Top Banner
The skin and mucosal surfaces of vertebrates are colo‑ nized by large numbers of microorganisms, includ‑ ing bacteria, fungi, parasites and viruses, commonly referred to as the microbiota. In humans, more than 100 trillion microorganisms, mostly bacteria, colonize the oral–gastrointestinal tract, and most of these micro‑ organisms reside in the distal intestine. Millions of years of co‑evolution between the host and microorganisms have led to a mutualistic relationship in which the micro‑ biota contributes to many host physiological processes and, in turn, the host provides niches and nutrients for microbial survival 1 . The main contributions of the microbiota to the host include the digestion and fermen‑ tation of carbohydrates, the production of vitamins, the development of gut-associated lymphoid tissues (GALTs), the polarization of gut‑specific immune responses and the prevention of colonization by pathogens 1–3 . In turn, gut immune responses that are induced by commensal populations regulate the composition of the micro‑ biota. Thus, a complex interplay between the host immune system and the microbiota is necessary for gut homeostasis. However, when the mutualistic rela‑ tionship between the host and microbiota is disrupted, the gut microbiota can cause or contribute to disease 4,5 . In this Review, we provide an overview of the current understanding of the dual role of the gut microbiota in health and disease. Microbiota-dependent lymphoid development GALTs. The study of germ-free mice led to the discov‑ ery that the gut microbiota is required for the normal generation and/or maturation of GALTs. GALTs are immune structures in which antigen can be taken up and presented by antigen‑presenting cells, and there‑ fore these structures have important roles in lympho‑ cyte functions that lead to inflammation or tolerance. GALTs include the Peyer’s patches, crypt patches and isolated lymphoid follicles (ILFs) 6–8 . In the fetus, lym‑ phoid tissue inducer (LTi) cells promote the devel‑ opment of Peyer’s patches in the absence of resident bacteria, although Peyer’s patches in germ‑free mice are smaller in size than those in specific-pathogen-free mice 9 . Unlike Peyer’s patches, the maturation of ILFs and crypt patches requires stimulation by the gut microbiota 7,10 . Specifically, incomplete maturation of ileal and colonic ILFs is observed in mice that are deficient in various pattern recognition receptors (PRRs) (BOX 1) that are acti‑ vated by bacterial stimuli, such as Toll‑like receptor 2 (TLR2), TLR4, and nucleotide‑binding oligomerization domain 2 (NOD2), and in their adaptor molecules, such as myeloid differentiation primary‑response protein 88 (MYD88) and TIR domain‑containing adaptor protein inducing IFNβ (TRIF; also known as TICAM1) 8 . The development of ILFs is also tightly regulated by NOD1, which is a bacterial sensor that responds to bacterial 1 Department of Pathology and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, Michigan, USA. 2 Department of Internal Medicine and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, Michigan, USA. Correspondence to G.N. e-mail: [email protected] doi:10.1038/nri3430 Gut-associated lymphoid tissues (GALTs). Lymphoid structures and aggregates that are associated with the intestinal mucosa, specifically the tonsils, Peyer’s patches, lymphoid follicles, appendix or cecal patch, and mesenteric lymph nodes. They are enriched in conventional and unconventional lymphocytes, and in specialized dendritic cell and macrophage subsets. Role of the gut microbiota in immunity and inflammatory disease Nobuhiko Kamada 1 , Sang-Uk Seo 1 , Grace Y. Chen 2 and Gabriel Núñez 1 Abstract | The mammalian intestine is colonized by trillions of microorganisms, most of which are bacteria that have co‑evolved with the host in a symbiotic relationship. The collection of microbial populations that reside on and in the host is commonly referred to as the microbiota. A principal function of the microbiota is to protect the intestine against colonization by exogenous pathogens and potentially harmful indigenous microorganisms via several mechanisms, which include direct competition for limited nutrients and the modulation of host immune responses. Conversely, pathogens have developed strategies to promote their replication in the presence of competing microbiota. Breakdown of the normal microbial community increases the risk of pathogen infection, the overgrowth of harmful pathobionts and inflammatory disease. Understanding the interaction of the microbiota with pathogens and the host might provide new insights into the pathogenesis of disease, as well as novel avenues for preventing and treating intestinal and systemic disorders. REVIEWS NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 321 © 2013 Macmillan Publishers Limited. All rights reserved
15

Role of the gut microbiota in immunity and inflammatory ...

Dec 09, 2021

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Role of the gut microbiota in immunity and inflammatory ...

The skin and mucosal surfaces of vertebrates are colo‑nized by large numbers of microorganisms, includ‑ing bacteria, fungi, parasites and viruses, commonly referred to as the microbiota. In humans, more than 100 trillion microorganisms, mostly bacteria, colonize the oral–gastrointestinal tract, and most of these micro‑organisms reside in the distal intestine. Millions of years of co‑evolution between the host and micro organisms have led to a mutualistic relationship in which the micro‑biota contributes to many host physiological processes and, in turn, the host provides niches and nutrients for microbial survival1. The main contributions of the microbiota to the host include the digestion and fermen‑tation of carbohydrates, the production of vitamins, the develop ment of gut-associated lymphoid tissues (GALTs), the polarization of gut‑specific immune responses and the prevention of colonization by pathogens1–3. In turn, gut immune responses that are induced by commensal populations regulate the composition of the micro‑biota. Thus, a complex interplay between the host immune system and the microbiota is necessary for gut homeostasis. However, when the mutualistic rela‑tionship between the host and microbiota is disrupted, the gut microbiota can cause or contribute to disease4,5. In this Review, we provide an overview of the current understanding of the dual role of the gut microbiota in health and disease.

Microbiota-dependent lymphoid developmentGALTs. The study of germ-free mice led to the discov‑ery that the gut microbiota is required for the normal generation and/or maturation of GALTs. GALTs are immune structures in which antigen can be taken up and presented by antigen‑presenting cells, and there‑fore these structures have important roles in lympho‑cyte functions that lead to inflammation or tolerance. GALTs include the Peyer’s patches, crypt patches and isolated lymphoid follicles (ILFs)6–8. In the fetus, lym‑phoid tissue inducer (LTi) cells promote the devel‑opment of Peyer’s patches in the absence of resident bacteria, although Peyer’s patches in germ‑free mice are smaller in size than those in specific-pathogen-free mice9. Unlike Peyer’s patches, the maturation of ILFs and crypt patches requires stimulation by the gut microbiota7,10. Specifically, incomplete maturation of ileal and colonic ILFs is observed in mice that are deficient in various pattern recognition receptors (PRRs) (BOX 1) that are acti‑vated by bacterial stimuli, such as Toll‑like receptor 2 (TLR2), TLR4, and nucleotide‑binding oligomerization domain 2 (NOD2), and in their adaptor molecules, such as myeloid differentiation primary‑response protein 88 (MYD88) and TIR domain‑containing adaptor protein inducing IFNβ (TRIF; also known as TICAM1)8. The development of ILFs is also tightly regulated by NOD1, which is a bacterial sensor that responds to bacterial

1Department of Pathology and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, Michigan, USA.2Department of Internal Medicine and Comprehensive Cancer Center, University of Michigan, Ann Arbor 48109, Michigan, USA.Correspondence to G.N.e-mail: [email protected]:10.1038/nri3430

Gut-associated lymphoid tissues(GALTs). Lymphoid structures and aggregates that are associated with the intestinal mucosa, specifically the tonsils, Peyer’s patches, lymphoid follicles, appendix or cecal patch, and mesenteric lymph nodes. They are enriched in conventional and unconventional lymphocytes, and in specialized dendritic cell and macrophage subsets.

Role of the gut microbiota in immunity and inflammatory diseaseNobuhiko Kamada1, Sang-Uk Seo1, Grace Y. Chen2 and Gabriel Núñez1

Abstract | The mammalian intestine is colonized by trillions of microorganisms, most of which are bacteria that have co‑evolved with the host in a symbiotic relationship. The collection of microbial populations that reside on and in the host is commonly referred to as the microbiota. A principal function of the microbiota is to protect the intestine against colonization by exogenous pathogens and potentially harmful indigenous microorganisms via several mechanisms, which include direct competition for limited nutrients and the modulation of host immune responses. Conversely, pathogens have developed strategies to promote their replication in the presence of competing microbiota. Breakdown of the normal microbial community increases the risk of pathogen infection, the overgrowth of harmful pathobionts and inflammatory disease. Understanding the interaction of the microbiota with pathogens and the host might provide new insights into the pathogenesis of disease, as well as novel avenues for preventing and treating intestinal and systemic disorders.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 321

© 2013 Macmillan Publishers Limited. All rights reserved

Page 2: Role of the gut microbiota in immunity and inflammatory ...

Germ-free miceMice that are born and raised in isolators, without exposure to microorganisms.

Peyer’s patchesCollections of lymphoid tissue that are located in the mucosa of the small intestine. They have an outer epithelial layer consisting of specialized epithelial cells called M cells. Peyer’s patches consist of a dome area, B cell follicles and interfollicular T cell areas. High endothelial venules are present mainly in the interfollicular areas.

Specific-pathogen-free miceMice raised in conditions whereby an increasing number of pathogens are excluded or eradicated from the colony. These animals are maintained in the absence of most of the known chronic and latent persistent pathogens. Although this enables better control of experimental conditions related to immunity and infection, it also sets apart such animal models from pathogen-exposed humans or non-human primates, whose immune systems are in constant contact with pathogens.

peptidoglycan molecules8. However, although it is clear from studies using germ‑free mice that GALT develop‑ment requires the presence of microbiota, the require‑ment for specific PRRs is less clear, as these studies have been confounded by differences between mouse strains that can result in environmental alterations in the microbiota, which, in turn, might affect lymphoid development independently of a specific gene defect.

TH17 cells. It is now well known that the resident micro‑biota regulates the development of specific lymphocyte subsets in the gut. T helper 17 (TH17) cells are a specific lineage of CD4+ TH cells that are crucial for host defence and that have a role in the development of autoimmune disease by producing the pro‑inflammatory cytokines interleukin‑17A (IL‑17A), IL‑17F and IL‑22 (REF. 11). Unlike other subsets of CD4+ TH cells, such as TH1 and TH2 cells, TH17 cells preferentially accumulate in the intestine, which indicates that the development of TH17 cells might be regulated by gut‑intrinsic mechanisms. Consistent with this hypothesis, the presence of intes‑tinal TH17 cells is greatly reduced in antibiotic‑treated or germ‑free mice12–16, which shows that the microbiota has a crucial role in TH17 cell development. In fact, a particular species of Clostridia‑related bacteria, called segmented filamentous bacteria (SFB), promotes the generation of TH17 cells in mice13–15 (FIG. 1a). The precise mechanism by which SFB promote TH17 cell develop‑ment remains to be fully elucidated. The adhesion of SFB to the host epithelium upregulates serum amyloid A protein (SAA) production, which, in turn, promotes

IL‑6 and IL‑23 production by CD11c+ lamina propria dendritic cells (DCs) and the subsequent induction of TH17 cell differentiation14 (FIG. 1a). Likewise, luminal ATP, which is provided by commensal bacteria but not by pathogenic bacteria, promotes TH17 cell development through a mechanism that is distinct to that elicited by SFB12. However, whether SAA and ATP are required for TH17 cell development in vivo remains unclear. IL‑1β is also crucial for TH17 cell differentiation and is specifi‑cally induced by the gut microbiota16 (FIG. 1a). Although reconstitution of germ‑free mice with the microbiota of conventionally raised mice rescues the development of TH17 cells in the gut, colonization with rat or human microbiota does not17. This indicates that the microbiota might regulate the development of TH17 cells in the gut in a species‑specific manner.

TReg cells. Forkhead box P3 (FOXP3)+ regulatory T (TReg) cells comprise a subset of CD4+ TH cells that accumu‑late in the intestine, where they help to maintain gut homeostasis. The depletion of TReg cells results in the abnormal expansion of CD4+ TH cells expressing com‑mensal bacteria‑specific T cell receptors (TCRs); conse‑quently, intestinal inflammation ensues18. Importantly, the development of peripherally derived TReg cells is partly dependent on the gut microbiota, as the number of TReg cells is greatly decreased in the colonic lamina propria of germ‑free mice19,20 (FIG. 1a). The induction of TReg cells is triggered by specific populations of commensal bac‑teria. The accumulation of IL‑10‑producing TReg cells in the colon is promoted by colonization of germ‑free mice with one of the following bacterial populations: a mix‑ture of 46 Clostridium spp. cluster IV and XIVa strains; altered Schaedler flora (ASF), which consists of a cock‑tail of eight defined commensal bacteria; or the human commensal bacterium Bacteroides fragilis19–21 (FIG. 1a). However, the mechanism by which specific bacterial populations induce the development of TReg cells remains poorly understood.

IgA-producing B cells and plasma cells. A close relation‑ship also exists between the microbiota and gut‑specific B cell responses. IgA is a major class of immunoglobu‑lin that is produced in mucosal tissues, including in the intestine. In the intestinal lumen, IgA is produced as pol‑ymeric IgA at high concentrations22 (FIG. 1b). Polymeric IgA is transported via the polymeric immunoglobulin receptor (pIgR) that is expressed on intestinal epithelial cells and is released into the intestinal lumen as secreted IgA (SIgA)23 (FIG. 1b). SIgA coats commensal bacteria and soluble antigens to inhibit their binding to the host epi‑thelium and their penetration into the lamina propria22. Thus, SIgA promotes intestinal barrier function and helps to maintain host–commensal mutualism (FIG. 1b).

In addition, IgA has been shown to regulate the com‑position and the function of the gut microbiota24. For example, deficiency or dysfunction of IgA as a result of deficiency or mutation of activation-induced cytidine deaminase (AID), respectively, leads to alterations in the composition of the gut microbiota25,26. Moreover, bind‑ing of IgA to the commensal Bacteroides thetaiotaomicron

Box 1 | Pattern recognition receptors

Pattern recognition receptors (PRRs) are receptor molecules expressed in immune cells. PRRs recognize pathogen-associated molecular patterns (PAMPs), such as lipopolysaccharide, flagellin, bacterial DNA and RNA, and they have crucial roles in innate immunity and protection against pathogens. In addition to pathogens, the gut-resident microbiota contains PAMPs that are recognized by PRRs. Most PRRs fall into three families: the Toll-like receptors (TLRs), NOD-like receptors (NLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs).

TLR family members are expressed on the cell surface and in endosomes, and stimulation of TLRs by bacterial components activates downstream adaptor molecules such as myeloid differentiation primary response 88 protein (MYD88) and/or TIR domain- containing adaptor protein inducing IFNβ (TRIF). TLR activation results in the upregulation of pro-inflammatory mediators that facilitate host immune responses. NLRs are cytoplasmic proteins that regulate inflammatory and apoptotic responses. The NLRs nucleotide-binding oligomerization domain 1 (NOD1) and NOD2 share the downstream adaptor molecule receptor-interacting serine/threonine kinase (RICK), which activates innate immunity through nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs). A specific subset of NLRs is involved in the formation of the inflammasome, which activates caspase 1. These NLRs include NLRP3 (NOD-, LRR- and pyrin domain-containing 3), NLRP1 and NLRC4 (NOD-, LRR- and CARD-containing 4), and they contain specific domains (a pyrin domain or a CARD) that assist in the recruitment of the adaptor protein ASC, which forms an oligomeric complex with caspase 1. Activation of the inflammasome and caspase 1 leads to the secretion of mature interleukin-1β (IL-1β) and IL-18, which are involved in host defence and in the pathogenesis of multiple autoimmune diseases. RLRs are cytoplasmic proteins that are involved in intracellular virus recognition but have been demonstrated to also be important in gut immunity and in disease pathogenesis166,167. The RLRs RIG-I and melanoma differentiation-associated protein 5 (MDA5) recognize double-stranded RNA. Another RLR member, LGP2 (also known DHX58), lacks amino-terminal caspase-recruitment domains and functions as a regulator of RIG-I and MDA5 signalling.

R E V I E W S

322 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 3: Role of the gut microbiota in immunity and inflammatory ...

Nature Reviews | Immunology

Epithelial cell

a

b

FOXP3+

TReg cell

Retinoic acid andIL-10

SAA

ATP

• SFB• Other microbiota

• PSA• Other factors?

• ↑ AID• Class switching

REGIIIγ

REGIIIγ

Laminapropria

Gut lumen

TH17 cell

IL-22IL-1β,IL-6 andIL-23

IL-12(?)TH1 cell

BAFFandAPRIL

TSLP

TLR

BAFF,APRIL

IL-17RB+ DCRORγt+ ILC3

IL-22

IgA

SIgA

APRIL, BAFF, NO, retinoic acid and TNF

pDC, TIP DC andTLR5+ DC

CD70+CD11clow DCand CX3CR1+CD11c+ DC

CD103+ DCand TLR5+ DC

IL-25

CD11b+CD11c– macrophage

CD11b+CD11c– macrophage

• Clostridium cluster IV and XIVa strains

• PSA+ Bacteroides fragilis• Other microbiota

B cell

IgA+ plasma cell

IgA+ B cell

FDC

c

TGFβ

Pattern recognition receptors(PRRs). Germline-encoded receptors (such as the Toll-like receptors (TLRs) and the NOD-like receptors (NLRs)) that can sense pathogen- associated molecular patterns and initiate signalling cascades that lead to an innate immune response. They can be membrane-bound (such as TLRs and C-type lectin receptors (CLRs)) or soluble cytoplasmic receptors (such as retinoic acid-inducible gene I (RIG-I), melanoma differentiation- associated protein 5 (MDA5) and NLRs).

Peripherally derived TReg cellsA subset of forkhead box P3 (FOXP3)+ regulatory T (TReg) cells that are induced in the periphery and mediate their suppressive effects through the secretion of cytokines such as interleukin-10 and transforming growth factor-β, which inhibit other T cells. Their role is to maintain self-tolerance.

Activation-induced cytidine deaminase(AID). An enzyme that is required for two crucial events in the germinal centre: somatic hypermutation and class-switch recombination.

Figure 1 | The gut microbiota-mediated development of the intestinal immune system. a | Segmented filamentous bacteria (SFB) and other commensal microorganisms activate lamina propria dendritic cells (DCs) and macrophages to induce T helper 17 (T

H17) cells and T

H1 cells through the production of interleukin‑1β (IL‑1β), IL‑6 and IL‑23 in the case

of TH17 cells, and possibly IL‑12 in the case of T

H1 cells (although the role of IL‑12 in T

H1 development in vivo in the gut

remains to be confirmed). TH17 cells regulate the gut microbiota community in an IL‑22‑ and regenerating islet‑derived

protein 3γ (REGIIIγ)‑dependent manner. Clostridium spp. clusters IV and XIVa, polysaccharide A (PSA)+ Bacteroides fragilis and other microbiota stimulate intestinal epithelial cells, T cells, and lamina propria DCs and macrophages to promote the development and/or the activation of forkhead box P3 (FOXP3)+ regulatory T (T

Reg) cells. b | The microbiota stimulates

intestinal epithelial cells and DCs to promote IgA-producing B cell and plasma cell differentiation in the lamina propria. Toll-like receptor (TLR) activation on intestinal epithelial cells induces the secretion of B cell-activating factor (BAFF) and a proliferation‑inducing ligand (APRIL), which promote the differentiation of IgA‑producing plasma cells. Intestinal epithelial cells also produce thymic stromal lymphoprotein (TSLP) to promote BAFF and APRIL expression by DCs. Various types of DCs, such as plasmacytoid DCs (pDCs), TIP DCs (TNF and inducible nitric oxide synthase (iNOS)‑producing DCs) and TLR5+ DCs secrete BAFF, APRIL, nitric oxide (NO), retinoic acid and tumour necrosis factor (TNF) to facilitate the expression of activation-induced cytidine deaminase (AID) and IgA class-switching in B cells. Follicular DCs (FDCs) also induce the differentiation of IgA‑producing plasma cells in Peyer’s patches and isolated lymphoid follicles. IgA that is produced by lamina propria B cells is secreted into the intestinal lumen (SIgA), where it alters microbiota composition and function. c | Innate lymphoid cells (ILCs) that express retinoic acid receptor‑related orphan receptor‑γt (RORγt) and produce IL‑22 (termed ILC3s) regulate the gut microbiome through the induction of REGIIIγ in intestinal epithelial cells. The microbiota positively regulates the production of IL‑22 by RORγt+ ILC3s via an unknown mechanism. In addition, the microbiota induces IL‑25 secretion by endothelial cells, which acts on lamina propria IL‑17 receptor B (IL‑17RB)+ DCs, and the IL‑25‑activated DC subset suppresses IL‑22 production by RORγt+ ILC3s. CX

3CR1, CX

3C‑chemokine receptor 1;

SAA, serum amyloid A protein; TGFβ, transforming growth factor‑β.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 323

© 2013 Macmillan Publishers Limited. All rights reserved

Page 4: Role of the gut microbiota in immunity and inflammatory ...

Follicular DCs(FDCs). Specialized non-haematopoietic stromal cells that reside in the follicles and germinal centres. These cells possess long dendrites, but are not related to dendritic cells, and carry intact antigen on their surface.

inhibits innate immune responses by affecting bacterial gene expression27. The gut microbiota also regulates IgA production, as the number of IgA‑producing cells in the intestine is markedly decreased in germ‑free mice22. Although the precise mechanisms by which commensal bacteria promote the development of IgA‑producing cells remain poorly understood, bacterial recognition through MYD88 in follicular DCs (FDCs) is known to be important for the generation of IgA28 (FIG. 1b). Notably, for a subset of lamina propria DCs, commensal bacteria‑derived flagellin promotes the synthesis of retinoic acid, which is an important molecule that facilitates the differ‑entiation of IgA‑producing B cells29,30 (FIG. 1b). Likewise, commensal bacteria promote the expression of factors that are involved in the induction of IgA+ B cells, such as tumour necrosis factor (TNF), inducible nitric oxide synthase (iNOS; also known as NOS2), B cell activating factor (BAFF; also known as TNFSF13B) and a prolifer‑ation‑inducing ligand (APRIL; also known as TNFSF13) in lamina propria DCs31,32. In addition to TNF‑ and iNOS‑producing (TIP) DCs, intestinal plasma cells express TNF and iNOS following microbial exposure, which further promotes the IgA secretory function of intestinal B cells33 (FIG. 1b). Thus, the microbiota instructs lamina propria DCs and/or FDCs to induce the differentiation of IgA‑producing B cells, and in turn, IgA regulates the func‑tion and composition of the gut microbiota to maintain mutualism between the host and the microbiota.

ILCs. Innate lymphoid cells (ILCs) are increasingly recog‑nized as innate immune cells that share functional charac‑teristics with T cells (reviewed in REFS 34,35). ILCs arise from a common lymphoid precursor cell but differenti‑ate into multiple lineages on the basis of the expression of specific transcriptional factors. Currently, ILCs have been categorized into three main groups: T‑bet+ ILCs (termed ILC1s) GATA‑binding protein 3 (GATA3)+ ILCs (termed ILC2s), and retinoic acid receptor‑related orphan receptor‑γt (RORγt)+ ILCs (termed ILC3s)34–37.

The role of the microbiota in the development and the function of ILCs has been controversial. Although some studies report that the microbiota is required for the differentiation of ILCs and for their production of IL‑22 (REFS 38,39) (FIG. 1c), another study suggested that the microbiota suppressed IL‑22 production by RORγt+ ILCs40 (FIG. 1c). A crucial function of IL‑22 is to promote antimicrobial peptide production by intestinal epithe‑lial cells. Specifically, IL‑22 induces the expression of the C‑type lectin antimicrobial peptides regenerating islet‑derived protein 3β (REGIIIβ) and REGIIIγ, which can affect the gut microbiota41,42. Depletion of either IL‑22 or IL‑22‑producing ILCs leads to overgrowth and/or dis‑semination of potentially pathogenic bacteria, such as Alcaligenes xylosoxidans, which might increase the risk of subsequent intestinal damage and systemic inflamma‑tion43. Thus, the regulation of gut‑specific immune cells by commensal bacteria has far‑reaching effects that are not limited to immune homeostasis, but also include the regulation of the balance between beneficial and poten‑tially pathogenic commensals in the microbiota, and resistance to intestinal pathology.

Microbiota and resistance to pathogensIt has been known for many years that germ‑free mice are more susceptible to infection than conventionally raised mice44,45. Furthermore, treatment with antibiot‑ics is associated with increased colonization of patho‑gens in both mice and humans46–49. These observations indicate that an important function of the indigenous microbiota might be to protect the host from infection. The mechanisms by which commensal bacteria achieve this remain poorly understood, but there is evidence to indicate that both direct and indirect mechanisms might be involved.

Direct competition for nutrients. Several studies have shown that commensal bacteria can inhibit pathogen colonization by successfully competing for the limited supply of nutrients within the intestine. For example, Escherichia coli competes with enterohaemorrhagic E. coli (EHEC; an enteric pathogen that causes substan‑tial morbidity and mortality worldwide) for organic acids, amino acids and other nutrients50–53 (FIG. 2). As different commensal E. coli strains have distinct meta‑bolic profiles, each strain can differentially compete with pathogens53. Although our understanding of how commensal bacteria outcompete pathogens is still poor, studies thus far indicate that pathogen eradica‑tion might be most effective with bacteria that are meta‑bolically related to the pathogen. For example, E. coli, but not Bacteroides spp., can effectively outcompete the metabolically related pathogen Citrobacter rodentium, which is a mouse bacterium that models infection by enteropathogenic E. coli (EPEC) and EHEC54. The ability of E. coli to outcompete C. rodentium is partly mediated by competition for simple sugars that are used by both bacteria in the intestine54 (FIG. 2).

Likewise, the enteric pathogen Salmonella enteria subsp. enterica serovar Typhimurium is capable of colo‑nizing the intestine of mice that have been harbouring a limited microbiota for several weeks, but it is rapidly eradicated after co‑housing with conventionally raised mice, which indicates that not all commensal bacteria have the ability to out‑compete the pathogen55. The metabolic activity of the microbiota can also affect pathogen colonization by a mechanism that is distinct from nutritional competition. The commensal bacterial strain B. thetaiotaomicron produces multiple fucosidases that generate fucose from host‑derived glycans, such as mucin, and this modulates the expression of EHEC viru‑lence genes through the activation of the fucose sensor FusKR56 (FIG. 2).

Pathogens, in turn, have also evolved strategies to over‑come competition by commensal bacteria. One strategy is to use nutrients that are not primarily metabolized by commensal bacteria. Carbohydrate usage can differ substantially between pathogenic and commensal E. coli strains52,57. For example, EHEC, but not certain commen‑sal E. coli strains, can use galactose, hexuronates, man‑nose, ribose and ethanolamine, which is released into the intestine during epithelial cell turnover, as a carbon or nitrogen source52,58. Use of ethanolamine can be explained by the presence of the ethanolamine utilization (eut)

R E V I E W S

324 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 5: Role of the gut microbiota in immunity and inflammatory ...

Nature Reviews | Immunology

Mic

robi

ota

stra

tegi

es

Virulence factorexpression

Nutrientsspecifically used by pathogens

Competitionfor nutrients

Fucose

Pathogen Pathogen

Pathogencolonization

• Pathogen colonization• Systemic translocation

Direct

Commensalmicrobiota

Path

ogen

str

ateg

ies

Indirect

Neutrophil

Macrophage

B cell

TH1 orTH17 cell

ILC3

IL-1βandIL-23

IL-1β

Induce pro-IL-1β

Inflamedepithelial cell IL-22

IL-22

Plasma cell

REGIIIγMucus

SCFAsEpithelialcell

Killing (T6SS)

REGIIIγ IgA

Laminapropria

Gut lumen

↑ Barrier function

Type IV secretion system(T6SS). An export system that is used by Gram-negative bacteria to deliver effector proteins to other cells in a cell contact-dependent manner.

operon in the genome of the EHEC strain and not in the commensal E. coli strain58. Thus, pathogens have evolved to use nutritional resources that are not consumed by commensal bacteria to acquire a growth advantage. Another strategy used by pathogens is the induction of inflammation by their virulence factors. For example, infection by S. Typhimurium results in the production of reactive oxygen species by neutrophils, which facili‑tates the conversion of endogenous thio sulphate into tetrathionate, thereby selectively promoting the growth of S. Typhimurium59,60 (FIG. 2). Moreover, virulence factors such as intimin allow C. rodentium to attach to the host

epithelium where commensal bacteria do not normally reside54. Localization to epithelium‑associated niches might also allow certain pathogens such as EPEC and EHEC to use nutrients that are available at or near the epithelium and to escape from direct competition with resident microorganisms (FIG. 2). Furthermore, certain Gram‑negative enteric pathogens, such as Serratia marce‑scens and C. rodentium, can directly kill their commensal competitors through the expression of the type VI secretion system (T6SS)61,62 (FIG. 2). Thus, the outcome of infection by pathogens is ultimately determined by both host and commensal bacterial factors.

Figure 2 | Direct and indirect resistance of the microbiota to colonization by enteric pathogens. Direct competition between pathogens and commensal bacteria is shown on the left‑hand side. Resident microorganisms directly inhibit the colonization and/or the proliferation of incoming enteric pathogens. Commensal microorganisms can outcompete pathogens for shared nutrients, such as carbohydrates, amino acids and organic acids. In addition, commensal bacterial strains such as Bacteroides thetaiotaomicron catabolize mucin to produce fucose, which inhibits virulence factor expression by pathogenic Escherichia coli. Enteric pathogens have evolved strategies to overcome competition by commensal bacteria. Some pathogens can directly kill their commensal competitors through their type VI secretion system (T6SS). Pathogen‑induced inflammation, which leads to increased epithelial cell turnover, provides nutrients that selectively promote the growth of pathogens. Moreover, pathogens can localize to epithelium‑associated niches that are devoid of commensal bacteria and use nutrients near the epithelium to escape direct competition with resident microorganisms. Indirect mechanisms of competition between commensal bacteria and pathogens are shown on the right‑hand side. Commensal bacteria catabolize polysaccharides to generate short‑chain fatty acid (SCFAs), such as acetate, which enhances intestinal epithelial cell barrier function. In addition, commensal microbiota promotes the production of mucus and the release of antimicrobial peptides such as regenerating islet‑derived protein 3γ (REGIIIγ) from epithelial cells to limit pathogen colonization and proliferation. Innate immune cells, such as intestinal resident macrophages, neutrophils and some group 3 innate lymphoid cells (namely ILC3s), as well as T helper 1 (T

H1) cells, T

H17 cells and IgA-producing B cells

and plasma cells, are also activated by the microbiota to limit pathogen colonization. IL, interleukin.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 325

© 2013 Macmillan Publishers Limited. All rights reserved

Page 6: Role of the gut microbiota in immunity and inflammatory ...

InflammasomeA molecular complex of several proteins, including a NOD-like receptor (NLR), the adaptor protein ASC and a caspase, that, upon assembly, cleaves pro-interleukin-1β (pro-IL-1β) and pro-IL-18, thereby producing mature IL-1β and IL-18.

Aryl hydrocarbon receptor(AHR). A cytosolic, ligand-dependent transcription factor that translocates to the nucleus following the binding of specific ligands, which include dietary and microbial metabolites. AHR participates in the differentiation of regulatory T cells, T helper 17 (TH17) cells and intraepithelial intestinal T cells, and it is required for the secretion of interleukin-22 by TH17 cells. More recently, AHR has been shown to have crucial roles in the development and function of group 3 innate lymphoid cells, including lymphoid tissue inducer (LTi) cells and ILC3s.

Commensal bacteria promote mucosal barrier function. The attachment of pathogens to the surface of the intes‑tinal epithelium is a crucial initial step for infection to occur. As a defence mechanism, the epithelium produces mucus and antimicrobial molecules to inhibit pathogen invasion. In the colon, the mucus layer forms a strong bar‑rier against both pathogens and commensal bacteria63,64. In the small and large intestines, the inner mucus layer near the epithelium is devoid of commensal bacteria63,64. However, the mucus layer in germ‑free mice is much thinner than that in conventionally raised mice, which indicates that the microbiota might contribute to mucus production65 (FIG. 2). Consistent with this hypothesis, the thickness of the mucus layer of germ‑free mice can be restored to normal by oral administration of the bacterial products lipopolysaccharide (LPS) and peptidoglycan65.

The microbiota also contributes to the production of antimicrobial molecules by epithelial cells of the small and large intestines (FIG. 2). For example, REGIIIγ and REGIIIβ that are contained in Paneth cell granules are released into the lumen to regulate host–bacte‑ria inter actions through their antimicrobial activity66. Epithelium‑intrinsic deletion of MYD88 impairs the production of REGIIIγ and REGIIIβ; therefore, it seems that recognition of the microbiota by TLRs medi‑ates the expression of these antimicrobial peptides42,66. Importantly, the impaired production of REGIIIγ and REGIIIβ increases the susceptibility of mice to infection by enteric pathogens including Listeria mono cytogenes, Yersinia pseudotuberculosis and S. enterica subsp. enterica serovar Enteritidis67–69.

The microbiota can also enhance epithelial barrier function through the production of metabolites. For instance, short‑chain fatty acids (SCFAs), especially acetate, produced by Bifidobacterium spp. act on the epi‑thelium to inhibit the translocation of Shiga toxin that is produced by E. coli O157:H7 (REF. 45) (FIG. 2). The precise signalling mechanism by which bacterial products and metabolites enhance epithelial defence, and how this mutually affects the microbiota and the host, remains to be elucidated.

The microbiota enhances innate immunity to patho-gens. Mononuclear phagocytes, such as macrophages and DCs, are located in the lamina propria and promote immunological unresponsiveness to commensal bacteria, which is important for maintaining gut homeostasis70,71. Specifically, gut‑resident phagocytes are hyporespon‑sive to microbial ligands and commensal bacteria, and they do not produce biologically significant levels of pro‑inflammatory molecules upon stimulation70–72. However, the microbiota is essential for upregulating the production of pro‑IL‑1β, the precursor to IL‑1β, in resident mononuclear phagocytes71. Under steady‑state conditions when the epithelial barrier is intact, resident commensal bacteria cannot also induce the processing of pro‑IL‑1β into biologically active mature IL‑1β and thus a state of hyporesponsiveness is maintained71. By contrast, infection by enteric pathogens such as S. Typhimurium and Pseudomonas aeruginosa can induce the processing of pro‑IL‑1β by promoting the activation of caspase 1

via the NLRC4 (NOD‑, LRR‑ and CARD‑containing 4) inflammasome71. Unlike commensal bacteria, these patho‑gens are capable of activating the NLRC4 inflammasome to produce IL‑1β because they express a type III secretion system, which allows the transfer of the NLRC4 agonist flagellin into the host cytosol71. Consequently, BALB/c mice that are deficient in NLRC4 or in the IL‑1 receptor (IL‑1R) are highly susceptible to orogastric infection with S. Typhimurium71. The protective role of IL‑1β in intesti‑nal immunity is, at least partly, mediated by its ability to induce the expression of endothelial adhesion molecules that contribute to neutrophil recruitment and pathogen clearance in the intestine. Thus, NLRC4‑dependent IL‑1β production by resident phagocytes represents a specific response that can discriminate between pathogenic and commensal bacteria.

The intestinal microbiota also promotes immunity through the production of IL‑22 by ILC3s38. Germ‑free mice show impaired intestinal production of IL‑22, which indicates that there might be a requirement for commen‑sal bacteria or their metabolites16. Mice with defects in IL‑22‑expressing cells exhibit increased susceptibility to C. rodentium infection, which indicates that commen‑sal bacterial‑driven IL‑22 that is produced by ILC3s is important for protection against infectious pathogens38,73,74. Consistent with this idea, administration of IL‑22 to aryl hydrocarbon receptor (Ahr)−/− mice, which exhibit impaired IL‑22 production, provides protection against C. rodentium infection74. Taken together, these results indicate that commensal bacteria might also promote host defence by stimulating ILCs to produce IL‑22 (FIG. 2).

The microbiota promotes adaptive immunity. As men‑tioned above, specific commensal bacteria promote the generation of different T cell subsets in the intes‑tine12,14,19–21,75 that have unique roles during pathogen infection (FIG. 2). For instance, TH17 cell differentiation that is induced by SFB colonization facilitates protec‑tion against C. rodentium infection14. Likewise, micro‑biota‑induced TReg cells attenuate intestinal damage that is caused by exaggerated immune responses against infectious pathogens. Specifically, B. fragilis promotes IL‑10‑producing TReg cells that protect against Helicobacter hepaticus infection21,76, and Bifidobacterium infantis increases the number of TReg cells that attenuate intes‑tinal disease severity following S. Typhimurium infec‑tion77. Commensal bacteria also induce specific immune responses, including IgA production and the generation of CD4+ T cells, that are directed against their own anti‑gens78–80. Although the precise role of commensal bacte‑ria‑specific adaptive immunity against invasive pathogens remains poorly understood, there is evidence indicating that it is important for limiting the systemic dissemination of commensal bacteria81. The induction of this ‘firewall’ function of the adaptive immune system by the micro‑biota might prevent collateral damage — which could be caused by the translocation of indigenous bacteria — that is often associated with pathogen infection and epithelial barrier disruption. In addition, it might contribute to the elimination of pathogens through opsonization or other immune mechanisms.

R E V I E W S

326 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 7: Role of the gut microbiota in immunity and inflammatory ...

Inflammatory bowel disease(IBD). Immune-mediated inflammation of the bowel. There are two main forms: Crohn’s disease, which is a granulomatous segmental inflammation affecting any part of the intestine; and ulcerative colitis, which is a mucosal inflammation involving the rectum and extending for a variable distance along the colon. In developed countries, the incidence of inflammatory bowel disease is approximately one in 50,000. It usually starts in early adult life and continues afterwards with a relapsing, remitting course.

SAMP1/yit miceA mutant mouse strain that spontaneously develops a chronic intestinal inflammation that is mainly localized in the terminal ileum.

AutophagyAn evolutionarily conserved process in which acidic double-membrane vacuoles sequester intracellular contents (such as damaged organelles and macromolecules) and target them for degradation through fusion to secondary lysosomes.

α-defensinsOne subset of defensin proteins. Defensins and cathelicidins are members of a family of small antimicrobial polypeptides that are abundant in neutrophils and epithelial cells. They contribute to host defence by disrupting the cytoplasmic membrane of microorganisms.

Protective role of the commensal microbiota against sys-temic infection. The mechanism by which the micro‑biota promotes systemic pathogen eradication is poorly understood. Pretreatment of germ‑free mice with LPS induces pro‑inflammatory cytokine production and neutrophil recruitment, and can prevent systemic bac‑terial infection82. This observation indicates that com‑mensal bacteria might, at least partly, promote host defence at distant sites through the release of microbial molecules. Consistent with this hypothesis, peptido‑glycan molecules that are derived from the microbiota are found in the periphery and can prime peripheral blood neutrophils to facilitate their bactericidal capacity via NOD1 signalling83. This neutrophil priming effect, which is presumably induced by intestinal bacteria, enhances host defence against systemic infection with Streptococcus pneumoniae83.

The microbiota might also have an important role in systemic antiviral immune responses. Germ‑free mice and antibiotic‑treated mice show reduced innate and adaptive immune responses to influenza virus, which results in increased viral loads in their tissues84,85. The microbiota might enhance antiviral immunity through the inflammasome and the production of innate cytokines that are required for optimal immune responses against viruses84. For example, the microbiota promotes type I interferon (IFN) production by macrophages and the subsequent IFN‑priming of natural killer (NK) cells, which is important for protection against infection with mouse cytomegalovirus and lymphocytic chorio‑meningitis virus86. Moreover, the gut microbiota induces intestinal TLR‑dependent DC activation and inhibits systemic parasitic infection87. Recent studies also indicate that commensal bacteria might provide tissue‑specific defence mechanisms, as has been demonstrated by the protection provided by resident skin bacteria against local pathogen infection88.

Taken together, these studies using different infection models show a protective effect of the microbiota against systemic infection; however, additional studies are nec‑essary to identify the underlying mechanisms as well as the relative contributions of host defence mechanisms that are promoted by the microbiota of specific tissues such as the skin or the respiratory tract. It is also impor‑tant to note that commensal bacteria do not always pro‑tect against pathogenic infection and in certain contexts they can facilitate it (as discussed below)89–91.

The microbiota and intestinal diseaseThe microbiota contributes to IBD. The gut micro‑biota is essential for triggering or enhancing chronic intestinal inflammation in various inflammatory bowel disease (IBD)‑prone mouse strains (TABLE 1). Under germ‑free conditions, Il10−/− or Tcra−/− mice, as well as transgenic rats that have been engineered to overexpress HLA‑B27 and human β2 microglobulin, do not develop colitis92–95. In Il2−/− or SAMP1/yit mice, although sponta‑neous intestinal inflammation occurs even under germ‑free conditions ,symptoms are attenuated compared with conventionally raised mutant mice96,97. In many spontaneous colitis models, intestinal inflammation is

abrogated if mice also have a deficiency in individual TLRs or in MYD88. For example, spontaneous coli‑tis that develops in Il10−/− mice, in conditional signal transducer and activator of transcription 3 (Stat3)−/− mice or in mice with a deletion in inhibitor of nuclear factor‑κB kinase‑γ (IKKγ; also known as NEMO) spe‑cifically in intestinal epithelial cells (NEMOΔIEC mice) is reversed when these mice also lack TLRs or MYD88 (REFS 98–101). However, Il2−/− mice that are deficient in MYD88 still develop spontaneous colitis99. In this model, the microbiota might drive colitis through the promo‑tion of inflammatory TH17 cell responses12 that occur independently of MYD88.

Similar to mice, human intestinal mononuclear phagocytes show hyporesponsiveness to microbial stim‑ulation under steady‑state conditions102,103. However, in patients with IBD, intestinal mononuclear phagocytes robustly respond to microbial products and to the resi‑dent bacteria, which results in the production of large amounts of pro‑inflammatory cytokines such as TNF and IL‑23, as occurs in IBD‑prone mice103. Thus, the abnormal activation of resident intestinal mononuclear phagocytes by commensal bacteria might facilitate the development or persistence of intestinal inflammation in IBD. Consistent with this idea, intestinal macrophages isolated from Il10−/− mice robustly respond to gut bac‑teria, whereas wild‑type intestinal macrophages are hyporesponsive70–72. However, the increased production of pro‑inflammatory molecules by intestinal phagocytes might also reflect the activity of recruited monocytes in areas of infection or inflammation.

There is genetic evidence showing that the impaired recognition and killing of commensal bacteria also con‑tributes to IBD development, as has been suggested by the fact that many of the identified IBD‑susceptibility genes regulate host–microbial interactions104. NOD2, which is an intracellular sensor of bacterial peptidog‑lycan, was identified as a susceptibility gene for Crohn’s disease, and Crohn’s disease‑associated NOD2 muta‑tions are associated with a loss of function of the pro‑tein105–107. Mutations in the autophagy regulatory genes autophagy related gene 16‑like 1 (ATG16L1) and immu‑nity‑related GTPase family M (IRGM) are also linked to Crohn’s disease108,109, and autophagy dysfunction is associated with defects in bacterial killing110–112. In addi‑tion, NOD2 and autophagy proteins regulate the func‑tion of Paneth cells, which release granules containing antimicrobial peptides in response to bacteria113. NOD2 is highly expressed in Paneth cells, and some studies report diminished expression of Paneth cell α-defensins in individuals with Crohn’s disease‑associated NOD2 mutations114,115. Nod2−/− mice have impaired antimicro‑bial functions in Paneth cells, resulting in the accumu‑lation of ileal bacteria, which might contribute to IBD pathogenesis116. Atg16l1‑mutant (Atg16l1HM) mice show abnormal granule formation in Paneth cells, which is also observed in patients with Crohn’s disease who have homozygous mutations in ATG16L1 (REF. 117). In Atg16l1HM mice this abnormality can be triggered by enteric viral infection (for example, by murine noro‑virus) and is associated with increased susceptibility

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 327

© 2013 Macmillan Publishers Limited. All rights reserved

Page 8: Role of the gut microbiota in immunity and inflammatory ...

to chemically induced colitis118. Thus, defects in host mechanisms that recognize and clear bacteria are associated with the development of human IBD. How genetic defects lead to chronic colitis in patients with IBD remains unknown, but it is possible that impaired NOD2 or autophagy function might result in the accu‑mulation of intestinal commensal bacteria that have the capacity to locally invade the intestinal mucosa and to trigger an abnormal inflammatory response.

The role of the pathobiont in IBD. As non‑pathogenic symbionts, the gut microbiota derives nutritional ben‑efits from the host and help to maintain gut homeostasis. However, under certain conditions, particular bacterial populations that are typically found in very low abun‑dance can acquire pathogenic properties. These condi‑tions include inherent immune defects as well as changes in diet and/or acute inflammation, and can result in the disruption of the normal balanced state of the gut

Table 1 | Role of the gut microbiota in the protection or induction of IBD

Commensal bacterium Host Genotype Disease Receptors Possible mechanism Refs

Commensal microbiota that protect against IBD

Clostridium spp. clusters IV and XIVa

Mouse Wild‑type DSS colitis Unknown Induction of TReg

cells 19

Altered Schaedler flora Mouse Wild‑type DSS colitis MYD88 and TRIF

Induction of TReg

cells 20

Bacteroides fragilis Mouse Wild‑type TNBS colitis TLR2, MYD88 Induction of TReg

cells via polysaccharide A

21

Bacteroides vulgatus Mouse Il2−/− Spontaneous colitis Unknown Suppression of Escherichia coli- triggered colitis in Il2−/− mice

120

Faecalibacterium prausnitzii

Human NA IBD Unknown Induction of IL‑10 by PBMCs 148

Commensal microbiota that promote the development of IBD

E. coli Mouse Il10−/− Spontaneous colitis Unknown Monocolonization of germ‑free mice induces colitis

119

E. coli Mouse Il2−/− Spontaneous colitis Unknown Monocolonization of germ‑free mice induces colitis

96, 120

Enterococcus faecalis Mouse Il10−/− Spontaneous colitis Unknown Monocolonization of germ‑free mice induces colitis

119

B. vulgatus Rat HLA-B27–B2m transgenic

Spontaneous colitis Unknown Monocolonization of germ‑free rats induces colitis

96

B. vulgatus Mouse Il10r2−/−Tgfbr2−/− Spontaneous colitis Unknown Colonization of antibiotic‑ treated mice triggers colitis

122

Bacteroides thetaiotaomicron

Mouse Il10r2−/–Tgfbr2−/− Spontaneous colitis Unknown Colonization of antibiotic‑treated mice triggers colitis

122

B. thetaiotaomicron Rat HLA-B27–B2m transgenic

Spontaneous colitis Unknown Monocolonization of germ‑free rats induces colitis

95

Bacteroides unifirmatis Mouse Il10r2−/− Tgfbr2−/− Spontaneous colitis Unknown Colonization of antibiotic‑treated mice triggers colitis

122

Klebsiella pneumoniae*‡ Mouse Tbx21−/−Rag2−/− Spontaneous colitis Unknown Other commensal bacteria are required for the induction of colitis

123,124

Proteus mirabilis*‡ Mouse Tbx21−/−Rag2−/− Spontaneous colitis Unknown Other commensal bacteria are required for the induction of colitis

123,124

Helicobacter typhlonius* Mouse Tbx21−/−Rag2−/− Spontaneous colitis Unknown Transmissible to non‑colitogenic TRUC mice

125

Prevotellaceae*‡ Mouse Nlrp6−/−, Asc−/− or Casp1−/−

DSS colitis Unknown Impaired IL‑18 signalling promotes pathobiont expansion

126

TM7*‡ Mouse Nlrp6−/−, Asc−/− or Casp1−/−

DSS colitis Unknown Impaired IL‑18 signalling promotes pathobiont expansion

126

Bilophila wadsworthia Mouse Il10−/− Spontaneous colitis Unknown Consumption of a diet composed of milk‑derived fat induces pathobiont expansion

128

B2m, gene encoding β2‑microglobulin; Casp1, caspase 1; DSS, dextran‑sulphate sodium; IBD, inflammatory bowel disease; IL, interleukin; MYD88, myeloid differentiation primary‑response protein 88; NA, not applicable; Nlrp6, NOD‑, LRR‑ and pyrin domain‑containing 6; PBMCs, peripheral blood mononuclear cells; Rag2, recombination‑activating gene 2; Tbx21, T‑box 21 (encodes T‑bet); Tgfbr2, transforming growth factor‑β receptor 2; TLR, Toll‑like receptor; TNBS, 2,4,6‑trinitrobenzene sulphonic acid; T

Reg cell, regulatory T cell; TRIF, TIR domain-containing adaptor protein inducing IFNβ; TRUC, Tbx21−/−Rag2−/− mice.

*Transmissable to mutant hosts. ‡Transmissable to wild‑type hosts.

R E V I E W S

328 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 9: Role of the gut microbiota in immunity and inflammatory ...

Nature Reviews | Immunology

Unclassified bacteria

• GALT development• ↑ TH17 and TH1 cells• ↑ Barrier function• ↑ REGIIIγ

Chronic inflammation

Homeostasis

Beneficialbacteria

Pathobionts

Gut lumen

Laminapropria

Symbiont

Homeostasis Inflammatory bowel disease

Dysbiosis

Transmissableto healthy host

Unknowncontribution

Genetic factors (for example, mutations in Nod2, Atg16I1 and Il23r)

Environmental factors(diet, stress and infection)

• ↑ TReg cell• ↑ IL-10• ↑ REGIIIγ

• ↓ TReg cells• ↓ IL-10• ↓ REGIIIγ

• ↑↑↑ TH17 cells• ↑↑↑ TH1 cells

RagRecombinase activating genes; Rag1and Rag2 are expressed in developing lymphocytes. Mice that are deficient in either of these genes fail to produce B or T cells owing to a developmental block in the gene rearrangement that is necessary for receptor expression.

microbiota, which is referred to as dysbiosis5. Dysbiosis involves the abnormal accumulation or increased viru‑lence of certain commensal populations of bacteria, thereby transforming former symbionts into ‘patho‑bionts’ (FIG. 3). Pathobionts are typically colitogenic in that they can trigger intestinal inflammation; therefore, there has been considerable interest in the identification of pathobionts to understand the pathogenesis of IBD.

Colitogenic pathobionts have been identified using rodent models of spontaneous colitis (TABLE 1). For exam‑ple, colonization of germ‑free Il10−/− or Il2−/− mice with one of several particular E. coli or Enterococcus faecalis strains, but not with Bacteroides vulgatus, induces colitis119–121. By contrast, B. vulgatus, but not E. coli, triggers colitis in HLA‑B27‑transgenic rats94,95. The differential colitogenic capacity of these bacteria cannot be merely explained by differences in the host species (mouse versus rat), as B. vulgatus, but not E. coli, induces intestinal inflamma‑tion in mice that are deficient in both IL‑10R2 and TGFβ receptor 2 (REF. 122). These findings indicate that specific immune defects in the host might determine the ability of individual commensal bacteria to trigger colitis (FIG. 3). However, the physiological relevance of these findings remains unclear because monocolonization of germ‑free mice does not mimic the normal complex gut ecosystem.

Mice that have only innate immune cells with a defi‑ciency in T‑bet (Tbx21−/− (gene encoding T‑bet) Rag−/− mice; also known as TRUC mice) develop spontaneous ulcerative colitis‑like inflammation123. This colitis is trans‑missible to T‑bet‑sufficient Rag−/− mice and even to wild‑type mice through co‑housing, which indicates that impaired T‑bet signalling in innate immune cells leads to the overgrowth of pathobionts that cause disease123. Indeed, the analysis of gut microbial communities in TRUC mice showed that Klebsiella pneumoniae and Proteus mirabilis, both of which are found in very low abundance in healthy mice, accumulate in the guts of TRUC mice124. These bac‑teria can trigger colitis in T‑bet‑sufficient Rag−/− mice and in wild‑type mice, which indicates that they can function as pathobionts. Interestingly, although K. pneumoniae or P. mirabilis can induce colitis in healthy mice, mono‑ or dual‑colonization of germ‑free TRUC mice with these bacteria does not, which indicates that interactions with additional commensal bacteria are required to actually trigger inflammation124. The proteobacterium Helicobacter typhlonius also accumulates in colitic TRUC mice, and oral administration of the bacterium is sufficient to induce colitis in non‑colitic TRUC mice, but not in T‑bet‑sufficient Rag−/− mice125. Similarly, the colitis induced by H. typhlonius is not transmissible to wild‑type mice125.

Figure 3 | Protective and pathogenic role of the gut microbiota in IBD. During homeostasis (left‑hand side), the gut microbiota has important roles in the development of intestinal immunity. Beneficial subsets of commensal bacteria tend to have anti‑inflammatory activities. Pathobionts that are colitogenic are directly suppressed by beneficial commensal bacteria partly through the induction of regulatory immune responses, involving regulatory T (T

Reg) cells, interleukin‑10

(IL‑10) and regenerating islet‑derived protein 3γ (REGIIIγ). In inflammatory bowel disease (IBD) (right‑hand side), a combination of genetic factors (for example, mutations in nucleotide‑binding oligomerization domain 2 (Nod2), autophagy‑related gene 16‑like 1 (Atg16l1) and interleukin‑23 receptor (Il23r)) and environmental factors (such as infection, stress and diet) result in disruption of the microbial community structure, a process termed dysbiosis. Dysbiosis results in a loss of protective bacteria and/or in the accumulation of colitogenic pathobionts, which leads to chronic inflammation involving hyperactivation of T helper 1 (T

H1) and T

H17 cells. Dashed line shows that that the suppression of

pathobionts by beneficial bacteria is diminished. In certain contexts, pathobionts can be transferred to the host and can cause disease without the host having a predisposing genetic susceptibility. It is unknown whether unclassified bacteria have a role in the pathogenesis of IBD. GALT, gut‑associated lymphoid tissue.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 329

© 2013 Macmillan Publishers Limited. All rights reserved

Page 10: Role of the gut microbiota in immunity and inflammatory ...

iNKT cellsInvariant natural killer (iNKT) cells; a population of cells that are thought to be particularly important in bridging the innate and adaptive immune responses. iNKT cells are typified by a capacity for self-recognition and rapid release of cytokines such as interferon-γ.

There is increasing evidence that defects in the NOD‑like receptor (NLR) family of proteins are associated with dysbiosis. For example, Nlrp6−/− mice have impaired IL‑18 production, which is associated with an abnormal expan‑sion of the commensal bacterial species Prevotellaceae and the candidate division TM7 (REF. 126). Although Nlrp6−/− mice do not develop spontaneous colitis, these mice have an increased susceptibility to dextran sulphate sodium (DSS)‑induced colitis, and this phenotype is transmis‑sible to wild‑type mice by co‑housing126. This study demonstrates a correlation between the accumulation of Prevotellaceae and TM7, as well as DSS‑induced colitis susceptibility, but whether these bacteria are the direct cause of this susceptibility and whether their accumu‑lation is indeed NLRP6‑dependent remain to be deter‑mined. Similarly, bacterial genomic sequencing shows that Nod2−/− mice, which have an increased susceptibility to DSS‑induced colitis, have an altered microbiota that can be transferred to wild‑type mice to increase dis‑ease susceptibility127. However, the specific pathobionts involved have not yet been identified.

Diet has a considerable effect on the composition of microbial communities and can lead to the expansion of pathobionts. For instance, a diet rich in saturated milk fats, but not in polyunsaturated vegetable oil fats, induces dysbiosis and expansion of Bilophila wadsworthia, which is a hydrogen‑consuming, sulphite‑reducing commen‑sal bacterium. This intestinal bacterium promotes pro‑inflammatory TH1 immunity and exacerbates colitis in IBD‑prone Il10−/− mice but not in wild‑type mice128. Although further studies are necessary, the increase in taurine conjugation of hepatic bile acids might stimulate the growth of sulphite‑reducing microorganisms such as B. wadsworthia, which promote harmful inflammation via mechanisms that remain to be elucidated128.

Taken together, these observations indicate that defects in the immune system and/or changes in diet can trigger dysbiosis, which results in the accumulation of pathobionts that promote transmissible or non‑trans‑missible colitis (FIG. 3). There is increasing evidence that dysbiosis affects susceptibility to developing not only IBD but also colitis‑associated colon cancer in mouse models, as well as colon cancer in humans127,129–133. The role of commensal bacteria in intestinal cancer has been recently reviewed134 and is not discussed here.

Protective effect of the microbiota in IBD. Although colitogenic pathobionts promote the development of IBD, commensal bacteria are also crucial for reducing IBD susceptibility, and this has generated considerable interest in the development of probiotic approaches to prevent IBD (TABLE 1). The protective effect of com‑mensal bacteria is evident from studies using germ‑free mice, which are more susceptible to DSS‑induced colitis than conventionally housed mice135–137. Consistent with these observations, deficiency in TLR2, TLR4, TLR9 or MYD88 are all associated with increased susceptibility to DSS‑induced colitis138–140. Tlr5−/− mice also develop spon‑taneous colitis in certain mouse housing facilities, which strongly indicates that the microbiota might affect disease susceptibility141,142. However, the precise mechanisms by

which the microbiota mediates resistance against the development of colitis through TLR signalling remain unclear. Administration of LPS (a TLR4 ligand) or CpG (a TLR9 ligand) protects mice from colitis by promoting epithelial barrier function following the upregulation of cytoprotective heat shock proteins or by the induction of type I IFN production, respectively138,139,143. In addi‑tion, the production of protective IgA and IgM suppresses the harmful dissemination of commensal bacteria after colonic damage through MYD88 signalling in B cells140. The human commensal bacterium B. fragilis also pro‑tects mice from colitis by promoting the accumulation of IL‑10‑producing TReg cells in the colon76,144. This protective effect is mediated by the outer membrane polysaccharide A (PSA) of B. fragilis76,144. TLR2‑mediated internalization of PSA into DCs, followed by its surface presentation, activates IL‑10‑producing TReg cells145,146. Commensal bacterial stimulation through MYD88 also limits the trafficking of CX3CR1+ mononuclear phagocytes that capture luminal non‑invasive bacteria to the mesenteric lymph nodes (MLNs)147. Consequently, disruption of the commensal microbiota and/or MYD88 signalling results in excessive B and T cell stimulation in the MLNs, which might contribute to the development of IBD.

It has also been reported that the microbiota protects against colitis through TLR‑independent mechanisms. For example, the colonization of neonatal germ‑free mice with whole microbiota results in decreased hyper‑methylation of the CXC‑chemokine ligand 16 (Cxcl16) gene75. This epigenetic modification of the Cxcl16 gene reduces CXCL16 expression and CXCL16‑mediated accumulation of iNKT cells that can contribute to colitis development75. Clostridium spp. (clusters IV and XIVa) and ASF also promote the development of FOXP3+ TReg cells, which can protect against chemically induced colitis19,20 (FIG. 3). Differentiation of TReg cells that are induced by Clostridium spp. (clusters IV and XIV) is independent of NOD1, NOD2, caspase‑recruitment domain‑containing protein 9 (CARD9; a signalling molecule downstream of Dectin‑1) and MYD88 sig‑nalling19; however, the induction of TReg cells by ASF requires MYD88–TRIF signalling20. Intestinal commen‑sal bacteria can also exert protective anti‑inflammatory effects through their production of metabolites, such as SCFAs136. Likewise, numbers of Faecalibacterium praus‑nitzii, a principal member of the phylum Firmicutes, are reduced in patients with Crohn’s disease. Culture super‑natants of F. prausnitzii containing secreted metabolites exhibit anti‑inflammatory effects on human epithelial cell lines in vitro, and they decrease the susceptibility of mice to chemically induced colitis in vivo, although it is unclear whether these effects are TLR‑independent148. Thus, commensal bacteria and their products have activ‑ities that can protect the host against inflammation by different mechanisms (FIG. 3).

Microbiota and extra-intestinal diseasesMultiple sclerosis. The gut microbiota has the capacity to affect the development of autoimmune central nerv‑ous system (CNS) disorders. Broad‑spectrum antibi‑otics are given orally to mice reduce the symptoms of

R E V I E W S

330 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 11: Role of the gut microbiota in immunity and inflammatory ...

Nature Reviews | Immunology

BasophilB cell

iNKT cell

Gut lumen

Laminapropria

IgE

Pancreas Lungs

↓ Firmicutes ↑ Bacteroidetes

TReg cell

IL-1β

CNS Joint

TH17 cell IL-1R

antagonist

TH17 cell

TH17 cell

Peripheral blood

Multiple sclerosisEAE

Type 1 diabetesPancreatitis

Allergicinflammation

Arthritis

Beneficial commensal bacteria

SFB

Experimental autoimmune encephalomyelitis(EAE). An experimental model of multiple sclerosis that is induced by immunization of susceptible animals with myelin-derived antigens, such as myelin basic protein, proteolipid protein or myelin oligodendrocyte glycoprotein.

K/BxN transgenic miceMice formed by crossing non-obese diabetic (NOD)/Lt mice with KRN T cell receptor-transgenic mice on the C57BL/6 background. As the KRN receptor on T cells recognizes a peptide from the autoantigen glucose-6-phosphate isomerase, these mice develop an arthritis that is mediated, and transferable, by circulating antibodies against glucose-6- phosphate isomerase.

experimental autoimmune encephalomyelitis (EAE)149. In one model of multiple sclerosis, mice are immunized with the self antigen myelin oligodendrocyte glyco‑protein (MOG) in complete Freund’s adjuvant (CFA). Disease symptoms in either MOG–CFA‑induced EAE or in a spontaneous EAE mouse model are reduced when the mice are housed under germ‑free conditions150,151. Monocolonization of germ‑free mice with SFB results in an increase in the number of TH17 cells in both the intestinal lamina propria and the CNS, which results in severe EAE150 (FIG. 4). Thus, SFB‑enhanced TH17 cell‑mediated inflammation might contribute to EAE exac‑erbation. However, it is unclear whether the disease is caused by the migration of SFB‑specific TH17 cells into the CNS or by the expansion of pathogenic autoantigen‑specific T cells that are promoted by intestinal TH17 cell responses (FIG. 4). By contrast, certain populations of commensal bacteria are capable of attenuating CNS inflammation. For example, PSA+ B. fragilis, which

induces FOXP3+ TReg cell differentiation, can prevent EAE symptoms152 (FIG. 4). Thus, the pathogenesis of CNS disorders might ultimately depend on the balance of different community members in the gut microbiota.

Arthritis. Autoimmune arthritis, such as rheumatoid arthritis, is a systemic inflammatory disease that pri‑marily affects the joints but can also affect other parts of the body. The events that trigger the development of autoimmune arthritis remains unknown. However, in mouse models, the gut microbiota contributes to disease symptoms. Arthritis symptoms in K/BxN transgenic mice are reduced in a germ‑free environment136,153. As in EAE, TH17 cell responses are implicated in promoting disease, and SFB‑mediated enhancement of TH17 cell immunity stimulates autoantibody production by B cells, which leads to arthritic symptoms153 (FIG. 4). TH17 cell immunity is also a key factor in spontaneous rheumatoid arthri‑tis in IL‑1R antagonist (Il1rn)−/− mice, which exhibit

Figure 4 | Gut microbiota affects extra-intestinal autoimmune diseases. Segmented filamentous bacteria (SFB) colonization induces T helper 17 (T

H17) cell development in the intestine. These T

H17 cells might migrate to the periphery

to affect systemic and central nervous system (CNS) immunity; increased intestinal TH17 cells enhance the expansion

of pathogenic autoantigen-specific T cells in the intestine and cause inflammation in the CNS. By contrast, ‘beneficial’ commensal bacteria can attenuate CNS inflammation through the induction of forkhead box P3 (FOXP3)+ regulatory T (T

Reg)

cells. Induced TH17 cells can also promote autoimmune arthritis by facilitating autoantibody production by B cells (not

shown). In addition, microbiota‑induced interleukin‑1β (IL‑1β) signalling participates in the development of rheumatoid arthritis through the induction of T

H17 cells. The IL‑1 receptor (IL‑1R) antagonist blocks IL‑1β signalling and abrogates

joint inflammation. Balance in the microbial community also determines susceptibility to type 1 diabetes. A decreased Firmicutes/Bacteroidetes ratio as a result of a deficiency in myeloid differentiation primary‑response protein 88 (MYD88) in non‑obese diabetic mice is associated with an attenuated risk of type 1 diabetes. SFB‑induced T

H17 cells protect the host

against type 1 diabetes development by an unknown mechanism. Finally, exposure to microorganisms in neonatal, but not adult, life decreases the accumulation of invariant natural killer T (iNKT) cells in the gut, which results in protection against allergic inflammation in the lungs. In addition, microbial compounds stimulate peripheral B cells through B cell-intrinsic MYD88 signalling and inhibit IgE production. Decreased levels of peripheral IgE result in decreased numbers of basophils, and attenuate the risk of allergic airway inflammation. EAE, experimental autoimmune encephalomyelitis.

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 331

© 2013 Macmillan Publishers Limited. All rights reserved

Page 12: Role of the gut microbiota in immunity and inflammatory ...

excessive IL‑1 signalling, enhanced IL‑17 expression and increased arthritic symptoms154. Similar to K/BxN mice, arthritic symptoms in Il1rn−/− mice are diminished when the mice are housed in germ‑free conditions, which indicates that the microbiota might be essential for the progression of arthritis in this model, although whether SFB are involved is unclear155 (FIG. 4). Together with the observation that IL‑1β signalling is essential for TH17 cell differentiation16, these results indicate that the microbiota is involved in the pathogenesis of rheumatoid arthritis partly through the promotion of TH17 cell responses.

T1D. Type 1 diabetes (T1D) is an autoimmune disorder that results in the destruction of insulin‑producing cells in the pancreas. Recent studies with NOD mice, which have been used to model this disease, have provided some insights into how the gut microbiota can affect the devel‑opment of T1D. Diabetes in NOD mice is ameliorated in the absence of MYD88 (REF. 156). However, protection against diabetes in Myd88−/− NOD mice is abrogated by antibiotic treatment and in germ‑free conditions156. These data indicate that commensal bacteria might be impor‑tant for reducing disease susceptibility in this model. Indeed, 16S ribosomal RNA sequencing showed altera‑tions in the composition of the microbiota in Myd88−/− NOD mice compared with their Myd88‑sufficient NOD littermates. These alterations were characterized by a lower Firmicutes/Bacteroides ratio on a phylum level and indicate that certain bacterial populations might contribute to protection against T1D (FIG. 4). As it has been recently demonstrated that deficiencies in MYD88 or individual TLRs alone do not affect the composition of the gut microbiota157, it is possible that the genetic background of the NOD mice contributes to the effect of MYD88 deficiency on microbiota composition.

Interestingly, SFB has also been implicated in pro‑tection against T1D, which indicates that intestinal TH17 cells in certain contexts might be protective, despite their link to autoimmunity158 (FIG. 4). SFB‑mediated pro‑tection against T1D seems to be gender‑specific: male NOD mice with SFB have a lower incidence of T1D than their female counterparts158. Furthermore, the microbiota in male NOD mice is distinct from that in females and it contributes to increased testosterone levels, which are associated with protection against T1D159. Indeed, trans‑ferring the microbiota from male into female NOD mice results in increased levels of testosterone and reduced susceptibility to T1D in female NOD mice159. Much work remains to be done to understand the complexities of TH17 cell responses and the mechanisms by which the gut microbiota contributes to the development of T1D.

Allergic inflammation. Under germ‑free conditions, host immune responses are TH2‑biased17. Restoration of the gut microbiota in germ‑free mice results in an increase in TH1 and TH17 cells and a reduction of TH2‑type responses to levels that are observed in conven‑tionally housed mice17. This indicates that the microbi‑ota might be important in maintaining balance in TH cell responses. The mechanism by which the microbiota achieves this is still unclear. Germ‑free mice or mice

treated with antibiotics have an expansion of basophils in the peripheral blood as well as increased serum IgE levels160 (FIG. 4). The expansion of basophils as a result of microbiota depletion exacerbates the allergic airway inflammation that is triggered by exposure to house dust mite allergen160. The microbiota is directly sensed by B cells and activates MYD88 signalling to limit IgE class‑switching in B cells. As CpG stimulation alone can prevent IgE production by B cells, the TLR9–MYD88 axis might have a crucial role in IgE responses that can lead to allergic inflammation160.

Consistent with this idea, allergic responses to food antigens are aggravated in antibiotic‑treated mice or in Tlr4−/− mice, but are ameliorated by CpG treatment161. Thus, exposure to microorganisms that stimulate TLR signalling might be important in reducing the develop‑ment of allergic inflammation by the promotion of TH1 rather than TH2 cell polarization. A recent study dem‑onstrated that neonatal colonization of the gut micro‑biota is essential for reducing numbers of iNKT cells in the intestine, and iNKT cells have been implicated in mediating allergic responses in the lungs75 (FIG. 4). Taken together, the essential role of the gut microbiota in regu‑lating immune balance in the intestine might explain its ability to influence susceptibility to allergies. These find‑ings support the hygiene hypothesis, which arose from the observation that early exposure to bacteria during infancy is associated with a reduced incidence of allergic diseases.

Other extra-intestinal diseases. Systemic dissemination of the gut microbiota has a crucial role in the develop‑ment of severe acute pancreatitis162. Cerulein‑induced pancreatitis is dramatically reduced in Nod1−/− mice, which indicates that there might be an important role for commensal bacteria‑mediated NOD1 activation in the development of pancreatitis163. NOD1 stimulation induces CC‑chemokine ligand 2 (CCL2) production, thereby promoting the recruitment of CCR2+ monocytes that facilitate pancreatitis163.

Intestinal dysbiosis might also be linked to the pathogenesis of cystic fibrosis, as mice with cystic fibrosis‑like disease exhibit an increased abundance of Mycobacteriaceae spp., Enterobacteriaceae spp., Clostridaceae spp., and B. fragilis164. However, the pre‑cise mechanisms by which a dysbiotic microbiota affects the pathophysiology of cystic fibrosis still remains to be elucidated.

PerspectivesThe gut microbiota has been studied for more than a century; however, recent studies have shown ever‑expanding roles for these microscopic organisms in health and disease. Despite the complexity of the gut microbiota, there is a delicate balance in bacterial popu‑lations such that any disruption in this balance leads to dysbiosis and, consequently, to decreased resistance to pathogen colonization, to the favoured growth of patho‑bionts and to pathological immune responses. Although the association of dysbiosis with disease pathogenesis has become evident, it is still largely unknown which

NOD miceNon-obese diabetic (NOD) mice spontaneously develop type 1 diabetes as a result of autoreactive T cell-mediated destruction of pancreatic β-islet cells.

Hygiene hypothesisThis hypothesis originally stated that the increased incidence of atopic diseases in westernized countries was a consequence of living in an overly clean environment, which resulted in an understimulated immune system that responded inappropriately to harmless antigens. More recently it has been proposed that an absence of exposure to pathogens, in particular helminths, might predispose individuals to both increased allergy and autoimmune disease.

R E V I E W S

332 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 13: Role of the gut microbiota in immunity and inflammatory ...

factors are important in triggering dysbiosis. Obviously, both genetic and environmental factors are important in shaping the microbiota; however, the relative contribu‑tions of these two groups of factors, and the mechanism by which they interact to lead to dysbiosis, remain areas of active investigation.

Other questions that need to be further addressed are whether dysbiosis is disease‑specific and whether the timing with which dysbiosis occurs during the lifetime of the host is important for disease pathogenesis, espe‑cially as microbiota colonization early in life is crucial for the optimal development and function of the immune

system. Given the importance of the gut microbiota and dysbiosis in disease pathogenesis, targeting the micro‑biota is an attractive therapeutic approach. In fact, faecal microbiota transplantation (FMT) has been highly effec‑tive in the treatment of Clostridium difficile infection165, which indicates that the correction of dysbiosis might be a valid approach for reducing susceptibility to other diseases, including IBD. Clearly, much work remains to be done not only to achieve a deeper understanding of host–microbial relationships but also to establish the best way to use that relationship to prevent or to treat diseases within and outside the intestine.

1. Hooper, L. V. & Macpherson, A. J. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nature Rev. Immunol. 10, 159–169 (2010).

2. Renz, H., Brandtzaeg, P. & Hornef, M. The impact of perinatal immune development on mucosal homeostasis and chronic inflammation. Nature Rev. Immunol. 12, 9–23 (2011).

3. Stecher, B. & Hardt, W. D. Mechanisms controlling pathogen colonization of the gut. Curr. Opin. Microbiol. 14, 82–91 (2011).

4. Littman, D. R. & Pamer, E. G. Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microbe 10, 311–323 (2011).

5. Honda, K. & Littman, D. R. The microbiome in infectious disease and inflammation. Annu. Rev. Immunol. 30, 759–795 (2012).

6. Gordon, H. A., Bruckner-Kardoss, E. & Wostmann, B. S. Aging in germ-free mice: life tables and lesions observed at natural death. J. Gerontol. 21, 380–387 (1966).

7. Hamada, H. et al. Identification of multiple isolated lymphoid follicles on the antimesenteric wall of the mouse small intestine. J. Immunol. 168, 57–64 (2002).

8. Bouskra, D. et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 456, 507–510 (2008).

9. Moreau, M. C. & Corthier, G. Effect of the gastrointestinal microflora on induction and maintenance of oral tolerance to ovalbumin in C3H/HeJ mice. Infect. Immun. 56, 2766–2768 (1988).

10. Pabst, O. et al. Adaptation of solitary intestinal lymphoid tissue in response to microbiota and chemokine receptor CCR7 signaling. J. Immunol. 177, 6824–6832 (2006).

11. Littman, D. R. & Rudensky, A. Y. Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140, 845–858 (2010).

12. Atarashi, K. et al. ATP drives lamina propria TH17 cell differentiation. Nature 455, 808–812 (2008).

13. Ivanov, I. I. et al. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4, 337–349 (2008).

14. Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).This study identifies SFB as a main driver of intestinal TH17 cell differentiation, which can protect the host against enteric pathogen infection.

15. Gaboriau-Routhiau, V. et al. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31, 677–689 (2009).

16. Shaw, M. H., Kamada, N., Kim, Y. G. & Nunez, G. Microbiota-induced IL-1β, but not IL-6, is critical for the development of steady-state TH17 cells in the intestine. J. Exp. Med. 209, 251–258 (2012).

17. Chung, H. et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 149, 1578–1593 (2012).

18. Powrie, F., Leach, M. W., Mauze, S., Caddle, L. B. & Coffman, R. L. Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. Int. Immunol. 5, 1461–1471 (1993).

19. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).In this study, the authors identify a cocktail of 46 strains of Clostridium spp. that efficiently induce the development of peripherally derived FOXP3+ TReg cells in the colon, which are important for preventing intestinal inflammation and allergy.

20. Geuking, M. B. et al. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 34, 794–806 (2011).

21. Round, J. L. et al. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 332, 974–977 (2011).

22. 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. 28, 243–273 (2010).

23. Strugnell, R. A. & Wijburg, O. L. The role of secretory antibodies in infection immunity. Nature Rev. Microbiol. 8, 656–667 (2010).

24. Macpherson, A. J., Geuking, M. B. & McCoy, K. D. Homeland security: IgA immunity at the frontiers of the body. Trends Immunol. 33, 160–167 (2012).

25. Fagarasan, S. et al. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science 298, 1424–1427 (2002).This report shows that intestinal IgA responses can shape the commensal bacterial community.

26. Wei, M. et al. Mice carrying a knock-in mutation of Aicda resulting in a defect in somatic hypermutation have impaired gut homeostasis and compromised mucosal defense. Nature Immunol. 12, 264–270 (2011).

27. Peterson, D. A., McNulty, N. P., Guruge, J. L. & Gordon, J. I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2, 328–339 (2007).

28. Suzuki, K. et al. The sensing of environmental stimuli by follicular dendritic cells promotes immunoglobulin A generation in the gut. Immunity 33, 71–83 (2010).

29. Mora, J. R. et al. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 314, 1157–1160 (2006).

30. Uematsu, S. et al. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nature Immunol. 9, 769–776 (2008).

31. Tezuka, H. et al. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature 448, 929–933 (2007).

32. Tezuka, H. et al. Prominent role for plasmacytoid dendritic cells in mucosal T cell-independent IgA induction. Immunity 34, 247–257 (2011).

33. Fritz, J. H. et al. Acquisition of a multifunctional IgA+ plasma cell phenotype in the gut. Nature 481, 199–203 (2011).

34. Walker, J. A., Barlow, J. L. & McKenzie, A. N. Innate lymphoid cells — how did we miss them? Nature Rev. Immunol. 13, 75–87 (2013).

35. Sonnenberg, G. F. & Artis, D. Innate lymphoid cell interactions with microbiota: implications for intestinal health and disease. Immunity 37, 601–610 (2012).

36. Cella, M. et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 457, 722–725 (2009).In this report, the authors identify IL‑22‑producing ILC3s in mucosal tissues, including the intestine, in both mice and humans.

37. Spits, H. & Di Santo, J. P. The expanding family of innate lymphoid cells: regulators and effectors of immunity and tissue remodeling. Nature Immunol. 12, 21–27 (2011).

38. Satoh-Takayama, N. et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 29, 958–970 (2008).

39. Sanos, S. L. et al. RORγt and commensal microflora are required for the differentiation of mucosal interleukin 22-producing NKp46+ cells. Nature Immunol. 10, 83–91 (2009).

40. Sawa, S. et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nature Immunol. 12, 320–326 (2011).

41. Zheng, Y. et al. Interleukin-22 mediates early host defense against attaching and effacing bacterial pathogens. Nature Med. 14, 282–289 (2008).

42. Vaishnava, S. et al. The antibacterial lectin RegIIIγ promotes the spatial segregation of microbiota and host in the intestine. Science 334, 255–258 (2011).In this study, the authors demonstrate that bacterial stimuli from commensal microbiota promote the production of the antimicrobial molecule REGIIIγ by the epithelium in a MYD88‑dependent process. REGIIIγ functions as an additional barrier against the microbiota.

43. Sonnenberg, G. F. et al. Innate lymphoid cells promote anatomical containment of lymphoid-resident commensal bacteria. Science 336, 1321–1325 (2012).

44. Osawa, N. & Mitsuhashi, S. Infection of germfree mice with Shigella flexneri 3A. Jpn J. Exp. Med. 34, 77–80 (1964).

45. Fukuda, S. et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 469, 543–547 (2011).

46. Bohnhoff, M., Drake, B. L. & Miller, C. P. Effect of streptomycin on susceptibility of intestinal tract to experimental Salmonella infection. Proc. Soc. Exp. Biol. Med. 86, 132–137 (1954).

47. Hentges, D. J. & Freter, R. In vivo and in vitro antagonism of intestinal bacteria against Shigella flexneri I. Correlation between various tests. J. Infect. Dis. 110, 30–37 (1962).

48. Lawley, T. D. et al. Antibiotic treatment of Clostridium difficile carrier mice triggers a supershedder state, spore-mediated transmission, and severe disease in immunocompromised hosts. Infect. Immun. 77, 3661–3669 (2009).

49. Rupnik, M., Wilcox, M. H. & Gerding, D. N. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nature Rev. Microbiol. 7, 526–536 (2009).

50. Momose, Y., Hirayama, K. & Itoh, K. Competition for proline between indigenous Escherichia coli and E. coli O157:H7 in gnotobiotic mice associated with infant intestinal microbiota and its contribution to the colonization resistance against E. coli O157:H7. Antonie Van Leeuwenhoek 94, 165–171 (2008).

51. Momose, Y., Hirayama, K. & Itoh, K. Effect of organic acids on inhibition of Escherichia coli O157:H7 colonization in gnotobiotic mice associated with infant intestinal microbiota. Antonie Van Leeuwenhoek 93, 141–149 (2008).

52. Fabich, A. J. et al. Comparison of carbon nutrition for pathogenic and commensal Escherichia coli strains in the mouse intestine. Infect. Immun. 76, 1143–1152 (2008).

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 333

© 2013 Macmillan Publishers Limited. All rights reserved

Page 14: Role of the gut microbiota in immunity and inflammatory ...

53. Leatham, M. P. et al. Precolonized human commensal Escherichia coli strains serve as a barrier to E. coli O157:H7 growth in the streptomycin-treated mouse intestine. Infect. Immun. 77, 2876–2886 (2009).

54. Kamada, N. et al. Regulated virulence controls the ability of a pathogen to compete with the gut microbiota. Science 336, 1325–1329 (2012).This study demonstrates the importance of commensal bacteria in eradicating pathogens by competing for shared nutrients. The expression of virulence factors by the pathogen allows it to localize to the epithelium as a means of escaping nutritional competition by the microbiota.

55. Endt, K. et al. The microbiota mediates pathogen clearance from the gut lumen after non-typhoidal Salmonella diarrhea. PLoS Pathog. 6, e1001097 (2010).

56. Pacheco, A. R. et al. Fucose sensing regulates bacterial intestinal colonization. Nature 492, 113–117 (2012).

57. Le Bouguenec, C. & Schouler, C. Sugar metabolism, an additional virulence factor in enterobacteria. Int. J. Med. Microbiol. 301, 1–6 (2010).

58. Bertin, Y. et al. Enterohaemorrhagic Escherichia coli gains a competitive advantage by using ethanolamine as a nitrogen source in the bovine intestinal content. Environ. Microbiol. 13, 365–377 (2011).

59. Winter, S. E. et al. Gut inflammation provides a respiratory electron acceptor for Salmonella. Nature 467, 426–429 (2010).This study demonstrates that an enteric pathogen acquires a growth advantage in the presence of intestinal inflammation. Pathogen‑induced inflammation promotes neutrophil recruitment and the conversion of a commensal metabolite to a form that can be used by the pathogen.

60. Thiennimitr, P. et al. Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. Proc. Natl Acad. Sci. USA 108, 17480–17485 (2011).

61. Murdoch, S. L. et al. The opportunistic pathogen Serratia marcescens utilizes type VI secretion to target bacterial competitors. J. Bacteriol. 193, 6057–6069 (2011).

62. Gueguen, E. & Cascales, E. Promoter swapping unveils the role of the Citrobacter rodentium CTS1 type VI secretion system in interbacterial competition. Appl. Environ. Microbiol. 79, 32–38 (2013).

63. Johansson, M. E. et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc. Natl Acad. Sci. USA 105, 15064–15069 (2008).

64. Bergstrom, K. S. et al. Muc2 protects against lethal infectious colitis by disassociating pathogenic and commensal bacteria from the colonic mucosa. PLoS Pathog. 6, e1000902 (2010).

65. Petersson, J. et al. Importance and regulation of the colonic mucus barrier in a mouse model of colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 300, G327–G333 (2010).

66. Cash, H. L., Whitham, C. V., Behrendt, C. L. & Hooper, L. V. Symbiotic bacteria direct expression of an intestinal bactericidal lectin. Science 313, 1126–1130 (2006).

67. Brandl, K., Plitas, G., Schnabl, B., DeMatteo, R. P. & Pamer, E. G. MyD88-mediated signals induce the bactericidal lectin RegIIIγ and protect mice against intestinal Listeria monocytogenes infection. J. Exp. Med. 204, 1891–1900 (2007).

68. Dessein, R. et al. Toll-like receptor 2 is critical for induction of Reg3β expression and intestinal clearance of Yersinia pseudotuberculosis. Gut 58, 771–776 (2009).

69. van Ampting, M. T. et al. Intestinally secreted C-type lectin Reg3β attenuates salmonellosis but not listeriosis in mice. Infect. Immun. 80, 1115–1120 (2012).

70. Denning, T. L., Wang, Y. C., Patel, S. R., Williams, I. R. & Pulendran, B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nature Immunol. 8, 1086–1094 (2007).

71. Franchi, L. et al. NLRC4-driven production of IL-1β discriminates between pathogenic and commensal bacteria and promotes host intestinal defense. Nature Immunol. 13, 449–456 (2012).This study demonstrates that NLRC4 selectively recognizes pathogenic, but not commensal, bacteria and that it promotes protective immunity against pathogens via induction of mature IL‑1β.

72. Kamada, N. et al. 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. 175, 6900–6908 (2005).

73. Kiss, E. A. et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334, 1561–1565 (2011).

74. Qiu, J. et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104 (2012).

75. Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 336, 489–493 (2012).

76. Mazmanian, S. K., Round, J. L. & Kasper, D. L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453, 620–625 (2008).In this study, the authors report that a human commensal bacterium, B. fragilis, protects against pathobiont‑induced intestinal inflammation, and that the PSA in its capsule is sufficient for this protection. Specifically, PSA induces IL‑10 production from CD4+ T cells that protect against intestinal inflammation.

77. O’Mahony, C. et al. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-κB activation. PLoS Pathog. 4, e1000112 (2008).

78. Macpherson, A. J. et al. A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science 288, 2222–2226 (2000).

79. Lathrop, S. K. et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254 (2011).

80. Hand, T. W. et al. Acute gastrointestinal infection induces long-lived microbiota-specific T cell responses. Science 337, 1553–1556 (2012).

81. Slack, E. et al. Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science 325, 617–620 (2009).

82. Fagundes, C. T. et al. Transient TLR activation restores inflammatory response and ability to control pulmonary bacterial infection in germfree mice. J. Immunol. 188, 1411–1420 (2012).

83. Clarke, T. B. et al. Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature Med. 16, 228–231 (2010).

84. Ichinohe, T. et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl Acad. Sci. USA 108, 5354–5359 (2011).

85. Abt, M. C. et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 37, 158–170 (2012).

86. Ganal, S. C. et al. Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity 37, 171–186 (2012).

87. Benson, A., Pifer, R., Behrendt, C. L., Hooper, L. V. & Yarovinsky, F. Gut commensal bacteria direct a protective immune response against Toxoplasma gondii. Cell Host Microbe 6, 187–196 (2009).

88. Naik, S. et al. Compartmentalized control of skin immunity by resident commensals. Science 337, 1115–1119 (2012).

89. Giel, J. L., Sorg, J. A., Sonenshein, A. L. & Zhu, J. Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PLoS ONE 5, e8740 (2010).

90. Kane, M. et al. Successful transmission of a retrovirus depends on the commensal microbiota. Science 334, 245–249 (2011).

91. Kuss, S. K. et al. Intestinal microbiota promote enteric virus replication and systemic pathogenesis. Science 334, 249–252 (2011).

92. Sellon, R. K. et al. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect. Immun. 66, 5224–5231 (1998).

93. Dianda, L. et al. T cell receptor-α β-deficient mice fail to develop colitis in the absence of a microbial environment. Am. J. Pathol. 150, 91–97 (1997).

94. Taurog, J. D. et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J. Exp. Med. 180, 2359–2364 (1994).

95. Rath, H. C., Wilson, K. H. & Sartor, R. B. Differential induction of colitis and gastritis in HLA-B27 transgenic rats selectively colonized with Bacteroides vulgatus or Escherichia coli. Infect. Immun. 67, 2969–2974 (1999).

96. Schultz, M. et al. IL-2-deficient mice raised under germfree conditions develop delayed mild focal intestinal inflammation. Am. J. Physiol. 276, G1461–G1472 (1999).

97. Bamias, G. et al. Commensal bacteria exacerbate intestinal inflammation but are not essential for the development of murine ileitis. J. Immunol. 178, 1809–1818 (2007).

98. Kobayashi, M. et al. Toll-like receptor-dependent production of IL-12p40 causes chronic enterocolitis in myeloid cell-specific Stat3-deficient mice. J. Clin. Invest. 111, 1297–1308 (2003).

99. Rakoff-Nahoum, S., Hao, L. & Medzhitov, R. Role of Toll-like receptors in spontaneous commensal-dependent colitis. Immunity 25, 319–329 (2006).

100. Hoshi, N. et al. MyD88 signalling in colonic mononuclear phagocytes drives colitis in IL-10-deficient mice. Nature Commun. 3, 1120 (2012).

101. Nenci, A. et al. Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 446, 557–561 (2007).

102. Smythies, L. E. et al. Human intestinal macrophages display profound inflammatory anergy despite avid phagocytic and bacteriocidal activity. J. Clin. Invest. 115, 66–75 (2005).

103. Kamada, N. et al. Unique CD14 intestinal macrophages contribute to the pathogenesis of Crohn disease via IL-23/IFN-γ axis. J. Clin. Invest. 118, 2269–2280 (2008).

104. Cho, J. H. The genetics and immunopathogenesis of inflammatory bowel disease. Nature Rev. Immunol. 8, 458–466 (2008).

105. Ogura, Y. et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 411, 603–606 (2001).

106. Hugot, J. P. et al. Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn’s disease. Nature 411, 599–603 (2001).

107. Inohara, N. et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J. Biol. Chem. 278, 5509–5512 (2003).

108. Hampe, J. et al. A genome-wide association scan of nonsynonymous SNPs identifies a susceptibility variant for Crohn disease in ATG16L1. Nature Genet. 39, 207–211 (2007).

109. Parkes, M. et al. Sequence variants in the autophagy gene IRGM and multiple other replicating loci contribute to Crohn’s disease susceptibility. Nature Genet. 39, 830–832 (2007).

110. Travassos, L. H. et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nature Immunol. 11, 55–62 (2010).

111. Cooney, R. et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nature Med. 16, 90–97 (2010).

112. Homer, C. R., Richmond, A. L., Rebert, N. A., Achkar, J. P. & McDonald, C. ATG16L1 and NOD2 interact in an autophagy-dependent antibacterial pathway implicated in Crohn’s disease pathogenesis. Gastroenterology 139, 1630–1641.e2 (2010).

113. Bevins, C. L. & Salzman, N. H. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nature Rev. Microbiol. 9, 356–368 (2011).

114. Ogura, Y. et al. Expression of NOD2 in Paneth cells: a possible link to Crohn’s ileitis. Gut 52, 1591–1597 (2003).

115. Wehkamp, J. et al. NOD2 (CARD15) mutations in Crohn’s disease are associated with diminished mucosal α-defensin expression. Gut 53, 1658–1664 (2004).

116. Petnicki-Ocwieja, T. et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc. Natl Acad. Sci. USA 106, 15813–15818 (2009).

117. Cadwell, K. et al. A key role for autophagy and the autophagy gene Atg16l1 in mouse and human intestinal Paneth cells. Nature 456, 259–263 (2008).

118. Cadwell, K. et al. Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell 141, 1135–1145 (2010).

119. Kim, S. C. et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology 128, 891–906 (2005).

R E V I E W S

334 | MAY 2013 | VOLUME 13 www.nature.com/reviews/immunol

© 2013 Macmillan Publishers Limited. All rights reserved

Page 15: Role of the gut microbiota in immunity and inflammatory ...

120. Waidmann, M. et al. Bacteroides vulgatus protects against Escherichia coli-induced colitis in gnotobiotic interleukin-2-deficient mice. Gastroenterology 125, 162–177 (2003).

121. Bohn, E. et al. Host gene expression in the colon of gnotobiotic interleukin-2-deficient mice colonized with commensal colitogenic or noncolitogenic bacterial strains: common patterns and bacteria strain specific signatures. Inflamm. Bowel Dis. 12, 853–862 (2006).

122. Bloom, S. M. et al. Commensal Bacteroides species induce colitis in host-genotype-specific fashion in a mouse model of inflammatory bowel disease. Cell Host Microbe 9, 390–403 (2011).

123. Garrett, W. S. et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 131, 33–45 (2007).In this study, the authors show that colitogenic pathobionts can accumulate in genetically susceptible mice and can transmit disease to wild‑type mice.

124. Garrett, W. S. et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe 8, 292–300 (2010).

125. Powell, N. et al. The transcription factor T-bet regulates intestinal inflammation mediated by interleukin-7 receptor+ innate lymphoid cells. Immunity 37, 674–684 (2012).

126. Elinav, E. et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 145, 745–757 (2011).This report demonstrates that impaired NLRP6 signalling and IL‑18 production is associated with the development of dysbiosis and the accumulation of potentially colitogenic pathobionts.

127. Couturier-Maillard, A. et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J. Clin. Invest. 123, 700–711 (2013).

128. Devkota, S. et al. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10−/− mice. Nature 487, 104–108 (2012).In this study, the authors show that a milk‑derived fat diet increases the level of bile acids in the intestine, which results in the accumulation of colitogenic pathobionts in genetically susceptible IBD‑prone mice.

129. Lowe, E. L. et al. Toll-like receptor 2 signaling protects mice from tumor development in a mouse model of colitis-induced cancer. PLoS ONE 5, e13027 (2010).

130. Chen, G. Y., Shaw, M. H., Redondo, G. & Nunez, G. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res. 68, 10060–10067 (2008).

131. Wu, S. et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nature Med. 15, 1016–1022 (2009).

132. Arthur, J. C. et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338, 120–123 (2012).

133. Kostic, A. D. et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 22, 292–298 (2012).

134. Hajishengallis, G., Darveau, R. P. & Curtis, M. A. The keystone–pathogen hypothesis. Nature Rev. Microbiol. 10, 717–725 (2012).

135. Kitajima, S., Morimoto, M., Sagara, E., Shimizu, C. & Ikeda, Y. Dextran sodium sulfate-induced colitis in germ-free IQI/Jic mice. Exp. Anim. 50, 387–395 (2001).

136. Maslowski, K. M. et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282–1286 (2009).

137. Saha, S. et al. Peptidoglycan recognition proteins protect mice from experimental colitis by promoting normal gut flora and preventing induction of interferon-γ. Cell Host Microbe 8, 147–162 (2010).

138. Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S. & Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 118, 229–241 (2004).

139. Lee, J. et al. Maintenance of colonic homeostasis by distinctive apical TLR9 signalling in intestinal epithelial cells. Nature Cell Biol. 8, 1327–1336 (2006).

140. Kirkland, D. et al. B cell-intrinsic MyD88 signaling prevents the lethal dissemination of commensal bacteria during colonic damage. Immunity 36, 228–238 (2012).

141. Vijay-Kumar, M. et al. Deletion of TLR5 results in spontaneous colitis in mice. J. Clin. Invest. 117, 3909–3921 (2007).

142. Carvalho, F. A. et al. Interleukin-1β (IL-1β) promotes susceptibility of Toll-like receptor 5 (TLR5) deficient mice to colitis. Gut 61, 373–384 (2012).

143. Katakura, K. et al. Toll-like receptor 9-induced type I IFN protects mice from experimental colitis. J. Clin. Invest. 115, 695–702 (2005).

144. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl Acad. Sci. USA 107, 12204–12209 (2010).

145. Mazmanian, S. K., Liu, C. H., Tzianabos, A. O. & Kasper, D. L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 122, 107–118 (2005).

146. Shen, Y. et al. Outer membrane vesicles of a human commensal mediate immune regulation and disease protection. Cell Host Microbe 12, 509–520 (2012).

147. Diehl, G. E. et al. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells. Nature 494, 116–120 (2013).

148. Sokol, H. et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl Acad. Sci. USA 105, 16731–16736 (2008).

149. Ochoa-Reparaz, J. et al. Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis. J. Immunol. 183, 6041–6050 (2009).

150. Lee, Y. K., Menezes, J. S., Umesaki, Y. & Mazmanian, S. K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl Acad. Sci. USA 108, S4615–S4622 (2011).

151. Berer, K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).

152. Ochoa-Reparaz, J. et al. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 3, 487–495 (2010).

153. Wu, H. J. et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32, 815–827 (2010).

154. Nakae, S. et al. IL-17 production from activated T cells is required for the spontaneous development of destructive arthritis in mice deficient in IL-1 receptor antagonist. Proc. Natl Acad. Sci. USA 100, 5986–5990 (2003).

155. Abdollahi-Roodsaz, S. et al. Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse model of arthritis. J. Clin. Invest. 118, 205–216 (2008).

156. Wen, L. et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature 455, 1109–1113 (2008).This report demonstrates that altered microbial communities in MYD88‑deficient NOD mice prevent the development of T1D, which indicates that certain subsets of commensal microbiota might have the capacity to protect against the development of T1D.

157. Ubeda, C. et al. Familial transmission rather than defective innate immunity shapes the distinct intestinal microbiota of TLR-deficient mice. J. Exp. Med. 209, 1445–1456 (2012).This study indicates that differences in bacterial communities in mice that are deficient in individual TLRs or MYD88 are more strongly related to maternal transmission and cage effects than to an inherent genetic susceptibility.

158. Kriegel, M. A. et al. Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice. Proc. Natl Acad. Sci. USA 108, 11548–11553 (2011).

159. Markle, J. G. et al. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science 339, 1084–1088 (2013).

160. Hill, D. A. et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nature Med. 18, 538–546 (2012).

161. Bashir, M. E., Louie, S., Shi, H. N. & Nagler-Anderson, C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J. Immunol. 172, 6978–6987 (2004).

162. Frossard, J. L., Steer, M. L. & Pastor, C. M. Acute pancreatitis. Lancet 371, 143–152 (2008).

163. Tsuji, Y. et al. Sensing of commensal organisms by the intracellular sensor NOD1 mediates experimental pancreatitis. Immunity 37, 326–338 (2012).

164. Lynch, S. V. et al. Cystic fibrosis transmembrane conductance regulator knockout mice exhibit aberrant gastrointestinal microbiota. Gut Microbes 4, 41–47 (2013).

165. Borody, T. J. & Khoruts, A. Fecal microbiota transplantation and emerging applications. Nature Rev. Gastroenterol. Hepatol. 9, 88–96 (2011).

166. Li, X. D. et al. Mitochondrial antiviral signaling protein (MAVS) monitors commensal bacteria and induces an immune response that prevents experimental colitis. Proc. Natl Acad. Sci. USA 108, 17390–17395 (2011).

167. Negishi, H. et al. Essential contribution of IRF3 to intestinal homeostasis and microbiota-mediated Tslp gene induction. Proc. Natl Acad. Sci. USA 109, 21016–21021 (2012).

AcknowledgementsThis work is supported by grants from the US National Institutes of Health and the Bill & Melinda Gates Foundation, USA. N.K. is supported by a Research Fellowship from the Crohn’s and Colitis Foundation of America.

Competing interests statementThe authors declare no competing financial interests.

FURTHER INFORMATIONGabriel Núñez’s homepage: http://www.med.umich.edu/immprog/faculty/nunezg.htm

ALL LINKS ARE ACTIVE IN THE ONLINE PDF

R E V I E W S

NATURE REVIEWS | IMMUNOLOGY VOLUME 13 | MAY 2013 | 335

© 2013 Macmillan Publishers Limited. All rights reserved