, respectively. This microbial restructuring positively correlated with intestinal health status. Cui Wenzhi [140] observed that dietary supplementation with swine-derived probiotics significantly reduced diarrhea incidence by 36% (p < 0.01) in weaned piglets. Further investigations revealed that probiotic microorganisms enhance gut microbiota α-diversity (Shannon index increase: 1.8–2.3), elevate the relative abundance of Firmicutes (+15.4%), and suppress overproliferation of Bacteroidetes and Proteobacteria (decreases of 12.1% and 8.9%, respectively), collectively optimizing microbial architecture and maintaining microecological homeostasis (Figure 3). While these changes suggest improved ecological balance, it is important to note that microbial responses are context-dependent and may vary with diet, environment, and host genotype.

5. Application of Swine Beneficial Strains and Their Metabolites as Antibiotic Alternatives

In the field of antibiotic-alternative feed additives, swine-derived Lactobacillus demonstrates promising mechanisms, though single-strain probiotics are often limited by challenges in stability and storage viability. Consequently, composite probiotic preparations that leverage synergistic functional advantages have emerged as a more effective strategy [141,142]. Studies indicate that mixed fermentation using multiple swine-derived Lactobacillus strains can produce enhanced antimicrobial effects, achieving significantly greater inhibition against pathogens such as Escherichia coli and Salmonella spp. compared to fermentates from single strains, with some studies reporting increases in inhibition zone diameter of 35–48% (p < 0.01) under specific experimental conditions [143,144] (Figure 3). It should be noted that such pronounced effects may vary with experimental design.
From a veterinary standpoint, the strategic application of probiotics is most effective when aligned with the specific physiological challenges and health management goals of each stage of the swine life cycle. Probiotics have shown substantial benefits in both sow and piglet production [145]. Supplementation during gestation and lactation has been found to improve sow body condition, increase milk quality and yield, and enhance immunoglobulin content in colostrum, thereby promoting piglet growth and survival rates [146]. Composite fermented feeds may further enhance nutritional value through microbial metabolism, overcoming species barriers while simultaneously boosting antibacterial activity and growth performance [147].
The weaning period represents a high-stress transition associated with intestinal dysbiosis and increased susceptibility to diarrhea. Probiotic intervention at this stage plays a critical role in alleviating post-weaning stress, supporting the development of intestinal barrier function, and reducing the incidence of digestive disorders. Studies report improvements in the feed conversion ratio (FCR) by 12–15% and average daily gain (ADG) by 18–22% in weaned piglets receiving composites [148,149]. Furthermore, studies have shown that trials with specific composite probiotics have reported a 40–45% reduction in diarrhea incidence and significant improvements in growth metrics, likely mediated through enhanced intestinal development and antioxidant capacity [150].
During the growing and finishing phases, the primary goals shift toward optimizing growth performance and feed conversion. The use of composite probiotics and fermented feeds has been shown to enhance nutrient availability and improve growth metrics [151]. Li Genghui [152] confirmed that fermented feed increased finishing pig average daily gain by 43.5% (p < 0.01) and reduced feed: gain ratio by 28.8% compared to conventional feed.
Beyond swine-derived probiotics bacteria, other antibiotic alternatives like yeast-based supplements operate through distinct yet complementary mechanisms. Whereas swine-derived probiotics often directly modulate the gut microbiota and strengthen barrier function, yeast-based alternatives may exert benefits by altering the gut environment. For instance, Cao Di [153] reported that yeast-based antibiotic alternatives in late-gestation and lactating sow diets significantly increased total volatile fatty acids (VFA) in ileal chyme (+32.7%, p < 0.05), with butyrate proportion rising by 18.3 percentage points—thereby helping inhibit pathogenic microbial proliferation.
Despite these promising results, the practical application of probiotic additives faces several important challenges beyond technical efficacy. Economic feasibility must be considered, as production costs of composite probiotics—including fermentation, stabilization, and delivery technologies—may be higher than those of conventional additives, though potentially offset by improved performance and reduced veterinary costs. Regulatory aspects also play a critical role, as approval processes for probiotic feed additives vary across regions and may require substantial evidence of safety and efficacy. Additionally, consumer acceptance represents another key factor, with growing preference for natural and antibiotic-free animal products creating market opportunities for probiotic-based solutions [154].
Collectively, composite swine derived from porcine sources show substantial potential as antibiotic alternatives. They may enhance swine growth performance, reproductive efficiency, and health status by improving feed nutritional quality, precisely modulating gut microecology, and strengthening immune function (Table 5). However, their practical application faces several challenges, including production cost, standardization of probiotic consortia, stability during feed processing and storage, and inconsistent responses across different production systems. Future efforts should focus on large-scale validation and addressing these variabilities to fully realize their potential in supporting sustainable livestock production.

6. Conclusions

Current findings underscore that composite probiotic additives, by capitalizing on synergistic interactions among microbial strains, offer superior efficacy compared to single-strain formulations, marking a clear direction for future development in this field. Evidence indicates that incorporating swine-derived fermented feed into piglet diets contributes to notable health and productivity improvements, including reduced diarrhea incidence, enhanced growth performance and feed efficiency, as well as elevated serum immunoglobulin concentrations. However, several technical challenges remain, particularly in maintaining strain viability, standardizing fermentation protocols, and establishing consistent quality control systems for such fermented products.
Future research should efforts should focus on: (1) Elucidating the molecular mechanisms underlying bacterial metabolite interactions with host physiology; (2) Optimizing high-density fermentation parameters to enhance yield and functionality; (3) Developing scientific quality evaluation frameworks for standardized product assessment; and (4) integrating probiotics with precision nutrition approaches and other antibiotic alternatives, such as phytogenics and organic acids, for synergistic effects. Furthermore, long-term field studies are essential to comprehensively validate efficacy, safety, and economic benefits in diverse production systems. By addressing these challenges and research priorities, swine-derived probiotics and fermented feeds can advance toward sustainable, large-scale implementation in swine production, contributing to reduced antibiotic dependence and improved animal health management.

Author Contributions

Conceptualization, M.Z. and Z.L.; methodology, B.C.; software, S.P. and Z.L.; formal analysis, G.M. and M.L.; investigation, M.Z. and F.L.; data curation, M.Z. and T.S.; writing—original draft preparation, M.Z. and Z.L.; writing—review and editing, Z.L. and M.Z.; funding acquisition, Z.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Fujian Provincial Competitive Public Welfare Project (2023R1076).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Leng, S.M. Formation and control of antibiotic resistance under intensive breeding mode. Chin. J. Anim. Husb. Vet. Med. 2024, 12, 25–27. [Google Scholar]
  2. Checcucci, A.; Trevisi, P.; Luise, D.; Modesto, M.; Blasioli, S.; Braschi, I.; Mattarelli, P. Exploring the Animal Waste Resistome: The Spread of Antimicrobial Resistance Genes Through the Use of Livestock Manure. Front. Microbiol. 2020, 11, 1416. [Google Scholar] [CrossRef] [PubMed]
  3. Li, Y.X. The impact of antibiotic drug resistance on pig farming industry. China Anim. Health 2022, 24, 3–4. [Google Scholar]
  4. Li, C.Y. Five years of resistance: Challenges and innovations in China’s livestock industry. GDF 2024, 33, 15–17. [Google Scholar]
  5. Guo, H.W. Common feed replacement products: Effects and advantages and disadvantages. J. Anim. Sci. Vet. Sci. Inform. 2023, 3, 189–191. [Google Scholar]
  6. Zhang, J.S.; Zhu, Y.F.; Hai, L.; Nan, J.D.; Chen, G.W.; Hao, C.H.; Jia, B.; Zhang, B.; Wang, L.K.; Zhang, G.H.; et al. Advances in the Mechanism of Action and Application of Probiotics in Pig Production. J. Anim. Feed Sci. 2024, 45, 29–34. [Google Scholar]
  7. Kelsic, E.D.; Zhao, J.; Vetsigian, K.; Kishony, R. Counteraction of antibiotic production and degradation stabilizes microbial communities. Nature 2015, 521, 516–519. [Google Scholar] [CrossRef] [PubMed]
  8. Chen, X.S.; Yang, Y.Y.; Zhang, Y.L.; Liu, M.Y.; Hou, Y.T.; Han, D.D.; Meng, X.B. The Application of Microecological Preparations in Livestock Production. Shandong J. Anim. Sci. Vet. Med. 2025, 46, 36–40. [Google Scholar]
  9. Cai, Y.; Wu, G.; Yang, H.; Fang, Y.; Cheng, Z.; Zhan, P. The Mechanism and Application Status of Probiotics in Animal Health Management. China Anim. Husb. Vet. Med. 2025, 52, 2101–2114. [Google Scholar]
  10. Wang, Q.; Sun, Q.; Qi, R.; Wang, J.; Qiu, X.; Liu, Z.; Huang, J. Effects of Lactobacillus plantarum on the intestinal morphology, intestinal barrier function and microbiota composition of suckling piglets. J. Anim. Physiol. Anim. Nutr. 2019, 103, 1908–1918. [Google Scholar] [CrossRef]
  11. Gao, H.; Li, X.; Chen, X.; Hai, D.; Wei, C.; Zhang, L.; Li, P. The Functional Roles of Lactobacillus acidophilus in Different Physiological and Pathological Processes. J. Microbiol. Biotechnol. 2022, 32, 1226–1233. [Google Scholar] [CrossRef] [PubMed]
  12. Walsh, C.J.; Healy, S.; O’Toole, P.W.; Murphy, E.F.; Cotter, P.D. The probiotic L. casei LC-XCAL™ improves metabolic health in a diet-induced obesity mouse model without altering the microbiome. Gut Microbes. 2020, 12, 1704141. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, L.H.; Wang, M.F.; Chang, C.C.; Huang, S.Y.; Pan, C.H.; Yeh, Y.T.; Huang, C.H.; Chan, C.H.; Huang, H.Y. Lacticaseibacillus paracasei PS23 Effectively Modulates Gut Microbiota Composition and Improves Gastrointestinal Function in Aged SAMP8 Mice. Nutrients 2021, 13, 1116. [Google Scholar] [CrossRef]
  14. Tong, L.; Zhang, X.; Hao, H.; Liu, Q.; Zhou, Z.; Liang, X.; Liu, T.; Gong, P.; Zhang, L.; Zhai, Z.; et al. Lactobacillus rhamnosus GG Derived Extracellular Vesicles Modulate Gut Microbiota and Attenuate Inflammatory in DSS-Induced Colitis Mice. Nutrients 2021, 13, 3319. [Google Scholar] [CrossRef]
  15. Martoni, C.J.; Srivastava, S.; Damholt, A.; Leyer, G.J. Efficacy and dose response of Lactiplantibacillus plantarum in diarrhea-predominant irritable bowel syndrome. World J. Gastroenterol. 2023, 29, 4451–4465. [Google Scholar] [CrossRef]
  16. Cheng, J.; Laitila, A.; Ouwehand, A.C. Bifidobacterium animalis subsp. lactis HN019 Effects on Gut Health: A Review. Front. Nutr. 2021, 8, 790561. [Google Scholar]
  17. Moreno-Muñoz, J.A.; Ojeda, J.D.; López, J.J. A Probiotic Bacterium with Activity against the Most Frequent Bacteria and Viruses Causing Pediatric Diarrhea: Bifidobacterium longum subsp. infantis CECT 7210 (B. infantis IM1). Microorganisms 2024, 12, 1183. [Google Scholar]
  18. Li, Y.; Li, Q.; Yuan, R.; Wang, Y.; Guo, C.; Wang, L. Bifidobacterium breve-derived indole-3-lactic acid ameliorates colitis-associated tumorigenesis by directing the differentiation of immature colonic macrophages. Theranostics 2024, 14, 2719–2735. [Google Scholar] [CrossRef]
  19. Zhang, T.; Zhang, C.; Zhang, J.; Sun, F.; Duan, L. Efficacy of Probiotics for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Front. Cell. Infect. Microbiol. 2022, 12, 859967. [Google Scholar] [CrossRef]
  20. Stülke, J.; Grüppen, A.; Bramkamp, M.; Pelzer, S. Bacillus subtilis, a Swiss Army Knife in Science and Biotechnology. J. Bacteriol. 2023, 205, e0010223. [Google Scholar] [CrossRef]
  21. Acosta-Rodríguez-Bueno, C.P.; Abreu y Abreu, A.T.; Guarner, F.; Guno, M.J.V.; Pehlivanoğlu, E.; Perez, M., 3rd. Bacillus clausii for Gastrointestinal Disorders: A Narrative Literature Review. Adv. Ther. 2022, 39, 4854–4874. [Google Scholar]
  22. Zhou, X.; Willems, R.J.L.; Friedrich, A.W.; Rossen, J.W.A.; Bathoorn, E. Enterococcus faecium: From microbiological insights to practical recommendations for infection control and diagnostics. Antimicrob. Resist. Infect. Control 2020, 9, 130. [Google Scholar] [CrossRef]
  23. Pais, P.; Almeida, V.; Yılmaz, M.; Teixeira, M.C. Saccharomyces boulardii: What Makes It Tick as Successful Probiotic? J. Fungi 2020, 6, 78. [Google Scholar] [CrossRef]
  24. Karim, A.; Gerliani, N.; Aïder, M. Kluyveromyces marxianus: An emerging yeast cell factory for applications in food and biotechnology. Int. J. Food Microbiol. 2020, 333, 108818. [Google Scholar] [CrossRef]
  25. Pang, Y.; Xu, X.; Li, D.; Ge, Y.; Liang, J.; Xiong, X.; Wen, S.; Li, X.; Wang, Y.; Li, X.; et al. Impact of Clostridium butyricum HADIG-CB003 dietary supplementation on the gut microbiota of Kunming mice. Appl. Microbiol. Biotechnol. 2025, 109, 237. [Google Scholar] [CrossRef] [PubMed]
  26. Aw, W.; Fukuda, S. Protective effects of bifidobacteria against enteropathogens. Microb. Biotechnol. 2019, 12, 1097–1100. [Google Scholar] [CrossRef] [PubMed]
  27. Peterson, S.B.; Bertolli, S.K.; Mougous, J.D. The Central Role of Interbacterial Antagonism in Bacterial Life. Curr. Biol. 2020, 30, R1203–R1214. [Google Scholar] [CrossRef]
  28. Zhou, H.; Yu, B.; Sun, J.; Liu, Z.; Chen, H.; Ge, L.; Chen, D. Short-chain fatty acids can improve lipid and glucose metabolism independently of the pig gut microbiota. J. Anim. Sci. Biotechnol. 2021, 12, 61. [Google Scholar] [CrossRef]
  29. Bao, G.; Yu, X.; Wang, X.; Ye, P.; Yin, H.; Hu, T.; Cao, Z. Study on the Isolation, Identification and Probiotic Characteristics of Lactic Acid Bacteria from Free-Range Local Pigs in Yunnan. Anim. Nutr. 2023, 35, 5418–5429. [Google Scholar]
  30. Dowarah, R.; Verma, A.K.; Agarwal, N. The use of Lactobacillus as an alternative of antibiotic growth promoters in pigs: A review. Anim. Nutr. 2017, 3, 1–6. [Google Scholar] [CrossRef]
  31. Yang, L.; Zeng, X.; Qiao, S. Advances in research on solid-state fermented feed and its utilization: The pioneer of private customization for intestinal microorganisms. Anim. Nutr. 2021, 7, 905–916. [Google Scholar] [CrossRef] [PubMed]
  32. Wu, H.; Xu, C.; Wang, J.; Hu, C.; Ji, F.; Xie, J.; Yang, Y.; Yu, X.; Diao, X.; Lv, R. Effects of Dietary Probiotics and Acidifiers on the Production Performance, Colostrum Components, Serum Antioxidant Activity and Hormone Levels, and Gene Expression in Mammary Tissue of Lactating Sows. Animals 2023, 13, 1536. [Google Scholar] [CrossRef]
  33. Zhang, Y.; Dong, G. Screening of Porcine Probiotics and research on their probiotic characteristics. Gansu Anim. Vet. Sci. 2024, 54, 77–80. [Google Scholar]
  34. Marchwińska, K.; Gwiazdowska, D. Isolation and probiotic potential of lactic acid bacteria from swine feces for feed additive composition. Arch. Microbiol. 2021, 204, 61. [Google Scholar] [CrossRef] [PubMed]
  35. Kim, P.I.; Jung, M.Y.; Chang, Y.-H.; Kim, S.; Kim, S.-J.; Park, Y.-H. Probiotic properties of Lactobacillus and Bifidobacterium strains isolated from porcine gastrointestinal tract. Appl. Microbiol. Biotechnol. 2007, 74, 1103–1111. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, H.B.; Isaacson, R.E. The pig gut microbial diversity: Understanding the pig gut microbial ecology through the next generation high throughput sequencing. Vet. Microbiol. 2015, 177, 242–251. [Google Scholar] [CrossRef]
  37. Liu, W.J.; Zhang, H.P. The Application of Probiotic genomics in the Screening and Functional Evaluation of Lactic acid Bacteria. J. Chin. Inst. Food Sci. Technol. 2024, 24, 1. [Google Scholar]
  38. Ma, C.F.; Zhang, J.B.; Zhang, H.N.; Chen, H.Y.; Yang, Q.; Zhang, Q.E. Metabolomics technology and its application in animal production. Prog. Vet. Med. 2025, 46, 120–124. [Google Scholar]
  39. Zhang, W.; Gao, X.; Cui, Q.; Chen, C.; Liu, Y.; Peng, Y. Analyze the research progress of pig muscle growth and development based on omics technology. J. Chin. J. Anim. Sci. 2024, 60, 14–20. [Google Scholar]
  40. Holman, D.B.; Kommadath, A.; Tingley, J.P.; Abbott, D.W. Novel insights into the pig gut microbiome using metagenome-assembled genomes. Microbiol. Spectr. 2022, 10, e0238022. [Google Scholar] [CrossRef]
  41. Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut 1987, 28, 1221–1227. [Google Scholar] [CrossRef]
  42. Hays, K.E.; Pfaffinger, J.M.; Ryznar, R. The interplay between gut microbiota, short-chain fatty acids, and implications for host health and disease. Gut Microbes 2024, 16, 2393270. [Google Scholar] [CrossRef] [PubMed]
  43. Wong, J.M.W.; de Souza, R.; Kendall, C.W.C.; Emam, A.; Jenkins, D.J.A. Colonic Health: Fermentation and Short Chain Fatty Acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  44. Clarke, D.J.; Morris, J.G. Butyricin 7423: A Bacteriocin Produced by Clostridium butyricum NCIB7423. J. Gen. Microbiol. 1976, 95, 67–77. [Google Scholar] [CrossRef]
  45. Schilderink, R.; Verseijden, C.; Seppen, J.; Muncan, V.; van den Brink, G.R.; Lambers, T.T.; van Tol, E.A.; de Jonge, W.J. The SCFA butyrate stimulates the epithelial production of retinoic acid via inhibition of epithelial HDAC. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G1138–G1146. [Google Scholar] [CrossRef]
  46. Park, S.Y.; Kim, S.; Lim, S.D. The Inhibitory Effect of L. plantarum Q180 on Adipocyte Differentiation in 3T3-L1 and Reduction of Adipocyte Size in Mice Fed High-fat Diet. Korean J. Food Sci. Anim. Resour. 2018, 38, 99–109. [Google Scholar]
  47. Den Besten, G.; Bleeker, A.; Gerding, A.; Van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.-J.; et al. Short-Chain Fatty Acids Protect Against High-Fat Diet–Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef]
  48. Vinolo, M.A.R.; Rodrigues, H.G.; Nachbar, R.T.; Curi, R. Regulation of Inflammation by Short Chain Fatty Acids. Nutrients 2011, 3, 858–876. [Google Scholar] [CrossRef]
  49. Diao, H.; Jiao, A.R.; Yu, B.; Mao, X.B.; Chen, D.W. Gastric infusion of short-chain fatty acids can improve intestinal barrier function in weaned piglets. Genes Nutr. 2019, 14, 4. [Google Scholar] [CrossRef] [PubMed]
  50. Laszczyńska, M.; Sipak, O.; Walczakiewicz, K.; Mizerski, A.; Rył, A.; Ratajczak, W. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [Google Scholar]
  51. Nogal, A.; Valdes, A.M.; Menni, C. The role of short-chain fatty acids in the interplay between gut microbiota and diet in cardio-metabolic health. Gut Microbes 2021, 13, 1897212. [Google Scholar] [CrossRef]
  52. Gao, W.; Fang, Z.; Lei, L.; Ju, L.; Jin, B.; Loor, J.J.; Liang, Y.; Shi, Z.; Shen, T.; Yu, H.; et al. Propionate alleviates palmitic acid–induced endoplasmic reticulum stress by enhancing autophagy in calf hepatic cells. J. Dairy Sci. 2021, 104, 9316–9326. [Google Scholar] [CrossRef]
  53. Deleu, S.; Machiels, K.; Raes, J.; Verbeke, K.; Vermeire, S. short chain fatty acids and its producing organisms: An overlooked therapy for ibd? Ebiomedicine 2021, 66, 103293. [Google Scholar] [CrossRef] [PubMed]
  54. Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef] [PubMed]
  55. Niu, D.; Feng, N.; Xi, S.; Xu, J.; Su, Y. Genomics-based analysis of four swine-derived lactic acid bacteria strains and their evaluation as potential probiotics. Mol. Genet. Genom. 2024, 299, 24. [Google Scholar] [CrossRef]
  56. Gillor, O.; Etzion, A.; Riley, M.A. The dual role of bacteriocins as anti- and probiotics. Appl. Microbiol. Biotechnol. 2008, 81, 591–606. [Google Scholar] [CrossRef]
  57. Liang, J.F.; Ren, L.X.; Liu, P.; Tian, J.J.; Xiao, Y.B.; Dong, Z.L. The influence of porcine probiotics on the growth performance of fattening pigs. North. Anim. Husb. 2019, 10, 20. [Google Scholar]
  58. Dinata, R.; Baindara, P. Laterosporulin25: A probiotically produced, novel defensin-like bacteriocin and its immunogenic properties. Int. Immunopharmacol. 2023, 121, 110500. [Google Scholar] [CrossRef] [PubMed]
  59. Bosák, J.; Hrala, M.; Micenková, L.; Šmajs, D. Non-antibiotic antibacterial peptides and proteins of Escherichia coli: Efficacy and potency of bacteriocins. Expert. Rev. Anti. Infect. Ther. 2020, 19, 309–322. [Google Scholar] [CrossRef]
  60. Kumariya, R.; Garsa, A.K.; Rajput, Y.S.; Sood, S.K.; Akhtar, N.; Patel, S. Bacteriocins: Classification, synthesis, mechanism of action and resistance development in food spoilage causing bacteria. Microb. Pathog. 2019, 128, 171–177. [Google Scholar] [CrossRef]
  61. Sugrue, I.; Ross, R.P.; Hill, C. Bacteriocin diversity, function, discovery and application as antimicrobials. Nat. Rev. Microbiol. 2024, 22, 556–571. [Google Scholar] [CrossRef]
  62. Du, K. Research Progress on the Antibacterial Characteristics of Lactic Acid Bacteria Bacteriocins and Their Applications in Food. China Brew. 2022, 41, 16–20. [Google Scholar]
  63. Baindara, P.; Mandal, S.M. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics 2023, 12, 1732. [Google Scholar] [CrossRef] [PubMed]
  64. Ra, Y.E.; Bang, Y.-J. Balancing Act of the Intestinal Antimicrobial Proteins on Gut Microbiota and Health. J. Microbiol. 2024, 62, 167–179. [Google Scholar] [CrossRef]
  65. Grande Burgos, M.; Pulido, R.; Del Carmen López Aguayo, M.; Gálvez, A.; Lucas, R. The Cyclic Antibacterial Peptide Enterocin AS-48: Isolation, Mode of Action, and Possible Food Applications. Int. J. Mol. Sci. 2014, 5, 22706–22727. [Google Scholar] [CrossRef] [PubMed]
  66. Li, Y.-X.; Zhong, Z.; Zhang, W.-P.; Qian, P.-Y. Discovery of cationic nonribosomal peptides as Gram-negative antibiotics through global genome mining. Nat. Commun. 2018, 9, 3273. [Google Scholar] [CrossRef]
  67. Cardoso, M.H.; Meneguetti, B.T.; Oliveira-Júnior, N.G.; Macedo, M.L.R.; Franco, O.L. Antimicrobial peptide production in response to gut microbiota imbalance. Peptides 2022, 157, 170865. [Google Scholar] [CrossRef]
  68. Darbandi, A.; Asadi, A.; Mahdizade Ari, M.; Ohadi, E.; Talebi, M.; Halaj Zadeh, M.; Darb Emamie, A.; Ghanavati, R.; Kakanj, M. Bacteriocins: Properties and potential use as antimicrobials. J. Clin. Lab. Anal. 2022, 36, e24093. [Google Scholar] [CrossRef] [PubMed]
  69. Madhu, M.; Kumar, D.; Sirohi, R.; Tarafdar, A.; Dhewa, T.; Aluko, R.E.; Badgujar, P.C.; Awasthi, M.K. Bioactive peptides from meat: Current status on production, biological activity, safety, and regulatory framework. Chemosphere 2022, 307 Pt 1, 135650. [Google Scholar] [CrossRef]
  70. Hynönen, U.; Palva, A. Lactobacillus surface layer proteins: Structure, function and applications. Appl. Microbiol. Biotechnol. 2013, 97, 5225–5243. [Google Scholar] [CrossRef]
  71. Salazar, N.; Gueimonde, M.; de los Reyes-Gavilán, C.G.; Ruas-Madiedo, P. Exopolysaccharides Produced by Lactic Acid Bacteria and Bifidobacteria as Fermentable Substrates by the Intestinal Microbiota. Crit. Rev. Food Sci. Nutr. 2015, 56, 1440–1453. [Google Scholar] [CrossRef] [PubMed]
  72. Kaur, N.; Dey, P. Bacterial exopolysaccharides as emerging bioactive macromolecules: From fundamentals to applications. Res. Microbiol. 2023, 174, 104024. [Google Scholar] [CrossRef] [PubMed]
  73. Zhou, Y.; Cui, Y.; Qu, X. Exopolysaccharides of lactic acid bacteria: Structure, bioactivity and associations: A review. Carbohydr. Polym. 2019, 207, 317–332. [Google Scholar] [CrossRef] [PubMed]
  74. Trabelsi, I.; Ktari, N.; Ben Slima, S.; Triki, M.; Bardaa, S.; Mnif, H.; Ben Salah, R. Evaluation of dermal wound healing activity and in vitro antibacterial and antioxidant activities of a new exopolysaccharide produced by Lactobacillus sp. Ca 6. Int. J. Biol. Macromol. 2017, 103, 194–201. [Google Scholar] [CrossRef]
  75. Bhagat, N.; Raghav, M.; Dubey, S.; Bedi, N. Bacterial Exopolysaccharides: Insight into Their Role in Plant Abiotic Stress Tolerance. J. Microbiol. Biotechnol. 2021, 31, 1045–1059. [Google Scholar] [CrossRef]
  76. Yang, Y.; Jiang, G.; Tian, Y. Biological activities and applications of exopolysaccharides produced by lactic acid bacteria: A mini-review. World J. Microbiol. Biotechnol. 2023, 39, 155. [Google Scholar] [CrossRef]
  77. Rahbar Saadat, Y.; Yari Khosroushahi, A.; Pourghassem Gargari, B. A comprehensive review of anticancer, immunomodulatory and health beneficial effects of the lactic acid bacteria exopolysaccharides. Carbohydr. Polym. 2019, 217, 79–89. [Google Scholar] [CrossRef]
  78. Angelin, J.; Kavitha, M. Exopolysaccharides from probiotic bacteria and their health potential. Int. J. Biol. Macromol. 2020, 162, 853–865. [Google Scholar] [CrossRef]
  79. Laiño, J.; Villena, J.; Kanmani, P.; Kitazawa, H. Immunoregulatory Effects Triggered by Lactic Acid Bacteria Exopolysaccharides: New Insights into Molecular Interactions with Host Cells. Microorganisms 2016, 4, 27. [Google Scholar] [CrossRef]
  80. Jurášková, D.; Ribeiro, S.C.; Silva, C.C.G. Exopolysaccharides Produced by Lactic Acid Bacteria: From Biosynthesis to Health-Promoting Properties. Foods 2022, 11, 156. [Google Scholar] [CrossRef]
  81. Caggianiello, G.; Kleerebezem, M.; Spano, G. Exopolysaccharides produced by lactic acid bacteria: From health-promoting benefits to stress tolerance mechanisms. Appl. Microbiol. Biotechnol. 2016, 100, 3877–3886. [Google Scholar] [CrossRef]
  82. Liang, S.; Wang, X.; Li, C.; Liu, L. Biological Activity of Lactic Acid Bacteria Exopolysaccharides and Their Applications in the Food and Pharmaceutical Industries. Foods 2024, 13, 1621. [Google Scholar] [CrossRef]
  83. Patten, D.A.; Laws, A.P. Lactobacillus-produced exopolysaccharides and their potential health benefits: A review. Benef. Microbes 2015, 6, 457–471. [Google Scholar] [CrossRef]
  84. Simpson, A.; Martinez, F.D. The role of lipopolysaccharide in the development of atopy in humans. Clin. Exp. Allergy 2010, 40, 209–223. [Google Scholar] [CrossRef]
  85. Eribo, O.A.; du Plessis, N.; Chegou, N.N. The Intestinal Commensal, Bacteroides fragilis, Modulates Host Responses to Viral Infection and Therapy: Lessons for Exploration during Mycobacterium tuberculosis Infection. Infect. Immun. 2022, 90, e0032121. [Google Scholar] [CrossRef] [PubMed]
  86. Castro-Bravo, N.; Wells, J.M.; Margolles, A.; Ruas-Madiedo, P. Interactions of Surface Exopolysaccharides From Bifidobacterium and Lactobacillus Within the Intestinal Environment. Front. Microbiol. 2018, 9, 2426. [Google Scholar] [CrossRef] [PubMed]
  87. LeBlanc, J.G.; Chain, F.; Martín, R.; Bermúdez-Humarán, L.G.; Courau, S.; Langella, P. Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria. Microb. Cell Fact. 2017, 16, 79. [Google Scholar] [CrossRef]
  88. Peterson, C.T.; Rodionov, D.A.; Osterman, A.L.; Peterson, S.N. B Vitamins and Their Role in Immune Regulation and Cancer. Nutrients 2020, 12, 3380. [Google Scholar] [CrossRef] [PubMed]
  89. Olesen, S.V.; Rajabi, N.; Svensson, B.; Olsen, C.A.; Madsen, A.S. An NAD+-Dependent Sirtuin Depropionylase and Deacetylase (Sir2La) from the Probiotic Bacterium Lactobacillus acidophilus NCFM. Biochemistry 2018, 57, 3903–3915. [Google Scholar] [CrossRef]
  90. Ashoori, M.; Saedisomeolia, A. Riboflavin (vitamin B2) and oxidative stress: A review. Br. J. Nutr. 2014, 111, 1985–1991. [Google Scholar] [CrossRef]
  91. Powers, H.J.; Hill, M.H.; Mushtaq, S.; Dainty, J.R.; Majsak-Newman, G.; Williams, E.A. Correcting a marginal riboflavin deficiency improves hematologic status in young women in the United Kingdom (RIBOFEM). Am. J. Clin. Nutr. 2011, 93, 1274–1284. [Google Scholar] [CrossRef]
  92. Kennedy, D. B Vitamins and the Brain: Mechanisms, Dose and Efficacy—A Review. Nutrients 2016, 8, 68. [Google Scholar] [CrossRef]
  93. Fusaro, M.; Cosmai, L.; Evenepoel, P.; Nickolas, T.L.; Cheung, A.M.; Aghi, A.; Tripepi, G.; Plebani, M.; Iervasi, G.; Vettor, R.; et al. Vitamin K and Kidney Transplantation. Nutrients 2020, 12, 2717. [Google Scholar] [CrossRef] [PubMed]
  94. Alonso, N.; Meinitzer, A.; Fritz-Petrin, E.; Enko, D.; Herrmann, M. Role of Vitamin K in Bone and Muscle Metabolism. Calcif. Tissue Int. 2022, 112, 178–196. [Google Scholar] [CrossRef] [PubMed]
  95. Kirschning, A. On the evolution of coenzyme biosynthesis. Nat. Prod. Rep. 2022, 39, 2175–2199. [Google Scholar] [CrossRef] [PubMed]
  96. Pereira, W.A.; Franco, S.M.; Reis, I.L.; Mendonça, C.M.N.; Piazentin, A.C.; Azevedo, P.O.S.; Tse, M.L.P.; De Martinis, E.C.P.; Gierus, M.; Oliveira, R.P.S. Beneficial effects of probiotics on the pig production cycle: An overview of clinical impacts and performance. Vet. Microbiol. 2022, 269, 109431. [Google Scholar] [CrossRef]
  97. Liu, G.; Gu, K.; Liu, X.; Jia, G.; Zhao, H.; Chen, X.; Wang, J. Dietary glutamate enhances intestinal immunity by modulating microbiota and Th17/Treg balance-related immune signaling in piglets after lipopolysaccharide challenge. Food Res. Int. 2023, 166, 112597. [Google Scholar] [CrossRef]
  98. Wang, W.; Dang, G.; Hao, W.; Li, A.; Zhang, H.; Guan, S.; Ma, T. Dietary Supplementation of Compound Probiotics Improves Intestinal Health by Modulated Microbiota and Its SCFA Products as Alternatives to In-Feed Antibiotics. Probiotics Antimicrob. Proteins 2024, 17, 1969–1984. [Google Scholar] [CrossRef]
  99. Ding, H.; Zhao, X.; Azad, M.A.K.; Ma, C.; Gao, Q.; He, J.; Kong, X. Dietary supplementation with Bacillus subtilis and xylo-oligosaccharides improves growth performance and intestinal morphology and alters intestinal microbiota and metabolites in weaned piglets. Food Funct. 2021, 12, 5837–5849. [Google Scholar] [CrossRef]
  100. Ma, S.; Yeom, J.; Lim, Y.-H. Specific activation of hypoxia-inducible factor-2α by propionate metabolism via a β-oxidation-like pathway stimulates MUC2 production in intestinal goblet cells. Biomed. Pharmacother. 2022, 155, 113672. [Google Scholar] [CrossRef]
  101. Burger-van Paassen, N.; Vincent, A.; Puiman, P.J.; van der Sluis, M.; Bouma, J.; Boehm, G.; van Goudoever, J.B.; van Seuningen, I.; Renes, I.B. The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: Implications for epithelial protection. Biochem. J. 2009, 420, 211–219. [Google Scholar] [CrossRef]
  102. Chakravortty, D.; Mao, X.; Gu, C.; Hu, H.; Tang, J.; Chen, D.; Yu, B.; He, J.; Yu, J.; Luo, J.; et al. Dietary Lactobacillus rhamnosus GG Supplementation Improves the Mucosal Barrier Function in the Intestine of Weaned Piglets Challenged by Porcine Rotavirus. PLoS ONE 2016, 11, e0146312. [Google Scholar]
  103. Yue, Y.X.; Wang, Y.Q.; Yan, F.F.; Li, N.; Li, B.L.; Huo, J.C. Research progress on the Production Methods of Butyric acid and its physiological functions in the Intestinal Tract. Sci. Technol. Food Ind. 2019, 40, 339–344. [Google Scholar]
  104. Kimura, I.; Inoue, D.; Maeda, T.; Hara, T.; Ichimura, A.; Miyauchi, S.; Kobayashi, M.; Hirasawa, A.; Tsujimoto, G. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41). Proc. Natl. Acad. Sci. USA 2011, 108, 8030–8035. [Google Scholar] [CrossRef]
  105. Kobayashi, M.; Mikami, D.; Kimura, H.; Kamiyama, K.; Morikawa, Y.; Yokoi, S.; Kasuno, K.; Takahashi, N.; Taniguchi, T.; Iwano, M. Short-chain fatty acids, GPR41 and GPR43 ligands, inhibit TNF-α-induced MCP-1 expression by modulating p38 and JNK signaling pathways in human renal cortical epithelial cells. Biochem. Biophys. Res. Commun. 2017, 486, 499–505. [Google Scholar] [CrossRef]
  106. Wu, Z.; Zhang, L.; Li, H.; Li, J.; Zhang, Z.; Tan, B.; Wang, J. Ningxiang Pig-Derived Parabacteroides distasonis HNAU0205 Alleviates ETEC-Induced Intestinal Apoptosis, Oxidative Damage, and Inflammation in Piglets. Animals 2024, 14, 2156. [Google Scholar] [CrossRef]
  107. Luu, M.; Visekruna, A. Short-chain fatty acids: Bacterial messengers modulating the immunometabolism of T cells. Eur. J. Immunol. 2019, 49, 842–848. [Google Scholar] [CrossRef] [PubMed]
  108. Fernández, A.; Rodríguez, J.M.; Bongaerts, R.J.; Gasson, M.J.; Horn, N. Nisin-Controlled Extracellular Production of Interleukin-2 inLactococcus lactisStrains, without the Requirement for a Signal Peptide Sequence. Appl. Environ. Microbiol. 2007, 73, 7781–7784. [Google Scholar] [CrossRef] [PubMed]
  109. Gresse, R.; Chaucheyras-Durand, F.; Fleury, M.A.; Van de Wiele, T.; Forano, E.; Blanquet-Diot, S. Gut Microbiota Dysbiosis in Postweaning Piglets: Understanding the Keys to Health. Trends Microbiol. 2017, 25, 851–873. [Google Scholar] [CrossRef] [PubMed]
  110. Bu, Y.; Liu, Y.; Zhang, T.; Liu, Y.; Zhang, Z.; Yi, H. Bacteriocin-Producing Lactiplantibacillus plantarum YRL45 Enhances Intestinal Immunity and Regulates Gut Microbiota in Mice. Nutrients 2023, 15, 3437. [Google Scholar] [CrossRef]
  111. Wang, K.; Cao, G.; Zhang, H.; Li, Q.; Yang, C. Effects of Clostridium butyricum and Enterococcus faecalison growth performance, immune function, intestinal morphology, volatile fatty acids, and intestinal flora in a piglet model. Food Funct. 2019, 10, 7844–7854. [Google Scholar] [CrossRef]
  112. Mizumachi, K.; Aoki, R.; Ohmori, H.; Saeki, M.; Kawashima, T. Effect of fermented liquid diet prepared with Lactobacillus plantarum LQ80 on the immune response in weaning pigs. Animal 2009, 3, 670–676. [Google Scholar] [CrossRef] [PubMed]
  113. Xia, W.; Han, J.; Zhu, S.; Wang, Y.; Zhang, W.; Wu, Z. Structural elucidation of the exopolysaccharide from Streptococcus thermophilus XJ53 and the effect of its molecular weight on immune activity. Int. J. Biol. Macromol. 2023, 230, 123177. [Google Scholar] [CrossRef]
  114. Kavitake, D.; Devi, P.B.; Delattre, C.; Reddy, G.B.; Shetty, P.H. Exopolysaccharides produced by Enterococcus genus—An overview. Int. J. Biol. Macromol. 2023, 226, 111–120. [Google Scholar] [CrossRef]
  115. Kaushik, A.; Kest, H.; Sood, M.; Steussy, B.; Thieman, C.; Gupta, S. Biofilm Producing Methicillin-Resistant Staphylococcus aureus (MRSA) Infections in Humans: Clinical Implications and Management. Pathogens 2024, 13, 76. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, X.Y.; Woodward, A.; Zijlstra, R.T.; Gänzle, M.G.; Griffiths, M.W. Exopolysaccharides Synthesized by Lactobacillus reuteri Protect against Enterotoxigenic Escherichia coli in Piglets. Appl. Environ. Microbiol. 2014, 80, 5752–5760. [Google Scholar] [CrossRef] [PubMed]
  117. Maalej, H.; Boisset, C.; Hmidet, N.; Colin-Morel, P.; Buon, L.; Nasri, M. Depolymerization of Pseudomonas stutzeri exopolysaccharide upon fermentation as a promising production process of antibacterial compounds. Food Chem. 2017, 227, 22–32. [Google Scholar] [CrossRef] [PubMed]
  118. Li, L.; Jiang, Y.-J.; Yang, X.-Y.; Liu, Y.; Wang, J.-Y.; Man, C.-X. Immunoregulatory effects on Caco-2 cells and mice of exopolysaccharides isolated from Lactobacillus acidophilus NCFM. Food Funct. 2014, 5, 3261–3268. [Google Scholar] [CrossRef]
  119. Engevik, M.A.; Versalovic, J.; Britton, R.A.; Cani, P.D. Biochemical Features of Beneficial Microbes: Foundations for Therapeutic Microbiology. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef]
  120. Järbrink-Sehgal, E.; Andreasson, A. The gut microbiota and mental health in adults. Curr. Opin. Neurobiol. 2020, 62, 102–114. [Google Scholar] [CrossRef]
  121. Knaus, U.G.; Hertzberger, R.; Pircalabioru, G.G.; Yousefi, S.P.M.; Branco dos Santos, F. Pathogen control at the intestinal mucosa–H2O2to the rescue. Gut microbes 2017, 8, 67–74. [Google Scholar] [CrossRef]
  122. Xi, M.; Zhao, P.; Li, F.; Bao, H.; Ding, S.; Ji, L.; Yan, J. MicroRNA-16 inhibits the TLR4/NF-kappaB pathway and maintains tight junction integrity in irritable bowel syndrome with diarrhea. J. Biol. Chem. 2022, 298, 102461. [Google Scholar] [CrossRef]
  123. Yin, H.; Wang, C.; Shuai, Y.; Xie, Z.; Liu, J. Pig-Derived Probiotic Bacillus tequilensis YB-2 Alleviates Intestinal Inflammation and Intestinal Barrier Damage in Colitis Mice by Suppressing the TLR4/NF-kappaB Signaling Pathway. Animals 2024, 14, 1989. [Google Scholar] [CrossRef]
  124. Zhang, R.; Qin, S.; Yang, C.; Niu, Y.; Feng, J. The protective effects of Bacillus licheniformis against inflammatory responses and intestinal barrier damage in broilers with necrotic enteritis induced by Clostridium perfringens. J. Sci. Food Agric. 2023, 103, 6958–6965. [Google Scholar] [CrossRef] [PubMed]
  125. Liao, S.F.; Nyachoti, M. Using probiotics to improve swine gut health and nutrient utilization. Anim. Nutr. 2017, 3, 331–343. [Google Scholar] [CrossRef] [PubMed]
  126. Yu, F. The functions of probiotics and their application in the pig breeding industry. China Anim. Health 2025, 27, 127–128. [Google Scholar]
  127. Zhou, Y.L.; Yao, M.X.; Yang, J.J.; Yin, X.H.; Zhang, Y.J.; Zhai, S.Q.; Wang, J. The effects of dietary supplementation with different levels of porcine Lactobacillus plantarum on the digestive function of weaned piglets. Feed Ind. 2025, 46, 51–57. [Google Scholar]
  128. Chen, W.J.; Luo, W.T.; Huang, J.M.; Chen, C.; Jiang, Y.J.; Wang, Z.D.; Zhao, L.M.; Zhang, H.T. The application of Clostridium butyricum in livestock, poultry and aquaculture. Guangdong J. Anim. Vet. Sci. 2025, 3, 82–89. [Google Scholar]
  129. Merlo, G.; Bachtel, G.; Sugden, S.G. Gut microbiota, nutrition, and mental health. Front. Nutr. 2024, 11, 1337889. [Google Scholar] [CrossRef] [PubMed]
  130. Tang, X. Probiotic Roles of Clostridium butyricum in Piglets: Considering Aspects of Intestinal Barrier Function. Animals 2024, 14, 1069. [Google Scholar] [CrossRef]
  131. Hou, C.; Zeng, X.; Yang, F.; Liu, H.; Qiao, S. Study and use of the probiotic Lactobacillus reuteri in pigs: A review. JASB 2015, 6, 14. [Google Scholar] [CrossRef]
  132. Hu, S.; Wang, L.; Jiang, Z. Dietary Additive Probiotics Modulation of the Intestinal Microbiota. Protein Pept. Lett. 2017, 24, 382–387. [Google Scholar] [CrossRef]
  133. Sen, U.; Lützhøft, D.O.; Bækgård, C.; Wimborne, E.; Straarup, E.M.; Pedersen, K.-M.; Swann, J.R.; Pedersen, H.D.; Kristensen, K.; Morgills, L.; et al. High fat diet is associated with gut microbiota dysbiosis and decreased gut microbial derived metabolites related to metabolic health in young Göttingen Minipigs. PLoS ONE 2024, 19, e0298602. [Google Scholar]
  134. Yun, Y.; Ji, S.; Yu, G.; Jia, P.; Niu, Y.; Zhang, H.; Zhang, X.; Wang, T.; Zhang, L. Effects of Bacillus subtilis on jejunal integrity, redox status, and microbial composition of intrauterine growth restriction suckling piglets. J. Anim. Sci. 2021, 99, skab255. [Google Scholar] [CrossRef] [PubMed]
  135. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2016, 61, 1600240. [Google Scholar] [CrossRef] [PubMed]
  136. Cai, L.; Zhao, Y.; Chen, W.; Li, Y.; Han, Y.; Zhang, B.; Pineda, L.; Li, X.; Jiang, X. Effect of an organic acid blend as an antibiotic alternative on growth performance, antioxidant capacity, intestinal barrier function, and fecal microbiota in weaned piglets. J. Anim. Sci. 2024, 102, skae149. [Google Scholar] [CrossRef]
  137. Wang, X.; Tian, Z.; Azad, M.A.K.; Zhang, W.; Blachier, F.; Wang, Z.; Kong, X. Dietary supplementation with Bacillus mixture modifies the intestinal ecosystem of weaned piglets in an overall beneficial way. J. Appl. Microbiol. 2020, 130, 233–246. [Google Scholar] [CrossRef]
  138. Bai, P.T.; Kong, J.M.; Pei, T.; Cheng, Z.X.; Ren, Y.H. The effects of compound microecological preparations on the growth performance, immune function and cecal flora structure of weaned piglets. China Anim. Husb. Vet. Med. 2022, 49, 942–952. [Google Scholar]
  139. Gao, J.; Li, Y.; Wan, Y.; Hu, T.; Liu, L.; Yang, S.; Gong, Z.; Zeng, Q.; Wei, Y.; Yang, W.; et al. A Novel Postbiotic From Lactobacillus rhamnosus GG With a Beneficial Effect on Intestinal Barrier Function. Front. Microbiol. 2019, 10, 477. [Google Scholar] [CrossRef]
  140. Liu, J.Y.; Wu, H.; Wang, W.S.; Chen, Z.X.; Xu, E.R.; Chen, L.; Zheng, R.T. The effects of compound microecological preparations on the production performance and nutrient digestion and metabolism of ruminants. Feed Ind. 2025, 46, 37. [Google Scholar]
  141. Zhao, C.; Li, Y.; Wang, H.; Solomon, A.I.; Wang, S.; Dong, X.; Song, B.; Ren, Z. Dietary supplementation with compound microecological preparations: Effects on the production performance and gut microbiota of lactating female rabbits and their litters. Microbiol. Spectr. 2025, 13, e0006724. [Google Scholar] [CrossRef]
  142. Tang, J.; Li, W.; Zhou, Q.; Fang, Z.; Lin, Y.; Xu, S.; Feng, B.; Zhuo, Y.; Jiang, X.; Zhao, H.; et al. Effect of heating, microbial fermentation, and enzymatic hydrolysis of soybean meal on growth performance, nutrient digestibility, and intestinal microbiota of weaned piglets. J. Anim. Sci. 2023, 101, skad384. [Google Scholar] [CrossRef]
  143. Xu, X.; Duarte, M.E.; Kim, S.W. Postbiotic effects of Lactobacillus fermentate on intestinal health, mucosa-associated microbiota, and growth efficiency of nursery pigs challenged with F18+Escherichia coli. J. Anim. Sci. 2022, 100, skac210. [Google Scholar] [CrossRef]
  144. Yu, J.; Zuo, B.; Li, Q.; Zhao, F.; Wang, J.; Huang, W.; Sun, Z.; Chen, Y. Dietary supplementation with Lactiplantibacillus plantarum P-8 improves the growth performance and gut microbiota of weaned piglets. Microbiol. Spectr. 2024, 12, e0234522. [Google Scholar] [CrossRef]
  145. Kiernan, D.P.; O’Doherty, J.V.; Sweeney, T. The effect of maternal probiotic or synbiotic supplementation on sow and offspring gastrointestinal microbiota, health, and performance. Animals 2023, 13, 2996. [Google Scholar] [CrossRef]
  146. Wang, T.; Zhong, Y.Q. Research Progress on the Application of Microbial Fermented Feed in Pig Breeding. China Anim. Ind. 2024, 12, 50–51. [Google Scholar]
  147. Ma, N.N.; Zhang, X.Q.; Yang, C.L.; Yang, D.D.; Li, J.L.; Wang, X.Z.; Cao, Q.J. Optimization and application evaluation of mixed bacteria compounding in Fermented Feed. Shandong Agric. Sci. 2020, 52, 112–117+125. [Google Scholar]
  148. Zhang, G.F.; Zeng, W.L.; Zheng, M.L.; Chen, Q.H. Research Progress on the Characteristics of Microbial Fermented Feed and Its Application in Animal Production. Feed Ind. 2023, 44, 79–85. [Google Scholar]
  149. Kenny, M.; Smidt, H.; Mengheri, E.; Miller, B. Probiotics–do they have a role in the pig industry? Animal 2011, 5, 462–470. [Google Scholar] [CrossRef] [PubMed]
  150. Liu, Y.; Jia, X.; Chang, J.; Jiang, X.; Che, L.; Lin, Y.; Zhuo, Y.; Feng, B.; Fang, Z.; Li, J.; et al. Effect of yeast culture supplementation in sows during late gestation and lactation on growth performance, antioxidant properties, and intestinal microorganisms of offspring weaned piglets. Front. Microbiol. 2023, 13, 1105888. [Google Scholar] [CrossRef]
  151. Vasquez, R.; Oh, J.K.; Song, J.H.; Kang, D.-K. Gut microbiome-produced metabolites in pigs: A review on their biological functions and the influence of probiotics. J. Anim. Sci. Technol. 2022, 64, 671–695. [Google Scholar] [CrossRef] [PubMed]
  152. Li, G.H.; Wang, Y.J.; Zheng, T.L.; Wang, S.B.; Wang, L.N.; Zheng, C.J.; Wu, R.F.; Jiang, Q.Y.; Shu, G. Screening and identification of endogenous Lactobacillus in porcine intestines and its application in liquid fermented feed. Acta Agric. Univ. Jiangxiensis 2025, 47, 152–166. [Google Scholar]
  153. Liu, M.; Luan, H.; Qiu, W.; Zhang, Y.; Feng, W.; Xu, W.; Wang, F.; Xuan, H.; Song, P. Antibiotic alternatives in livestock feeding. Sci. Total Environ. 2025, 989, 179867. [Google Scholar] [CrossRef] [PubMed]
  154. da Silva, T.F.; de Glória, R.A.; Americo, M.F.; Freitas, A.D.S.; de Jesus, L.C.L.; Barroso, F.A.L.; Laguna, J.G.; Coelho-Rocha, N.D.; Tavares, L.M.; le Loir, Y. Unlocking the potential of probiotics: A comprehensive review on research, production, and regulation of probiotics. Probiotics Antimicrob. Prot. 2024, 16, 1687–1723. [Google Scholar] [CrossRef]
Figure 1. Probiotics and their beneficial effects through multiple mechanisms.
Figure 1. Probiotics and their beneficial effects through multiple mechanisms.
Vetsci 12 01100 g001
Figure 2. Isolation and Identification of swine-derived Beneficial Strains. Note: The red arrow indicates an increase.
Figure 2. Isolation and Identification of swine-derived Beneficial Strains. Note: The red arrow indicates an increase.
Vetsci 12 01100 g002
Figure 3. Bioactive Components and Functions of swine-derived Beneficial Strain Metabolites. Note: The red arrow indicates an increase. The blue arrow represents a decrease.
Figure 3. Bioactive Components and Functions of swine-derived Beneficial Strain Metabolites. Note: The red arrow indicates an increase. The blue arrow represents a decrease.
Vetsci 12 01100 g003
Table 1. Bacteria that have demonstrated probiotic effects in the veterinary field.
Table 1. Bacteria that have demonstrated probiotic effects in the veterinary field.
GenusSpeciesEffect on Animal Health
BacteriaLactobacillusL. acidophilusA core probiotic species primarily residing in the small intestine; supports digestive health [11].
L. caseiWidely used in fermented foods; supports gut microbiota balance [12].
L. paracaseiCommonly researched for immune support [13].
L. rhamnosusOne of the most clinically documented strains; effective against antibiotic-associated diarrhea [14].
L. plantarumHighly adaptable; often studied for managing IBS symptoms [15].
BifidobacteriumB. animalis subsp. lactisExtremely common in dairy products; supports immune and digestive health [16].
B. longum subsp. longumA dominant inhabitant of the adult gut; researched for IBS and anti-inflammatory effects [17].
B. brevePrevalent in infants; studied for the prevention of necrotizing enterocolitis (NEC) [18].
BacillusB. coagulansSpore-forming bacterium; highly stable and resistant to heat and acid [19].
B. subtilisSpore-forming; used for its enzymatic activity and support for protein digestion and gut health [20].
B. clausiiSpore-forming; widely used, especially in Europe, for preventing antibiotic-associated diarrhea [21].
EnterococcusE. faeciumUsed for management of diarrhea. Note: Some strains are vancomycin-resistant (VRE), so careful selection is critical [22].
fungusSaccharomycesS. boulardiiThe most well-known probiotic yeast; effective against antibiotics [23].
KluyveromycesK. marxianusUsed in dairy fermentations; also researched for its potential probiotic properties in gut health [24].
Table 5. A comparison of the therapeutic effects of probiotics and antibiotics.
Table 5. A comparison of the therapeutic effects of probiotics and antibiotics.
Disease/DisorderTarget Population/SpeciesIntervention (Dosage/Duration)Key Efficacy Outcomes & Mechanisms
Necrotic EnteritisLivestock: Broiler chickens (Poultry).Antibiotic: Ionophores (Salinomycin) or therapeutic antibiotics (Bacitracin).
Probiotic: Bacillus subtilis or Clostridium butyricum spores. ~1–5 × 105 CFU/g of feed. Duration: Throughout the grow-out period.
Antibiotic: Effective for prevention and treatment but faces resistance and withdrawal issues.
Probiotic: Reduces mortality and intestinal lesions caused by Clostridium perfringens. Mechanism: Competitive exclusion, production of antimicrobial compounds (bacteriocins), stimulation of immune responses [137].
Acute Infectious DiarrheaHumans: Infants & Children.
Livestock: Neonatal piglets.
Antibiotic: Only for bacterial causes (e.g., E. coli scours).
Probiotic: L. rhamnosus GG or Bacillus clausii. ~1–10 Billion CFU/day for 5–7 days.
Antibiotic: Targets specific pathogens but may disrupt microbiota.
Probiotic: Shortens duration of diarrhea. Mechanism: Inhibition of pathogen adhesion, secretion of antibacterial substances, support of mucosal immunity [59].
Post-Weaning DiarrheaLivestock: Weaned piglets.Antibiotic: ZnO (pharmacological dose) is widely used, but being phased out in the EU. Therapeutic antibiotics (Colistin).
Probiotic: Enterococcus faecium or Bacillus licheniformis. ~1–5 × 109 CFU/kg feed. Duration: 2–4 weeks post-weaning.
Antibiotic: Effective but contributes to antimicrobial resistance (AMR).
Probiotic: Improves growth performance and reduces diarrhea incidence. Mechanism: Stabilizes gut microbiota during stress, enhances digestive enzyme activity, improves gut barrier integrity [101].
Helicobacter pylori InfectionHumans: Infected adults.Antibiotic: Standard triple/quadruple therapy (PPI + Amoxicillin + Clarithromycin).
Probiotic: As adjunct: Specific strains of Lactobacillus and Bifidobacterium. ~10–20 Billion CFU/day.
Antibiotic: Eradicates pathogen but has high side effect rate.
Probiotic: Increases eradication rates and reduces antibiotic side effects (especially diarrhea). Mechanism: May inhibit Hp adhesion, modulate local immune response [138].
MastitisLivestock: Dairy cows.Antibiotic: Intramammary infusions (Cephapirin) during the dry period or for treatment.
Probiotic: Intramammary infusions of lactic acid bacteria (e.g., Lactococcus lactis) or oral administration.
Antibiotic: Standard treatment but leads to milk discard periods.
Probiotic: Promising for prevention and treatment, reducing somatic cell count (SCC). Mechanism: Competitive exclusion of pathogens like S. aureus, modulation of the local immune response in the mammary gland [137].
General Growth Promotion/Health MaintenanceLivestock: Poultry and Swine.Antibiotic: Growth-Promoting Antibiotics (AGPs)—now banned/restricted in many regions.
Probiotic: Various (Bacillus, Lactobacillus, Enterococcus strains). ~0.5–2 × 109 CFU/kg feed.
Duration: Continuous.
Antibiotic: Historically improved growth rate and feed efficiency.
Probiotic: Modestly improves Average Daily Gain (ADG) and Feed Conversion Ratio (FCR). Mechanism: Competitive exclusion of pathogens, improved nutrient digestibility, enhanced intestinal health and morphology [126].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhao, M.; Chen, B.; Peng, S.; Mei, G.; Li, M.; Lin, F.; Sun, T.; Li, Z. Swine-Derived Probiotics and Their Metabolites as an Alternative to Veterinary Antibiotics. Vet. Sci. 2025, 12, 1100. https://doi.org/10.3390/vetsci12111100

AMA Style

Zhao M, Chen B, Peng S, Mei G, Li M, Lin F, Sun T, Li Z. Swine-Derived Probiotics and Their Metabolites as an Alternative to Veterinary Antibiotics. Veterinary Sciences. 2025; 12(11):1100. https://doi.org/10.3390/vetsci12111100

Chicago/Turabian Style

Zhao, Mengshi, Bihong Chen, Song Peng, Guiheng Mei, Meiqin Li, Fengqiang Lin, Tiecheng Sun, and Zhaolong Li. 2025. "Swine-Derived Probiotics and Their Metabolites as an Alternative to Veterinary Antibiotics" Veterinary Sciences 12, no. 11: 1100. https://doi.org/10.3390/vetsci12111100

APA Style

Zhao, M., Chen, B., Peng, S., Mei, G., Li, M., Lin, F., Sun, T., & Li, Z. (2025). Swine-Derived Probiotics and Their Metabolites as an Alternative to Veterinary Antibiotics. Veterinary Sciences, 12(11), 1100. https://doi.org/10.3390/vetsci12111100

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