Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 May 1.
Published in final edited form as: Gastroenterology. 2014 Feb 4;146(6):1477–1488. doi: 10.1053/j.gastro.2014.01.060

Regulation of the Immune System by the Resident Intestinal Bacteria

Nobuhiko Kamada 1, Gabriel Núñez 2
PMCID: PMC3995843  NIHMSID: NIHMS564550  PMID: 24503128

Abstract

The microbiota is an important factor in the development of the immune response. The interaction between the gastrointestinal tract and resident microbiota is well-balanced in healthy individuals, but its breakdown can lead to intestinal and extra-intestinal disease. We review current knowledge about the mechanisms that regulate the interaction between the immune system and the microbiota, focusing on the role of resident intestinal bacteria in the development of immune responses. We also discuss mechanisms that prevent immune responses against resident bacteria, and how the indigenous bacteria stimulate the immune system to protect against commensal pathobionts and exogenous pathogens. Unraveling the complex interactions between resident intestinal bacteria and the immune system could improve our understanding of disease pathogenesis and lead to new therapeutic approaches.

Keywords: Microbiota, Th17, regulatory T cell, IgA, innate lymphoid cells


The mammalian gastrointestinal (GI) tract is colonized by trillions of microorganisms, including bacteria, fungi, parasites, and viruses. This internal microbial community that is most abundant in the distal intestine is referred to as the microbiota 1, 2. In healthy individuals, the intestinal microbiota comprises more than 1000 species of bacteria that contribute to GI homeostasis3. One key role of the resident intestinal bacteria is to promote metabolism of nutrients. Some bacterial members of the microbiota is capable of fermenting diet- and/or host-derived undigested carbohydrates, as well as synthesizing vitamins involved in host energy acquisition3. In addition, the resident intestinal bacteria regulate immune homeostasis and pathogen clearance2, 4. The microbiota plays a critical role in the development and function of the GI immune system, but the immune system also regulates the composition and/or function of the microbiota. This bi-directional interaction between the microbiota and the immune system is well-balanced in healthy individuals, but its breakdown can lead to GI diseases, such as inflammatory bowel diseases, as well as extra-intestinal disorders, including metabolic disease2. We review the role of the microbiota, and in particular the resident intestinal bacteria, in the regulation of immune responses in the context of GI homeostasis, host defense, and pathogenesis of GI disease.

Resident Intestinal Bacteria in the Function and Development of the Immune System

Structural development of Gut-Associated Lymphoid Tissues (GALTs)

The role of the microbiota in the development of the immune system has been studied for more than a century. A major advance was the establishment of germ-free (GF) and gnotobiotic animal systems. Rearing of GF animals was first attempted in 1890s and succeeded in the early 1910s. Subsequent advances in the methodology eventually led to the establishment of the basic principles of GF technology in the 1940s5. Although the microbiota was found to be important for optimal acquisition of nutrients in the intestine, the average survival period of mice housed under GF conditions was longer than that of conventionally raised mice, indicating that the microbiota is not essential for animal survival6. However, GF animals exhibit impaired development of GALTs, such as Peyer’s patches (PPs) and isolated lymphoid follicles 68. Notably, a greater number of immunoglobulin (Ig)E+ B cells and lower number of IgA+ B cells were found in the PPs of GF mice, compared with conventionally raised mice9. Colonization of GF mice by commensal microorganisms induces immune reactions in lymphoid cell clusters called germinal centers and IgA development10, 11. Recent studies have begun to elucidate the roles of the microbial molecules, and host signaling pathways that mediate the development of GALTs. For example, NOD1-mediated signaling is required for the development of isolated lymphoid follicles8.

Effector T Cells

The microbiota also have an important role in balancing effector T-cell immune responses in the GI tract12. GF mice have reduced numbers of T-helper (Th)1 and Th17 cells, so the intestinal T-cell immune response in GF animals is primarily controlled by Th2 cells13, 14. The imbalance in Th-cell responses in GF mice can be reversed by reconstitution with conventional microbiota, indicating that microorganisms shape intestinal Th-cell mediated immunity14.

IL17 responses were first identified in the GI tract in association with Helicobacter pylori infection and subsequently an important advance was made when a particular species of commensal Clostridia-related bacteria, called segmented filamentous bacteria (SFB), were found to induce Th17-cell development in the small intestine 1518 (Table 1). Th17 cells are a lineage of CD4+ Th cells, which defend against extracellular microorganisms and are also involved in the development of autoimmune diseases19. GF mice or mice given antibiotics have greatly reduced numbers of Th17 cells in the small intestine16, 20, 21. Conventionally raised mice from colonies lacking SFB also have low numbers of Th17 cells and the numbers of Th17 cells increase upon SFB colonization16, 18, indicating that SFB induces the development of intestinal Th17 cells.

Table 1.

The resident intestinal bacteria regulate the development and function of intestinal immunity

Immune cell Bacteria (bacterial molecules) Host cell mediator Host receptor Mediator Note Ref
Effector T cells
Th17 Segmented filamentous bacteria Intestinal epithelium unknown Serum amyloid A MyD88, TRIF, RIP2 independent 16, 17, 18
Not identified LP DC and Macrophage unknown IL-1β MyD88 dependent 48
Not identified (ATP) CD70hiCD11clow DC P2XR, P2YR IL-6, IL-23 MyD88, TRIF, RIP2 independent 20
Regulatory T cells
Foxp3+ Treg Clostridium clusters IV and XIVa Intestinal epithelium unknown TGF-β MyD88, TRIF, RIP2, Card9 independent 25
Altered Schaedler flora unknown unknown unknown MyD88, TRIF dependent 22
Bacteroides fragilis (PSA) CD11c+ DC TLR2/MyD88 unknown 27, 28
Not identified (SCFAs) Treg cells GPR43 unknown Epigenetic modification of Foxp3 51, 52, 53
Human 17 Clostridium strains (SCFAs) Intestinal epithelium TGF-β unknown 24
Not identified (Butyrate, niacin) LP DC and Macrophage GPR109a RA, IL-10 Induce Foxp3 IL-10-producing Tr1 cells 60
Not identified (SCFAs) Treg cells GPR43 Direct Upregulate GPR15 61
IgA-producing B/plasma cells
IgA+ B cell Segmented filamentous bacteria unknown unknown unknown 11, 70
Not identified Follicular DC TLR/MyD88 TGF-β, CXCL13, BAFF Bacterial ligands synergize with RA 66
Not identified (flagellin) CD11c+CD11b+TLR5+ DC TLR5 RA 69
Not identified TNF-α/iNOS-producing DC TLR/MyD88 TGF-β, BAFF, APRIL Reduced in GF mice 67
Not identified LP DC unknown RA Synergize with IL-5, IL-6 68
CD11b+ IgA+ PC Not identified unknown MyD88 unknown CD11bIgA+PCs are microbe-independent 65
Group 3 Innate lymphoid cells
RORγt+ NKp46+CD127+NK1.1−/int ILC Not identified unknown unknown unknown Reduced in GF mice 33, 34
NKp46+ ILC Lactobacilli (indole-3-aldehyde) NKp46+ ILC AhR Direct Enhance IL-22 production 73
CD4+RORγt+CD127+ LTi cell Not identified CX3CR1+ DC unknown IL-23 Enhance IL-22 production 74, 75
c-kit+CD127+Sca-1NKp46CD4 ILC Not identified (flagellin) CD103+CD11b+ DC TLR5 IL-23 Enhance IL-22 production 76
RORγt+ LTi and RORγt+NKp46+ ILC Not identified Intestinal epithelium unknown IL-25 Activates IL-17BR+ DC to suppress IL-22 by ILC 37
RORγt+ NKp46+CD127+NK1.1low LTi Not identified Intestinal epithelium unknown IL-7 Stabilizes the expression of RORγt 36
Mono-, poly-morphonuclear phagocytes
CD11b+ macrophage Not identified unknown MyD88 unknown Induce pro-IL-1β 38
CD11b+ macrophage, CD11c+ DC Not identified unknown MyD88 Direct Induce pro-IL-1β 49
CD11b+ CD11c−/+ macrophage Not identified unknown unknown unknown Enhance IL-10 production 41
Neutrophil Not identified Neutrophils unknown Direct Induce pro-IL-1β 42
Not identified (SCFAs) Neutrophils GPR43 Direct Induce ROS, increase phagocytic activity 79
Not identified (PGN) Neutrophils NOD1 Direct Enhance bactericidal capacity 43

However, SFB are not the only bacteria that can induce Th17 cells. For example, colonization of GF mice with Altered Schaedler Flora (ASF), a cocktail of 8 defined commensals, also significantly increased the numbers of Th17 cells, although the ASF is less effective than SFB22. Enteric pathogens can also induce a Th17-mediated response, even in the absence of commensal microbes. For example, monocolonization of GF mice with the enteric pathogen Citrobacter rodentium, which is used to model enteropathogenic Escherichia coli and enterohemorrhagic E coli infection in mice, increases the numbers of intestinal Th17 cells23. Furthermore, colonization of GF mice with the human intestinal microbiota promotes development of Th17 cells, even though SFB is not included 14, 24. So, SFB, as well as other members of the human intestinal microbiota and certain bacterial pathogens, are strong inducers of Th17-cell development.

T Regulatory (Treg) cells

Similar to effector T cells, the resident intestinal bacteria is essential for the development and function of Foxp3+ Treg cells in the intestine. In the absence of the microbiota, the number of inducible Foxp3+ Helios Treg (iTreg) cells, but not Foxp3+Helios+ thymic-derived natural Treg cells, is significantly reduced in the colonic lamina propria (LP), but not the LP of the small intestine or mesenteric lymph nodes22, 25. Reduced development of iTreg cells in GF mice can be restored by re-colonization with the intestinal microbiota, indicating that commensals are involved in the generation of colonic Treg cells.

Certain bacterial species within the intestinal microbiota have been shown to possess Treg-inducing activity. For example, colonization of GF mice with Clostridia species resulted in robust accumulation of iTreg cells, whereas colonization with mouse intestinal Bacteroides spp., Lactobacillus, or SFB induced no or small numbers of iTreg cells in the colonic LP25. Atarashi et al., identified 46 strains of the Clostridium genus belonging to clusters IV and XIVa (also known as Clostridium leptum and coccoides groups, respectively) as Treg-inducing Clostridia in mice25 (Table 1). Colonization of GF mice with ASF is also sufficient to induce the development of iTreg cells in the intestine22 (Table 1). ASF contains 3 Clostridia species belonging to the Clostridium cluster XIV; these Clostridia species may be responsible for the Treg-inducing capacity of the ASF26. These Treg-inducing Clostridia species are present in the mouse and human intestine. In fact, Atarashi et al. further identified 17 Treg-inducing Clostridia strains in the microbiota of healthy humans24 (Table 1). These 17 strains belong to the Clostridium clusters IV, XIVa, and XVIII, so similar bacteria have Treg-inducing activities in humans and mice24.

Unlike the mouse intestinal microbiota, Clostridia are not the only bacterial species in the human microbiota that promotes Treg-cell development. For example, colonization of mice with Bacteroides fragilis, a human commensal, robustly facilitates the differentiation of Treg cells and interleukin (IL)10 production by Treg cells, whereas mouse commensal strains of Bacteroides only weakly induce development of Treg cells24, 27, 28 (Table 1). However, it is unclear whether B fragilis promotes induction of intestinal Treg cells in humans. Although commensal Clostridia are potent inducers of Treg cell development in the intestine of mice and humans, other bacterial species might also induce Treg-cell responses.

Development of B Cells

B cells protect against microbial infections by producing Igs29. Early stages of B-cell development occur not only in fetal liver and the bone marrow, but also in the intestinal mucosa30. Receptors on developing B cells within the intestine are edited differently from those on bone marrow B cells. Intestine-specific editing of B-cell receptors is regulated by extracellular signals induced by resident microorganisms30. The commensal microbiota therefore regulates development of intestine-specific B-cell receptor repertoires.

The microbiota also influences Ig responses within the intestinal mucosa. IgA is secreted primarily on the surface of mucosal tissues, including those of the GI tract, and regulates mucosal homeostasis. Within the GI tract, most IgA-producing B cells mature in PPs upon stimulation by commensal microorganisms31, although little is understood about the mechanisms of this process. One possible mechanism involves the effects of the microbiota on the development and organization of GALTs, where IgA-producing cells are generated. In systemic lymphoid organs such as the spleen, intestinal lymphoid follicles contain IgD+- IgM+-naïve B cells under physiological conditions. In response to antigen stimulation (associated with infection or vaccination), germinal centers form in lymphoid follicles. Here, B cells undergo maturation and proliferation, including class-switching recombination and somatic hypermutation, resulting in appropriate antigen-specific humoral responses31. Unlike in systemic lymphoid organs, germinal center formation is observed under steady-state conditions in GALTs—especially PPs, because the GI tract is continuously exposed to commensal bacterial antigens. In GF mice, the lack of microbiota-derived signals results in immature formation of germinal centers in the PPs and reduced generation of IgA-producing B cells31.

Innate Lymphoid Cells (ILCs)

ILCs are innate immune cells that have functional characteristics commonly associated with T cells32. ILCs arise from common lymphoid precursors; there are 3 subgroups, based on their different functional properties: T-bet+ (group 1), GATA-3+ (group 2), and RORγt+ (group 3)32. There is increasing evidence of the important roles for group 3 (RORγt+) ILCs in intestinal immunity. These ILCs resemble Th17 cells in their cytokine profile (production of IL22 and/or IL17 upon IL23 and IL1β stimulation)32. The extent to which the commensal microbiota influences the development of group 3 ILCs in the intestine is controversial12. Some studies have shown that the microbiota is required for the differentiation of group 3 ILCs33, 34. Consistently, the proportion of RORγt+NKp46+CD127+NK1.1 or RORγt+NKp46+CD127+NK1.1int ILCs is significantly reduced in GF mice33, 34 (Table 1). In the absence of the microbiota, production of IL22 by ILCs is reduced, indicating that the function of ILCs is regulated by commensals33 (Table 1). In contrast, other studies found that the microbiota does not affect development of RORγt+ ILCs3537.

Lamina Propria Phagocytes

Mononuclear phagocytes such as macrophages and dendritic cells (DCs), along with polymorphonuclear phagocytes (neutrophils), protect many organs against microbial infections, including the GI tract. The resident microbiota appears to regulate the functions of these phagocytes within the intestine. The microbiota is required to upregulate production of pro-IL1β (the precursor of IL1β) in resident macrophages38 (Table 1). However, full processing and secretion of pro-IL1β does not occur in phagocytes until they encounter pathogenic microorganisms. Pathogens such as Salmonella and Pseudomonas aeruginosa can rapidly induce secretion of the mature form of IL1β from intestinal macrophages, which contributes to the elimination of infectious pathogens38. Unlike commensals, these pathogens can inject flagellin into the cytosol via type III secretion systems, thereby activating the Nod-like receptor (NLR)C4 inflammasome to induce caspase-1–dependent processing of pro-IL1β38. Commensals therefore regulate the protective functions of intestinal macrophages by promoting constitutive expression of pro-IL1β.

The intestinal microbiota also contributes to the anti-inflammatory functions of intestinal macrophages. Intestinal macrophages spontaneously produce the anti-inflammatory cytokine IL10, which is important for GI homeostasis3941 and controlled by the microbiota41 (Table 1). In addition, the microbiota modulates the function of neutrophils. During intestinal infection with the pathogen Clostrodium difficile, commensals translocate to the intestinal tissue, where they induce expression of pro-IL1β in recruited neutrophils; this protects against C difficile infection-associated mortality, by promoting the removal of translocated commensals42 (Table 1). The intestinal microbiota also influences systemic neutrophil responses. For instance, peptidoglycan molecules derived from resident bacteria can prime circulating blood neutrophils to boost their bactericidal ability via NOD1 signaling, which protects against infection with Streptococcus pneumoniae43 (Table 1). The microbiota is therefore important for the regulation and function of intestinal phagocytes.

Regulation of the Immune System by Resident Intestinal Bacteria

Intestinal Th17 cells

In vitro studies reported that transforming growth factor (TGF)β and IL6 or IL21 regulate differentiation of Th17 cells, whereas their expansion is mediated by IL234448. However, intestinal Th17 cells develop in mice deficient in IL6, IL23, or the IL21 receptor, so other factors appear to contribute to this process in vivo 16, 49. Mice deficient in IL1β or the IL1 receptor have reduced numbers of intestinal Th17 cells49. IL1β production in resident intestinal phagocytes is reduced in GF mice, so commensal microorganisms might induce production of IL1β in the intestine to promote Th17 development 49. Administration of IL1β is sufficient to induce development of intestinal Th17 cells in GF mice49. The intestinal microbiota therefore appears to promote development of Th17 cells, at least in part, through the induction of IL1β (but not IL6)49 (Table 1).

How does the microbiota induce IL1β and Th17 cells in the intestines? Mice deficient in MyD88, an adaptor required for Toll-like receptor (TLR), IL1-like, and IL18 receptor signaling, produce lower levels of IL1β, and develop fewer Th17 cells in the intestine49. Commensals might therefore induce IL1β via TLR–MyD88 signaling in the innate immune response pathway50.

Atarashi et al. reported that luminal ATP produced by the microbiota promotes development of Th17 cells by activating DCs in the LP20 (Table 1). However, SFB-induced development of Th17 cells was shown to be independent of ATP 18. Ivanov et al. reported that colonization by SFB leads to production of serum amyloid A proteins by intestinal epithelial cells; this might induce Th17-cell development independently of ATP18 (Table 1). However, little is known about the in vivo roles of serum amyloid A proteins in inducing intestinal Th17 cells. Nonetheless, the microbiota shapes the balance of effector T cell populations—especially of Th17 cells. However, further research is needed to understand the mechanism by which commensals induce Th17 cells in the intestine.

Foxp3+-inducible Treg Cells

There is increasing evidence that commensals induce the development of Treg cells via multiple mechanisms. There is controversy over the involvement of host pattern recognition receptors in this process. Induction of colonic Treg cells by mouse Clostridium clusters IV and XIVa was found to be independent of TLRs, NODs, and Dectin125 (Table 1). However, ASF (containing 3 Clostridium cluster XIV strains)-induced development of Treg cells is reduced in mice deficient in MyD88 and TRIF22 (Table 1). Moreover, activation of TLR2 signaling by polysaccharide A was required for induction of Treg cells and production of IL10 in mice following colonization with B fragilis 28 (Table 1). Further studies are needed to determine the precise roles of pattern recognition receptors in the development of Treg cells.

Commensal metabolites were reported to induce the development of Treg cells. Among bacterial metabolites, short-chain fatty acids (SCFAs) have been proposed to regulate development and function of colonic Treg cells5153. In GF mice, reduced concentrations of luminal SCFAs (such as acetate, propionate, isobutyrate, and butyrate) correlated with impaired development of intestinal Treg cells5153. Importantly, reconstitution with commensal bacteria or administration of SCFAs restores the numbers of Treg cells in GF mice, supporting a role for bacterial metabolites in Treg cell development5153. Several G protein-coupled receptors (GPRs) expressed by intestinal epithelial cells and/or hematopoietic cells can be activated by SCFAs. So, SCFAs affect different types of cells in the intestine to regulate homeostasis in the GI tract54, 55. SCFAs affect Treg cell function via epigenetic regulation of the Foxp3 gene5153. SCFAs, particularly butyrate, can act directly on T cells, increasing acetylation of the Foxp3 locus5153 (Table 1). Consistently, mice deficient in Ffar2, which encodes Gpr43, have reduced numbers of Treg cells, which cannot be restored by SCFA administration51. In addition to their direct effects on Treg cells, SCFAs can also promote development of Treg cells via non-immune cells. For example, colonization of GF mice with 17 human Clostridium strains induced the luminal levels of acetate, propionate, isobutyrate, and butyrate to promote TGFβ production by the intestinal epithelium24. SCFA-induced production of TGFβ appears to be necessary for the induction of Treg cells by commensal Clostridia strains in humans and mice24, 25.

Although little is known about the mechanism by which SCFAs, via TGFβ, promote development of Treg cells, LP DCs appear to be involved. CD103-expressing subsets of DCs in the LP (including CD103+CD11b+CD11c+ and CD103+CD11bCD11c+) induce differentiation of Treg cell from naïve CD4+ T cells via TGFβ and retinoic acid (RA)5658. In addition to DC subsets, F4/80+CD11b+CD11c LP macrophages produce RA and induce the differentiation of intestinal Treg cells in the presence of TGFβ40. SCFA-induced TGFβ might therefore regulate intestinal Treg cells along with LP DCs and macrophages. Depletion of CD103-expressing LP DCs reduces numbers of intestinal Treg cells, even in the presence of the intestinal microbiota59. Consistent with these observations, it was recently reported that Gpr109a, a GPR family member, is expressed in intestinal macrophages and DCs 60. Notably, butyrate and niacin (vitamin B3) produced by commensal bacteria can activate Gpr109a-expressing DCs and macrophages via Gpr109a to promote the differentiation of Foxp3+ Treg cells and IL-10-producing Tr1 cells 60.

In addition to de novo generation of Treg cells, the intestinal microbiota can promote homing of Treg cells to the large intestine. Kim et al. found that the GPR15 receptor is primarily expressed on Treg cells in the large intestine, where it acts as a homing molecule for Treg cells24 (Table 1). Mice deficient in GPR15 are characterized by reduced numbers of Foxp3+ Treg cells in the large intestine, but not the small intestine or the spleen61. GPR15 expression on Treg cells is reduced when the resident microbiota is depleted with broad spectrum antibiotics61. GPR15 expression is induced by SCFA administration, so homing of Treg cells appears to also be regulated by bacterial metabolites51. Likewise, dietary folic acids regulate the survival of Foxp3+ Treg cells in the colon62. Mammals do not have synthetic pathways for de novo production of folic acids, so folic acids derived from commensals are likely to be involved in the induction of intestinal Treg cells63.

Induction of Treg cells by the microbiota protects mice from colitis, infection with enteric pathogens, and allergic diarrhea24, 25, 64. For example, Clostridium-induced Treg cells suppress colitis and allergic diarrhea in mice24, 25. Likewise, IL10-producing Treg cells, induced by B fragilis, reduce intestinal inflammation caused by Helicobacter hepaticus28, 64. These studies indicate that certain commensal species, particularly those belonging to the Clostridium genus, contribute to the de novo generation, homing, and functional maturation of Treg cells in the colon to help maintain GI homeostasis.

IgA-producing B Cells

There are several pathways by which the microbiota regulates development of IgA-producing cells in the intestine. One pathway involves MyD88 signaling. The intestines of mice deficient in MyD88 have reduced numbers of CD11b+ IgA+, but not CD11b IgA+, plasma cells65 (Table 1). By unclear mechanisms, commensals activate MyD88 signaling in LP DCs and follicular DCs—cells that promote the generation of IgA+ B cells. In response to microbial stimulation, follicular DCs in PPs secrete TGFβ, CXCL13, and B-cell activating factor (BAFF, a member of the TNF family), which stimulates class switching and IgA production66 (Table 1). Likewise, the microbiota activates MyD88 signaling in subsets of LP DCs, leading to their expression of RA, TGFβ, TNFα, inducible nitric oxide synthetase, BAFF, and proliferation-inducing ligand to promote the generation of IgA+ B cells6769 (Table 1).

Although the microbiota is important for the development and maturation of intestinal B cells, the specific bacteria that mediate these processes have not been identified. Monocolonization of GF mice with SFB, in addition to Th17 cells, can induce IgA production11, 70 (Table 1). However, the amount of IgA in SFB monocolonized mice is low compared to mice colonized with conventional microbiota, indicating that other bacterial species, or mixtures of commensals, are required for maximal induction of intestinal IgA11. On the other hand, transient monocolonization with a mutant strain of E coli that is unable to proliferate in vivo induces amounts of intestinal IgA comparable to those observed following colonization with multiple commensal bacteria71. Although the IgA induced is specific for this bacterium, exposure of the E coli-colonized mice to different bacteria limits the persistence of E coli-specific IgA memory response71. Continuous stimulation by bacteria might therefore be required for sustained production of commensal-specific IgA in the intestine.

Group 3 Innate Lymphoid Cells

The mechanisms by which commensal bacteria regulate the development and function of ILCs are also unclear. One mechanism could involve the direct activation of ILCs by bacterial components. Human CD127+ RORγt+ ILCs express TLR1, TLR2, and TLR6, and stimulation with TLR2 ligands induces the production of IL22 in these cells72. In addition, bacterial metabolites can activate ILCs. Indole-3-aldehyde, a tryptophan catabolite produced by the microbiota, stimulate ILCs through the aryl hydrocarbon receptor (AhR) to induce production of IL22 by ILCs73 (Table 1). Microbial products therefore appear to directly activate ILCs. There is also evidence that the intestinal microbiota regulates the function of ILCs indirectly, by activating other immune cells. For example, commensal bacteria can orchestrate the recruitment of CX3CR1+ intestinal macrophages to the intestine, which subsequently modulate IL-22 production by ILCs74, 75 (Table 1). Additionally, in response to bacterial products, CD103+CD11b+ LP DCs produce IL23, through stimulation of TLR5, leading to secretion of IL22 by RORγt+ ILCs76 (Table 1). On the other hand, GF mice have increased production of IL22 by RORγt+ ILCs, indicating that the microbiota could be a negative regulator of IL22 production37.

IL25, a cytokine generated by intestinal epithelial cells upon stimulation with commensals, activates LP DCs via the IL17BR to inhibit the production of IL22 by ILCs through an unknown mechanism37 (Table 1). The microbiota therefore acts as a positive and negative regulator of intestinal ILCs. The dual role of the microbiota in ILC activation and development might arise through differences in access of microbes to immune cells. Under steady-state conditions, the intestinal epithelium is intact and the microbiota may stimulate epithelial cells to repress ILC function. However, during inflammation, the epithelial barrier is disrupted, allowing the translocation of commensals and luminal products into the intestinal tissue, which activates LP immune cells, including ILCs.

Activation of ILC functions by commensals is important for protection against microbial infection. For example, IL22-mediated production of RegIIIγ, a secreted antimicrobial peptide, by the intestinal epithelium protects against enteric infection by C rodentium33, 77, 78. Likewise, administration of tryptophan catabolites increases production of IL22 by ILCs, which is associated with protection against fungal infection and dextran sodium sulfate-induced colitis73. In addition, the intestinal microbiota contributes to the stabilization of RORγt expression in group 3 ILCs by stimulating epithelial cells to produce IL7—a cytokine that stabilizes expression of RORγt36. GF mice express reduced levels of IL7 in the intestinal epithelium, which subsequently downregulates expression of the RORγt in ILCs36. Decreased RORγt expression in ILCs leads to the conversion of ILCs into IFNγ-producing pathogenic ILCs, which promote intestinal inflammation36 (Table 1). The microbiota can promote GI homeostasis and protect against infection and inflammation by regulating intestinal ILCs—particularly group 3 RORγt ILCs.

Lamina Propria Phagocytes

LP mononuclear phagocytes, including macrophages, express pro-IL1β even in the steady state, which is important for rapid production of mature IL1β in response to pathogen invasion38, 49. Accumulation of pro-IL1β in LP macrophages requires resident microbiota and commensal-induced MyD88 signaling38, 49 (Table 1). Likewise, the anti-inflammatory function of intestinal macrophages, such as spontaneous production of IL10, is also controlled by the microbiota, in a MyD88-independent manner41 (Table 1). In addition, the function of neutrophils is regulated by commensal by-products such as SCFAs, which directly activate neutrophils and increase their bactericidal activity through GPR4379 (Table 1).

Regulation of Resident Intestinal Bacteria by the Immune System

Compartmentalization of Intestinal Microbes

Although the resident intestinal bacteria are important for the development of intestinal immune responses, excessive immune stimulation by commensals can lead to inappropriate activation of immune cells and harmful intestinal inflammation. To avoid excessive stimulation by resident microbes, the intestinal immune system is separated from luminal microorganisms by a physical barrier. In the colon, the epithelial surface is covered by an inner mucus layer that shields the immune system from microbes and mechanical stress, whereas large numbers of commensal bacteria colonize the outer mucus layer80, 81 (Figure 1). Also, RegIIIγ limits the penetration of luminal bacteria into the intestinal mucosa and acts as an additional barrier82 (Figure 1). Group 3 ILCs contribute to the induction of RegIIIγ by epithelial cells by producing IL22. Depletion of IL22-producing ILCs leads to dissemination of the commensal Alcaligenes xylosoxidians, which promotes intestinal damage and systemic inflammation83.

Figure 1. Multiple Firewalls Limit Immune Responses Against Resident Intestinal Bacteria.

Figure 1

The intestine has firewalls that limit inappropriate immune responses against commensal bacteria. One mechanism involves compartmentalization of the resident bacteria by a thick mucus layer, anti-microbial peptides (such as RegIIIγ), or secreted IgA, which all prevent access of the resident bacteria to the epithelium. Neutrophils can migrate out to the intestinal lumen and build intraluminal cellular casts that encapsulate commensals, limiting the penetration of luminal microbes. A second mechanism involves microbial hyporesponsiveness of resident intestinal macrophages. In contrast to monocytes and macrophages in systemic tissues, intestinal macrophages are hypo-responsive to microbial stimuli and produce only limited amounts of inflammatory cytokines such as TNFα or IL12. Furthermore, resident phagocytes spontaneously produce anti-inflammatory cytokines, such as IL10. A third mechanism involves active suppression of microbe-reactive effector T cell responses by Foxp3+ iTreg cells and Foxp3 Tr1 cells via production of IL10. In addition, MHC-II+ ILCs present bacterial antigens, which limits activation of commensal-reactive CD4+ T cells.

Secreted IgA also has an important role in the compartmentalization of the commensal microbiota on the mucosal surface (Figure 1). Polymeric IgA is transported via the polymeric Ig receptor (pIgR), expressed on intestinal epithelial cells, and released into the intestinal lumen as secretory IgA84. Secretory IgA binds to luminal commensal bacteria and prevents their penetration across the epithelial barrier31, 85.

Neutrophils also contribute to the compartmentalization of commensal organisms. Although there is no evidence that neutrophils have a role in the compartmentalization of commensals under steady-state conditions, neutrophils migrate to the intestinal lumen after pathogen invasion and build intraluminal cellular casts that encapsulate commensals, thereby limiting their translocation 86 (Figure 1). Furthermore, neutrophils are recruited to the intestinal tissue and limit the translocation of commensals after epithelial damage from release of toxins by C difficile 42. The mucosal epithelium and the immune system therefore limit access of the commensal microbiota to host tissues, to maintain their symbiotic relationship.

Local Hyporesponsiveness Against Resident Intestinal Bacteria

In addition to compartmentalization of luminal microorganisms, the GI immune system has developed multiple mechanisms to avoid inappropriate immune responses to the commensal microbiota. Different subsets of regulatory Treg cells in the intestine (including Foxp3+ iTregs and IL10-producing Foxp3 Tr1 cells) 87 are thought to be required to suppress proliferation of commensal-reactive effector T cells. In the absence of these Treg cells, commensal-reactive T cells in the intestine proliferate, leading to colitis88, 89. Adoptive transfer of T cells specific for flagellin antigens derived from certain commensals, such as H hepaticus, causes colitis in mice, but not in GF mice90, 91. Different subsets of Treg cells (CD25+ and CD25 CD45RBlowCD4+ T cells) suppress proliferation of commensal-reactive T cells89, via IL10 but not TGFβ. These findings indicate that CD25+Foxp3+IL10+ Treg cells and CD25Foxp3 IL10+ Tr1 cells are both involved in this regulatory mechanism87, 89. So, suppression of commensal reactive effector T cells by Treg cells is important for prevention of immune reactivity against the commensal microbiota (Figure 1).

A recent study identified a unique role for ILCs in regulating effector T-cell responses against commensals (Figure 1). Hepworth et al. found that intestinal group 3 ILCs express MHC-II and are capable of processing and presenting antigens. Notably, MHC-II+ ILCs present commensal antigens to bacteria-reactive CD4+ T cells to inhibit their activation and to suppress pathologic immune responses92 (Figure 1). In addition to Treg cells, intestinal mononuclear phagocytes, such as macrophages, also contribute to intestinal homeostasis, by limiting excessive immune responses against commensal bacteria (Figure 1). Macrophages recognize and eliminate bacteria via phagocytic activity and induction of robust inflammatory responses. Beside their anti-microbial and inflammatory roles, macrophages can reduce inflammation by producing anti-inflammatory cytokines such as IL10 and TGFβ93. In the GI tract, intestinal macrophages are hypo-responsive to microbial ligands, and do not produce significant amounts of inflammatory molecules upon bacterial stimulation3840. Yet, intestinal macrophages retain their phagocytic and bacteriocidal functions and spontaneously secrete IL1039, 40, 94 (Figure 1). Intestinal macrophages can phagocytose luminal bacteria that penetrate the epithelial barrier without inducing excessive inflammatory responses (Figure 1). Taken together, the intestinal immune system has developed multiple regulatory mechanisms of tolerance against the resident intestinal bacteria.

The Immune System Shapes the Composition and Function of the Intestinal Bacteria

The intestinal immune system actively influences the structure and function of the commensal microbiota. Defective intestinal IgA responses, due to deficiency of or mutations in activation-induced cytidine deaminase, alter the composition of the gut microbiota95, 96 (Figure 2). Likewise, loss of inhibitory co-receptor programmed death-1 reduces the bacterial-binding capacity of IgA, which alters the intestinal bacterial community97. In addition to altering the composition of the resident intestinal bacteria, binding of IgA to the commensal Bacteroides thetaiotaomicron affects expression of bacterial genes98 (Figure 2). Importantly, an altered microbial community can facilitate outgrowth of potentially pathogenic microorganisms (pathobionts) or modify gene expression in the commensal bacteria, allowing harmless bacteria to become harmful. Defects in the function of group 3 ILCs can lead to abnormal expansion of SFB99 (Figure 2), which can increases responses of intestinal Th17 cells and promote the development of spontaneous colitis under certain conditions99 (Figure 2). This abnormal accumulation of SFB appears to require a lack of IL22 production by ILCs99. The composition of the microbiota in IL22-deficient mice is altered compared with wild-type mice; this alteration is transmissible and colitogenic100. Furthermore, T-bet deficiency in Rag-deficient mice leads to accumulation of potential pathobionts Klebsiella pneumoniae, Proteus mirabilis, and Helicobacter typhlonius, which are associated with spontaneous colitis 101103 (Figure 2).

Figure 2. Regulation of the Resident Intestinal Bacteria by the Immune System.

Figure 2

Direct binding of IgA to the commensal B thetaiotaomicron regulates bacterial gene expression. IgA also regulates the composition of the commensal bacterial community. IL22 produced by RORgt+ group 3 ILCs induces production of RegIIIγ by IECs, which limits the growth of SFB. Overgrowth of SFB increases numbers of Th17 cells and Th17-cell–mediated intestinal inflammation. RORγt+ ILCs limit expansion of the commensal pathobiont A xylosoxidians in PPs. Expression of T-bet by ILCs limits the accumulation of certain commensal pathobionts, such as K pneumoniae, P mirabilis, or H typhlonius. IL18 is induced by the intestinal epithelium via a NLRP3-dependent pathway. IL18 limits the overgrowth of the pathobionts Prevotellaceae and TM7 via unidentified mechanisms.

Defects in the NLR family have also been associated with dysbiosis. Mice deficient in NLRP6 have impaired production of IL18, which is associated with an abnormal expansion of the commensals Prevotellaceae and TM7104 (Figure 2). However, the observation that immune factors such as IL22 and host innate receptors regulate the composition of the intestinal microbiota should be taken with caution—recent studies evaluating TLR-deficient mice found that the composition of the microbiota largely depends on maternal transmission of the microbiota from isolated mouse colonies, rather than any specific TLR defect105. Because few of these studies were performed in littermates, the observed changes in mutant mice might result from differences in ancestry, rather than an immune defect.

Perspectives

Mounting evidence indicates that the intestinal microbiota regulates the development and function of the immune system, which in turn, shapes the microbial community and/or regulates immune responses against the microbiota. However, diet is an important determinant of the composition and distribution of the microbiota4. Bacterial species in the intestine have distinct nutritional requirements, based on their distinct metabolic pathways and abilities to access luminal nutrient sources. Commensal microorganisms regulate each other through competition as well as by providing nutrients based on their metabolic profiles23, 106. Therefore, competition for nutrients among different commensal species determines, at least in part, the balance and structure of the intestinal microbial community.

However, the immune system could, at least partially, regulate the composition and/or amounts of luminal nutrients. For instance, enteric inflammation may influence the intestinal microbial community by altering luminal metabolites and nutrients in the intestine. There is evidence that intestinal inflammation can affect luminal nutrient availability, leading to a microbial shift within the intestine107, 108. More research is needed to increase our understanding of how the immune system affects luminal metabolic profiles; this could provide new insights into the mechanisms by which immune cells control the intestinal microbiota. Moreover, novel nutritional therapies, based on better understanding of the metabolic pathways that regulate commensal populations, could be developed to correct disease-associated dysbiosis.

Acknowledgments

We thank Grace Chen, Peter Kuffa, and Melody Zeng for critical review of the manuscript. This work is supported by a Fellowship grant from the Crohn’s and Colitis Foundation of America (N.K.), the University of Michigan Gastrointestinal Peptide Research Center Grant 5 P30 DK034933 (N.K.) and grants from the National Institutes of Health DK61707, DK091191, DK095782 and the Bill & Melinda Gates Foundation (G.N.).

Footnotes

The authors disclose no conflicts

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Hooper LV, Macpherson AJ. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol. 2010;10:159–69. doi: 10.1038/nri2710. [DOI] [PubMed] [Google Scholar]
  • 2.Kamada N, Seo SU, Chen GY, Nunez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13:321–35. doi: 10.1038/nri3430. [DOI] [PubMed] [Google Scholar]
  • 3.Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–30. doi: 10.1038/nature11550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Kamada N, Chen GY, Inohara N, Nunez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14:685–90. doi: 10.1038/ni.2608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Reyniers JA, Trexler PC. Germfree research: a basic study in host-contaminant relationship. I. General and theoretical aspects of the problem. Bull N Y Acad Med. 1955;31:231–5. [PMC free article] [PubMed] [Google Scholar]
  • 6.Gordon HA, Bruckner-Kardoss E, Wostmann BS. Aging in germ-free mice: life tables and lesions observed at natural death. J Gerontol. 1966;21:380–7. doi: 10.1093/geronj/21.3.380. [DOI] [PubMed] [Google Scholar]
  • 7.Hamada H, Hiroi T, Nishiyama Y, Takahashi H, Masunaga Y, Hachimura S, Kaminogawa S, Takahashi-Iwanaga H, Iwanaga T, Kiyono H, Yamamoto H, Ishikawa H. Identification of multiple isolated lymphoid follicles on the antimesenteric wall of the mouse small intestine. J Immunol. 2002;168:57–64. doi: 10.4049/jimmunol.168.1.57. [DOI] [PubMed] [Google Scholar]
  • 8.Bouskra D, Brezillon C, Berard M, Werts C, Varona R, Boneca IG, Eberl G. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature. 2008;456:507–10. doi: 10.1038/nature07450. [DOI] [PubMed] [Google Scholar]
  • 9.Durkin HG, Bazin H, Waksman BH. Origin and fate of IgE-bearing lymphocytes. I. Peyer’s patches as differentiation site of cells. Simultaneously bearing IgA and IgE. J Exp Med. 1981;154:640–8. doi: 10.1084/jem.154.3.640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Shroff KE, Meslin K, Cebra JJ. Commensal enteric bacteria engender a self-limiting humoral mucosal immune response while permanently colonizing the gut. Infect Immun. 1995;63:3904–13. doi: 10.1128/iai.63.10.3904-3913.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Talham GL, Jiang HQ, Bos NA, Cebra JJ. Segmented filamentous bacteria are potent stimuli of a physiologically normal state of the murine gut mucosal immune system. Infect Immun. 1999;67:1992–2000. doi: 10.1128/iai.67.4.1992-2000.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kamada N, Nunez G. Role of the gut microbiota in the development and function of lymphoid cells. J Immunol. 2013;190:1389–95. doi: 10.4049/jimmunol.1203100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122:107–18. doi: 10.1016/j.cell.2005.05.007. [DOI] [PubMed] [Google Scholar]
  • 14.Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR, Umesaki Y, Mathis D, Benoist C, Relman DA, Kasper DL. Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 2012;149:1578–93. doi: 10.1016/j.cell.2012.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Luzza F, Parrello T, Monteleone G, Sebkova L, Romano M, Zarrilli R, Imeneo M, Pallone F. Up-regulation of IL-17 is associated with bioactive IL-8 expression in Helicobacter pylori-infected human gastric mucosa. J Immunol. 2000;165:5332–7. doi: 10.4049/jimmunol.165.9.5332. [DOI] [PubMed] [Google Scholar]
  • 16.Ivanov, de Frutos RL, Manel N, Yoshinaga K, Rifkin DB, Sartor RB, Finlay BB, Littman DR. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe. 2008;4:337–49. doi: 10.1016/j.chom.2008.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, Bridonneau C, Rochet V, Pisi A, De Paepe M, Brandi G, Eberl G, Snel J, Kelly D, Cerf-Bensussan N. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity. 2009;31:677–89. doi: 10.1016/j.immuni.2009.08.020. [DOI] [PubMed] [Google Scholar]
  • 18.Ivanov, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139:485–98. doi: 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Littman DR, Rudensky AY. Th17 and regulatory T cells in mediating and restraining inflammation. Cell. 2010;140:845–58. doi: 10.1016/j.cell.2010.02.021. [DOI] [PubMed] [Google Scholar]
  • 20.Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, Takeda K. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455:808–12. doi: 10.1038/nature07240. [DOI] [PubMed] [Google Scholar]
  • 21.Hall JA, Bouladoux N, Sun CM, Wohlfert EA, Blank RB, Zhu Q, Grigg ME, Berzofsky JA, Belkaid Y. Commensal DNA limits regulatory T cell conversion and is a natural adjuvant of intestinal immune responses. Immunity. 2008;29:637–49. doi: 10.1016/j.immuni.2008.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Geuking MB, Cahenzli J, Lawson MA, Ng DC, Slack E, Hapfelmeier S, McCoy KD, Macpherson AJ. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity. 2011;34:794–806. doi: 10.1016/j.immuni.2011.03.021. [DOI] [PubMed] [Google Scholar]
  • 23.Kamada N, Kim YG, Sham HP, Vallance BA, Puente JL, Martens EC, Nunez G. Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. Science. 2012;336:1325–9. doi: 10.1126/science.1222195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B, Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K. T induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013 doi: 10.1038/nature12331. [DOI] [PubMed] [Google Scholar]
  • 25.Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S, Ivanov, Umesaki Y, Itoh K, Honda K. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011;331:337–41. doi: 10.1126/science.1198469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Dewhirst FE, Chien CC, Paster BJ, Ericson RL, Orcutt RP, Schauer DB, Fox JG. Phylogeny of the defined murine microbiota: altered Schaedler flora. Appl Environ Microbiol. 1999;65:3287–92. doi: 10.1128/aem.65.8.3287-3292.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Round JL, Mazmanian SK. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci U S A. 2010;107:12204–9. doi: 10.1073/pnas.0909122107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332:974–7. doi: 10.1126/science.1206095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Macpherson AJ, Geuking MB, McCoy KD. Homeland security: IgA immunity at the frontiers of the body. Trends Immunol. 2012;33:160–7. doi: 10.1016/j.it.2012.02.002. [DOI] [PubMed] [Google Scholar]
  • 30.Wesemann DR, Portuguese AJ, Meyers RM, Gallagher MP, Cluff-Jones K, Magee JM, Panchakshari RA, Rodig SJ, Kepler TB, Alt FW. Microbial colonization influences early B-lineage development in the gut lamina propria. Nature. 2013;501:112–5. doi: 10.1038/nature12496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fagarasan S, Kawamoto S, Kanagawa O, Suzuki K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu Rev Immunol. 2010;28:243–73. doi: 10.1146/annurev-immunol-030409-101314. [DOI] [PubMed] [Google Scholar]
  • 32.Sonnenberg GF, Artis D. Innate lymphoid cell interactions with microbiota: implications for intestinal health and disease. Immunity. 2012;37:601–10. doi: 10.1016/j.immuni.2012.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Satoh-Takayama N, Vosshenrich CA, Lesjean-Pottier S, Sawa S, Lochner M, Rattis F, Mention JJ, Thiam K, Cerf-Bensussan N, Mandelboim O, Eberl G, Di Santo JP. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity. 2008;29:958–70. doi: 10.1016/j.immuni.2008.11.001. [DOI] [PubMed] [Google Scholar]
  • 34.Sanos SL, Bui VL, Mortha A, Oberle K, Heners C, Johner C, Diefenbach A. RORgammat and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nat Immunol. 2009;10:83–91. doi: 10.1038/ni.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Sawa S, Cherrier M, Lochner M, Satoh-Takayama N, Fehling HJ, Langa F, Di Santo JP, Eberl G. Lineage relationship analysis of RORgammat+ innate lymphoid cells. Science. 2010;330:665–9. doi: 10.1126/science.1194597. [DOI] [PubMed] [Google Scholar]
  • 36.Vonarbourg C, Mortha A, Bui VL, Hernandez PP, Kiss EA, Hoyler T, Flach M, Bengsch B, Thimme R, Holscher C, Honig M, Pannicke U, Schwarz K, Ware CF, Finke D, Diefenbach A. Regulated expression of nuclear receptor RORgammat confers distinct functional fates to NK cell receptor-expressing RORgammat(+) innate lymphocytes. Immunity. 2010;33:736–51. doi: 10.1016/j.immuni.2010.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Sawa S, Lochner M, Satoh-Takayama N, Dulauroy S, Berard M, Kleinschek M, Cua D, Di Santo JP, Eberl G. RORgammat+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol. 2011;12:320–6. doi: 10.1038/ni.2002. [DOI] [PubMed] [Google Scholar]
  • 38.Franchi L, Kamada N, Nakamura Y, Burberry A, Kuffa P, Suzuki S, Shaw MH, Kim YG, Nunez G. NLRC4-driven production of IL-1beta discriminates between pathogenic and commensal bacteria and promotes host intestinal defense. Nat Immunol. 2012;13:449–56. doi: 10.1038/ni.2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kamada N, Hisamatsu T, Okamoto S, Sato T, Matsuoka K, Arai K, Nakai T, Hasegawa A, Inoue N, Watanabe N, Akagawa KS, Hibi T. Abnormally differentiated subsets of intestinal macrophage play a key role in Th1-dominant chronic colitis through excess production of IL-12 and IL-23 in response to bacteria. J Immunol. 2005;175:6900–8. doi: 10.4049/jimmunol.175.10.6900. [DOI] [PubMed] [Google Scholar]
  • 40.Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007;8:1086–94. doi: 10.1038/ni1511. [DOI] [PubMed] [Google Scholar]
  • 41.Rivollier A, He J, Kole A, Valatas V, Kelsall BL. Inflammation switches the differentiation program of Ly6Chi monocytes from antiinflammatory macrophages to inflammatory dendritic cells in the colon. J Exp Med. 2012;209:139–55. doi: 10.1084/jem.20101387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hasegawa M, Kamada N, Jiao Y, Liu MZ, Nunez G, Inohara N. Protective Role of Commensals against Clostridium difficile Infection via an IL-1beta-Mediated Positive-Feedback Loop. J Immunol. 2012 doi: 10.4049/jimmunol.1200821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Clarke TB, Davis KM, Lysenko ES, Zhou AY, Yu Y, Weiser JN. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nat Med. 2010;16:228–31. doi: 10.1038/nm.2087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
  • 45.Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. 2006;441:231–4. doi: 10.1038/nature04754. [DOI] [PubMed] [Google Scholar]
  • 46.Ivanov, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126:1121–33. doi: 10.1016/j.cell.2006.07.035. [DOI] [PubMed] [Google Scholar]
  • 47.Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, Schluns K, Tian Q, Watowich SS, Jetten AM, Dong C. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. 2007;448:480–3. doi: 10.1038/nature05969. [DOI] [PubMed] [Google Scholar]
  • 48.Zhou L, Ivanov, Spolski R, Min R, Shenderov K, Egawa T, Levy DE, Leonard WJ, Littman DR. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat Immunol. 2007;8:967–74. doi: 10.1038/ni1488. [DOI] [PubMed] [Google Scholar]
  • 49.Shaw MH, Kamada N, Kim YG, Nunez G. Microbiota-induced IL-1beta, but not IL-6, is critical for the development of steady-state TH17 cells in the intestine. J Exp Med. 2012;209:251–8. doi: 10.1084/jem.20111703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hasegawa M, Osaka T, Tawaratsumida K, Yamazaki T, Tada H, Chen GY, Tsuneda S, Nunez G, Inohara N. Transitions in oral and intestinal microflora composition and innate immune receptor-dependent stimulation during mouse development. Infect Immun. 2010;78:639–50. doi: 10.1128/IAI.01043-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341:569–73. doi: 10.1126/science.1241165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, Takahashi M, Fukuda NN, Murakami S, Miyauchi E, Hino S, Atarashi K, Onawa S, Fujimura Y, Lockett T, Clarke JM, Topping DL, Tomita M, Hori S, Ohara O, Morita T, Koseki H, Kikuchi J, Honda K, Hase K, Ohno H. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013 doi: 10.1038/nature12721. [DOI] [PubMed] [Google Scholar]
  • 53.Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, Deroos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, Rudensky AY. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013 doi: 10.1038/nature12726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, Parmentier M, Detheux M. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278:25481–9. doi: 10.1074/jbc.M301403200. [DOI] [PubMed] [Google Scholar]
  • 55.Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011;3:858–76. doi: 10.3390/nu3100858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204:1757–64. doi: 10.1084/jem.20070590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204:1775–85. doi: 10.1084/jem.20070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, Cheroutre H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256–60. doi: 10.1126/science.1145697. [DOI] [PubMed] [Google Scholar]
  • 59.Welty NE, Staley C, Ghilardi N, Sadowsky MJ, Igyarto BZ, Kaplan DH. Intestinal lamina propria dendritic cells maintain T cell homeostasis but do not affect commensalism. J Exp Med. 2013 doi: 10.1084/jem.20130728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, Thangaraju M, Prasad PD, Manicassamy S, Munn DH, Lee JR, Offermanns S, Ganapathy V. Activation of gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity. 2014;40:128–39. doi: 10.1016/j.immuni.2013.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kim SV, Xiang WV, Kwak C, Yang Y, Lin XW, Ota M, Sarpel U, Rifkin DB, Xu R, Littman DR. GPR15-mediated homing controls immune homeostasis in the large intestine mucosa. Science. 2013;340:1456–9. doi: 10.1126/science.1237013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, Sakaguchi S, Takeda K. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol. 2012;189:2869–78. doi: 10.4049/jimmunol.1200420. [DOI] [PubMed] [Google Scholar]
  • 63.Rossi M, Amaretti A, Raimondi S. Folate production by probiotic bacteria. Nutrients. 2011;3:118–34. doi: 10.3390/nu3010118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453:620–5. doi: 10.1038/nature07008. [DOI] [PubMed] [Google Scholar]
  • 65.Kunisawa J, Gohda M, Hashimoto E, Ishikawa I, Higuchi M, Suzuki Y, Goto Y, Panea C, Ivanov, Sumiya R, Aayam L, Wake T, Tajiri S, Kurashima Y, Shikata S, Akira S, Takeda K, Kiyono H. Microbe-dependent CD11b(+) IgA(+) plasma cells mediate robust early-phase intestinal IgA responses in mice. Nat Commun. 2013;4:1772. doi: 10.1038/ncomms2718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Suzuki K, Maruya M, Kawamoto S, Sitnik K, Kitamura H, Agace WW, Fagarasan S. The sensing of environmental stimuli by follicular dendritic cells promotes immunoglobulin A generation in the gut. Immunity. 2010;33:71–83. doi: 10.1016/j.immuni.2010.07.003. [DOI] [PubMed] [Google Scholar]
  • 67.Tezuka H, Abe Y, Iwata M, Takeuchi H, Ishikawa H, Matsushita M, Shiohara T, Akira S, Ohteki T. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature. 2007;448:929–33. doi: 10.1038/nature06033. [DOI] [PubMed] [Google Scholar]
  • 68.Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, Otipoby KL, Yokota A, Takeuchi H, Ricciardi-Castagnoli P, Rajewsky K, Adams DH, von Andrian UH. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science. 2006;314:1157–60. doi: 10.1126/science.1132742. [DOI] [PubMed] [Google Scholar]
  • 69.Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, Nishiyama M, Sato S, Tsujimura T, Yamamoto M, Yokota Y, Kiyono H, Miyasaka M, Ishii KJ, Akira S. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat Immunol. 2008;9:769–76. doi: 10.1038/ni.1622. [DOI] [PubMed] [Google Scholar]
  • 70.Umesaki Y, Setoyama H, Matsumoto S, Imaoka A, Itoh K. Differential roles of segmented filamentous bacteria and clostridia in development of the intestinal immune system. Infect Immun. 1999;67:3504–11. doi: 10.1128/iai.67.7.3504-3511.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hapfelmeier S, Lawson MA, Slack E, Kirundi JK, Stoel M, Heikenwalder M, Cahenzli J, Velykoredko Y, Balmer ML, Endt K, Geuking MB, Curtiss R, 3rd, McCoy KD, Macpherson AJ. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science. 2010;328:1705–9. doi: 10.1126/science.1188454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Crellin NK, Trifari S, Kaplan CD, Satoh-Takayama N, Di Santo JP, Spits H. Regulation of cytokine secretion in human CD127(+) LTi-like innate lymphoid cells by Toll-like receptor 2. Immunity. 2010;33:752–64. doi: 10.1016/j.immuni.2010.10.012. [DOI] [PubMed] [Google Scholar]
  • 73.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, Carvalho A, Puccetti P, Romani L. Tryptophan Catabolites from Microbiota Engage Aryl Hydrocarbon Receptor and Balance Mucosal Reactivity via Interleukin-22. Immunity. 2013;39:372–85. doi: 10.1016/j.immuni.2013.08.003. [DOI] [PubMed] [Google Scholar]
  • 74.Manta C, Heupel E, Radulovic K, Rossini V, Garbi N, Riedel CU, Niess JH. CX(3)CR1(+) macrophages support IL-22 production by innate lymphoid cells during infection with Citrobacter rodentium. Mucosal Immunol. 2013;6:177–88. doi: 10.1038/mi.2012.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Niess JH, Adler G. Enteric flora expands gut lamina propria CX3CR1+ dendritic cells supporting inflammatory immune responses under normal and inflammatory conditions. J Immunol. 2010;184:2026–37. doi: 10.4049/jimmunol.0901936. [DOI] [PubMed] [Google Scholar]
  • 76.Kinnebrew MA, Buffie CG, Diehl GE, Zenewicz LA, Leiner I, Hohl TM, Flavell RA, Littman DR, Pamer EG. Interleukin 23 production by intestinal CD103(+)CD11b(+) dendritic cells in response to bacterial flagellin enhances mucosal innate immune defense. Immunity. 2012;36:276–87. doi: 10.1016/j.immuni.2011.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, Diefenbach A. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science. 2011;334:1561–5. doi: 10.1126/science.1214914. [DOI] [PubMed] [Google Scholar]
  • 78.Qiu J, Heller JJ, Guo X, Chen ZM, Fish K, Fu YX, Zhou L. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity. 2012;36:92–104. doi: 10.1016/j.immuni.2011.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, Schilter HC, Rolph MS, Mackay F, Artis D, Xavier RJ, Teixeira MM, Mackay CR. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–6. doi: 10.1038/nature08530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Johansson ME, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A. 2008;105:15064–9. doi: 10.1073/pnas.0803124105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bergstrom KS, Kissoon-Singh V, Gibson DL, Ma C, Montero M, Sham HP, Ryz N, Huang T, Velcich A, Finlay BB, Chadee K, Vallance BA. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog. 2010;6:e1000902. doi: 10.1371/journal.ppat.1000902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Vaishnava S, Behrendt CL, Ismail AS, Eckmann L, Hooper LV. Paneth cells directly sense gut commensals and maintain homeostasis at the intestinal host-microbial interface. Proc Natl Acad Sci U S A. 2008;105:20858–63. doi: 10.1073/pnas.0808723105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sonnenberg GF, Monticelli LA, Alenghat T, Fung TC, Hutnick NA, Kunisawa J, Shibata N, Grunberg S, Sinha R, Zahm AM, Tardif MR, Sathaliyawala T, Kubota M, Farber DL, Collman RG, Shaked A, Fouser LA, Weiner DB, Tessier PA, Friedman JR, Kiyono H, Bushman FD, Chang KM, Artis D. Innate lymphoid cells promote anatomical containment of lymphoid-resident commensal bacteria. Science. 2012;336:1321–5. doi: 10.1126/science.1222551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Strugnell RA, Wijburg OL. The role of secretory antibodies in infection immunity. Nat Rev Microbiol. 2010;8:656–67. doi: 10.1038/nrmicro2384. [DOI] [PubMed] [Google Scholar]
  • 85.Macpherson AJ, Gatto D, Sainsbury E, Harriman GR, Hengartner H, Zinkernagel RM. A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science. 2000;288:2222–6. doi: 10.1126/science.288.5474.2222. [DOI] [PubMed] [Google Scholar]
  • 86.Molloy MJ, Grainger JR, Bouladoux N, Hand TW, Koo LY, Naik S, Quinones M, Dzutsev AK, Gao JL, Trinchieri G, Murphy PM, Belkaid Y. Intraluminal Containment of Commensal Outgrowth in the Gut during Infection-Induced Dysbiosis. Cell Host Microbe. 2013;14:318–28. doi: 10.1016/j.chom.2013.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Maynard CL, Harrington LE, Janowski KM, Oliver JR, Zindl CL, Rudensky AY, Weaver CT. Regulatory T cells expressing interleukin 10 develop from Foxp3+ and Foxp3- precursor cells in the absence of interleukin 10. Nat Immunol. 2007;8:931–41. doi: 10.1038/ni1504. [DOI] [PubMed] [Google Scholar]
  • 88.Powrie F, Leach MW, Mauze S, Caddle LB, Coffman RL. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. Int Immunol. 1993;5:1461–71. doi: 10.1093/intimm/5.11.1461. [DOI] [PubMed] [Google Scholar]
  • 89.Kullberg MC, Jankovic D, Gorelick PL, Caspar P, Letterio JJ, Cheever AW, Sher A. Bacteria-triggered CD4(+) T regulatory cells suppress Helicobacter hepaticus-induced colitis. J Exp Med. 2002;196:505–15. doi: 10.1084/jem.20020556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kullberg MC, Andersen JF, Gorelick PL, Caspar P, Suerbaum S, Fox JG, Cheever AW, Jankovic D, Sher A. Induction of colitis by a CD4+ T cell clone specific for a bacterial epitope. Proc Natl Acad Sci U S A. 2003;100:15830–5. doi: 10.1073/pnas.2534546100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Feng T, Wang L, Schoeb TR, Elson CO, Cong Y. Microbiota innate stimulation is a prerequisite for T cell spontaneous proliferation and induction of experimental colitis. J Exp Med. 2010;207:1321–32. doi: 10.1084/jem.20092253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hepworth MR, Monticelli LA, Fung TC, Ziegler CG, Grunberg S, Sinha R, Mantegazza AR, Ma HL, Crawford A, Angelosanto JM, Wherry EJ, Koni PA, Bushman FD, Elson CO, Eberl G, Artis D, Sonnenberg GF. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature. 2013;498:113–7. doi: 10.1038/nature12240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–64. doi: 10.1038/nri1733. [DOI] [PubMed] [Google Scholar]
  • 94.Smythies LE, Sellers M, Clements RH, Mosteller-Barnum M, Meng G, Benjamin WH, Orenstein JM, Smith PD. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J Clin Invest. 2005;115:66–75. doi: 10.1172/JCI19229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Fagarasan S, Muramatsu M, Suzuki K, Nagaoka H, Hiai H, Honjo T. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science. 2002;298:1424–7. doi: 10.1126/science.1077336. [DOI] [PubMed] [Google Scholar]
  • 96.Wei M, Shinkura R, Doi Y, Maruya M, Fagarasan S, Honjo T. Mice carrying a knock-in mutation of Aicda resulting in a defect in somatic hypermutation have impaired gut homeostasis and compromised mucosal defense. Nat Immunol. 2011;12:264–70. doi: 10.1038/ni.1991. [DOI] [PubMed] [Google Scholar]
  • 97.Kawamoto S, Tran TH, Maruya M, Suzuki K, Doi Y, Tsutsui Y, Kato LM, Fagarasan S. The inhibitory receptor PD-1 regulates IgA selection and bacterial composition in the gut. Science. 2012;336:485–9. doi: 10.1126/science.1217718. [DOI] [PubMed] [Google Scholar]
  • 98.Peterson DA, McNulty NP, Guruge JL, Gordon JI. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe. 2007;2:328–39. doi: 10.1016/j.chom.2007.09.013. [DOI] [PubMed] [Google Scholar]
  • 99.Qiu J, Guo X, Chen ZM, He L, Sonnenberg GF, Artis D, Fu YX, Zhou L. Group 3 Innate Lymphoid Cells Inhibit T-Cell-Mediated Intestinal Inflammation through Aryl Hydrocarbon Receptor Signaling and Regulation of Microflora. Immunity. 2013;39:386–99. doi: 10.1016/j.immuni.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zenewicz LA, Yin X, Wang G, Elinav E, Hao L, Zhao L, Flavell RA. IL-22 deficiency alters colonic microbiota to be transmissible and colitogenic. J Immunol. 2013;190:5306–12. doi: 10.4049/jimmunol.1300016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Garrett WS, Lord GM, Punit S, Lugo-Villarino G, Mazmanian SK, Ito S, Glickman JN, Glimcher LH. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell. 2007;131:33–45. doi: 10.1016/j.cell.2007.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Garrett WS, Gallini CA, Yatsunenko T, Michaud M, DuBois A, Delaney ML, Punit S, Karlsson M, Bry L, Glickman JN, Gordon JI, Onderdonk AB, Glimcher LH. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe. 2010;8:292–300. doi: 10.1016/j.chom.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Powell N, Walker AW, Stolarczyk E, Canavan JB, Gokmen MR, Marks E, Jackson I, Hashim A, Curtis MA, Jenner RG, Howard JK, Parkhill J, Macdonald TT, Lord GM. The Transcription Factor T-bet Regulates Intestinal Inflammation Mediated by Interleukin-7 Receptor(+) Innate Lymphoid Cells. Immunity. 2012;37:674–84. doi: 10.1016/j.immuni.2012.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, Flavell RA. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145:745–57. doi: 10.1016/j.cell.2011.04.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Ubeda C, Lipuma L, Gobourne A, Viale A, Leiner I, Equinda M, Khanin R, Pamer EG. Familial transmission rather than defective innate immunity shapes the distinct intestinal microbiota of TLR-deficient mice. J Exp Med. 2012;209:1445–56. doi: 10.1084/jem.20120504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ng KM, Ferreyra JA, Higginbottom SK, Lynch JB, Kashyap PC, Gopinath S, Naidu N, Choudhury B, Weimer BC, Monack DM, Sonnenburg JL. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric pathogens. Nature. 2013 doi: 10.1038/nature12503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Winter SE, Thiennimitr P, Winter MG, Butler BP, Huseby DL, Crawford RW, Russell JM, Bevins CL, Adams LG, Tsolis RM, Roth JR, Baumler AJ. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature. 2010;467:426–9. doi: 10.1038/nature09415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Winter SE, Winter MG, Xavier MN, Thiennimitr P, Poon V, Keestra AM, Laughlin RC, Gomez G, Wu J, Lawhon SD, Popova IE, Parikh SJ, Adams LG, Tsolis RM, Stewart VJ, Baumler AJ. Host-derived nitrate boosts growth of E.coli in the inflamed gut. Science. 2013;339:708–11. doi: 10.1126/science.1232467. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES