Review Article

Gut Microbiota Coordinates with the Host Immunity against Bacterial Infections

by Paola Cuomo1, Chiara Medaglia2, Antonio Gentile1, Rosanna Capparelli3*, Domenico Iannelli1

1Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy

2Functional Genomics Research Center, Fondazione Human Technopole, Milan, Italy

3Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy

*Corresponding author: Rosanna Capparelli, Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy

Received Date: 20 March, 2024

Accepted Date: 29 March, 2024

Published Date: 02 April, 2024

Citation: Cuomo P, Medaglia C, Gentile A, Capparelli R, Iannelli D (2024) Gut Microbiota Coordinates with the Host Immunity against Bacterial Infections. J Community Med Public Health 8: 424. https://doi.org/10.29011/2577-2228.100424

Abstract

The coexistence of mammals and gut microbiota is essential for the host health. In this context, microbial metabolites play a critical role. This review describes the intricate interplay between the gut microbiota and the host immune system, in order to provide further insights into alternative therapies, based on the host metabolism modulation, to counteract bacterial infections.

Keywords: Gut Microbial Metabolites; Host Immunity; Pathogens; Immunometabolism

Introduction

Human body is a complex ecosystem of human and microbial cells. Each person carries trillions of symbiotic microbes (bacteria, virus, fungi, archaea and protozoa) [1], which collectively constitute the microbiota [2]. This community is extremely variable among individuals [3] and this variability starts in utero (vertical or maternal transmission) and continues during and post-partum [4,5]. Natural or caesarean delivery, as well as microbe colonization in the first days of life (horizontal transmission), are critical for the establishment and development of mature microbiota.

Microbiota, especially gut microbiota, is often considered as an independent organ and a second genome, providing the host with 100-fold more genes [6,7]. These genes are involved in processing indigestible dietary polysaccharides and producing primary metabolites [8,9].

The interaction between microbial and human metabolites is particularly relevant in the context of the immune system regulation [10]. This crosstalk is indispensable for energy metabolism and inflammatory response [8,11], highlighting the essential role of microbiota in human health [12].

Given the importance of microbiota, it is not surprising that its dysregulation (dysbiosis) - due to environmental factors such as diet and drug consumption - can affect the host homeostasis promoting serious diseases (Obesity, Malnutrition, Chronic Inflammatory Diseases and Cancer) [13]. In addition, dysbiosis of gut microbiota has also been correlated with an imbalanced immune regulation against self and non-self-bacteria.

The host has developed mechanisms of tolerance and resistance that enable symbiotic bacteria to support the immune system without causing tissue damage. However, in specific scenarios, such as opportunistic invasion by commensal bacteria into non-native tissues, this delicate equilibrium can be disrupted, leading to systemic infections [10,14].

Infectious diseases are - at the moment - one of the most important challenges for the global public health. The inappropriate use of antibiotics in humans and animals have significantly contributed to this concern, generating Drug-Resistant Pathogens (DRPs) [15]. It is estimated that in 2050, DRPs will cause more than 10 million of deaths every year, surpassing cancer in the list of the major causes of death [16]. In this perspective, novel therapeutical interventions are imperative.

Developing strategies targeting metabolic pathways of the host immune cells, through modulation of microbial metabolites, could represent a promising approach.

This review aims at providing a lesson on the reciprocal dynamics between the gut microbiota and the host immune system, elucidating the role of microbial metabolites in fostering host immune cells, in an attempt to exploit these knowledges to improve human health and control infectious diseases.

Host-Microbiota Co-Evolution: Immune Tolerance

The mammalian immune system has evolved to protect the host from pathogens, while nurturing tolerance towards beneficial symbiotic microbes. This intricate process, named immune tolerance, relies on the difficult interplay between innate and adaptive immune responses, which coordinate their activities against pathogens, while preserving self-components [17].

Mucosae are the primary site for innate and adaptive immune regulation. They coordinate the host immune defense through a pool of lymphoid tissues and cells, which collectively constitute the Mucosa-Associated Lymphoid Tissue (MALT) [18].

The Gut-Associated Lymphoid Tissue (GALT) is the largest reservoir of mucosa-associated immune cells, including B and T cells, macrophages, plasma cells and Dendritic Cells (DCs) [19]. These cells serve to set up a barrier against harmful antigens and mount a fine-tuned immune response, which protects the host without altering the gut microbial ecosystem [20].

To initiate the mucosal immune response, GALT utilizes specialized non-immune cells named Microfold (M) cells, which represent 10% of Follicle-Associated Epithelial (FAE) cells found in Peyer’s Patches (PPs), vermiform appendix and Isolated Lymphoid Follicles (ILFs) [21,22]. M cells play an essential role in transporting gut lumen antigens to intestinal Dendritic Cells (DCs), through transcytosis, phagocytosis or forming pores in inter-epithelial tight junctions [23]. Microbial transcytosis has been proven to stimulate Lipopolysaccharide (LPS)-mediated IL-1 release - and consequently - proliferation of T and B lymphocytes and IgA production [20,24]. IgA antibodies are critical in mounting an effective mucosal immune response against pathogens [25]. Further, they also limit mucosal penetration by commensal microorganisms, ensuring a tolerogenic response [25,26].

Similar to pathogenic bacteria, commensal microbes are recognized by innate immune receptors [27]. However, unlike their pathogenic counterparts, commensal microorganisms do not provoke an inflammatory response [26]. In opposition, they stimulate the release of anti-inflammatory mediators such as IL-10 and TGF-β1, which help preserve the integrity of the intestinal epithelial cell barrier and maintain a stable intestinal microbial community, contributing to overall gut health [27]. Growing evidences also demonstrate the role of RORγt+ FoxP3+ peripherally derived T regulatory cells (pTregs) in tolerogenic response due to IL-10 production [28], suggesting IL-10 as a critical immunoregulator mediator (Figure 1). Lack of IL-10, in fact, has been documented to increase the risk for chronic enterocolitis [29].

Figure 1: The role of IL-10 in immune tolerance and bacterial infection resistance. Commensal microbiota stimulates Lipopolysaccharide- (LPS) or Short Chain Fatty Acid- (SCFAs) mediated IL-10 production in the intestinal lamina propria. IL-10 is primarily produced by dendritic cells, macrophages and FoxP3+ T regulatory cells (Tregs). Upon release, IL-10 initiates cellular response by interacting with the heterotetrametric IL-10 Receptor (IL-10R) complex, which is characterized by two IL-10RA and IL-10RB subunits. The interaction between IL-10 and IL-10RA triggers a signalling cascade involving Jak1, Tyrosine kinase 2 (Tyk2) and the activator of transcription 3 (STAT-3). In detail, following IL-10 binding, both Jak1 and Tyk2 are phosphorylated, thus stimulating STAT3 recruitment and phosphorylation and allowing its translocation into the nucleus. Here, STAT-3 induces the SOCS-3 (suppressor of cytokine signalling 3), which, in turn, inhibits the NF-kB nuclear transcription and the pro-inflammatory gene expression [30].

In conclusion, whereas gut microbiota stimulates the immune system and induces immunogenic responses to harmful antigens, immune system inhibits unnecessary inflammatory response against innocuous gut microorganisms, suggesting a reciprocal balance between gut microbiota and immune system, which is critical for maintaining the host health.

Gut Microbe Metabolites: A Linking Point Between Microbiota and The Host Immune System

For a long time, it was believed that the immune system operates alone in identifying and eliminating harmful pathogens. This conclusion has been largely surpassed by current understanding of microbiota functions. Studies have demonstrated that germ-free animals exhibit underdeveloped immune systems characterized by absence of Th17 cells, reduced populations of αβ and γδ intraepithelial lymphocytes, and diminished levels of IgA antibodies [31,32]. This compromises the immune system and increases the susceptibility to infectious diseases, reducing life expectancy [31]. Interestingly, transplantation of mice microbiota into germ-free mice was found to restore immune functions. It provides microbial species which increase the expression of T cell markers genes (Cd4, Cd8a and Foxp3); CD8a+ intestinal lymphocytes and induce Th17 cells [33], thus enhancing the immune surveillance against pathogens.

To date, the precise mechanisms used by symbiotic microbes to influence the host physiology are still unclear. However, the emerging interest in high throughput technologies, such as metabolomics, has facilitated the acquisition of detailed information on how gut microbiota shape the host immune system for host defense.

The sophisticated relationship between gut microbiota and the host immune system depends on microbial metabolites. Gut microbes can generate metabolites through different approaches:

i) de novo synthesis; ii) digestion of dietary components (Short Chain Fatty Acids and indole derivatives) and iii) biochemical modification of host metabolites (secondary bile acids) [11,34]. Collectively, these metabolites represent approximately 10% of blood mammalian metabolites and together with the host metabolites contribute to the establishment of an immunological barrier against infections.

Short Chain fatty Acids

Short-chain fatty acids (SCFAs; acetate, propionate and butyrate) protects the host from pathogens, participating to the intestinal energy metabolism. They are produced by gut microbes through fermentation of dietary nondigestible fibres [11,35]. Thus, the diet can affect SCFAs production. High-Fat Diets (HFDs) decrease SCFAs production, compared to high-carbohydrate or high-protein diets (HCDs and HPDs, respectively) [36,37]. In particular, HPDs lead to production of  Branched-Chain Fatty Acids (BCFAs; isovalerate, isobutyrate and 2-methylbutyrate) through fermentation of proteins rich in Branched-Chain Amino Acids (BCAAs). In addition, fluctuations of the gut microbe composition due to age-related changes also affect the levels of SCFAs [38]. In the early stage of life, Bifidobacteria prevail and increase acetate, lactate ad formate (two minors SCFAs) production, resulting from Human Milk Oligosaccharides (HMO) fermentation [39]. Upon breastfeeding interruption, Firmicutes prevail and increase propionate production, resulting from sugar fermentation [38]. Finally, in the late stage of life, there is an increase in Proteobacteria and a decrease in Firmicutes and Bacteroides, which, in turn, results in reduction of butyrate and propionate production [38].

SCFAs exert their immunomodulatory function against pathogens through different mechanisms. They can affect the signalling transduction pathway of immune cells by: i) activating specific extracellular G-protein coupled receptors, including free fatty acid receptor 2 and 3 (FFAR2 or GPR43 and FFAR3 or GPR41, respectively) and hydroxycarboxylic acid receptor 2 (HCA2, GPR109A); ii) inhibiting histone deacetylases (HDACs) and iii) promoting the activity of the histone acetyltransferase (HAT) enzymes [37].

By sensing FFAR2 on dendritic cells and neutrophils, SCFAs can switch B-cells into plasma cells and stimulate neutrophil recruitment to the inflammatory site, thus increasing antibody and inflammatory responses against pathogens [40]. Acetate has been found to facilitate the immune response against Citrobacter rodentium by recruiting neutrophils and Th17 cells and increasing the expression of IL6, CXCL1 and CXCL2 genes, as well as the production of IgA and IgG [40,41]. Of note, recent studies have demonstrated that the activation of Formyl peptide receptors by pro-inflammatory ligands strongly potentiates the acetate FFAR2-mediated effects, increasing chemotaxis and ROS production, for a successful antimicrobial response. However, excessive and prolonged ROS production due to neutrophil priming may exacerbate inflammatory response, leading to tissue damage [42]. In such condition, acetate can interfere to mitigate the inflammatory response by inducing regulatory T cell (Treg) accumulation in a FFAR2-dependent manner [43]. In addition, butyrate and propionate have also been found to suppress the inflammatory response by directly inhibiting histone deacetylase and encouraging Treg cell differentiation and IL-10 release (Figure 1) [43,44]. Overall, these findings indicate the key regulatory role of SCFAs in mounting a defensive response against pathogens and, concurrently, ensuring a balance between pro- and antiinflammatory responses.

Indole derivatives

Indole (In) and indole derivatives (InDs) provide competitive advantages to gut microbes, supporting the host catabolism of tryptophan [46]. As an essential amino acid, changes in diet influence tryptophan abundance and, consequently, InD production. In addition, similar to SCFAs, gut microbe composition also affects InD production.

Clostridia are the major tryptophan-metabolizing microbial strains, involved in producing indole (In), indole-3-acetic acid (IAA), 3-indole acrylic acid (IA), Indole-3-Propionic Acid (IPA) and tryptamine (Roager & Licht, 2018). Clostridium colonization typically begins during childhood, specifically during the transition from breastfeeding to solid food intake, emphasizing the crucial role of diet in host homeostasis and infection susceptibility [46,47].

In Ds have been shown to inhibit the growth of different pathogens, including Salmonella enterica, Staphylococcus aureus, Lactobacillus plantarum and Penicillium strains [46]. Recent studies have also demonstrated that In- and InD-producing bacteria protect the host from Cryptosporidium infections 48]. This protective effect has been mainly attributed to the capacity of In in reverting the host mitochondrial reprogramming induced by Cryptosporidium to grow and replicate within the host [48]. In addition, the beneficial effects of In can also be attributed to the activation of the Aryl Hydrocarbon Receptor (AhRs) signalling pathway [49].

AhR belongs to the Periodic Circadian Protein (PER)AHR Nuclear Translocator (ARNT)-Single-Minded Protein (SIM) superfamily of transcription factors, which can recognize exogenous and endogenous ligands, including In and InDs, and modulate both innate and adaptive immunity.

According to the ligand and the cytokine context, AhR signalling pathway can stimulate Th17, Foxp3+ Treg or type 1 regulatory T (Tr1) cell differentiation [50]. Th17 differentiation is mediated by IL-6, TGF-β1or IL-21 cytokines and together with IL-23 or IL-1β-activated RORγt+ Innate Lymphoid Cells (ILC) stimulate IL-22 release [49,51], inducing production of antimicrobial peptides and pro-inflammatory molecules involved in host defense against pathogens. Recent studies revealed the essential role of IL-22 in conferring protection against Helicobacter pylori infection in immunized animals, likely due to the expression of the antimicrobial peptide RegIIIβ (regenerating islet-derived protein IIIβ) [52]. Tr1 differentiation instead, is mediated by IL27 and enhanced by IL-10, IL-21 and CD39, while Foxp3+ Treg differentiation is initiated by TGF-β1 [50]. Furthermore, AhR activation in dendritic cells also participates to T cell polarization, favouring Treg differentiation [49]. Both Tr1 and Foxp3+ Treg cells exert immunosuppressive functions [53,54].

Together, these findings suggest the pleiotropic role of InDs in mounting an effective immune response against pathogens, establishing their importance in host-microbe interactions and immune regulation.

Secondary Bile Acids

Gut microbial communities significantly influence the composition and dynamics of the bile acid pool [55,56]. Firmicutes, Bacteroides and Actinobacteria express hydrolase genes that catabolize approximately 5% of taurine or glycine-conjugated bile acids into taurine or glycine and primary bile acids (PBAs) [56,57]. The obtained amino acids are used for energy production, while PBAs undergo further biotransformation by gut microbes to produce secondary bile acids [SBAs; e.g., deoxycholic acid (DCA), lithocholate (LCA) and Urodeoxycholic Acid (UDCA)] (Figure 2) [58].

Figure 2: Primary Bile Acids (PBAs) biotransformation by gut microbes. Cholesterol is converted into PBAs [cholic acid (CA) and chenodeoxycholic acid (CDCA)] in the liver by the neutral or acidic pathway. Both CA and CDCA are successively conjugated with taurine or glycine to form bile salts, which are finally converted into secondary bile acids [SBAs; deoxycholic acid (DCA), Lithocholic Acid (LCA) and urodeoxycholic acid (UDCA)] by gut microbiota.

As ligands of the farnesoid-X-receptor (FXR), SBAs play critical roles in regulating immune responses. They have been shown to exert anti-inflammatory effects by suppressing inflammation through different mechanisms. SBAs inhibit the NF-kB-mediated pathway of TLR4 in macrophages, limiting the release of IL-6, TNF-α and IL-1β cytokines [59]. Additionally, they induce epigenetic changes and influence the transcription of innate immune genes [60]. Notably, SBAs have also been documented to inhibit Th17 cell differentiation, while stimulating the differentiation of FOXP3+ regulatory T (Treg) cells [61]. The capacity of SBAs to modulate both innate and adaptive immune responses, make these molecules effective in providing protection against opportunistic pathogens [62].

Recently, Burgess et al. demonstrated that the symbiotic Clostridium scindens protects the host from Entamoeba histolytica colonization and attributed this result to an increase in DCA production [63]. They showed that DCA administration stimulates granulocyte-monocyte progenitor expansion in vivo via epigenetic modifications, including H3K27me3 decrease and H3K4me3 increase [63]. Furthermore, DCA, combined with LCA, was found to protect the host against Clostridium difficile infections. This result was attributed to their ability of inhibiting pathogen growth, spore germination and toxin production [64].

In conclusion, these findings give reason for suggesting gut microbiota-metabolized bile acids as additional elements helpful in addressing the modern challenge of bacterial infections.

Gut Microbiota Dysbiosis and Host Resistance to Pathogens

For several years, it was thought that genetic variation in the host was the key determinant for microbial diversity. Recent evidences clearly confuted this hypothesis, suggesting environmental factors as main players involved in shaping microbial composition [65]. Investigations on monozygotic (MZ) and dizygotic (DZ) twins revealed that, even though MZ twins share a higher degree of similarity in microbiome composition compared to DZ twins [66,67], when living apart, MZ pairedtwins change their microbiome profile [68].

Lifestyle and most notably diet, strongly influence gut microbial ecosystem by altering the relative abundance of some of the most important bacterial taxa, including Bacteroides, Prevotella and Bifidobacteroides [65,69]. These changes compromise the physiological balance between host and gut microbiota, leading to dysbiosis, key factor for the occurrence of inflammatory and metabolic diseases [70,71]. In addition, dysbiosis may also predispose to infectious diseases [72]. Malnutrition is the major contributor to bacterial infections. It is widely recognized that under- or overnutrition strongly imbalance density and ratio of gut dominant bacterial taxa, which reflect an impaired production of bioactive metabolites [73]. In other words, dysbiosis associated with the nutritional status strongly influences the host resistance to infections.

Dietary pattern and dysbiosis

In humans, both long-term and short-term dietary changes (LTDCs and STDCs, respectively) significantly influence microbial composition [8].

David LA, et al. [69] proved that STDCs promptly influence both structure and activities of gut microbial communities, with noticeable changes occurring as early as day 1 after the diet modification. They observed that animal-based diet (ADt) increases fat and protein intake while reducing fiber intake compared to plant-based diet (PDt). Interestingly, ADt was found to diminish the similarity in gut microbiome composition between the analyzed samples. Specifically, ADt depleted bacteria belonging to the genus Prevotella, which contribute to carbohydrate degradation and Short-Chain Fatty Acid (SCFA) production, and enriched bacteria exhibiting Bile Salt Hydrolase (BSH) activity [74]. Consequently, ADt may predispose to enteric diseases by enlarging the population of pathogenic Enterobacteriaceae and increasing the production of secondary bile acids.

Growing evidences suggest that BSH and its derivatives, including DCA and LCA, although proposed as potential therapeutical targets for infectious and metabolic diseases (as previously specified), may also participate to colorectal cancer development and progression via activation of the Wnt/Beta-catenin signalling pathway, induction of M2 macrophage polarization and infiltration of Tregs in the tumor microenvironment [75,76]. In addition, alterations in gut microbiota-mediated bile acid signalling associated with consumption of a high-fat diet have also been correlated with obesity [77].

Mice fed a high-fat diet and prone to obesity showed decreased abundance of Clostridium scindens and Clostridium hylemonae and, accordingly, decreased levels of the non-12OH bile acids [ursodeoxycholate (UDCA), chenodeoxycholate (CDCA) and lithocholate (LCA)], which modulate the anorexogenic GLP-1 hormone [77,78] involved in insulin release and appetite control [79,80]. Similarly, reduction of SCFA levels due to high fat diet also impairs the insulin sensitivity and increase body weight and fat mass [81,82]. In this regard, emerging studies on mice and humans have demonstrated that fecal microbiota transplantation (FMT) from lean donors to obese acceptors can restore microbial metabolite dysregulation and improve insulin sensitivity, controlling both mass fat and body weight [83,84]. However, appropriate diet regimen is required. Thus, taken together these findings clearly indicate that gut microbiota rapidly responds to dietary changes and that gut microbiota manipulation through probiotic supplementation or FMT, by reconstituting a balanced gut microbiota, could represent a valid approach to reduce dysbiosis and the related bacterial infection susceptibility due to malnutrition or dietary pattern modifications.

Conclusion

Gut microbiota plays a critical role in regulating the host homeostasis by providing beneficial metabolites, which communicate with the host immune cells. Notably, gut microbiota influences the host immunometabolism, through SCFAs production, bile acid detoxification and tryptophan metabolism [12].

The extensive plasticity of gut microbiota could compromise the intracellular metabolism of host immune cells, leading to pathological events, including bacterial infections. Understanding how structural and functional perturbations in gut microbiota facilitate pathogen colonization is a critical aspect for developing valid strategies for disease control.

It is widely recognized that gut microbiota composition strongly reflects the individual dietary choices [81]. This finding paves the way to dietary interventions as an alternative therapy to modulate the host resistance to bacterial infections. Personalized dietary approaches emerge as promising strategies to regulate hostmicrobiota metabolism and favor microbial species producing helpful metabolites.

In conclusion, modulation of gut microbiota pathways might aid the host immune system in inhibiting pathogen colonization, representing a potential therapeutical intervention against bacterial infections. Nevertheless, further studies enrolling larger sample size are needed to deeper investigate the intricate relationship gut microbiota-host immunometabolism and explore the way to modulate microbiome metabolism for host benefit.

Conflict of interest

The authors declare no conflict of interest.

References

  1. Lavelle A, Sokol H (2020) Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 17: 223-237.
  2. Degnan PH, Taga ME, Goodman AL (2014) Vitamin B12 as a modulator of gut microbial ecology. Cell Metab 20: 769-778.
  3. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R (2012) Diversity, stability and resilience of the human gut microbiota. Nature 489: 220-230.
  4. Ferretti P, Pasolli E, Tett A, Asnicar F, Gorfer V, et al. (2018) Motherto-Infant Microbial Transmission from Different Body Sites Shapes the Developing Infant Gut Microbiome. Cell Host and Microbe 24: 133145.e5.
  5. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J (2017) A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 5: 48.
  6. Joice R, Yasuda K, Shafquat A, Morgan XC, Huttenhower C (2014) Determining microbial products and identifying molecular targets in the human microbiome. Cell Metab 20: 731-741.
  7. Brüssow H (2020) The relationship between the host genome, microbiome, and host phenotype. Environ Microbiol 22: 1170-1173.
  8. Sharon G, Garg N, Debelius J, Knight R, Dorrestein PC, et al. (2014) Specialized metabolites from the microbiome in health and disease. Cell Metab 20: 719-730.
  9. Zhao T, Zhang Y, Nan L, Zhu Q, Wang S, et al. (2023) Impact of structurally diverse polysaccharides on colonic mucin O-glycosylation and gut microbiota. NPJ Biofilms Microbiomes 9: 97.
  10. Hooper LV, Littman DR, Macpherson AJ (2012) Interactions between the microbiota and the immune system. Science 336: 1268-1273.
  11. Liu J, Tan Y, Cheng H, Zhang D, Feng W, et al. (2022) Functions of Gut Microbiota Metabolites, Current Status and Future Perspectives. Aging Dis 13: 1106-1126.
  12. Michaudel C, Sokol H (2020) The Gut Microbiota at the Service of Immunometabolism. Cell Metab 32: 514-523.
  13. Costello EK, Stagaman K, Dethlefsen L, Bohannan BJM, Relman DA (2012) The application of ecological theory toward an understanding of the human microbiome. Science 336: 1255-1262.
  14. Jordan CKI, Brown RL, Larkinson MLY, Sequeira RP, Edwards AM, et al. (2023) Symbiotic Firmicutes establish mutualism with the host via innate tolerance and resistance to control systemic immunity. Cell Host Microbe 31: 1433-1439.e9.
  15. Prestinaci F, Pezzotti P, Pantosti A (2015) Antimicrobial resistance: A global multifaceted phenomenon. Pathog Glob Health 109: 309-318.
  16. Murray CJ, Ikuta KS, Sharara F, Swetschinski L, Aguilar GR, Gray A, et al. (2022) Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet: 399: 629-655.
  17. Belkaid Y, Hand TW (2014) Role of the microbiota in immunity and inflammation. Cell 157: 121-141.
  18. Turner JR (2009) Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9: 799-809.
  19. Silva-Sanchez A, Randall TD (2019) Anatomical uniqueness of the mucosal immune system (GALT, NALT, iBALT) for the induction and regulation of mucosal immunity and tolerance. In Mucosal Vaccines: Innovation for Preventing Infectious Diseases. Mucosal Vaccines 2020: 21-54.
  20. Mörbe UM, Jørgensen PB, Fenton TM, von Burg N, Riis LB, et al. (2021) Human gut-associated lymphoid tissues (GALT); diversity, structure, and function. Mucosal Immunol 14: 793-802.
  21. Corr SC, Gahan CCGM, Hill C (2008) M-cells: Origin, morphology and role in mucosal immunity and microbial pathogenesis. FEMS Immunol Med Microbiol 52: 2-12.
  22. Mabbott NA, Donaldson DS, Ohno H, Williams IR, Mahajan A (2013) Microfold (M) cells: Important immunosurveillance posts in the intestinal epithelium. Mucosal Immunology 6: 666-677.
  23. Liang E, Kabcenell AK, Coleman JR, Robson J, Ruffles R, et al. (2001) Permeability measurement of macromolecules and assessment of mucosal antigen sampling using in vitro converted M cells. J Pharmacol Toxicol Methods 46: 93-101.
  24. Pappo J, Mahlman RT (1993) Follicle epithelial M cells are a source of interleukin-1 in Peyer’s patches. Immunology 78: 505-507.
  25. Huus KE, Petersen C, Finlay BB (2021) Diversity and dynamism of IgA−microbiota interactions. Nat Rev Immunol 21: 514-525.
  26. Macpherson AJ, Uhr T (2004) Induction of Protective IgA by Intestinal Dendritic Cells Carrying Commensal Bacteria. Science 303: 16621665.
  27. Semin I, Ninnemann J, Bondareva M, Gimaev I, Kruglov AA (2021) Interplay Between Microbiota, Toll-Like Receptors and Cytokines for the Maintenance of Epithelial Barrier Integrity. Front Med 8: 644333.
  28. Sakaguchi S, Yamaguchi T, Nomura T, Ono M (2008) Regulatory T cells and immune tolerance. Cell 133: 775-787.
  29. Nguyen HD, Aljamaei HM, Stadnyk AW (2021) The Production and Function of Endogenous Interleukin-10 in Intestinal Epithelial Cells and Gut Homeostasis. Cell Mol Gastroenterol Hepatol 12: 1343-1352.
  30. Saraiva M, Vieira P, O’Garra A (2020) Biology and therapeutic potential of interleukin-10. J Exp Med 217: e20190418.
  31. Fiebiger U, Bereswill S, Heimesaat MM (2016) Dissecting the interplay between intestinal microbiota and host immunity in health and disease: Lessons learned from germfree and gnotobiotic animal models. Eur J Microbiol Immunol 6: 253-271.
  32. Zheng D, Liwinski T, Elinav E (2020) Interaction between microbiota and immunity in health and disease. Cell Res 30: 492-506.
  33. Lundberg R, Toft MF, Metzdorff SB, Hansen CHF, Licht TR, et al. (2020) Human microbiota-transplanted C57BL/6 mice and offspring display reduced establishment of key bacteria and reduced immune stimulation compared to mouse microbiota-transplantation. Scientific Rep 10: 7805.
  34. Zhang Y, Chen R, Zhang DD, Qi S, Liu Y (2023) Metabolite interactions between host and microbiota during health and disease: Which feeds the other? Biomed Pharmacother 160: 114295.
  35. Russell WR, Gratz SW, Duncan SH, Holtrop G, Ince J, et al. (2011) High-protein, reduced-carbohydrate weight-loss diets promotemetabolite profiles likely to be detrimental to colonic health. Am J Clin Nutr 93: 1062-1072.
  36. Cornejo-Pareja I, Muñoz-Garach A, Clemente-Postigo M, Tinahones FJ (2019) Importance of gut microbiota in obesity. Eur J Clin Nutr 72: 26-37.
  37. Ney LM, Wipplinger M, Grossmann M, Engert N, Wegner VD, et al. (2023) Short chain fatty acids: key regulators of the local and systemic immune response in inflammatory diseases and infections. Open Biol 13: 230014.
  38. Fusco W, Lorenzo MB, Cintoni M, Porcari S, Rinninella E, et al. (2023) Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 15: 2211.
  39. Sela DA, Mills DA (2010) Nursing our microbiota: molecular linkages between bifidobacteria and milk oligosaccharides. Trends Microbiol 18: 298-307.
  40. Kim M, Qie Y, Park J, Kim CH (2016) Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 20: 202-214.
  41. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH (2013) Shortchain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 145: 396-406.e1-10.
  42. El-Benna J, Hurtado-Nedelec M, Marzaioli V, Marie JC, GougerotPocidalo MA, et al. (2016) Priming of the neutrophil respiratory burst: role in host defense and inflammation. Immunol Rev 273: 180-193.
  43. Arpaia N, Campbell C, Fan X, Dikiy S, Van Der Veeken J, et al. (2013) Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature, 504: 451-455.
  44. Zhang Z, Zhang H, Chen T, Shi L, Wang D, et al. (2022) Regulatory role of short-chain fatty acids in inflammatory bowel disease. Cell Commun Signal 20: 64.
  45. Roager HM, Licht TR (2018) Microbial tryptophan catabolites in health and disease. Nat Commun 9: 3294.
  46. Ye X, Li H, Anjum K, Zhong X, Miao S, et al. (2022) Dual Role of Indoles Derived from Intestinal Microbiota on Human Health. Front Immunol 13: 903526.
  47. Laursen MF, Andersen LBB, Michaelsen KF, Mølgaard C, Trolle E, et al. (2016) Infant Gut Microbiota Development Is Driven by Transition to Family Foods Independent of Maternal Obesity. mSphere 1: e0006915.
  48. Funkhouser-Jones LJ, Xu R, Wilke G, Fu Y, Schriefer LA, et al. (2023) Microbiota-produced indole metabolites disrupt mitochondrial function and inhibit Cryptosporidium parvum growth. Cell Rep 42: 112680.
  49. Rothhammer V, Quintana FJ (2019) The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol 19: 184-197.
  50. Gandhi R, Kumar D, Burns EJ, Nadeau M, Dake B, et al. (2010) Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3+ regulatory T cells. Nat Immunol 11: 846-853.
  51. Yeste A, Mascanfroni ID, Nadeau M, Burns EJ, Tukpah AM, et al. (2014) IL-21 induces IL-22 production in CD4+ T cells. Nat Commun 5: 3753.
  52. Moyat M, Bouzourene H, Ouyang W, Iovanna J, Renauld JC, et al. (2017) IL-22-induced antimicrobial peptides are key determinants of mucosal vaccine-induced protection against H. pylori in mice. Mucosal Immunol 10: 271-281.
  53. Li Z, Li D, Tsun A, Li B (2015) FOXP3+ regulatory T cells and their functional regulation. Cell Mol Immunol 12: 558-565.
  54. Song Y, Wang N, Chen L, Fang L (2021) Tr1 Cells as a Key Regulator for Maintaining Immune Homeostasis in Transplantation. Front Immunol 12: 671579.
  55. Chang PV (2020). Chemical Mechanisms of Colonization Resistance by the Gut Microbial Metabolome. ACS Chem Biol 15: 1119-1126.
  56. Staley C, Weingarden AR, Khoruts A, Sadowsky MJ (2017) Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol 101: 47-64.
  57. Winston JA, Theriot CM (2020) Diversification of host bile acids by members of the gut microbiota. Gut Microbes 11: 158-171.
  58. Larabi AB, Masson HLP, Bäumler AJ (2023) Bile acids as modulators of gut microbiota composition and function. Gut Microbes 15: 2172671.
  59. Vavassori P, Mencarelli A, Renga B, Distrutti E, Fiorucci S (2009) The bile acid receptor FXR is a modulator of intestinal innate immunity. J Immunol 183: 6251-6261.
  60. Ananthanarayanan M, Li SD, Balasubramaniyan N, Suchy FJ, Walsh MJ (2004) Ligand-dependent activation of the farnesoid X-receptor directs arginine methylation of histone H3 by CARM1. J Biol Chem 279: 54348-54357.
  61. Hang S, Paik D, Yao L, Kim E, Jamma T, et al. (2019) Bile acid metabolites control TH17 and Treg cell differentiation. Nature 576: 143148.
  62. Inagaki T, Moschetta A, Lee YK, Peng L, Zhao G, et al. (2006) Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor. Proc Natl Acad Sci U S A 103: 3920-3925.
  63. Burgess SL, Leslie JL, Uddin J, Oakland DN, Gilchrist C, et al. (2020) Gut microbiome communication with bone marrow regulates susceptibility to amebiasis. J Clin Invest 140: 4019-4024.
  64. Reed AD, Nethery MA, Stewart A, Barrangou R, Theriot CM (2020) Strain-dependent inhibition of clostridioides difficile by commensal clostridia carrying the bile acid-inducible (bai) operon. J Bacteriol 202: e00039-20.
  65. Vilchez-Vargas R, Skieceviciene J, Lehr K, Varkalaite G, Thon C, et al. (2022) Gut microbial similarity in twins is driven by shared environment and aging. EBioMedicine 79: 104011.
  66. Goodrich JK, Davenport ER, Beaumont M, Jackson MA, Knight R, et al. (2016) Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 19: 731-743.
  67. Goodrich JK, Di Rienzi SC, Poole AC, Koren O, Walters WA, et al. (2014) Conducting a microbiome study. Cell 158: 250-262.
  68. Xie H, Guo R, Zhong H, Feng Q, Lan Z, et al. (2016) Shotgun Metagenomics of 250 Adult Twins Reveals Genetic and Environmental Impacts on the Gut Microbiome. Cell Syst 3: 572-584.e3.
  69. -David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, et al. (2014) Diet rapidly and reproducibly alters the human gut microbiome. Nature 505: 559-563.
  70. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E (2017) Dysbiosis and the immune system. Nat Rev Immunol 17: 219-232.
  71. Matzaras R, Nikopoulou A, Protonotariou E, Christaki E (2022) Gut Microbiota Modulation and Prevention of Dysbiosis as an Alternative Approach to Antimicrobial Resistance: A Narrative Review. Yale J Biol Med 95: 479-494.
  72. Gupta A, Singh V, Mani I (2022) Dysbiosis of human microbiome and infectious diseases. In Progress in Molecular Biology and Translational Science 192: 33-51.
  73. Yang F, Yang Y, Zeng L, Chen Y, Zeng G (2021) Nutrition Metabolism and Infections. Infectious Microbes and Diseases 3: 134-141.
  74. Ecklu-Mensah G, Gilbert J, Devkota S (2022) Dietary Selection Pressures and Their Impact on the Gut Microbiome. Cell Mol Gastroenterol Hepatol 13: 7-18.
  75. Sun L, Zhang Y, Cai J, Rimal B, Rocha ER, et al. (2023) Bile salt hydrolase in non-enterotoxigenic Bacteroides potentiates colorectal cancer. Nat Commun 14: 755.
  76. Yang R, Qian L (2022) Research on Gut Microbiota-Derived Secondary Bile Acids in Cancer Progression. Integr Cancer Ther 21: 15347354221114100.
  77. Sarmiento-Andrade Y, Suárez R, Quintero B, Garrochamba K, Chapela SP (2022) Gut microbiota and obesity: New insights. Front Nutr 9: 1018212.
  78. Wei M, Huang F, Zhao L, Zhang Y, Yang W, et al.  (2020) A dysregulated bile acid-gut microbiota axis contributes to obesity susceptibility. EBioMedicine 55: 102766.
  79. Popoviciu MS, Păduraru L, Yahya G, Metwally K, Cavalu S (2023) Emerging Role of GLP-1 Agonists in Obesity: A Comprehensive Review of Randomised Controlled Trials. Int J Mol Sci 24: 10449.
  80. Wang JY, Wang QW, Yang XY, Yang W, Li DR, et al. (2023) GLP−1 receptor agonists for the treatment of obesity: Role as a promising approach. Front Endocrinol 14: 1085799.
  81. Ecklu-Mensah G, Choo-Kang C, Maseng MG, Donato S, Bovet P, et al. (2023) Gut microbiota and fecal short chain fatty acids differ with adiposity and country of origin: the METS-microbiome study. Nat Commun 14: 5160.
  82. Lu Y, Fan C, Li P, Lu Y, Chang X, et al. (2016) Short Chain Fatty Acids Prevent High-fat-diet-induced Obesity in Mice by Regulating G Protein-coupled Receptors and Gut Microbiota. Sci Rep 6: 37589.
  83. Lahtinen P, Juuti A, Luostarinen M, Niskanen L, Liukkonen T, et al. (2022) Effectiveness of Fecal Microbiota Transplantation for Weight Loss in Patients with Obesity Undergoing Bariatric Surgery: A Randomized Clinical Trial. JAMA Netw Open 5: e2247226.
  84. Walker AW, Parkhill J (2013) Microbiology. Fighting obesity with bacteria. Science 341: 1069-1070.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Community Medicine & Public Health

rumus slot mahjongrtp slot gacorfitur slot mahjong winsrekomendasi slot pragmartp live slotpola gates of gatotkacaapk cheat slotzeus godwrath maxwinmitra slot dana resmihabanero anti gagalserver kamboja gacordaftar link togelslot pg mahjongtrik pola zeus x500slot gacor mudah menangslot mahjong pragmaticpola trik slot mahjongrtp fortune dragonrtp slot speed winnerslot kamboja mahjong waystrik mantap slot olympusnaga hitam mahjongslot tergacor mahjongtrik jitu cuan mahjongpola slot mahjong winsrtp tinggi pragmaticslot mahjong onlineslot gacor hari inislot bonanza gacorfreebet mahjong winsserver jp rtp tinggigame resmi pragmatic terbaiktaktik efektif mahjongpola mahjong rekomendasi googleaztec gems boskututorial mahjong ways2starlight princess hari inipola starlight princessrtp fortune tigerrtp pg softrtp starlight princesstrik mahjong waysperkalian x5000 banditoslot mahjong waysslot terbaik olympusslot gates of olympusdaftar slot dana maxwinbocoran pola olympusmaxwin slot bonanzabocoran rtp tinggislot samurai codemetode slot starlightslot zeusrtp slot gacor pragmaticrtp slot pg softcara menang slot onlinescatter slot mahjongslot gacor server luar rekomendasi link olympusgerbang gatot kacartp kakek olympusslot gacor andalanrtp slot pgslot mahjongslot mahjong server jepangperkalian besar starlightrtp ways of qilinslot terbaik mahjongmahjong bulan mudastrategi permainan pragmaticcheat engine gacorjackpot auto cuanmahjong mekanik tinggitrik slot mahjongtips main slotslot server thailandpola mahjong unguslot gacor menangpg soft scatterslot olympusbocoran togel terpercayaamantotorm1131aman toto