Gut microbiota can interact with the immune system through direct or indirect pathways. In the indirect pathway, gut microbiota produces metabolites such as short chain fatty acids (SCFAs), which may modulate the immune response. SCFAs reduce inflammation, repair intestinal barrier, and induce propagation of specific immune cells, e.g., T regulatory cells (Treg), which can suppress reactive cells such as macrophage and dendritic cells (DCs). As one of the most dominant members of microbiota, Clostridium produces SCFAs. As one of SCFA members, butyrate plays an important role in the modulation of immune cells. Through butyrate production, Clostridium helps to generate aryl hydrocarbon receptor (AhR). AhR interacts with many proteins inside the cytoplasm including Heat Shock Protein 90 (HSP 90), HSP 23, and chaperone. Activation of AhR leads to its translocation inside the nucleus and gene expression, which yields cell differentiation, energy metabolism, microbial defense, and immune cell propagation. Moreover, it may interact with other cells like B-cell and epithelial cell, which are responsible for modulation and maturation, respectively. AhR causes upregulation in the co-stimulatory marker in the DCs and interacts with nuclear factor KB (NF-ĸB) to modulate cell function. Butyrate induces Treg (iTreg) propagation and upregulates the Forkhead box p3 (FOXP3) as a special marker of Treg cell. It may also yield signaling through G-protein coupled receptors (GPRs) which, in turn, facilitates polymorphonuclear (PMN) chemotaxis.
The interaction between microbiota and non-immune cells, such as Paneth cells, leads to the secretion of antimicrobial substance, erection of barriers against bacterial pathogens, and regulation of microbiota composition via feedback effect. In addition, the components released from microbiota, such as peptidoglycan, reinforce the maturation of both the immune system and non-immune tissue development. Moreover, microbiota can directly activate the effector cells, e.g., macrophage, to secrete cytokines and propagate Treg cells.
Moeller AH , et al. Transmission modes of the mammalian gut microbiota. Science 2018; 362(6413): 453–457.
Lengfelder I , et al. Complex bacterial consortia reprogram the colitogenic activity of Enterococcus faecalis in a gnotobiotic mouse model of chronic, immune-mediated colitis. Front Immunol 2019; 10: 1420.
Hold GL , et al. Assessment of microbial diversity in human colonic samples by 16S rDNA sequence analysis. FEMS Microbiol Ecol 2002; 39(1): 33–39.
COLLINS MD , et al. The phylogeny of the genus Clostridium: proposal of five new genera and eleven new species combinations. Int J Syst Evol Microbiol 1994; 44(4): 812–826.
Guo P , et al. Clostridium species as probiotics: potentials and challenges. J Anim Sci Biotechnol 2020; 11(1): 1–10.
Kim B-C , et al. Caproiciproducens galactitolivorans gen. nov., sp. nov., a bacterium capable of producing caproic acid from galactitol, isolated from a wastewater treatment plant. Int J Syst Evol Microbiol 2015; 65(Pt_12): 4902–4908.
Tang Y , et al. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int J Cancer 2011; 128(4): 847–856.
Kamada N , et al. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol 2013; 14(7): 685–690.
Albenberg LG , Wu GD . Diet and the intestinal microbiome: associations, functions, and implications for health and disease. Gastroenterology 2014; 146(6): 1564–1572.
Alipour MJ , et al. The composition of the perinatal intestinal microbiota in cattle. Scientific Rep 2018; 8(1): 1–14.
Bruggisser J , et al. CD31 (PECAM-1) serves as the endothelial cell-specific receptor of Clostridium perfringens β-toxin. Cell host & microbe 2020; 28(1): 69–78.
Macfarlane S , Macfarlane GT . Regulation of short-chain fatty acid production. Proc Nutr Soc 2003; 62(1): 67–72.
Miller TL , Wolin MJ . Pathways of acetate, propionate, and butyrate formation by the human fecal microbial flora. Appl Environ Microbiol 1996; 62(5): 1589.
Dion C , Chappuis E , Ripoll C . Does larch arabinogalactan enhance immune function? A review of mechanistic and clinical trials. Nutr Metab 2016; 13: 28–28.
Mortensen PB , Clausen MR . Short-chain fatty acids in the human colon: relation to gastrointestinal health and disease. Scand J Gastroenterol 1996; 31(sup216): 132–148.
Penner GB . Short chain fatty acid absorption and regulation of ruminal pH; 2019.
Ley RE , et al. Evolution of mammals and their gut microbes. Science 2008; 320(5883): 1647–1651.
Cheung SG , et al. Systematic review of gut microbiota and major depression. Front Psychiatry 2019; 10: 34.
Chinen T , Rudensky AY . The effects of commensal microbiota on immune cell subsets and inflammatory responses. Immunological Rev 2012; 245(1): 45–55.
Stecher B , et al. Like will to like: abundances of closely related species can predict susceptibility to intestinal colonization by pathogenic and commensal bacteria. PLoS Pathog 2010; 6(1).
Stecher B , et al. Like will to like: abundances of closely related species can predict susceptibility to intestinal colonization by pathogenic and commensal bacteria. PLoS Pathog 2010; 6(1): e1000711-e1000711.
Byndloss MX , et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017; 357(6351): 570.
Sun CM , et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med 2007; 204(8): 1775–1785.
Feng W , et al. Sodium butyrate attenuates diarrhea in weaned piglets and promotes tight junction protein expression in colon in a GPR109A-dependent manner. Cell Physiol Biochem 2018; 47(4): 1617–1629.
Yan H , Ajuwon KM . Butyrate modifies intestinal barrier function in IPEC-J2 cells through a selective upregulation of tight junction proteins and activation of the Akt signaling pathway. PloS one 2017; 12(6): e0179586–e0179586.
Tsai LC , et al. Apoptosis induced by the sodium butyrate in human gastric cancer TMK-1 cells. Anticancer Res 2000; 20(4): 2441–2448.
Gómez-Hurtado I , et al. Gut microbiota dysbiosis is associated with inflammation and bacterial translocation in mice with CCl4-induced fibrosis. PloS one 2011; 6(7): e23037–e23037.
Kamp ME , et al. G protein-coupled receptor 43 modulates neutrophil recruitment during acute inflammation. PloS one 2016; 11(9): e0163750–e0163750.
Kim Y-G , et al. Neonatal acquisition of Clostridia species protects against colonization by bacterial pathogens. Science 2017; 356(6335): 315.
Rodrigues PA , Ferrari RG , Conte-Junior CA . Application of molecular tools to elucidate the microbiota of seafood. J Appl Microbiol 2018; 124(6): 1347–1365.
Sciuto K , Moro I . Cyanobacteria: the bright and dark sides of a charming group. Biodiversity and Conservation 2015; 24(4): 711–738.
Chahorm K , Prakitchaiwattana C . Application of Reverse Transcriptase-PCR-DGGE as a rapid method for routine determination of Vibrio spp. in foods. Int J Food Microbiol 2018; 264: 46–52.
Lee H-W , et al. Identification of microbial communities, with a focus on foodborne pathogens, during kimchi manufacturing process using culture-independent and-dependent analyses. LWT-Food Sci Technol 2017; 81: 153–159.
Beste KJ , et al. Culture-independent and dependent evaluation of the equine paranasal sinus microbiota in health and disease. Equine Vet J 2020; 52(3): 455–463.
Fusco V , Quero GM . Culture-dependent and culture-independent nucleic-acid-based methods used in the microbial safety assessment of milk and dairy products. Compr Rev Food Sci Food Saf 2014; 13(4): 493–537.
Shang Y , et al. Chicken gut microbiota: importance and detection technology. Front Vet Sci 2018; 5: 254.
Xue Z , Kable ME , Marco M . Impact of DNA sequencing and analysis methods on 16S rRNA gene bacterial community analysis of dairy products. Msphere 2018; 3(5): e00410–e00418.
Wu GD , et al. Sampling and pyrosequencing methods for characterizing bacterial communities in the human gut using 16S sequence tags. BMC Microbiol 2010; 10(1): 1–14.
Tabit FT . Advantages and limitations of potential methods for the analysis of bacteria in milk: a review. J Food Sci Technol 2016; 53(1): 42–49.
Fasoli S , et al. Bacterial composition of commercial probiotic products as evaluated by PCR-DGGE analysis. Int J Food Microbiol 2003; 82(1): 59–70.
Rajilić-Stojanović M , et al. Development and application of the human intestinal tract chip, a phylogenetic microarray: analysis of universally conserved phylotypes in the abundant microbiota of young and elderly adults. Environ Microbiol 2009; 11(7): 1736–1751.
Sandholm M . Unsupervised learning from HITChip microarray data; 2011.
Spitaels F , Wieme AD , Vandamme P . MALDI-TOF MS as a novel tool for dereplication and characterization of microbiota in bacterial diversity studies. In: Applications of mass spectrometry in microbiology. Springer; 2016. p. 235–256.
Greppi A , Rantsiou K . Methodological advancements in foodborne pathogen determination: from presence to behavior. Curr Opin Food Sci 2016; 8: 80–88.
Duygan BDÖ , et al. Rapid detection of microbiota cell type diversity using machine-learned classification of flow cytometry data. Commun Biol 2020; 3(1): 1–13.
Chen MX , et al. Metabolome analysis for investigating host-gut microbiota interactions. J Formos Med Assoc 2019; 118: S10–S22.
Hsu Y-L , et al. Evaluation and optimization of sample handling methods for quantification of short-chain fatty acids in human fecal samples by GC–MS. J Proteome Res 2019; 18(5): 1948–1957.
Taminiau B , et al. Validation of real-time PCR for detection of six major pathogens in seafood products. Food Control 2014; 44: 130–137.
Ruijter JM , et al. Evaluation of qPCR curve analysis methods for reliable biomarker discovery: bias, resolution, precision, and implications. Methods 2013; 59(1): 32–46.
Siebert A , et al. Amplicon-sequencing of raw milk microbiota: impact of DNA extraction and library-PCR. Appl Microbiol Biotechnol 2021: 1–13.
Eisenstein M . Microbiology: making the best of PCR bias. Nat Methods 2018; 15(5): 317–320.
Moffitt JR , Zhuang X . RNA imaging with multiplexed error-robust fluorescence in situ hybridization (MERFISH). Methods Enzymol 2016; 572: 1–49.
Namsolleck P , et al. Molecular methods for the analysis of gut microbiota. Microb Ecol Health Dis 2004; 16(2–3): 71–85.
Lee KS , et al. An automated Raman-based platform for the sorting of live cells by functional properties. Nat Microbiol 2019; 4(6): 1035–1048.
Sarao LK , Arora M . Probiotics, prebiotics, and microencapsulation: a review. Crit Rev Food Sci Nutr 2017; 57(2): 344–371.
Aguilar-Toalá JE , et al. Postbiotics: an evolving term within the functional foods field. Trends Food Sci Technol 2018; 75: 105–114.
Veiga P , et al. Moving from probiotics to precision probiotics. Nat Microbiol 2020; 5(7): 878–880.
Li J , et al. Toxin plasmids of Clostridium perfringens. Microbiol Mol Biol Rev 2013; 77(2): 208–233.
Lee SM , et al. Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 2013; 501(7467): 426–429.
Rutten N , et al. Long term development of gut microbiota composition in atopic children: impact of probiotics. PloS one 2015; 10(9): e0137681.
van Zanten GC , et al. Synbiotic Lactobacillus acidophilus NCFM and cellobiose does not affect human gut bacterial diversity but increases abundance of lactobacilli, bifidobacteria and branched-chain fatty acids: a randomized, double-blinded cross-over trial. FEMS Microbiol Ecol 2014; 90(1): 225–236.
Egan M , et al. Metabolism of sialic acid by Bifidobacterium breve UCC2003. Appl Environ Microbiol 2014; 80(14): 4414–4426.
Nishiyama K , et al. Two extracellular sialidases from Bifidobacterium bifidum promote the degradation of sialyl-oligosaccharides and support the growth of Bifidobacterium breve. Anaerobe 2018; 52: 22–28.
Ueda Y , et al. Commensal microbiota induce LPS hyporesponsiveness in colonic macrophages via the production of IL-10. Int Immunol 2010; 22(12): 953–962.
Artis D . Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat Rev Immunol 2008; 8(6): 411–420.
Becker DJ , Lowe JB . Fucose: biosynthesis and biological function in mammals. Glycobiology 2003; 13(7): 41R–53R.
Bäumler AJ , Sperandio V . Interactions between the microbiota and pathogenic bacteria in the gut. Nature 2016; 535(7610): 85–93.
Jiang F , et al. The symbiotic bacterial surface factor polysaccharide A on Bacteroides fragilis inhibits IL-1β-induced inflammation in human fetal enterocytes via toll receptors 2 and 4. PLoS One 2017; 12(3).
Hall JA , et al. Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity 2008; 29(4): 637–649.
Frank DN , et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci 2007; 104(34): 13780–13785.
Ott SJ , et al. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut 2004; 53(5): 685–693.
Bauché D , Marie JC . Transforming growth factor β: a master regulator of the gut microbiota and immune cell interactions. Clin Translational Immunol 2017; 6(4): e136.
Travis MA , et al. Loss of integrin αvβ8 on dendritic cells causes autoimmunity and colitis in mice. Nature 2007; 449(7160): 361–365.
Cazac BB , Roes J . TGF-β receptor controls B cell responsiveness and induction of IgA in vivo. Immunity 2000; 13(4): 443–451.
Kim CH . B cell-helping functions of gut microbial metabolites. Microb Cell (Graz, Austria) 2016; 3(10): 529–531.
Kawamoto S , et al. Foxp3+ T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 2014; 41(1): 152–165.
Lochner M , et al. In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORγt+ T cells. J Exp Med 2008; 205(6): 1381–1393.
Shi LZ , et al. HIF1α–dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med 2011; 208(7): 1367–1376.
Mascanfroni ID , et al. Metabolic control of type 1 regulatory T cell differentiation by AHR and HIF1-α. Nat Med 2015; 21(6): 638.
Sano T , et al. An IL-23R/IL-22 circuit regulates epithelial serum amyloid A to promote local effector Th17 responses. Cell 2015; 163(2): 381–393.
Atarashi K , et al. Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell 2015; 163(2): 367–380.
Stefka AT , et al. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci 2014; 111(36): 13145–13150.
Mortha A , et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014; 343(6178): 1249288.
Kumar H , Kawai T , Akira S . Pathogen recognition by the innate immune system. Int Rev Immunol 2011; 30(1): 16–34.
Christ A , et al. Western diet triggers NLRP3-dependent innate immune reprogramming. Cell 2018; 172(1–2): 162–175.
Smale ST , Tarakhovsky A , Natoli G . Chromatin contributions to the regulation of innate immunity. Annu Rev Immunol 2014; 32: 489–511.
Smale ST , Natoli G . Transcriptional control of inflammatory responses. Cold Spring Harbor Perspect Biol 2014; 6(11): a016261.
Long EO . Ready for prime time: NK cell priming by dendritic cells. Immunity 2007; 26(4): 385–387.
Ganal SC , et al. Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity 2012; 37(1): 171–186.
Ramirez-Carrozzi VR , et al. A unifying model for the selective regulation of inducible transcription by CpG islands and nucleosome remodeling. Cell 2009; 138(1): 114–128.
Benjamin JL , et al. Intestinal epithelial autophagy is essential for host defense against invasive bacteria. Cell Host & Microbe 2013; 13(6): 723–734.
Kobayashi KS , et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science 2005; 307(5710): 731–734.
Ostaff MJ , Stange EF , Wehkamp J . Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5(10): 1465–1483.
Caruso R , et al. NOD1 and NOD2: signaling, host defense, and inflammatory disease. Immunity 2014; 41(6): 898–908.
Vaishnava S , et al. The antibacterial lectin RegIIIγ promotes the spatial segregation of microbiota and host in the intestine. Science 2011; 334(6053): 255.
Bouskra D , et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 2008; 456(7221): 507–510.
Tsuji M , et al. Requirement for lymphoid tissue-inducer cells in isolated follicle formation and T cell-independent immunoglobulin A generation in the gut. Immunity 2008; 29(2): 261–271.
Hergott CB , et al. Peptidoglycan from the gut microbiota governs the lifespan of circulating phagocytes at homeostasis. Blood J Am Soc Hematol 2016; 127(20): 2460–2471.
Kashyap DR , et al. Bactericidal peptidoglycan recognition protein induces oxidative stress in Escherichia coli through a block in respiratory chain and increase in central carbon catabolism. Mol Microbiol 2017; 105(5): 755–776.
Duerr CU , et al. Control of intestinal Nod2-mediated peptidoglycan recognition by epithelium-associated lymphocytes. Mucosal Immunol 2011; 4(3): 325–334.
Dabrowski AN , et al. Peptidoglycan recognition protein 4 limits bacterial clearance and inflammation in lungs by control of the gut microbiota. Front Immunol 2019; 10: 2106.
Scott MJ , et al. Hepatocytes express functional NOD1 and NOD2 receptors: a role for NOD1 in hepatocyte CC and CXC chemokine production. J Hepatol 2010; 53(4): 693–701.
Griseri T , et al. Dysregulated hematopoietic stem and progenitor cell activity promotes interleukin-23-driven chronic intestinal inflammation. Immunity 2012; 37(6): 1116–1129.
Wolf AJ , Underhill DM . Peptidoglycan recognition by the innate immune system. Nat Rev Immunol 2018; 18(4): 243–254.
Huang Z , et al. Antibody neutralization of microbiota-derived circulating peptidoglycan dampens inflammation and ameliorates autoimmunity. Nat Microbiol 2019; 4(5): 766–773.
Charroux B , et al. Cytosolic and secreted peptidoglycan-degrading enzymes in Drosophila respectively control local and systemic immune responses to microbiota. Cell Host & Microbe 2018; 23(2): 215–228. e4.
Arentsen T , et al. The bacterial peptidoglycan-sensing molecule Pglyrp2 modulates brain development and behavior. Mol Psychiatry 2017; 22(2): 257–266.
Salminen S , Isolauri E . Intestinal colonization, microbiota, and probiotics. J Pediatr 2006; 149(5, Supplement): S115–S120.
Nava GM , Friedrichsen HJ , Stappenbeck TS . Spatial organization of intestinal microbiota in the mouse ascending colon. ISME J 2011; 5(4): 627–638.
Tralongo P , et al. The role of butyric acid as a protective agent against inflammatory bowel diseases. Euromediterranean Biomed J 2014; 9.
Macfarlane GT , Macfarlane S . Bacteria, colonic fermentation, and gastrointestinal health. J AOAC Int 2012; 95(1): 50–60.
Falony G , et al. In vitro kinetics of prebiotic inulin-type fructan fermentation by butyrate-producing colon bacteria: implementation of online gas chromatography for quantitative analysis of carbon dioxide and hydrogen gas production. Appl Environ Microbiol 2009; 75(18): 5884–5892.
Louis P , Flint HJ . Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett 2009; 294(1): 1–8.
Jiang H , et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun 2015; 48: 186–194.
Bajaj JS , et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inflammation. Am J Physiology-Gastrointestinal Liver Physiol 2012; 303(6): G675–G685.
Ling Z , et al. Impacts of infection with different toxigenic Clostridium difficile strains on faecal microbiota in children. Scientific Rep 2014; 4: 7485.
Umesaki Y , et al. Differential roles of segmented filamentous bacteria and clostridia in development of the intestinal immune system. Infect Immun 1999; 67(7): 3504–3511.
Le Poul E , et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem 2003; 278(28): 25481–25489.
Schulthess J , et al. The short chain fatty bcid butyrate imprints an antimicrobial program in macrophages. Immunity 2019; 50(2): 432–445. e7.
Binder HJ . Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev Physiol 2010; 72: 297–313.
Park S , Dong B , Matsumura F . Rapid activation of c-src kinase by dioxin is mediated by the Cdc37− HSP90 complex as part of ah receptor signaling in MCF10A cells. Biochemistry 2007; 46(3): 899–908.
Moura-Alves P , et al. AhR sensing of bacterial pigments regulates antibacterial defence. Nature 2014; 512(7515): 387–392.
Kimura A , et al. Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production. Int Immunol 2013; 26(4): 209–220.
Wada T , et al. Aryl hydrocarbon receptor modulates NADPH oxidase activity via direct transcriptional regulation of p40phox expression. Mol Pharmacol 2013; 83(5): 1133–1140.
Boutros PC , et al. Dioxin-responsive AHRE-II gene battery: identification by phylogenetic footprinting. Biochem Biophysical Res Commun 2004; 321(3): 707–715.
Kim DW , et al The RelA NF-kappaB subunit and the aryl hydrocarbon receptor (AhR) cooperate to transactivate the c-myc promoter in mammary cells. Oncogene 2000; 19(48): 5498–5506.
Vogel CFA , et al. RelB, a new partner of Aryl hydrocarbon receptor-mediated transcription. Mol Endocrinol 2007; 21(12): 2941–2955.
Vogel CFA , et al. Aryl hydrocarbon receptor signaling regulates NF-κB RelB activation during dendritic-cell differentiation. Immunol Cell Biol 2013; 91(9): 568–575.
Kimura A , et al. Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 2009; 206(9): 2027–2035.
Na YR , et al. Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD. Nat Rev Gastroenterol Hepatol 2019; 16(9): 531–543.
Hadis U , et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity 2011; 34(2): 237–246.
Muller PA , et al. Crosstalk between muscularis macrophages and enteric neurons regulates gastrointestinal motility. Cell 2014; 158(2): 300–313.
Singh V , et al. Histone deacetylase inhibitors valproic acid and sodium butyrate enhance prostaglandins release in lipopolysaccharide-activated primary microglia. Neuroscience 2014; 265: 147–157.
Gordon S , Plüddemann A . Macrophage clearance of apoptotic cells: a critical assessment. Front Immunol 2018; 9: 127.
Stempelj M , et al. Essential role of the JAK/STAT1 signaling pathway in the expression of inducible nitric-oxide synthase in intestinal epithelial cells and its regulation by butyrate. J Biol Chem 2007; 282(13): 9797–9804.
Scott SA , Fu J , Chang PV . Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc Natl Acad Sci 2020; 117(32): 19376–19387.
Bittinger MA , Nguyen LP , Bradfield CA . Aspartate aminotransferase generates proagonists of the aryl hydrocarbon receptor. Mol Pharmacol 2003; 64(3): 550–556.
Dou L , et al. The cardiovascular effect of the uremic solute indole-3 acetic acid. J Am Soc Nephrol 2015; 26(4): 876–887.
Dziarski R , Ulmer AJ , Gupta D . Interactions of CD14 with components of gram-positive bacteria. In: CD14 in the inflammatory response. Karger Publishers; 2000. p. 83–107.
Akira S , Takeda K , Kaisho T . Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol 2001; 2(8): 675–680.
Yamada S , et al. RLRs and TLRs signaling pathways cause aberrant production of inflammatory cytokines/chemokines in an SFTSV infection mouse model. J Virol 2018.
Lin H-Y , et al. Peptidoglycan induces interleukin-6 expression through the TLR2 receptor, JNK, c-Jun, and AP-1 pathways in microglia. J Cell Physiol 2011; 226(6): 1573–1582.
Atarashi K , et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011; 331(6015): 337–341.
Thornton AM , et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol 2010; 184(7): 3433–3441.
Furusawa Y , et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013; 504(7480): 446–450.
Lu Y , et al. Expression profiles of genes in toll-like receptor-mediated signaling of broilers Infected with Clostridium perfringens. Clin Vaccin Immunol 2009; 16(11): 1639.
Collier CT , et al. Coccidia-induced mucogenesis promotes the onset of necrotic enteritis by supporting Clostridium perfringens growth. Vet Immunol Immunopathology 2008; 122(1): 104–115.
Logue CM , et al. In: Swayne DE , editor. Clostridial diseases. Diseases of poultry, 13th ed. Hoboken, NJ, USA: John Wiley & Sons, Ltd.; 2013. p. 943–970.
Rea MC , et al. Classification of bacteriocins from Gram-positive bacteria. In: Prokaryotic antimicrobial peptides. Springer; 2011. p. 29–53.
Draper LA , et al. Lantibiotic resistance. Microbiol Mol Biol Rev 2015; 79(2): 171.
Marcille F , et al. Distribution of genes encoding the trypsin-dependent lantibiotic Ruminococcin A among bacteria isolated from human fecal microbiota. Appl Environ Microbiol 2002; 68(7): 3424.
Crost EH , et al. Production of an antibacterial substance in the digestive tract involved in colonization-resistance against Clostridium perfringens. Anaerobe 2010; 16(6): 597–603.
Benjdia A , Balty C , Berteau O . Radical SAM enzymes in the biosynthesis of ribosomally synthesized and post-translationally modified peptides (RiPPs). Front Chem 2017; 5: 87.
Repka LM , et al. Mechanistic understanding of lanthipeptide biosynthetic enzymes. Chem Rev 2017; 117(8): 5457–5520.
Crost EH , et al. Ruminococcin C, a new anti-Clostridium perfringens bacteriocin produced in the gut by the commensal bacterium Ruminococcus gnavus E1. Biochimie 2011; 93(9): 1487–1494.
Balty C , et al. Ruminococcin C, an anti-clostridial sactipeptide produced by a prominent member of the human microbiota Ruminococcus gnavus. J Biol Chem 2019; 294(40): 14512–14525.
Lin Y , et al. Disruption in the cecal microbiota of chickens challenged with Clostridium perfringens and other factors was alleviated by Bacillus licheniformis supplementation. PloS one 2017; 12(8): e0182426–e0182426.
Li Z , et al. Effects of Lactobacillus acidophilus on gut microbiota composition in broilers challenged with Clostridium perfringens. PloS one 2017; 12(11): e0188634–e0188634.
Bortoluzzi C , et al. Supplementation of protected sodium butyrate alone or in combination with essential oils modulated the cecal microbiota of broiler chickens challenged with coccidia and Clostridium perfringens. Front Sustain Food Syst 2018; 2(72).
Cree BAC , et al. Gut microbiome analysis in neuromyelitis optica reveals overabundance of Clostridium perfringens. Ann Neurol 2016; 80(3): 443–447.