Abstract
The interaction between the gut microbiota and the host immune system is pivotal in maintaining health or driving disease pathogenesis. The gut microbiota directly or indirectly modulates immune cells activation and inflammatory cytokines secretion through microbial metabolites, including short-chain fatty acids (SCFAs), tryptophan metabolites, bile acids, and polyamines. Conversely, the immune system regulates microbial community composition by maintaining the integrity of the epithelial barrier. In addition, antibiotics and probiotics can further regulate the inflammatory response by altering gut microbiota structure and microbial metabolites levels. This review systematically examines the bidirectional regulatory mechanisms among the gut microbiota, microbial metabolites, and inflammatory cytokines, and explores the impact of antibiotics and probiotics on this interaction network. These insights provide new targets for immune-related diseases.
1. Introduction
The gut microbiota, often referred to as the human body’s “second brain,” is a diverse and dynamic microbial community colonizing the gastrointestinal tract [,]. Its composition and functional capacity are critically important to human health. Under physiological conditions, the gut microbiota contributes to host well-being by metabolizing dietary components, synthesizing essential nutrients, resisting pathogen colonization, and modulating immune responses [,,]. Much like other organs, the composition and diversity of the gut microbiota are closely linked to host physiological functions such as nutrient metabolism, biosynthesis, barrier protection, and immune homeostasis []. According to the International Code of Nomenclature of Prokaryotes (ICNP), the gut microbiota is classified into various taxonomic ranks []. Major phyla include Bacillota, Bacteroidota, Actinomycetota, Fusobacteriota, Pseudomonadota, and Verrucomicrobiota. The composition of the gut microbiota is not static; rather, its stability is co-regulated by both host genetics and external environmental factors. Among these external influences, dietary patterns (e.g., high-fiber or high-fat diets), antibiotic use, lifestyle, stress, and disease states can significantly influence gut microbial diversity [,].
The gut microbiota plays a crucial role in maintaining immune defense functions. Under normal physiological conditions, it exhibits strong ecological resilience, enabling it to restore its original composition through self-regulation mechanisms after minor disturbances []. However, long-term antibiotic abuse or changes in dietary patterns can lead to irreversible alterations in the gut microbiota structure, a condition known as dysbiosis []. Dysbiosis is primarily characterized by reduced microbial diversity, a decline in beneficial bacteria, and an increase in potentially pathogenic bacteria [,]. This imbalance can trigger systemic low-grade inflammation through gut microbiota-immune interactions, manifesting as weakened anti-inflammatory capacity and enhanced pro-inflammatory responses []. Consequently, dysbiosis drives the initiation and progression of various chronic diseases, including inflammatory bowel disease (IBD), metabolic syndrome, autoimmune disorders, and neurodegenerative diseases. Furthermore, structural disruption of the gut microbiota alters its metabolic output, reducing the synthesis of beneficial metabolites [] (e.g., short-chain fatty acids [SCFAs] like butyrate and propionate) while increasing production of harmful metabolites (e.g., lipopolysaccharides, LPS) []. These changes directly impair intestinal epithelial barrier function, induce “leaky gut,” and disrupt local and systemic immune responses [].
Conversely, the immune system also regulates microbial community composition by maintaining epithelial barrier function. This immunomodulatory is mediated through three major synergistic mechanisms: First, inflammatory cytokines (e.g., TNF-α, IL-4, and IL-13) produced by the immune system modulate the expression of mucins (e.g., MUC2) in goblet cells [,]. These mucins, upon glycosylation, form glycan structures that serve as key carbon sources for certain gut bacteria (e.g., Akkermansia muciniphila and Bacteroides spp.) []. Changes in the availability of mucin glycans directly affect the abundance of glycan-dependent bacteria, thereby reshaping the overall composition of the gut microbiota []. Second, immune cells secrete IgA antibodies that precisely coat and suppress mucosa-associated bacteria, particularly potential pathogens, limiting their adhesion, colonization, and translocation. This process helps maintain the spatial distribution and ecological balance of the microbiota []. Third, immune signaling enhances the expression of epithelial tight junction proteins and promotes Paneth cell secretion of antimicrobial peptides (e.g., defensins), further strengthening physical and chemical barrier functions []. These actions shape the microbial microenvironment and enable deep regulation of the microbiota’s structure and function []. Overall, the immune system employs these multiple mechanisms to synergistically maintain intestinal epithelial barrier integrity, directly or indirectly regulating the composition of the gut microbiota.
In summary, there exists a close and complex bidirectional regulatory relationship among the gut microbiota, its metabolites, and the immune system’s inflammatory cytokines []. On one hand, the gut microbiota and its metabolites directly regulate immune cell functions and inflammatory cytokine expression [,]. On the other hand, the immune system feedback-modulates microbial structure and ecological stability through cytokines and immune mediators []. This intricate crosstalk is central to maintaining gut homeostasis. Therefore, this review discusses the close interactions among the gut microbiota, microbial metabolites (e.g., SCFAs, bile acids, and tryptophan metabolites), and inflammatory cytokines, providing strategies for developing gut microbiota-based interventions (e.g., probiotics), with the aim of enhancing host immune function.
2. Gut Microbiota Modulate the Release of Inflammatory Cytokines
The intestinal immune system is primarily composed of the gut mucosa, immune cells, and associated metabolites []. The gut microbiota interacts directly or indirectly with host immune cells through microbial metabolites, leading to the production of inflammatory cytokines []. These cytokines serve as key signaling molecules that stimulate B cells, T cells, or lymphocytes to release specific antibodies in response to pathogenic insults, thereby triggering immune responses to maintain intestinal homeostasis [,]. In the context of intestinal immunity and inflammatory responses, cytokines can be broadly classified into two functional categories [,]: pro-inflammatory and anti-inflammatory. Typical pro-inflammatory cytokines, such as IL-6, IL-8, IL-12, TNF-α, and IFN-γ, are primarily produced by immune cells (e.g., macrophages and T cells) and epithelial cells []. Their functions include activating immune responses, mediating chemotaxis, and facilitating pathogen clearance. In contrast, anti-inflammatory cytokines such as IL-10 and TGF-β are mainly secreted by regulatory T cells and dendritic cells [], and function to suppress excessive inflammation, promote tissue repair, and maintain immune homeostasis []. IL-6, a pleiotropic cytokine, exhibits both pro- and anti-inflammatory properties, with its specific role depending on the gut microenvironment and relevant receptor signaling []. A systematic summary of the cellular origins and physiological functions of key intestinal inflammatory cytokines will help further elucidate the interaction mechanisms between gut microbiota and inflammatory cytokines (Table 1).
Table 1.
Major cellular sources and physiological functions of inflammatory cytokines in the gut.
The gut microbiota serves as a critical regulatory hub by modulating the release of inflammatory cytokines, thereby playing a central role in the transition between systemic homeostasis and disease pathogenesis [,]. Accumulating evidence has established gut dysbiosis as a key factor disrupting the inflammatory cytokine network. Accompanied by damage to the intestinal mucosal barrier, dysbiosis disrupts the balance of inflammatory cytokines []. Dysregulation of inflammatory mediators—characterized by excessive pro-inflammatory cytokine production and/or impaired anti-inflammatory mechanisms—directly promotes the initiation and progression of various chronic diseases, including autoimmune conditions, metabolic syndrome, and neurodegenerative disorders [] (Table 2). For instance, in diabetic patients, decreased abundance of Roseburia, Faecalibacterium, Bifidobacterium, and Akkermansia contributes to intestinal barrier dysfunction [,,,,]. This permits bacteria or bacterial products such as lipopolysaccharide (LPS) to enter the circulation. Translocated LPS binds to TLR4 receptors on adipocytes, activating NF-κB signaling and stimulating the production of pro-inflammatory cytokines, including TNF-α and IL-6 []. These cytokines inhibit insulin signaling pathways, induce insulin resistance, and propagate an inflammatory cascade []. In sepsis patients, elevated levels of Enterococcus, Streptococcus, and Staphylococcus correlate with increased systemic concentrations of cytokines such as IL-6 and IL-10 []. Similarly, Chen et al. observed that adolescents with depression exhibit reduced abundance of Bifidobacterium, Escherichia, Lactobacillus, and Bacteroides, along with heightened levels of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-6 []. Moreover, increased gut epithelial permeability facilitates the translocation of other immunogenic substances—such as intact proteins, gluten, and food antigens—into the bloodstream. Upon systemic dissemination, these antigens trigger an immune response culminating in the production of autoantibodies, exaggerated immune activation, and the development of autoimmune diseases, including celiac disease, systemic lupus erythematosus (SLE), and autoimmune hepatitis [,].
Table 2.
Associations between diseases, gut microbiota alterations, and inflammatory cytokines.
Research on the gut microbiota and its metabolites offers novel perspectives for innovating strategies in disease diagnosis and treatment. For instance, in the field of inflammatory bowel disease (IBD), the gut microbiota has emerged as a critical therapeutic target []. Modulation of the gut microbiota is also considered a promising treatment approach for hypertension []. In the management of gastrointestinal cancers and infectious diseases, microbial metabolites demonstrate considerable potential by engaging multi-dimensional regulatory mechanisms []. Targeting these metabolites allows for effective modulation of host immune responses, thereby slowing disease progression. Inflammatory cytokines act as key messengers in cardiometabolic diseases and various chronic conditions, playing a central role in disease pathogenesis. Essentially, regulating the gut microbiota and its metabolites can indirectly modulate cytokine levels, offering innovative avenues for preventing and treating these diseases [].
Research confirmed that the gut microbiota modulates the release of inflammatory cytokines by either directly colonizing or indirectly adhering to epithelial cells (Figure 1). Firstly, certain gut microorganisms, such as invasive Klebsiella species, can directly colonize intestinal epithelial cells. These organisms are phagocytosed by dendritic cells (DCs), stimulating the release of IL-6, TNF, and IL-12 []. Secondly, the surface components of gut microbes, including extracellular polysaccharides (EPS) and pili, enhance their adhesion to intestinal epithelial cells []. For instance, Bifidobacterium with a thicker EPS layer not only directly regulates the secretion of IL-10 and TNF-α by macrophages but also modulates the secretion of IL-6 by DCs via the Toll-like receptor 2 (TLR2) receptor []. Additionally, Bifidobacterium with pili enhances the adhesion to intestinal epithelial cells and regulates the secretion of TNF-α by macrophages compared to no pili control strains []. Thirdly, segmented filamentous bacteria (SFB) stimulate the secretion of TGF-β, IL-12, IL-23, and IL-17 by CD11c+ DC cells, mediated by the release of serum amyloid A (SAA) from epithelial cells. Fourthly, specific gut microbes, such as Bacteroides species or Clostridium clusters IV and XIVa, can directly regulate CD103+ DC cells through goblet cells, promoting the secretion of TGF-β and IL-10 []. Furthermore, Firmicutes with substrate-binding protein (SBP) and Bacteroidetes with tetratricopeptide repeat lipoprotein (TPRL) can selectively recognize T-cell receptors (TCRs), thereby regulating the secretion of TNF-α and IFN-γ []. In summary, the gut microbiota can directly or indirectly regulate the release of inflammatory cytokines, thereby influencing host immune function.
Figure 1.
The gut microbiota and metabolites regulate the release of inflammatory cytokines. The gut microbiota regulates inflammatory cytokine release through immune cells mainly by (1) direct adhesion to intestinal epithelial cells (e.g., ① invasive Klebsiella species with invasive ability; ② Bifidobacterium with extracellular polysaccharides (EPS) and pili); and (2) indirectly through goblet cells. (e.g., ③ segmented filamentous bacteria (SFB) that stimulate serum amyloid A (SAA) release; ④ Bacteroides species or Clostridium clusters IV and XIVa that pass through goblet cells). Interestingly, gut microbiota metabolites including: short-chain fatty acids (SCFA), tryptophan metabolites, bile acids (BAs), lipopolysaccharide (LPS), polyamines, etc., all directly or indirectly modulate the relevant receptors, which further stimulate the release of inflammatory cytokines from immune cells such as B cells, T cells (Tregs), dendritic cells (DCs).
3. Microbial Metabolites Modulate the Release of Inflammatory Cytokines
The gut microbiota can directly or indirectly modulate host immune responses through microbial metabolites, thereby contributing to the maintenance of immune homeostasis. Recent studies have revealed that the majority of systemic metabolites are gut-derived and influence immune reactivity by regulating the differentiation, proliferation, and apoptosis of immune cells []. In parallel, advances in both non-targeted and targeted metabolomics technologies have enabled the identification and detection of an increasing number of small-molecule metabolites associated with the gut microbiota. These metabolites can be categorized into three main groups (Figure 1). (1) Metabolites produced from dietary precursors via indispensable gut microbial biotransformation (e.g., short-chain fatty acids and tryptophan metabolites; not obtainable directly from diet) []. (2) Metabolites from host-microbial co-metabolism: host-synthesized molecules that require structural modification by gut microbial enzymes to alter their biological functions, as exemplified by bile acids []. (3) Microbially de novo synthesized metabolites: metabolites directly produced and released by gut microbiota (e.g., LPS; polyamines, primarily derived from microbes in the gut) []. Furthermore, it is now widely acknowledged among experts that these metabolites influence host immune function by modulating inflammatory factors through interactions with immune cells.
3.1. Short-Chain Fatty Acids (SCFAs)
Short-chain fatty acids (SCFAs) are byproducts of the gut microbiota’s metabolism of undigested carbohydrates. Among these, acetate, propionate, and butyrate are the most abundant SCFAs []. These organic acids play significant roles in immune regulation through several mechanisms. Firstly, SCFAs serve as energy sources for intestinal epithelial cells and can directly bind to G protein-coupled receptors (GPCRs) such as GPR43, GPR41, and GPR109A, thereby triggering the release of the pro-inflammatory cytokine IL-18 []. Secondly, butyrate, specifically, can induce the differentiation of Foxp3+ regulatory T cells (Tregs) by modulating histone deacetylase (HDAC) activity, thereby influencing IL-18 expression []. Thirdly, butyrate binds to GPCRs on DC cells, inducing the release of IL-10 []. Butyrate also directly influences the plasticity of type 3 innate lymphoid cells (ILC3s), enhancing their production of the barrier-protective cytokine IL-22 [].
3.2. Tryptophan Metabolites
The gut microbiota, including Escherichia coli, Proteus vulgaris, Clostridium, and Ruminococcus, can metabolize tryptophan into various metabolites such as indole, indole ethanol (IE), indole propionic acid (IPA), indole lactic acid (ILA), indole acetic acid (IAA), indole aldehyde (IAld), indole acrylic acid (IA), skatole, and tryptamine. This process occurs through the catalytic enzymes involved in tryptophan metabolism or via decarboxylation reactions [,]. Tryptophan metabolites primarily regulate the levels of IL-10 and IL-22 by acting on B cells through the aryl hydrocarbon receptor (AhR) []. For example, Bacteroides and Taylorella species facilitate the production of tryptophan-derived metabolites, such as IAA and IPA, which act as agonists of AhR [,,]. Upregulation of AhR promotes the differentiation of naïve T cells into Th17 and regulatory T (Treg) cells, thereby upregulating the expression of the anti-inflammatory cytokine IL-10 while suppressing the production of pro-inflammatory cytokines such as IFN-γ and IL-17. This mechanism has been demonstrated to exert anti-inflammatory effects in dextran sulfate sodium (DSS)-induced colitis models []. In rodent models of diabetes-associated depression, activation of indoleamine 2,3-dioxygenase (IDO)—a key enzyme in tryptophan metabolism—results in decreased hippocampal serotonin (5-HT) levels and elevated pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6 []. Nevertheless, current research on the regulation of inflammatory cytokines by these metabolites remains limited, and other potential signaling pathways involved warrant further exploration.
3.3. Bile Acids (BAs)
BAs encompass two categories: primary and secondary bile acids. Approximately 5% of primary BAs are transformed into secondary BAs through metabolism by the gut microbiota, notably by Clostridium scindens within the Firmicutes phylum, via the action of 7α-hydroxylase []. Several key mechanisms underlie how BAs modulate inflammatory cytokines. For instance, secondary BAs like lithocholic acid (LCA) can directly stimulate Th17 and Treg cells, thereby influencing the levels of inflammatory cytokines []. Additionally, BAs can bind to VDR receptors on Treg cells, stimulating the release of inflammatory cytokines []. BAs can also bind to the FXR receptor on DC cells or the TGR5 receptor on macrophages, further regulating the concentration of inflammatory cytokines [].
3.4. Polyamines
Polyamines mainly include spermine, spermidine, and putrescine. Gut microbiota, such as Bacteroides and Clostridium, are major producers of polyamines []. Carriche G. M. et al. confirmed that dietary spermidine supplementation can promote the steady-state differentiation of Treg cells in the gut, but its regulatory effect on inflammatory cytokines has not been studied in more detail [].
3.5. Microbe-Associated Molecular Patterns (MAMPs)
Lipopolysaccharide (LPS) functions as a crucial priming agent for the activation of the NLRP3 inflammasome. It initiates the activation of caspase-1, which subsequently catalyzes the maturation and release of the pro-inflammatory cytokines IL-1β and IL-18, thereby coordinating a targeted innate immune response []. This pathway represents a fundamental aspect of host defense and is also significantly involved in the pathogenesis of various inflammatory diseases, including Alzheimer’s disease, sepsis, and autoimmune disorders [,]. Similarly, other microbe-associated molecular patterns contribute to immune activation through distinct pathways. Lipoteichoic acid, a key component of the Gram-positive bacterial cell wall, triggers inflammation via the TLR2 signaling pathway []. Correspondingly, ubiquitous peptidoglycan fragments are detected by the intracellular pattern recognition receptors NOD1 and NOD2 []. This recognition subsequently initiates the activation of the NF-κB pathway and drives inflammatory responses []. Furthermore, bacterial extracellular vesicles (BEVs)—nanoscale particles released by bacteria—function as central hubs for immune coordination. These vesicles synergistically activate both TLR and NOD signaling pathways while also serving as potent activators of the NLRP3 inflammasome, significantly enhancing the maturation and release of IL-1β [,,].
4. Inflammatory Cytokines Remodel the Gut Microbiota Composition
The composition of gut microbiota can be either directly or indirectly regulated by host immune cells, such as macrophages, T cells, and B cells, through the action of inflammatory cytokines (Figure 2). Firstly, the composition of the gut microbiota can be regulated by the secretion of two antimicrobial peptides (AMPs), α-defensin or β-defensin, by macrophages, natural killer T cells (NKT), and B cells []. For instance, α-defensins can regulate the overgrowth of Clostridium difficile []. While β-defensin-hBD-1 is only active against Gram-positive bacteria, β-defensin HBD-2, 3, and 4 exhibit antibacterial action against Escherichia coli (E. coli), Pseudomonas aeruginosa, Staphylococcus aureus, and Streptococcus pyogenes. Secondly, TL1A and IL-18 release can be regulated by TCRγδ(+), CD8(+) T cells, and muco-associated immutable T cells (MAIT), which in turn can alter the ratio of Bacillota/Bacteroidetes, as well as the contents of Lactobacillus and E. coli [,]. Thirdly, NKT can selectively identify lipid antigens presented by CD1d on DC cells and intestinal epithelial cells. Additionally, NKT can control the makeup of symbiotic microbes such as Lactobacillus gasseri, Staphylococcus aureus, and E. coli []. Lastly, polymerized immunoglobulin receptors (pIgR) allow IgA antibodies made by B cells and follicular helper T (Tfh) cells to enter the intestinal lumen and control the composition of the gut microbiota [].
Figure 2.
Inflammatory cells and inflammatory cytokines reshape the gut microbiota composition. The regulation of gut microbiota composition by host immune cells mainly includes ① direct regulation through inflammatory cytokines; ② regulation through antimicrobial peptides (AMP); ③ regulation through specific recognition of antigens; and ④ regulation through the production of specific antibodies.
5. The Evolutionary Pressure by Antibiotics on the Communication Network
Antibiotics have long been considered effective agents for treating bacterial infections; however, their widespread and often indiscriminate use can exert long-lasting impacts on the gut microbiota through drug-specific mechanisms, thereby increasing the host’s susceptibility to various diseases [,]. Studies indicate that both short-term and long-term antibiotic treatments can disrupt the gut microbiota, leading to reduced microbial diversity, loss of ecological balance, and overgrowth of pathogenic bacteria such as Clostridium difficile []. For instance, the use of β-lactams, glycopeptides, and macrolides has been associated with decreased abundance of beneficial bacteria including Bifidobacterium and Lactobacillus []. Research in animal models has revealed that early-life administration of antibiotics like tylosin and amoxicillin reduces the abundance of taxa such as Muribaculaceae, *S24-7*, α-proteobacteria, and δ-proteobacteria. These alterations increase susceptibility to pathogens and exacerbate the severity of diseases in adulthood, including diarrhea, hemorrhagic colitis, Crohn’s disease, and colorectal tumors []. Clinical studies further confirm that even a brief four-day course of combined meropenem, gentamicin, and vancomycin can induce a substantial expansion of Enterobacteriaceae, Enterococcus faecalis, and Fusobacterium nucleatum, alongside a concurrent reduction in Bifidobacterium and butyrate-producing bacterial populations []. Moreover, exposure to antibiotics during early life, especially in infancy, may cause long-term disruptions to the composition of the gut microbiota. Evidence from a Finnish pediatric cohort revealed that early macrolide use led to decreased abundance of Actinobacteria and increased levels of Bacteroidetes and Proteobacteria. These microbial alterations are associated with elevated risks of asthma and obesity in later childhood []. In addition to the direct adverse outcomes of microbiota disruption and disease induction, antibiotic administration poses a more profound and enduring threat: the emergence and dissemination of antibiotic resistance []. The powerful selective pressure exerted by these drugs promotes the rapid enrichment and ecological dominance of resistant bacterial strains within the host [,,]. These resistant strains can also rapidly propagate resistance genes across different species via horizontal gene transfer mechanisms such as conjugation, transduction, and transformation []. This enables the widespread dissemination of resistance traits throughout microbial communities, further compromising treatment efficacy and facilitating the rise in multidrug-resistant infections [].
Beyond directly altering the microbial composition, antibiotics also significantly impact the levels of microbial metabolites. Zhang et al. [] demonstrated that after three weeks of vancomycin treatment, the fecal contents of mice showed significant reductions in short- and long-chain fatty acids, bile acids, L-arginine, L-tyrosine, and phosphatidylcholines. Further research [] revealed that treatment with enrofloxacin, vancomycin, and polymyxin B upregulates the expression of colonic cytokine-related genes and significantly disrupts the biosynthesis pathways of valine, leucine, and isoleucine. Moreover, the combined administration of vancomycin with ciprofloxacin–metronidazole markedly reduced the concentrations of alanine, branched-chain amino acids, and aromatic amino acids in the colonic contents of female mice [].
Antibiotic-induced disturbances in gut microbiota and metabolite profiles are often accompanied by shifts in inflammatory cytokine production. Notably, the expression levels of inflammatory cytokines such as IL-17A, IL-22, IL-1β, and IL-12 are affected by antibiotics [,]. Wang et al. [] identified positive correlations between Enterococcus and Klebsiella genera and pro-inflammatory mediators such as TNF-α, IL-12, and IL-1β. Kathryn A Knoop et al. [] found that levels of the inflammatory cytokines CXCL 1, IL-17, and IFN γ were significantly elevated after sustained antibiotic use. Collectively, these findings highlight a tightly intertwined relationship among antibiotic usage, gut microbial community structure, metabolite dynamics, and inflammatory immune responses.
6. Foods Intervene in the Immune-Inflammatory Response by Modulating Inflammatory Cytokines
6.1. Probiotics
Probiotics, defined by the World Health Organization (WHO) and the Food and Agriculture Organization (FAO) of the United Nations as “live microorganisms which when administered in adequate amounts confer a health benefit on the host,” [] represent a low-cost and well-tolerated intervention strategy for managing inflammatory conditions. In recent years, probiotic strains, particularly Bifidobacterium and Lactobacillus, have emerged as promising microbiota-targeting therapies [,]. They are now widely used as adjunctive or alternative treatments for a range of immune-related inflammatory diseases [].
Probiotics exert their beneficial effects through multiple synergistic mechanisms to maintain intestinal homeostasis. Firstly, as live microorganisms, they are capable of colonizing and proliferating within the host intestine, thereby increasing the abundance of beneficial bacteria and establishing ecological dominance [,]. Secondly, through competitive exclusion, probiotics compete with pathogens for nutrients and adhesion sites. This competition facilitates the expansion of commensal bacteria while suppressing the growth of harmful microbes []. Thirdly, probiotics produce metabolites such as SCFAs, which lower the luminal pH and create an unfavorable environment for enteropathogens [,]. Certain strains also secrete antimicrobial substances—including bacteriocins, lactic acid, and defensins—that directly inhibit pathogen colonization []. In terms of immunomodulation, probiotics fine-tune host immunity via direct interactions with immune cells []. Strains from different genera can engage with dendritic cells (DCs), prompting the differentiation of naïve T cells into various subsets such as Th1, Th2, or regulatory T cells (Tregs) []. This polarization, in turn, stimulates the production of specific inflammatory cytokines. For instance, Lactobacillus species can directly modulate T-cell responses by enhancing the secretion of cytokines including TGF-β, IL-10, and IL-8, thereby precisely calibrating the intensity and direction of immune activation []. Through such multi-layered crosstalk with the immune system, probiotics not only enhance innate and adaptive immunity but also help suppress excessive inflammation and mitigate the development of inflammation-associated disorders.
Regarding the immunomodulatory functions of probiotics within cytokine networks, accumulating evidence indicates that their beneficial effects are largely mediated through a dual regulatory mechanism. This involves the suppression of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-8, coupled with the promotion of anti-inflammatory cytokine secretion in the gut []. Sun et al. [] utilized untargeted metabolomics to investigate the anti-inflammatory properties of a postbiotic produced by Lactobacillus paracasei K56—a strain initially isolated from infant gut microbiota. Their study demonstrated that the cell-free supernatant (CFS) of L. paracasei K56 significantly reduced TNF-α expression in macrophage RAW 264.7 cells, effectively mitigating cellular inflammation. These anti-inflammatory effects were further validated in both a high-fat diet-induced zebrafish model and a DSS-induced murine model of ulcerative colitis. The anti-inflammatory properties of probiotics and postbiotics have been consistently demonstrated across various animal disease models. The DSS-induced mouse colitis model has demonstrated that Lactobacillus rhamnosus GG suppresses inflammation by upregulating IL-10 expression in Ly6C+ monocyte []. Similarly, Bifidobacterium infantis promotes Treg cell differentiation and enhances the secretion of IL-10 and TGF-β1 []. Furthermore, L. rhamnosus ZFM231 significantly alleviates the pathological progression of colitis by modulating the TGF-β/TGF-α balance and regulating the microbiota structure []. This immunomodulatory effect has been further validated in clinical trials. Bifidobacterium longum ES1 significantly reduces serum levels of IL-6, IL-8, and TNF-α in patients with irritable bowel syndrome (IBS), while restoring intestinal permeability and barrier function []. In the context of immune inflammation, Lactiplantibacillus plantarum HM-22 effectively corrects the Th1/Th2 immune imbalance in an α-lactalbumin (α-LA)-induced allergy model by markedly upregulating the expression of anti-inflammatory factors IL-10, IFN-γ, and TGF-β, while inhibiting Immunoglobulin E (IgE) production and the secretion of the Th2-type cytokine IL-4 []. A clinical trial involving patients with atopic dermatitis demonstrated that oral administration of L. plantarum IS-10506 significantly reduced serum levels of IL-4 and IL-17, while markedly increasing levels of IFN-γ and Foxp3+ []. In the field of neurodegenerative diseases, research on Parkinson’s disease (PD) has demonstrated that L. salivarius LS01 and L. acidophilus significantly reduce levels of TNF-α, IL-6, and IL-17A, while enhancing the expression of IL-4 and IL-10 through the stimulation of peripheral blood mononuclear cells (PBMCs) in PD patients []. Additionally, in the MPTP-induced PD mouse model, interventions with L. plantarum CRL 2130 and Streptococcus thermophilus (CRL 807/808) resulted in decreased serum levels of IL-6 and TNF-α, along with an increase in IL-10, which was significantly associated with the improvement of PD symptoms []. A diverse collection of probiotic strains, tested across various experimental systems—including in vitro studies, animal models, and clinical trials—demonstrates therapeutic potential in preventing and treating immune-related diseases via modulation of inflammatory cytokines (Table 3).
Table 3.
Effect and mechanism of probiotics on inflammatory cytokines.
Driven by the rapid advancement of gene editing technologies, therapeutic strategies for immune-related diseases continue to expand. In the future, interventional approaches will no longer be limited to conventional probiotics; engineered probiotics are emerging as a more precise and efficient treatment modality [,]. As live biotherapeutic products, they demonstrate significant potential for targeting specific diseases. These programmed probiotics can secrete bioactive metabolites such as SCFAs, which precisely interact with disease markers and related effector molecules, thereby modulating specific pathological pathways []. Leveraging this mechanism, engineered probiotics have been applied in various fields, including metabolic disorders, behavioral abnormalities, and cancer therapy [,]. For instance, Praveschotinunt et al. [] employed genetic engineering techniques to enable Escherichia coli Nissle 1917 (EcN) to express the anti-inflammatory cytokine IL-10. This modified strain not only promoted the expansion of beneficial microbial populations and improved the gut microenvironment but also markedly attenuated inflammatory responses, effectively alleviating symptoms associated with various intestinal diseases []. This example underscores the practical value and broad prospects of engineered probiotics in disease treatment.
6.2. Other Foods
A diverse range of interventions beyond probiotics—including prebiotics, postbiotics, synbiotics, fecal microbiota transplantation (FMT), and dietary modulation—has shown considerable potential in alleviating inflammatory diseases through the regulation of gut microbiota []. Prebiotics, selectively utilized by host microorganisms, promote the growth and colonization of beneficial bacteria via fermentation-derived metabolites such as SCFAs []. Research has demonstrated that prebiotics can mitigate periodontitis by downregulating pro-inflammatory cytokines, including TNF-α and IL-1β []. Postbiotics, characterized by their well-defined composition, stability, and safety, have exhibited superior efficacy in colitis models. Research in a DSS-induced mouse colitis model demonstrates that postbiotics from Saccharomyces boulardii more effectively lower serum TNF-α and IL-6 levels than live bacteria []. Complementing this, heat-inactivated Lactiplantibacillus argentoratensis BBLB001 boosts colonic mucin and secretory IgA, underscoring the critical function of postbiotics in reinforcing intestinal defense and microbial regulation []. Moreover, supernatants from Lactobacillus acidophilus and Lactobacillus casei suppress the release of the pro-inflammatory cytokine TNF-α and promote that of the anti-inflammatory IL-10 in macrophages, thereby enhancing intestinal barrier integrity and fine-tuning local immunity []. Synbiotics, which integrate probiotics and prebiotics in a complementary manner, have demonstrated therapeutic potential in inflammatory bowel disease []. In contrast, fecal microbiota transplantation (FMT) restores a healthy gut microbial ecosystem and has proven effective in treating recurrent Clostridioides difficile infections and metabolic syndromes [,]. Collectively, these integrated microecological strategies not only expand the understanding of gut–immune axis regulation but also facilitate the development of novel dietary interventions based on functional foods, underscoring their promising clinical applicability.
7. Conclusions
Numerous studies have been conducted on the relationships among inflammatory cytokines, gut microbiota, and gut metabolites. One possible therapeutic strategy is to control immunity through gut microbiota and its metabolites. Interestingly, a large number of studies on probiotics have confirmed that probiotics can regulate the release of inflammatory cytokines through gut microbiota and metabolites, but different types of probiotics, different doses, and different intervention times have different therapeutic effects. The gut microbiota and the immune system together form a complex network, and their relationship requires further investigation through in vitro or in vivo studies, including animal or clinical studies. Meanwhile, consideration must be given to the diverse immune cell types, the multiple levels of gut microbiota classification, and the varying conditions required for metabolite identification. In summary, further research into the interrelationships among these three aspects is expected to provide a robust foundation for enhancing immunity through gut microbiota modulation.
Author Contributions
Conceptualization, R.W., J.C., and Q.Z.; software, G.W. and Y.W.; investigation, G.W., Y.W., and S.S.; writing—original draft preparation, G.W., R.W., and Q.Z.; writing—review and editing, J.C. and Q.Z. All authors have read and agreed to the published version of the manuscript.
Funding
This research was funded by the National Natural Science Foundation of China (Grant No. 32402113; Q.Z. and G.W.) and the National Key R&D Program of China (Grant No. 2021YFD1600204; R.W. and Y.W.).
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
The original contributions presented in the study are included in the article; further inquiries can be directed to the corresponding authors.
Conflicts of Interest
The authors declare no conflicts of interest.
Abbreviations
The following abbreviations are used in this manuscript:
| IL | Interleukin |
| TNF | Tumor necrosis factor |
| TGF | Transforming growth factor |
| PRRs | Pattern recognition receptors |
| GPR | G protein-coupled receptor |
| AHR | Aryl hydrocarbon receptor |
| HDAC | Histone deacetylase |
| TLR | Toll-like receptor |
| TGR | Takeda G-protein-coupled receptor |
| TPRL | tetratricopeptide repeat lipoprotein |
| TCRs | T-cell receptors |
| FXR | Farnesoid X receptor |
| VDR | Vitamin D receptor |
| SAA | Serum amyloid A |
| SBP | Substrate-binding protein |
| SFB | Segmented filamentous bacteria |
| GPCRs | G protein-coupled receptors |
| IELs | Intraepithelial lymphocytes |
| NKT | Natural killer T |
| DCs | Dendritic cells |
| EPS | Extracellular polysaccharides |
| SCFAs | Short-chain fatty acids |
| BAs | Bile acids |
| DSS | Dextran sulfate sodium |
| LPS | Lipopolysaccharide |
| PD | Parkinson’s disease |
References
- Shutov, E.V.; Bolshakov, S.A.; Makarova, T.A.; Fedoseeva, I.A.; Teplyuk, D.A.; Pavlov, C.S.; Sorokoletov, S.M. Gut Microbiota and Kidney Diseases. Literature review. Nephrol. Dial. 2024, 26, 283–302. [Google Scholar] [CrossRef]
- Rejuba, P. The Gut Microbiome: The Complex Ecosystem within us. Int. J. Sci. Res. Arch. 2023, 10, 329–341. [Google Scholar] [CrossRef]
- Thursby, E.; Juge, N. Introduction to the Human Gut Microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
- Feng, X.; Luo, C.; Che, J. Diet Modulates Host Health through Gut Microbiota Derived Extracellular Vesicles: A Short Review. Aceh J. Anim. Sci. 2023, 8, 58–61. [Google Scholar] [CrossRef]
- Zhao, M.; Chu, J.; Feng, S.; Guo, C.; Xue, B.; He, K.; Li, L. Immunological Mechanisms of Inflammatory Diseases Caused by Gut Microbiota Dysbiosis: A Review. Biomed. Pharmacother. 2023, 164, 114985. [Google Scholar] [CrossRef]
- Metwally, H.; Kishimoto, T. Gut Microbiome Metabolites and the Intestinal Homeostasis. J. Clin. Res. Rep. 2024, 15, 01–06. [Google Scholar] [CrossRef]
- Parker, C.T.; Tindall, B.J.; Garrity, G.M. International Code of Nomenclature of Prokaryotes. Int. J. Syst. Evol. Microbiol. 2019, 69, S1–S111. [Google Scholar] [CrossRef]
- Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The Gut Microbiota and Inflammation: An Overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef]
- Adak, A.; Khan, M.R. An Insight into Gut Gicrobiota and Its Functionalities. Cell. Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef]
- Dogra, S.K.; Dore, J.; Damak, S. Gut Microbiota Resilience: Definition, Link to Health and Strategies for Intervention. Front. Microbiol. 2020, 11, 572921. [Google Scholar] [CrossRef]
- Cristofori, F.; Dargenio, V.N.; Dargenio, C.; Miniello, V.L.; Barone, M.; Francavilla, R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front. Immunol. 2021, 12, 578386. [Google Scholar] [CrossRef] [PubMed]
- Weiss, G.A.; Hennet, T. Mechanisms and Consequences of Intestinal Dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef] [PubMed]
- Kostic, A.D.; Xavier, R.J.; Gevers, D. The Microbiome in Inflammatory Bowel Disease: Current Status and the Future Ahead. Gastroenterology 2014, 146, 1489–1499. [Google Scholar] [CrossRef]
- Hotamisligil, G.S. Inflammation, Metaflammation and Immunometabolic Disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
- Malesza, I.J.; Malesza, M.; Walkowiak, J.; Mussin, N.; Walkowiak, D.; Aringazina, R.; Bartkowiak-Wieczorek, J.; Mądry, E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells 2021, 10, 3164. [Google Scholar] [CrossRef]
- Ghosh, S.S.; Wang, J.; Yannie, P.J.; Ghosh, S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J. Endocr. Soc. 2020, 4, bvz039. [Google Scholar] [CrossRef]
- Di Vincenzo, F.; Del Gaudio, A.; Petito, V.; Lopetuso, L.R.; Scaldaferri, F. Gut microbiota, Intestinal Permeability, and Systemic Inflammation: A Narrative Review. Intern. Emerg. Med. 2024, 19, 275–293. [Google Scholar] [CrossRef]
- Kim, A.J.; Moreau, F.; Gorman, H.; Chadee, K. Colonic Goblet Cells Produce Augmented Levels of Pro-inflammatory Cytokines due to Metabolically Stressful MUC2 Mucin Biosynthesis. FASEB J. 2022, 36, S1.0R878. [Google Scholar] [CrossRef]
- Contreras-Ruiz, L.; Ghosh-Mitra, A.; Shatos, M.A.; Dartt, D.A.; Masli, S. Modulation of Conjunctival Goblet Cell Function by Inflammatory Cytokines. Mediat. Inflamm. 2013, 2013, 636812. [Google Scholar] [CrossRef]
- 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]
- Yang, S.; Yu, M. Role of Goblet Cells in Intestinal Barrier and Mucosal Immunity. J. Inflamm. Res. 2021, 14, 3171–3183. [Google Scholar] [CrossRef]
- Adolph, T.E.; Mayr, L.; Grabherr, F.; Tilg, H. Paneth Cells and their Antimicrobials in Intestinal Immunity. Curr. Pharm. Des. 2018, 24, 1121–1129. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between Microbiota and Immunity in Health and Disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
- Schirmer, M.; Smeekens, S.P.; Vlamakis, H.; Jaeger, M.; Oosting, M.; Franzosa, E.A.; Horst, R.T.; Jansen, T.; Jacobs, L.; Bonder, M.J.; et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell 2016, 167, 1897. [Google Scholar] [CrossRef]
- Alswat, A.S. The Influence of the Gut Microbiota on Host Health: A Focus on the Gut–Lung Axis and Therapeutic Approaches. Life 2024, 14, 1279. [Google Scholar] [CrossRef]
- Kim, S.; Seo, S.U.; Kweon, M.N. Gut Microbiota-derived Metabolites Tune Host Homeostasis Fate. Semin. Immunopathol. 2024, 46, 2. [Google Scholar] [CrossRef]
- Shim, J.A.; Ryu, J.H.; Jo, Y.; Hong, C. The Role of Gut Microbiota in T cell Immunity and Immune Mediated Disorders. Int. J. Biol. Sci. 2023, 19, 1178–1191. [Google Scholar] [CrossRef]
- Vancamelbeke, M.; Vermeire, S. The Intestinal Barrier: A Fundamental Role in Health and Disease. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 821–834. [Google Scholar] [CrossRef]
- Mendes, V.; Galvao, I.; Vieira, A.T. Mechanisms by Which the Gut Microbiota Influences Cytokine Production and Modulates Host Inflammatory Responses. J. Interferon Cytokine Res. 2019, 39, 393–409. [Google Scholar] [CrossRef]
- Ullah, H.; Arbab, S.; Tian, Y.; Chen, Y.; Liu, C.Q.; Li, Q.; Li, K. Crosstalk between Gut Microbiota and Host Immune System and Its Response to Traumatic Injury. Front. Immunol. 2024, 15, 1413485. [Google Scholar] [CrossRef]
- Saini, A.; Dalal, P.; Sharma, D. Deciphering the Interdependent Labyrinth between Gut Microbiota and the Immune System. Lett. Appl. Microbiol. 2022, 75, 1122–1135. [Google Scholar] [CrossRef]
- Liu, C.; Chu, D.; Kalantar-Zadeh, K.; George, J.; Young, H.A.; Liu, G. Cytokines: From Clinical Significance to Quantification. Adv. Sci. 2021, 8, e2004433. [Google Scholar] [CrossRef]
- Bamias, G.; Cominelli, F. Cytokines and Intestinal Inflammation. Curr. Opin. Gastroenterol. 2016, 32, 437–442. [Google Scholar] [CrossRef]
- Hu, X.; Yuan, X.; Zhang, G.; Song, H.; Ji, P.; Guo, Y.; Liu, Z.; Tian, Y.; Shen, R.; Wang, D. The Intestinal Macrophage–Intestinal Stem Cell Axis in Inflammatory Bowel Diseases: From Pathogenesis to Therapy. Life Sci. 2024, 344, 122452. [Google Scholar] [CrossRef]
- Dubinsky, M. Targeting Cytokines in Inflammatory Bowel Disease. Gastroenterol. Hepatol. 2023, 19, 550–552. [Google Scholar]
- Li, S.; Li, W.; Wu, X.; Zhang, B.; Liu, L.; Yin, L. Immune Cell-derived Extracellular Vesicles for Precision Therapy of Inflammatory-related Diseases. J. Control. Release 2024, 368, 533–547. [Google Scholar] [CrossRef]
- Guo, Y.; Wang, B.; Wang, T.; Gao, L.; Yang, Z.J.; Wang, F.F.; Shang, H.W.; Hua, R.; Xu, J.D. Biological Characteristics of IL-6 and Related Intestinal Diseases. Int. J. Biol. Sci. 2021, 17, 204–219. [Google Scholar] [CrossRef]
- Jang, D.-I.; Lee, A.-H.; Shin, H.-Y.; Song, H.-R.; Park, J.-H.; Kang, T.-B.; Lee, S.-R.; Yang, S.-H. The Role of Tumor Necrosis Factor Alpha (TNF-α) in Autoimmune Disease and Current TNF-α Inhibitors in Therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef]
- Verstockt, B.; Salas, A.; Sands, B.E.; Abraham, C.; Leibovitzh, H.; Neurath, M.F.; Vande Casteele, N.; Alimentiv Translational Research Consortium (ATRC). IL-12 and IL-23 pathway inhibition in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 433–446. [Google Scholar] [CrossRef]
- Ritzmann, F.; Lunding, L.P.; Bals, R.; Wegmann, M.; Beisswenger, C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022, 11, 2132. [Google Scholar] [CrossRef]
- Krueger, J.G.; Eyerich, K.; Kuchroo, V.K.; Ritchlin, C.T.; Abreu, M.T.; Elloso, M.M.; Fourie, A.; Fakharzadeh, S.; Sherlock, J.P.; Yang, Y.W.; et al. IL-23 Past, Present, and Future: A Roadmap to Advancing IL-23 Science and Therapy. Front. Immunol. 2024, 15, 1331217. [Google Scholar] [CrossRef]
- Ding, H.; Wang, G.; Yu, Z.; Sun, H.; Wang, L. Role of Interferon-gamma (IFN-gamma) and IFN-gamma Receptor 1/2 (IFNgammaR1/2) in Regulation of Immunity, Infection, and Cancer Development: IFN-gamma-dependent or Independent Pathway. Biomed. Pharmacother. 2022, 155, 113683. [Google Scholar] [CrossRef]
- Deng, Z.; Fan, T.; Xiao, C.; Tian, H.; Zheng, Y.; Li, C.; He, J. TGF-beta Signaling in Health, Disease, and Therapeutics. Signal Transduct. Target. Ther. 2024, 9, 61. [Google Scholar] [CrossRef]
- West, N.R. Coordination of Immune-Stroma Crosstalk by IL-6 Family Cytokines. Front. Immunol. 2019, 10, 1093. [Google Scholar] [CrossRef]
- Hu, B.; Ren, J.; Luo, Y.; Keith, B.; Young, R.M.; Scholler, J.; Zhao, Y.; June, C.H. Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep. 2017, 20, 3025–3033. [Google Scholar] [CrossRef]
- Valeri, M.; Raffatellu, M. Cytokines IL-17 and IL-22 in the Host Response to Infection. Pathog. Dis. 2016, 74, ftw111. [Google Scholar] [CrossRef]
- Maciel-Fiuza, M.F.; Muller, G.C.; Campos, D.M.S.; do Socorro Silva Costa, P.; Peruzzo, J.; Bonamigo, R.R.; Veit, T.; Vianna, F.S.L. Role of Gut Microbiota in Infectious and Inflammatory Diseases. Front. Microbiol. 2023, 14, 1098386. [Google Scholar] [CrossRef]
- Ma, T.; Shen, X.; Shi, X.; Sakandar, H.A.; Quan, K.Y.; Li, Y.L.; Jin, H.; Kwok, L.Y.; Zhang, H.P.; Sun, Z.H. Targeting Gut Microbiota and Metabolism as the Major Probiotic Mechanism-An Evidence-based Review. Trends Food Sci. Technol. 2023, 138, 178–198. [Google Scholar] [CrossRef]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The Gut Microbiota and Host Health: A New Clinical Frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef] [PubMed]
- Nikola, L.; Iva, L. Gut Microbiota as a Modulator of Type 1 Diabetes: A Molecular Perspective. Life Sci. 2024, 359, 123187. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.X.; Tian, L.M.; Fabi, J.P.; de Vos, P. The Potential of Prebiotics, Probiotics, and Synbiotics for Ameliorating Intestinal Barrier Dysfunction and Modulating Inflammatory Responses as Dietary Supplements in Diabetes Mellitus Management. Food Biosci. 2025, 72, 107539. [Google Scholar] [CrossRef]
- Li, B.Y.; Xu, X.Y.; Gan, R.Y.; Sun, Q.C.; Meng, J.M.; Shang, A.; Mao, Q.Q.; Li, H.B. Targeting Gut Microbiota for the Prevention and Management of Diabetes Mellitus by Dietary Natural Products. Foods 2019, 8, 440. [Google Scholar] [CrossRef]
- Zygmunt, A.E.; Drabczyk, M.; Karoń, K.; Karoń, Ł.M.; Grabowski, W.; Pedrycz, D.; Drapała, G.; Pedrycz, E.; Karoń, S. Gut Microbiota and Insulin Resistance: Mechanisms, Therapeutic Strategies, and Future Directions. J. Educ. Health Sport. 2025, 80, 58309. [Google Scholar] [CrossRef]
- Fliegerova, K.O.; Mahayri, T.M.; Sechovcova, H.; Mekadim, C.; Mrazek, J.; Jarosikova, R.; Dubsky, M.; Fejfarova, V. Diabetes and Gut Microbiome. Front. Microbiol. 2024, 15, 1451054. [Google Scholar] [CrossRef]
- Chen, L.; Chen, R.; Wang, H.; Liang, F. Mechanisms Linking Inflammation to Insulin Resistance. Int. J. Endocrinol. 2015, 2015, 508409. [Google Scholar] [CrossRef]
- Lyv, Z.Y.; Wang, L.J.; Xu, M.X.; Bai, X.F.; Cao, L.J. Association between the Structure of Intestinal Flora and Inflammatory Response in Children with Sepsis: A Prospective Cohort Study. Chin. J. Contemp. Pediatr. 2024, 26, 567–574. [Google Scholar] [CrossRef]
- Chen, X.; Shi, S.; Sun, C.; Li, S. A Study of the Relationship between Inflammatory Immune Function and Intestinal Flora in Adolescent Patients with First-Episode Depression. Actas Esp. Psiquiatr. 2024, 52, 1–9. [Google Scholar]
- Shen, Y.; Fan, N.; Ma, S.X.; Cheng, X.; Yang, X.; Wang, G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm 2025, 6, e70168. [Google Scholar] [CrossRef]
- Paray, B.A.; Albeshr, M.F.; Jan, A.T.; Rather, I.A. Leaky Gut and Autoimmunity: An Intricate Balance in Individuals Health and the Diseased State. Int. J. Mol. Sci. 2020, 21, 9770. [Google Scholar] [CrossRef]
- Vich Vila, A.; Imhann, F.; Collij, V.; Jankipersadsing, S.A.; Gurry, T.; Mujagic, Z.; Kurilshikov, A.; Bonder, M.J.; Jiang, X.; Tigchelaar, E.F.; et al. Gut microbiota Composition and Functional Changes in Inflammatory Bowel Disease and Irritable Bowel Syndrome. Sci. Transl. Med. 2018, 10, eaap8914. [Google Scholar] [CrossRef]
- Tatiya-Aphiradee, N.; Chatuphonprasert, W.; Jarukamjorn, K. Immune Response and Inflammatory Pathway of Ulcerative Colitis. J. Basic Clin. Physiol. Pharmacol. 2018, 30, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Al-Bayati, L.; Nayeri Fasaei, B.; Merat, S.; Bahonar, A.; Ghoddusi, A. Quantitative Analysis of the Three Gut Microbiota in UC and Non-UC Patients Using Real-time PCR. Microb. Pathog. 2023, 181, 106198. [Google Scholar] [CrossRef]
- Zhang, J.; Chen, S.L.; Li, L.B. Correlation between Intestinal Flora and Serum Inflammatory Factors in Patients with Crohn’s Disease. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4913–4917. [Google Scholar] [PubMed]
- Zhou, G.; Yang, J. Correlations of Gastrointestinal Hormones with Inflammation and Intestinal Flora in Patients with Gastric Cancer. J. BUON 2019, 24, 1595–1600. [Google Scholar]
- Li, L.; Yan, Q.; Ma, N.; Chen, X.; Li, G.; Liu, M. Analysis of Intestinal Flora and Inflammatory Cytokine Levels in Children with Non-infectious Diarrhea. Transl. Pediatr. 2021, 10, 1340–1345. [Google Scholar] [CrossRef]
- Wang, S.S.; Li, X.H.; Liu, P.; Li, J.; Liu, L. The Relationship between Alzheimer’s Disease and Intestinal Microflora Structure and Inflammatory Factors. Front. Aging Neurosci. 2022, 14, 972982. [Google Scholar] [CrossRef]
- Liu, B.; Ding, Z.; Xiong, J.; Heng, X.; Wang, H.; Chu, W. Gut Microbiota and Inflammatory Cytokine Changes in Patients with Ankylosing Spondylitis. Biomed. Res. Int. 2022, 2022, 1005111. [Google Scholar] [CrossRef]
- Zhang, J.; Wang, C.; Wang, J.; Zhang, F. Relationship between Intestinal Flora and Inflammatory Factors in Patients with Nonalcoholic Steatohepatitis. Exp. Ther. Med. 2018, 15, 723–726. [Google Scholar] [CrossRef]
- Zhang, Y.; Li, T.; Yuan, H.; Pan, W.; Dai, Q. Correlations of Inflammatory Factors with Intestinal Flora and Gastrointestinal Incommensurate Symptoms in Children with Asthma. Med. Sci. Monit. 2018, 24, 7975–7979. [Google Scholar] [CrossRef]
- Wang, X.; Peng, J.; Cai, P.; Xia, Y.; Yi, C.; Shang, A.; Akanyibah, F.A.; Mao, F. The Emerging Role of the Gut Microbiota and Its Application in Inflammatory Bowel Disease. Biomed. Pharmacother. 2024, 179, 117302. [Google Scholar] [CrossRef]
- Mahgoup, E.M. Gut Microbiota as a Therapeutic Target for Hypertension: Challenges and Insights for Future Clinical Applications Gut Microbiota and Hypertension Therapy. Curr. Hypertens. Rep. 2025, 27, 14. [Google Scholar] [CrossRef]
- Anwer, E.K.E.; Ajagbe, M.; Sherif, M.; Musaibah, A.S.; Mahmoud, S.; ElBanbi, A.; Abdelnaser, A. Gut Microbiota Secondary Metabolites: Key Roles in GI Tract Cancers and Infectious Diseases. Biomedicines 2025, 13, 100. [Google Scholar] [CrossRef] [PubMed]
- Yuan, L.; Li, Y.; Chen, M.; Xue, L.; Wang, J.; Ding, Y.; Gu, Q.; Zhang, J.; Zhao, H.; Xie, X.; et al. Therapeutic Applications of Gut Microbes in Cardiometabolic Diseases: Current State and Perspectives. Appl. Microbiol. Biotechnol. 2024, 108, 156. [Google Scholar] [CrossRef]
- Atarashi, K.; Suda, W.; Luo, C.; Kawaguchi, T.; Motoo, I.; Narushima, S.; Kiguchi, Y.; Yasuma, K.; Watanabe, E.; Tanoue, T.; et al. Ectopic Colonization of Oral Bacteria in the Intestine Drives T(H)1 Cell Induction and Inflammation. Science 2017, 358, 359–365. [Google Scholar] [CrossRef]
- Verma, R.; Lee, C.; Jeun, E.J.; Yi, J.; Kim, K.S.; Ghosh, A.; Byun, S.; Lee, C.G.; Kang, H.J.; Kim, G.C.; et al. Cell Surface Polysaccharides of Bifidobacterium bifidum Induce the Generation of Foxp3(+) Regulatory T Cells. Sci. Immunol. 2018, 3, eaat6975. [Google Scholar] [CrossRef]
- Hickey, A.; Stamou, P.; Udayan, S.; Ramón-Vázquez, A.; Esteban-Torres, M.; Bottacini, F.; Woznicki, J.A.; Hughes, O.; Melgar, S.; Ventura, M.; et al. Bifidobacterium breve Exopolysaccharide Blocks Dendritic Cell Maturation and Activation of CD4+ T Cells. Front. Microbiol. 2021, 12, 653587. [Google Scholar] [CrossRef]
- Turroni, F.; Serafini, F.; Foroni, E.; Duranti, S.; Motherway, M.O.; Taverniti, V.; Mangifesta, M.; Milani, C.; Viappiani, A.; Roversi, T.; et al. Role of Sortase-dependent Pili of Bifidobacterium bifidum PRL2010 in Modulating Bacterium-host Interactions. Proc. Natl. Acad. Sci. USA 2013, 110, 11151–11156. [Google Scholar] [CrossRef]
- Brown, E.M.; Kenny, D.J.; Xavier, R.J. Gut Microbiota Regulation of T Cells During Inflammation and Autoimmunity. Annu. Rev. Immunol. 2019, 37, 599–624. [Google Scholar] [CrossRef]
- Nagashima, K. Blockbuster T cells in the gut: A High-resolution View of Immune Modulation by the Gut Microbiome Is Presented. Science 2024, 385, 36–37. [Google Scholar] [CrossRef]
- Li, Z.; Xiong, W.; Liang, Z.; Wang, J.; Zeng, Z.; Kolat, D.; Li, X.; Zhou, D.; Xu, X.; Zhao, L. Critical Role of the Gut Microbiota in Immune Responses and Cancer Immunotherapy. J. Hematol. Oncol. 2024, 17, 33. [Google Scholar] [CrossRef]
- Takeuchi, T.; Nakanishi, Y.; Ohno, H. Microbial Metabolites and Gut Immunology. Annu. Rev. Immunol. 2024, 42, 153–178. [Google Scholar] [CrossRef]
- Guzior, D.V.; Quinn, R.A. Review: Microbial Transformations of Human Bile Acids. Microbiome 2021, 9, 140. [Google Scholar] [CrossRef]
- Candelli, M.; Franza, L.; Pignataro, G.; Ojetti, V.; Covino, M.; Piccioni, A.; Gasbarrini, A.; Franceschi, F. Interaction between Lipopolysaccharide and Gut Microbiota in Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2021, 22, 6242. [Google Scholar] [CrossRef]
- Mann, E.R.; Lam, Y.K.; Uhlig, H.H. Short-chain Fatty Acids: Linking Diet, the Microbiome and Immunity. Nat. Rev. Immunol. 2024, 24, 577–595. [Google Scholar] [CrossRef]
- van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol. 2021, 29, 700–712. [Google Scholar] [CrossRef] [PubMed]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal Microbe-derived Butyrate Induces the Differentiation of Colonic Regulatory T Cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Wu, W.; Sun, M.; Chen, F.; Cao, A.T.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota Metabolite Short-Chain Fatty Acid Acetate Promotes Intestinal IgA Response to Microbiota Which Is Mediated by GPR43. Mucosal Immunol. 2017, 10, 946–956. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal Microbiota-derived Short-chain Fatty Acids Regulation of Immune Cell IL-22 Production and Gut Immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
- Dodd, D.; Spitzer, M.H.; Van Treuren, W.; Merrill, B.D.; Hryckowian, A.J.; Higginbottom, S.K.; Le, A.; Cowan, T.M.; Nolan, G.P.; Fischbach, M.A.; et al. A Gut Bacterial Pathway Metabolizes Aromatic Amino Acids into Nine Circulating Metabolites. Nature 2017, 551, 648–652. [Google Scholar] [CrossRef]
- Roager, H.M.; Licht, T.R. Microbial Tryptophan Catabolites in Health and Disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
- Renga, G.; Nunzi, E.; Pariano, M.; Puccetti, M.; Bellet, M.M.; Pieraccini, G.; D’Onofrio, F.; Santarelli, I.; Stincardini, C.; Aversa, F.; et al. Optimizing Therapeutic Outcomes of Immune Checkpoint Blockade by a Microbial Tryptophan Metabolite. J. Immunother. Cancer 2022, 10, e003725. [Google Scholar] [CrossRef] [PubMed]
- Riazati, N.; Kable, M.E.; Newman, J.W.; Adkins, Y.; Freytag, T.; Jiang, X.; Stephensen, C.B. Associations of Microbial and Indoleamine-2,3-dioxygenase-derived Tryptophan Metabolites with Immune Activation in Healthy Adults. Front. Immunol. 2022, 13, 917966. [Google Scholar] [CrossRef]
- Li, X.; Lu, C.; Fan, D.; Lu, X.; Xia, Y.; Zhao, H.; Xu, H.; Zhu, Y.; Li, J.; Liu, H.; et al. Human Umbilical Mesenchymal Stem Cells Display Therapeutic Potential in Rheumatoid Arthritis by Regulating Interactions Between Immunity and Gut Microbiota via the Aryl Hydrocarbon Receptor. Front. Cell Dev. Biol. 2020, 8, 131. [Google Scholar] [CrossRef]
- Liu, M.; Nieuwdorp, M.; de Vos, W.M.; Rampanelli, E. Microbial Tryptophan Metabolism Tunes Host Immunity, Metabolism, and Extraintestinal Disorders. Metabolites 2022, 12, 834. [Google Scholar] [CrossRef]
- Li, K.Y.; Hao, Z.H.; Du, J.Y.; Gao, Y.M.; Yang, S.Y.; Zhou, Y.L. Bacteroides thetaiotaomicron Relieves Colon Inflammation by Activating Aryl Hydrocarbon Receptor and Modulating CD4(+) T Cell Homeostasis. Int. Immunopharmacol. 2021, 90, 107183. [Google Scholar] [CrossRef]
- Dias, I.C.D.; Carabelli, B.; Ishii, D.K.; de Morais, H.; de Carvalho, M.C.; de Souza, L.E.R.; Zanata, S.M.; Brandao, M.L.; Cunha, T.M.; Ferraz, A.C.; et al. Indoleamine-2,3-Dioxygenase/Kynurenine Pathway as a Potential Pharmacological Target to Treat Depression Associated with Diabetes. Mol. Neurobiol. 2016, 53, 6997–7009. [Google Scholar] [CrossRef]
- Perino, A.; Schoonjans, K. Metabolic Messengers: Bile Acids. Nat. Metab. 2022, 4, 416–423. [Google Scholar] [CrossRef]
- Hang, S.; Paik, D.; Yao, L.; Kim, E.; Trinath, J.; Lu, J.; Ha, S.; Nelson, B.N.; Kelly, S.P.; Wu, L.; et al. Bile Acid Metabolites Control T(H)17 and T(reg) Cell Differentiation. Nature 2019, 576, 143–148. [Google Scholar] [CrossRef]
- Jia, W.; Xie, G.; Jia, W. Bile Acid-microbiota Crosstalk in Gastrointestinal Inflammation and Carcinogenesis. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 111–128. [Google Scholar] [CrossRef]
- Sanchez-Jimenez, F.; Medina, M.A.; Villalobos-Rueda, L.; Urdiales, J.L. Polyamines in Mammalian Pathophysiology. Cell Mol. Life Sci. 2019, 76, 3987–4008. [Google Scholar] [CrossRef]
- Carriche, G.M.; Almeida, L.; Stuve, P.; Velasquez, L.; Dhillon-LaBrooy, A.; Roy, U.; Lindenberg, M.; Strowig, T.; Plaza-Sirvent, C.; Schmitz, I.; et al. Regulating T-cell Differentiation through the Polyamine Spermidine. J. Allergy Clin. Immunol. 2021, 147, 335–348.e11. [Google Scholar] [CrossRef]
- Chen, Y.; Ye, X.; Escames, G.; Lei, W.; Zhang, X.; Li, M.; Jing, T.; Yao, Y.; Qiu, Z.; Wang, Z.; et al. The NLRP3 Inflammasome: Contributions to Inflammation-related Diseases. Cell. Mol. Biol. Lett. 2023, 28, 51. [Google Scholar] [CrossRef]
- Bai, H.; Zhang, Q. Activation of NLRP3 Inflammasome and Onset of Alzheimer’s Disease. Front. Immunol. 2021, 12, 701282. [Google Scholar] [CrossRef] [PubMed]
- Ren, W.; Sun, Y.; Zhao, L.; Shi, X. NLRP3 Inflammasome and Its Role in Autoimmune Diseases: A Promising Therapeutic Target. Biomed. Pharmacother. 2024, 175, 116679. [Google Scholar] [CrossRef]
- Kang, S.S.; Sim, J.R.; Yun, C.H.; Han, S.H. Lipoteichoic Acids As a Major Virulence Factor Causing Inflammatory Responses via Toll-like Receptor 2. Arch. Pharm. Res. 2016, 39, 1519–1529. [Google Scholar] [CrossRef]
- Mukherjee, T.; Hovingh, E.S.; Foerster, E.G.; Abdel-Nour, M.; Philpott, D.J.; Girardin, S.E. NOD1 and NOD2 in Inflammation, Immunity and Disease. Arch. Biochem. Biophys. 2019, 670, 69–81. [Google Scholar] [CrossRef]
- Wolf, A.J.; Underhill, D.M. Peptidoglycan Recognition by the Innate Immune System. Nat. Rev. Immunol. 2018, 18, 243–254. [Google Scholar] [CrossRef]
- Wen, M.; Wang, J.; Ou, Z.; Nie, G.; Chen, Y.; Li, M.; Wu, Z.; Xiong, S.; Zhou, H.; Yang, Z.; et al. Bacterial Extracellular Vesicles: A Position Paper by the Microbial Vesicles Task Force of the Chinese Society for Extracellular Vesicles. Interdiscip. Med. 2023, 1, e20230017. [Google Scholar] [CrossRef]
- Shi, Y.; Zheng, Z.; Wang, W.; Hu, H. Harnessing the Therapeutic Potential of Bacterial Extracellular Vesicles via Functional Peptides. Interdiscip. Med. 2025, 3, e20240125. [Google Scholar] [CrossRef]
- Xie, J.H.; Li, Q.Q.; Nie, S.P. Bacterial Extracellular Vesicles: An Emerging Postbiotic. Trends Food Sci. Technol. 2024, 143, 104275. [Google Scholar] [CrossRef]
- Gubatan, J.; Holman, D.R.; Puntasecca, C.J.; Polevoi, D.; Rubin, S.J.S.; Rogalla, S. Antimicrobial Peptides and the Gut Microbiome in Inflammatory Bowel Disease. World J. Gastroent. 2021, 27, 7402–7422. [Google Scholar] [CrossRef]
- Nuding, S.; Frasch, T.; Schaller, M.; Stange, E.F.; Zabel, L.T. Synergistic Effects of Antimicrobial Peptides and Antibiotics against. Antimicrob. Agents Chemother. 2014, 58, 5719–5725. [Google Scholar] [CrossRef]
- Tougaard, P.; Skov, S.; Pedersen, A.E.; Krych, L.; Nielsen, D.S.; Bahl, M.I.; Christensen, E.G.; Licht, T.R.; Poulsen, S.S.; Metzdorff, S.B.; et al. TL1A Regulates TCRγδ+ Intraepithelial Lymphocytes and Gut Microbial Composition. Eur. J. Immunol. 2015, 45, 865–875. [Google Scholar] [CrossRef]
- Le Bourhis, L.; Martin, E.; Péguillet, I.; Guihot, A.; Froux, N.; Coré, M.; Lévy, E.; Dusseaux, M.; Meyssonnier, V.; Premel, V.; et al. Antimicrobial Activity of Mucosal-associated Invariant T Cells. Nat. Immunol. 2010, 11, 701–708. [Google Scholar] [CrossRef]
- Hapil, F.Z.; Wingender, G. The Interaction between Invariant Natural Killer T Cells and the Mucosal Microbiota. Immunology 2018, 155, 164–175. [Google Scholar] [CrossRef]
- Saha, P.; Mell, B.; Golonka, R.M.; Bovilla, V.R.; Abokor, A.A.; Mei, X.; Yeoh, B.S.; Doris, P.A.; Gewirtz, A.T.; Joe, B.; et al. Selective IgA Deficiency in Spontaneously Hypertensive Rats with Gut Dysbiosis. Hypertension 2022, 79, 2239–2249. [Google Scholar] [CrossRef]
- Becattini, S.; Taur, Y.; Pamer, E.G. Antibiotic-Induced Changes in the Intestinal Microbiota and Disease. Trends Mol. Med. 2016, 22, 458–478. [Google Scholar] [CrossRef]
- Kim, S.; Covington, A.; Pamer, E.G. The Intestinal Microbiota: Antibiotics, Colonization Resistance, and Enteric Pathogens. Immunol. Rev. 2017, 279, 90–105. [Google Scholar] [CrossRef]
- Ianiro, G.; Mullish, B.H.; Kelly, C.R.; Kassam, Z.; Kuijper, E.J.; Ng, S.C.; Iqbal, T.H.; Allegretti, J.R.; Bibbò, S.; Sokol, H.; et al. Reorganisation of Faecal Microbiota Transplant Services during the COVID-19 Pandemic. Gut 2020, 69, 1555–1563. [Google Scholar] [CrossRef]
- Duan, H.; Yu, L.L.; Tian, F.W.; Zhai, Q.X.; Fan, L.P.; Chen, W. Antibiotic-induced Gut Dysbiosis and Barrier Disruption and the Potential Protective Strategies. Crit. Rev. Food Sci. Nutr. 2022, 62, 1427–1452. [Google Scholar] [CrossRef]
- Carson, D.; Barry, R.; Hopkins, E.G.D.; Roumeliotis, T.I.; García-Weber, D.; Mullineaux-Sanders, C.; Elinav, E.; Arrieumerlou, C.; Choudhary, J.S.; Frankel, G. Citrobacter Rodentium Induces Rapid and Unique Metabolic and Inflammatory Responses in Mice Suffering from Severe Disease. Cell. Microbiol. 2020, 22, e13126. [Google Scholar] [CrossRef]
- Palleja, A.; Mikkelsen, K.H.; Forslund, S.K.; Kashani, A.; Allinm, K.H.; Nielsen, T.; Hansen, T.H.; Liang, S.S.; Feng, Q.; Zhang, C.C.; et al. Recovery of Gut Microbiota of Healthy Adults following Antibiotic Exposure. Nat. Microbiol. 2018, 3, 1255–1265. [Google Scholar] [CrossRef] [PubMed]
- Korpela, K.; Salonen, A.; Virta, L.J.; Kekkonen, R.A.; Forslund, K.; Bork, P.; de Vos, W.M. Intestinal Microbiome Is Related to Lifetime Antibiotic Use in Finnish Pre-school Children. Nat. Commun. 2016, 7, 10410. [Google Scholar] [CrossRef]
- Halawa, E.M.; Fadel, M.; Al-Rabia, M.W.; Behairy, A.; Nouh, N.A.; Abdo, M.; Olga, R.; Fericean, L.; Atwa, A.M.; El-Nablaway, M.; et al. Antibiotic Action and Resistance: Updated Review of Mechanisms, Spread, Influencing Factors, and Alternative Approaches for Combating Resistance. Front. Pharmacol. 2023, 14, 1305294. [Google Scholar] [CrossRef]
- Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
- Dongre, D.S.; Saha, U.B.; Saroj, S.D. Exploring the Role of Gut Microbiota in Antibiotic Resistance and Prevention. Ann. Med. 2025, 57, 2478317. [Google Scholar] [CrossRef]
- Wuethrich, I.; Pelzer, B.W.; Khodamoradi, Y.; Vehreschild, M.J. The Role of the Human Gut Microbiota in Colonization and Infection with Multidrug-resistant Bacteria. Gut Microbes 2021, 13, 1911279. [Google Scholar] [CrossRef]
- Moura de Sousa, J.; Lourenco, M.; Gordo, I. Horizontal Gene Transfer Among Host-associated Microbes. Cell Host Microbe 2023, 31, 513–527. [Google Scholar] [CrossRef]
- Zhang, N.; Liu, J.; Chen, Z.; Chen, N.; Gu, F.; He, Q. Integrated Analysis of the Alterations in Gut Microbiota and Metabolites of Mice Induced After Long-Term Intervention with Different Antibiotics. Front. Microbiol. 2022, 13, 832915. [Google Scholar] [CrossRef]
- Sun, L.; Zhang, X.; Zhang, Y.; Zheng, K.; Xiang, Q.; Chen, N.; Chen, Z.; Zhang, N.; Zhu, J.; He, Q. Antibiotic-Induced Disruption of Gut Microbiota Alters Local Metabolomes and Immune Responses. Front. Cell. Infect. Microbiol. 2019, 9, 99. [Google Scholar] [CrossRef]
- Gao, H.; Shu, Q.; Chen, J.; Fan, K.; Xu, P.; Zhou, Q.; Li, C.; Zheng, H. Antibiotic Exposure Has Sex-Dependent Effects on the Gut Microbiota and Metabolism of Short-Chain Fatty Acids and Amino Acids in Mice. mSystems 2019, 4, 10–1128. [Google Scholar] [CrossRef]
- Wu, Y.; Tang, X.; Hu, F.; Zhu, T.; Liu, H.; Xiong, Y.; Zuo, X.; Xu, A.; Zhuang, X. Long-term Use of Broad-spectrum Antibiotics Affects Ly6C(hi) Monocyte Recruitment and IL-17A and IL-22 Production through the Gut Microbiota in Tumor-bearing Mice Treated with Chemotherapy. Immunol. Res. 2022, 70, 829–843. [Google Scholar] [CrossRef]
- Gao, W.; Liu, X.; Zhang, S.; Wang, J.; Qiu, B.; Shao, J.; Huang, W.; Huang, Y.; Yao, M.; Tang, L.L. Alterations in Gut Microbiota and Inflammatory Cytokines after Administration of Antibiotics in Mice. Microbiol. Spectr. 2024, 12, e0309523. [Google Scholar] [CrossRef]
- Wang, J.; Xiang, Q.; Gu, S.; Gu, Y.; Yao, M.; Huang, W.; Gao, W.; Tang, L.L. Short- and Long-Term Effects of Different Antibiotics on the Gut Microbiota and Cytokines Level in Mice. Infect. Drug Resist. 2022, 15, 6785–6797. [Google Scholar] [CrossRef]
- Si, W.; Zhao, X.; Li, R.; Li, Y.; Ma, C.; Zhao, X.; Bugno, J.; Qin, Y.; Zhang, J.; Liu, H.; et al. Lactobacillus rhamnosus GG Induces STING-dependent IL-10 in Intestinal Monocytes and Alleviates Inflammatory Colitis in Mice. J. Clin. Investig. 2025, 135, e174910. [Google Scholar] [CrossRef]
- Ganguly, N.K.; Bhattacharya, S.K.; Sesikeran, B.; Nair, G.B.; Ramakrishna, B.S.; Sachdev, H.P.S.; Batish, V.K.; Kanagasabapathy, A.S.; Muthuswamy, V.; Kathuria, S.C.; et al. ICMR-DBT Guidelines for Evaluation of Probiotics in Food. Indian J. Med. Res. 2011, 134, 22–25. [Google Scholar]
- Abdollahi, S.; Mehrabian, S.; Pourshafie, M.R.; Rohani, M. The Preventive and Therapeutic Role of Native Probiotic Lactobacillus Species in Controlling Inflammation. Folia Microbiol. 2025, 3, 1–9. [Google Scholar] [CrossRef]
- Jouriani, F.H.; Torkamaneh, M.; Torfeh, M.; Ashrafian, F.; Aghamohammad, S.; Rohani, M. Native Lactobacillus and Bifidobacterium Probiotics Modulate Autophagy Genes and Exert Anti-inflammatory Effect. Sci. Rep. 2025, 15, 25006. [Google Scholar] [CrossRef]
- Mishra, V.H.; Gupta, P.; Khade, A.M.; Deshkar, A.T. Use of Probiotics in Managing Gastrointestinal, Metabolic, and Immune-related Diseases. J. Datta Meghe Inst. Med. Sci. Univ. 2025, 20, 214–217. [Google Scholar] [CrossRef]
- Dicks, L.M.T.; Geldenhuys, J.; Mikkelsen, L.S.; Brandsborg, E.; Marcotte, H. Our Gut Microbiota: A Long Walk to Homeostasis. Benef. Microbes 2018, 9, 3–19. [Google Scholar] [CrossRef]
- Hu, S.L.; Wang, L.; Jiang, Z.Y. Dietary Additive Probiotics Modulation of the Intestinal Microbiota. Protein Pept. Lett. 2017, 24, 382–387. [Google Scholar] [CrossRef]
- Monteagudo-Mera, A.; Rastall, R.A.; Gibson, G.R.; Charalampopoulos, D.; Chatzifragkou, A. Adhesion Mechanisms Mediated by Probiotics and Prebiotics and Their Potential Impact on Human Health. Appl. Microbiol. Biotechnol. 2019, 103, 6463–6472. [Google Scholar] [CrossRef] [PubMed]
- Bu, Z.C.; Xia, Y.J.; Ai, L.Z.; Xiong, Z.Q.; Song, X.; Wang, G.Q. Synthetic Pathway and Functional Study of Short Chain Fatty Acids in Probiotics. Food Ferment. Ind. 2022, 48, 286–291, 302. [Google Scholar]
- da Silva, M.F.; de Lima, M.D.S.F.; Converti, A. Effect of Short-Chain Fatty Acids Produced by Probiotics. In Lactic Acid Bacteria; CRC Press: Boca Raton, FL, USA, 2020; pp. 124–141. [Google Scholar]
- Anjana, A.; Tiwari, S.K. Bacteriocin-Producing Probiotic Lactic Acid Bacteria in Controlling Dysbiosis of the Gut Microbiota. Front. Cell. Infect. Microbiol. 2022, 12, 851140. [Google Scholar] [CrossRef]
- Wang, X.Z.; Zhang, P.; Zhang, X. Probiotics Regulate Gut Microbiota: An Effective Method to Improve Immunity. Molecules 2021, 26, 6076. [Google Scholar] [CrossRef]
- Shvets, Y.; Khranovska, N.; Senchylo, N.; Ostapchenko, D.; Tymoshenko, I.; Onysenko, S.; Kobyliak, N.; Falalyeyeva, T. Microbiota Substances Modulate Dendritic Cells Activity: A Critical View. Heliyon 2024, 10, e27125. [Google Scholar] [CrossRef]
- Zheng, D.Z.; Wang, Z.R.; Sui, L.; Xu, Y.G.; Wang, L.; Qiao, X.Y.; Cui, W.; Jiang, Y.P.; Zhou, H.; Tang, L.J.; et al. Lactobacillus johnsonii Activates Porcine Monocyte Derived Dendritic Cells Maturation to Modulate Th Cellular Immune Response. Cytokine 2021, 144, 155581. [Google Scholar] [CrossRef]
- Mazziotta, C.; Tognon, M.; Martini, F.; Torreggiani, E.; Rotondo, J.C. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023, 12, 184. [Google Scholar] [CrossRef]
- Sun, Y.H.; Sun, Z.; Fang, B.; Wang, R.; Liu, Y.; Li, J.Y.; Lan, H.L.; Zhao, W.; Hung, W.L.; Zhang, M. Exploring the Anti-inflammatory Potential of Lacticaseibacillus paracasei Postbiotics: Mechanistic Insights and Functional Components. Food Biosci. 2025, 65, 106105. [Google Scholar] [CrossRef]
- Zhou, L.Y.; Liu, D.Y.; Xie, Y.; Yao, X.J.; Li, Y. Bifidobacterium infantis Induces Protective Colonic PD-L1 and Foxp3 Regulatory T Cells in an Acute Murine Experimental Model of Inflammatory Bowel Disease. Gut Liver 2019, 13, 430–439. [Google Scholar] [CrossRef]
- Wan, C.; Qian, W.W.; Liu, W.; Pi, X.G.; Tang, M.T.; Wang, X.L.; Gu, Q.; Li, P.; Zhou, T. Exopolysaccharide from Lactobacillus rhamnosus ZFM231 alleviates DSS-induced colitis in mice by regulating gut microbiota. J. Sci. Food Agric. 2022, 102, 7087–7097. [Google Scholar] [CrossRef]
- Caviglia, G.P.; Tucci, A.; Pellicano, R.; Fagoonee, S.; Rosso, C.; Abate, M.L.; Olivero, A.; Armandi, A.; Vanni, E.; Saracco, G.M.; et al. Clinical Response and Changes of Cytokines and Zonulin Levels in Patients with Diarrhoea-Predominant Irritable Bowel Syndrome Treated with Bifidobacterium Longum ES1 for 8 or 12 Weeks: A Preliminary Report. J. Clin. Med. 2020, 9, 2353. [Google Scholar] [CrossRef]
- Jiang, S.; Hou, Y.; Meng, L.; Pu, X.; Zhu, X.; Tuo, Y.; Qian, F.; Mu, G. Effect of Lactiplantibacillus plantarum HM-22 on ImmunoRegulation and Intestinal Microbiota in Alpha-lactalbumin-induced Allergic Mice. Food Funct. 2021, 12, 8887–8898. [Google Scholar] [CrossRef] [PubMed]
- Prakoeswa, C.R.S.; Bonita, L.; Karim, A.; Herwanto, N.; Umborowati, M.A.; Setyaningrum, T.; Hidayati, A.N.; Surono, I.S. Beneficial Effect of Lactobacillus plantarum IS-10506 Supplementation in Adults with Atopic Dermatitis: A Randomized Controlled Trial. J. Dermatol. Treat. 2022, 33, 1491–1498. [Google Scholar] [CrossRef]
- Magistrelli, L.; Amoruso, A.; Mogna, L.; Graziano, T.; Cantello, R.; Pane, M.; Comi, C. Probiotics May Have Beneficial Effects in Parkinson’s Disease: In vitro Evidence. Front. Immunol. 2019, 10, 969. [Google Scholar] [CrossRef]
- Visñuk, D.P.; de Giori, G.S.; LeBlanc, J.G.; de LeBlanc, A.D. Neuroprotective Effects Associated with Immune Modulation by Selected Lactic Acid Bacteria in a Parkinson’s Disease Model. Nutrition 2020, 79-80, 110995. [Google Scholar] [CrossRef]
- Hu, T.T.; Fan, Y.; Long, X.Y.; Pan, Y.N.; Mu, J.F.; Tan, F.; Zhao, X. Protective Effect of Lactobacillus plantarum YS3 on Dextran Sulfate Sodium-induced Colitis in C57BL/6J Mice. J. Food Biochem. 2021, 45, e13632. [Google Scholar] [CrossRef]
- Bai, L.; Gao, M.; Cheng, X.; Kang, G.; Cao, X.; Huang, H. Engineered Butyrate-Producing Bacteria Prevents High Fat Diet-induced Obesity in Mice. Microb. Cell Fact. 2020, 19, 94. [Google Scholar] [CrossRef]
- Wang, L.; Cheng, X.; Bai, L.; Gao, M.; Kang, G.; Cao, X.; Huang, H. Positive Interventional Effect of Engineered Butyrate-Producing Bacteria on Metabolic Disorders and Intestinal Flora Disruption in Obese Mice. Microbiol. Spectr. 2022, 10, e0114721. [Google Scholar] [CrossRef]
- Zhang, L.; Chen, N.; Chen, H.; Tang, C.; Wang, J.; Wang, Y.; Zhang, Y.; Guo, H.; Yuan, J. Recent Advances of Engineered Probiotics for Therapeutic Applications. BioDesign Res. 2025, 7, 100039. [Google Scholar] [CrossRef]
- Huang, H.; Yin, H.; Zhang, X. Editorial: Engineering Probiotics for Multiple Interventions on Intestinal Diseases. Front. Cell Infect. Microbiol. 2023, 13, 1138998. [Google Scholar] [CrossRef]
- Gurbatri, C.R.; Arpaia, N.; Danino, T. Engineering Bacteria as Interactive Cancer Therapies. Science 2022, 378, 858–864. [Google Scholar] [CrossRef]
- Praveschotinunt, P.; Duraj-Thatte, A.M.; Gelfat, I.; Bahl, F.; Chou, D.B.; Joshi, N.S. Engineered E. coli Nissle 1917 for the Delivery of Matrix-tethered Therapeutic Domains to the Gut. Nat. Commun. 2019, 10, 5580. [Google Scholar] [CrossRef] [PubMed]
- Yan, X.; Liu, X.Y.; Zhang, D.; Zhang, Y.D.; Li, Z.H.; Liu, X.; Wu, F.; Chen, G.Q. Construction of a Sustainable 3-hydroxybutyrate-producing Probiotic Escherichia coli for Treatment of Colitis. Cell. Mol. Immunol. 2021, 18, 2344–2357. [Google Scholar] [CrossRef]
- Al-Habsi, N.; Al-Khalili, M.; Haque, S.A.; Elias, M.; Olqi, N.A.; Al Uraimi, T. Health Benefits of Prebiotics, Probiotics, Synbiotics, and Postbiotics. Nutrients 2024, 16, 3955. [Google Scholar] [CrossRef]
- Erhardt, R.; Harnett, J.E.; Steels, E.; Steadman, K.J. Functional Constipation and the Effect of Prebiotics on the Gut Microbiota: A Review. Br. J. Nutr. 2023, 130, 1015–1023. [Google Scholar] [CrossRef]
- Levi, Y.; Novais, G.S.; Dias, R.B.; Andraus, R.A.C.; Messora, M.R.; Neto, H.B.; Ervolino, E.; Santinoni, C.S.; Maia, L.P. Effects of the Prebiotic Mannan Oligosaccharide on the Experimental Periodontitis in Rats. J. Clin. Periodontol. 2018, 45, 1078–1089. [Google Scholar] [CrossRef]
- Xu, X.; Wu, J.; Jin, Y.; Huang, K.; Zhang, Y.; Liang, Z. Both Saccharomyces boulardii and Its Postbiotics Alleviate Dextran Sulfate Sodium-Induced Colitis in Mice, Association with Modulating Inflammation and Intestinal Microbiota. Nutrients 2023, 15, 1484. [Google Scholar] [CrossRef]
- Itoh, T.; Miyazono, D.; Sugata, H.; Mori, C.; Takahata, M. Anti-inflammatory Effects of Heat-killed Lactiplantibacillus argentoratensis BBLB001 on a Gut Inflammation Co-culture Cell Model and Dextran Sulfate Sodium-induced Colitis Mouse Model. Int. Immunopharmacol. 2024, 143, 113408. [Google Scholar] [CrossRef]
- De Marco, S.; Sichetti, M.; Muradyan, D.; Piccioni, M.; Traina, G.; Pagiotti, R.; Pietrella, D. Probiotic Cell-Free Supernatants Exhibited Anti-Inflammatory and Antioxidant Activity on Human Gut Epithelial Cells and Macrophages Stimulated with LPS. Evid. Based Complement. Altern. Med. 2018, 2018, 1756308. [Google Scholar] [CrossRef]
- Zhang, X.F.; Guan, X.X.; Tang, Y.J.; Sun, J.F.; Wang, X.K.; Wang, W.D.; Fan, J.M. Clinical Effects and Gut Microbiota Changes of Using Probiotics, Prebiotics or Synbiotics in Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Eur. J. Nutr. 2021, 60, 2855–2875. [Google Scholar] [CrossRef]
- Yadegar, A.; Bar-Yoseph, H.; Monaghan, T.M.; Pakpour, S.; Severino, A.; Kuijper, E.J.; Smits, W.K.; Terveer, E.M.; Neupane, S.; Nabavi-Rad, A.; et al. Fecal Microbiota Transplantation: Current Challenges and Future Landscapes. Clin. Microbiol. Rev. 2024, 37, e0006022. [Google Scholar] [CrossRef]
- Belvoncikova, P.; Maronek, M.; Gardlik, R. Gut Dysbiosis and Fecal Microbiota Transplantation in Autoimmune Diseases. Int. J. Mol. Sci. 2022, 23, 10729. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).