Nature the Immune System and the Gut

Embed Size (px)

Citation preview

  • 8/16/2019 Nature the Immune System and the Gut

    1/10

    Initially, all microorganisms were viewed aspathogens that cause and propagate infectiousdiseases and, as a field, immunology was builtaround the paradigm that the host immunesystem should recognize and eliminate theseintruders (non-self) while tolerating self-molecules to preserve homeostasis. However,the persistent association of animal and plantspecies with obligate and facultative symbionts now shows that both bacteria and theireukaryote hosts benefit from their coopera-tive relationships. These benefits suggest thatco-evolution has selected mechanisms thatpromote and maintain associations betweenbacteria and eukaryotes. In humans, trillionsof bacteria are distributed in complex andsite-specific communities on the skinand at mucosal surfaces, and the largestcommunity is found in the distal gut. As thesebacteria encode hundreds of genes that are

    absent in the human genome1, the idea hasemerged that together with our microbiota,we form superorganisms in which energy andmetabolites can be exchanged2 and homeo-stasis is maintained by the immune system3.Therefore, a new paradigm proposes that theimmune system has evolved to accommodatecolonization by symbiotic bacterial commu-nities of increasing complexity while retainingthe capacity to fight pathogens.

    The gastrointestinal tract is the primarysite of interactions between the host and themicrobiota. How bacterial colonization of

    the gut might influence the developmentand functions of the immune system hasbecome a major focus of interest. A prevalenttheory, derived from hypotheses that werefirst postulated by Metchnikoff a centuryago, proposes that individual members ofthe microbiota might influence the balancebetween pro-inflammatory and regulatoryhost responses and that alterations in thecomposition of the microbiota (a process thatis known as dysbiosis) could jeopardize hostimmune responses and promote the devel-opment of various inflammatory disorders.Here, we discuss the principles that governthe interactions between the intestinal micro-biota and the host immune system, both inhealth and in disease. Moreover, we stresshow the complexity of the gut ecological sys-tem and the reciprocal nature of the regula-tion of the immune system and of microbial

    community structures must be consideredbefore one can draw any conclusions aboutthe role of the microbiota in disease andpropose therapeutic interventions.

    The host–microbiota interaction in the gut

    The intestine is an open ecological systemthat is colonized immediately after birthby a microbial population that reachesan impressive density of 1012 bacteria pergram of luminal content in the distal gut.Colonization is initiated by maternallyacquired bacteria during birth; these

    bacteria are then followed by hundredsof environmentally acquired species,which differ between individuals butmainly belong to two bacterial phylotypes,Firmicutes spp. and Bacteroidetes spp.2,4.A growing number of studies supportthe view that eukaryotic hosts and theirsymbionts have co-evolved towards mutu-alistic interactions that are based on thenutritional benefits that each partner gainsfrom the association5 (BOX 1). However,the huge collection of bacteria at the gutsurface is also a major threat to host integ-

    rity and has driven the selection of highlyflexible defence mechanisms, which enableeukaryotic hosts to cope with their micro-bial environment and compensate for theirless rapid genetic adaptation.

    The gut immune system. Recent reviewshave highlighted how the microbiota elicitsinnate and adaptive immune mechanismsthat cooperate to protect the host and main-tain intestinal homeostasis6,7. Epithelial cellsare a central component of the immunesystem of the gut. In a similar manner toimmune cells, epithelial cells express recep-tors for microbial-associated molecularpatterns (MAMPs). These receptors activatesignalling cascades that finely tune epithelialcell production of antimicrobial productsand chemokines, depending on the signalsthat are delivered by the microbiota (FIG. 1).Thus, gut epithelial cells form a potent andinducible physico-chemical barrier, whichlimits microbial growth and access to thegut surface. They can also recruit leukocytesto complement their barrier function or toparticipate in the activation of gut adap-tive immune responses. In mammals, the

    development of gut-associated lymphoidtissues (GALTs) is initiated before birth bya genetic programme8. However, GALTmaturation and the recruitment of IgA-secreting plasma cells and activated T cellsto mucosal sites only occurs after birth andis strictly dependent on microbiota-derivedsignals; these signals influence the crosstalkbetween epithelial cells and gut dendriticcells (DCs), thereby modulating the natureand intensity of intestinal B and T cellresponses7,9 (FIG. 2). In immunocompetentmice, intestinal colonization stimulates

    O P I N I O N

    The immune system and the gutmicrobiota: friends or foes?

    Nadine Cerf-Bensussan and Valérie Gaboriau-Routhiau

    Abstract | The mammalian intestine is home to a complex community of trillions of

    bacteria that are engaged in a dynamic interaction with the host immune system.

    Determining the principles that govern host–microbiota relationships is the focus

    of intense research. Here, we describe how the intestinal microbiota is able toinfluence the balance between pro-inflammatory and regulatory responses and

    shape the host’s immune system. We suggest that improving our understanding of

    the intestinal microbiota has therapeutic implications, not only for intestinal

    immunopathologies but also for systemic immune diseases.

    PERSPECTIVES

    NATURE REVIEWS | IMMUNOLOGY VOLUME 10 | OCTOBER 2010 | 735

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    2/10

    the production of secretory IgA, the dif-ferentiation of effector T helper 1 (T

    H1),

    TH2 and T

    H17 cells, and the development of

    regulatory T (TReg

    ) cells10.It is increasingly clear how these adap-

    tive immune elements cooperate with innateimmune cells to strengthen the gut barrierand protect the host from invading patho-gens. An outstanding issue now is to definehow individual members of the microbiotaor microbiota-derived products can affectthe balance between pro-inflammatory and

    regulatory immune responses, and to establishwhether the composition of the microbiotacan influence the development of inflamma-tory diseases in and beyond the gut. Beforeconsidering the possible role of the microbiotain disease, we will first highlight how the dif-ferent colonization strategies of individualmembers of the microbiota can influence thedevelopment and function of the gut immunesystem and show that, ultimately, it is the hostimmune system that determines whether abacterium is a friend or a foe.

     Anti-inflammatory roles of the microbiota. Current results indicate that a trade-off isestablished between the host immune sys-tem and the bulk of the microbiota, so thatin a healthy individual, intestinal coloniza-tion stimulates host production of micro-bicidal peptides11 and secretory IgA, whichin turn contain the microbiota within theintestinal lumen and neutralize MAMPs12.These mechanisms protect the host fromthe systemic translocation of bacteria orbacterial products and from the outburst ofpro-inflammatory cascades in intestinal epi-thelial and innate cells13. Conversely, the resi-dent bacteria also benefit from the symbioticrelationship and can thrive in the mucus,thus minimizing destruction by host-derivedinflammatory mediators. Hosts and bacteriahave evolved additional strategies to main-tain ‘friendly’ relationships. Thus, signallingcascades that occur downstream of Toll-like

    receptors (TLRs) can be desensitized bycontinuous exposure to lipopolysaccharide(LPS)14 or can be attenuated by other solublemediators that are produced by the micro-biota (FIG. 1). Furthermore, some microbiota-derived soluble products can promote thefunctions of T

    Reg cells15,16.

    The mechanism that maintains thisfriendly relationship has been elucidatedin the case of Bacteroides fragilis, which is acommon culturable member of the micro-biota15. This bacterium possesses an unusualcapsular polysaccharide A (PSA) that is ableto drive the differentiation of interleukin-10(IL-10)-secreting T

    Regcells. Colonization by

    a wild-type B. fragilis, but not by a mutantstrain that lacks PSA, protected mice fromthe severe experimental colitis that is induced

    Box 1 | Mutualistic relationships between hosts and their intestinal microbiota

    The human intestine harbours an estimated 100 trillion bacteria, 70–80% of which cannot yet be

    cultured. Each individual is thought to host several hundred species of bacteria from only 7 to 9

    phylotypes; these are mainly Gram-positive Firmicutes spp. (most notablyClostridium spp.,

    Enterococcus spp. and Lactobacillus spp.) and Gram-negative Bacteroidetes  spp. Recent

    metagenomics studies predict a core of ~1,200 prevalent species and a total intestinal

    microbiome that contains 150-fold more genes than the human genome4. The gut microbiome

    encodes a core of redundant bacterial genes that are likely to be needed to resist stressfulconditions in the host intestine63 and to harvest nutrients that are necessary for bacterial

    growth2,4. Competition between bacteria with distinct metabolic requirements might explain the

    massive and rapid shifts in the structure of the intestinal microbial community that are provoked

    by changes in host diet64. In addition to the genes that are necessary for microbial adaptation to

    the host environment, the gut microbiome encodes multiple biosynthetic pathways that are

    predicted to greatly increase the host’s capacity to metabolize glycans and xenobiotics and to

    synthesize vitamins2,4. Moreover, studies in gnotobiotic mice have shown the broad influence of

    the gut microbiota on host physiology. Intestinal colonizaton induces a spectrum of intestinal

    and metabolic changes, which promote the digestion and absorption of nutrients and stimulate

    fat storage65,66, accelerate gut epithelial renewal and alter epithelial locomotor activity67. The

    signalling pathways that are involved remain largely elusive, but recent observations suggest

    that overlapping mechanisms have been selected during host–microbiota co-evolution that

    simultaneously control host metabolic and innate immune responses to the microbiota. In mice,

    inactivation of Toll-like receptor 5 (TLR5), which is a receptor for bacterial flagellin that has an

    established role in host innate immune responses, results in severe obesity and profoundalterations in the microbiota structure68. Furthermore, peroxisome proliferator-activated

    receptor-γ (PPARγ), which is a transcription factor that has a central role in glucidolipidicmetabolism, can control the production of microbicidal peptides by colonocytes and serves

    as a feedback mechanism for the activation of nuclear factor-κB (NF-κB) in enterocytes69.

    Glossary

     Ankylosing enthesopathy

    An inflammatory autoimmune disease of the joints

    that naturally occurs in mice on a C57BL/10 genetic

    background; the disease is similar to human ankylosing

    spondylitis. The pathology is characterized by the

    proliferation of cartilage and connective tissue, which

    culminates in ankylosis of the joints.

    Germinal centres

    Highly specialized and dynamic microenvironments that

    are located in secondary lymphoid tissues and give rise

    to secondary B cell follicles during an immune response.

    Germinal centres are the main sites of B cell proliferation

    and differentiation, which leads to the generation of

    memory B cells and plasma cells that produce high-affinity

    antibodies.

    Gnotobiotic mice

    Germ-free mice are born and raised in sterile isolators

    and are devoid of colonization by any microorganisms,

    but after they have been experimentally colonized by

    known bacteria, they are said to be gnotobiotic. They

    are kept in isolators to control their bacterial status.

    IgE-associated allergies

    Type 1 hypersensitivity reactions that are mediated by

    IgE, which induces mast cell activation and degranulation.

    Such immune reactions are seen in asthma, allergic rhinitis,

    systemic anaphylaxis and food allergies.

    Obligate and facultative symbionts

    Obligate microbial symbionts need to colonize a hostto develop and multiply, unlike facultative microbial

    symbionts, which can also develop outside a host.

    Pathobionts

    Microbial symbionts that can cause defined disease in

    predisposed hosts following changes in the gastrointestinal

    environment.

    Microbiome

    The whole genome of all of the microorganisms that

    colonize a specific environment.

    Peyer’s patches

    Collections of lymphoid follicles that are located in the

    intestinal mucosa and are particularly abundant in the ileal

    mucosa. Together with mesenteric lymph nodes, they form

    the inductive compartment for intestinal immune responses.

    Proteobacteria

    Gram-negative microorganisms that colonize very distinct

    environments and are the second largest group of bacteria

    on earth. Proteobacteria that colonize the intestine include

    commensal, pathogenic and opportunistic species, such asSalmonella, Shigella and Helicobacter  spp. and Escherichia 

    coli  strains. In healthy adults, proteobacteria represent less

    than 1% of the enteric microbiota, but they are a major

    cause of intestinal and extraintestinal diseases.

    Type VI secretion system

    (T6SS). Like T3SS and T4SS, T6SS is a multi-subunit

    complex that acts like a ‘needle and syringe’ to

    translocate bacterial products across the

    double-membrane of Gram-negative bacteria into

    the cytoplasm of eukaryotic cells.

    Xenobiotics

    Chemical compounds that are foreign to a living organism

    and that can be toxic, even at low concentrations.

    P E R S P E C T I V E S

    736 | OCTOBER 2010 | VOLUME 10 www.nature.com/reviews/immunol

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    3/10

      | 

    Intestinal lumen

    Intestinalepithelial cellMYD88

    TLR 

    PAMP

    p50 p65

    p50 p65

    p65

    PPAR γ

    PPAR γ

    NF-κB

    IκB

    IRAK1Expression of IRAK1decreased by LPSfrom microbiota

    Nuclear translocationof NF-κB

    NF-κB transcribespro-inflammatorycytokines andchemokines anddefensins

    IκB ubiquitylated andtargeted to the proteasomefor degradation

    ROS induced bymicrobiota inhibitubiquitin ligases

    PPAR γ induced in

    response to microbiotaLPS diverts NF-κBfrom nucleus

    PPAR γ upregulatescolonic β-defensinsto sustain gut barrier

    Pathway impairedin patients withCrohn’s disease

    Bacterium

    by Helicobacter hepaticus15,17. Furthermore,administration of PSA reduced the sever-ity of disease in a model of trinitrobenzenesulphonic acid (TNBS)-induced colitis17.Therefore, B. fragilis may  represent a proto-type peace-keeper strain. Yet the outcomeof host–microbiota interactions cannot bepredicted from only the bacterium itself, anda bacterium that is beneficial for an immuno-competent host can become a dangerous foewhen the immune system is weakened. Thisis illustrated by the fact that B. fragilis causessevere sepsis in immunocompromised hosts.Likewise, normally harmless members of themicrobiota can initiate intestinal inflamma-tion in individuals who cannot mount effi-cient intestinal humoral responses18,19 and inindividuals with impaired intestinal immuno-regulation, most notably in those who lack afunctional IL-10 signalling pathway 20,21.

    Promotion of effector immune responses. Although certain members of the microbiotahave adopted peace-keeper activities to colo-nize the intestine, others are, undoubtedly,endowed with pro-inflammatory properties.One such group, which has recently attractedmuch attention, is segmented filamentousbacteria (SFB). These unculturable speciessettle in the rodent intestine at the time ofweaning and stimulate the postnatal matu-ration of immune responses in the mousegut. Mice that are colonized by a microbiotathat lacks SFB have weaker IgA antibodyresponses22 and much poorer intestinalT cell responses compared with mice thatare colonized with SFB. Notably, mice that arecolonized by an SFB-deficient microbiotalack mucosal T

    H17 cells10,23. Furthermore,

    these animals cannot control colonization bythe invasive pathogen Citrobacter rodentium,which suggests that microbiota-inducedimmune responses participate in the bar-rier function of the flora23. This hypothesisis also supported by recent work that showsthat the destruction of the microbiota fol-lowing treatment with antibiotics can jeop-ardize innate immune responses in the gut

    and promote colonization by pathogens24.A striking feature of SFB is their strong

    adherence to the surface epithelium of theileum and the Peyer’s patches shortly afterweaning25. This is in contrast with most othermembers of the microbiota, which remainentrapped within the mucus and have littleor no physical contact with host epithelium25.This attachment, which is perhaps necessaryto initiate the replication of SFB, is likely tofacilitate the sampling and presentation ofSFB antigens to T cells by DCs in the Peyer’spatches and to stimulate pro-inflammatory

    signalling pathways in epithelial cells andDCs, resulting in robust innate and adap-tive immune responses in the intestine10,23.Such behaviour is characteristic of bona fidepathogens, which use host inflammatoryresponses to eliminate the resident flora andto colonize the remaining niches26,27. Thisattachment might enable SFB to settle in the

    mouse intestine, but it also benefits the hostby strengthening the gut barrier. Strikingly,on the basis of morphological studies, SFBhave been detected in all species studied fromarthropods to mammals, including humans,and closely related 16S rRNA sequences havebeen found in chickens, trout and rodents25,28.Therefore, it is tempting to speculate that this

    Figure 1 | Modulation of intestinal epithelial cell pro-inflammatory responses by the microbiota.

    In a similar manner to immune cells, epithelial cells detect microbes through pattern-recognition recep-tors, including Toll-like receptors (TLRs). Upon TLR ligation, adaptor proteins, such as myeloid differen-

    tiation primary-response protein 88 (MYD88), are recruited and activate signalling cascades, notably

    the nuclear factor-κB (NF-κB) pathway, which stimulates the transcription of antimicrobial proteins,pro-inflammatory cytokines and chemokines. In resting cells, NF-κB is sequestered in the cytoplasm byits inhibitor IκB. Following TLR activation, IκB is phosphorylated, ubiquitylated and degraded by theproteasome, which allows nuclear translocation of NF-κB and transcription of NF-κB target genes. Thispathway can be modulated by microbiota-derived factors, preventing excessive and potentially delete-

    rious host pro-inflammatory responses. Immediately after birth, expression of the interleukin-1 receptor-

    associated kinase 1 (IRAK1), which is the proximal activator of the NF-κB cascade, is downregulated bymicrobiota-derived lipopolysaccharide (LPS)14. The polyubiquitylation and degradation of IκB can beinhibited by commensal bacteria, which inhibit a common ubiquitin ligase by inducing reactive oxygen

    species (ROS)70. Peroxisome proliferator- activated receptor-γ (PPARγ), which is induced in response toTLR4 activation by LPS71, can also divert NF-κB from the nucleus72. Checkpoints that are controlled bythe microbiota are indicated by T bars. Interestingly, PPARγ positively controls the expression of the

    colonic microbicidal peptide defensin 1 (REF. 69) and thus can simultaneously sustain the gut barrierand prevent excessive inflammation. This mechanism may be impaired in a subset of patients with

    colonic Crohn’s disease69. PAMP, pathogen-associated molecular pattern.

    P E R S P E C T I V E S

    NATURE REVIEWS | IMMUNOLOGY VOLUME 10 | OCTOBER 2010 | 737

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    4/10

      | 

    Intestinal epithelium

    Lamina propria

    Bacterium

    CD103+ DC

    Peyer’s patch and CD103+ DCsmigrate to MLNs and initiateadaptive immune responses

    Peyer’s patch

    Intestinal lumen

    Microbiota-derived productsactivate TLRs

    BAFF and APRILpromote T cell-dependent andT cell-independentIgA class-switching

    IgA+ plasma cell

    IgA molecules aretranscytosed by polymericIgA receptors and retainbacteria in the mucus

    DimericsecretoryIgA

    Mucus layer

    TLR 

    M cell

    Naive T cell

    TCR 

    MHC

    Peptide

    Epithelialcell

    BAFF,APRIL

    a

    cb

    TSLP,TGFβ,retinoicacid

    Tolerogenic DC FOXP3+

    TReg cell

    TH17 cell TH1 cell

    ATPSAA

    InflammatoryDCs

    IL-1, IL-6,IL-23

    IL-12

    SFB

    B. fragilis capsularpolysaccharide A

    B. fragilis

    TReg cellinduction

    TReg cells maintaintolerance to foodand commensals

    TH1 and TH17 cells sustain intestinal barrierby recruiting macrophages and neutrophilsand inducing antibacterial defensins

    Bacterial ATP andbacteria-inducedSAA activate DCs

    SFB induceIL-12-producingDCs by unknownmechansims

    CX3CR1-expressingphagocyte

    Direct sampling of bacteria in lumen?Antigen transfer?

    CD103+ DCs acquirebacterial antigens

    Transcytosed bacteria areacquired by DCs, which activateadaptive immune responses

    Figure 2 | | Modulation of adaptive immune responses in the gut by the

    microbiota. a | Intestinal adaptive immune responses can be initiated in

    Peyer’s patches or in mesenteric lymph nodes (MLNs). Activated T and B cells

    subsequently leave these lymphoid tissues and home to the intestinal lamina

    propria via the bloodstream. Bacteria are mainly sampled by Peyer’s patch

    dendritic cells (DCs) after transcytosis across the specialized epithelium that

    overlays these lymphoid organs. It has also been suggested that a population

    of CX3C-chemokine receptor 1 (CX3CR1)+ lamina propria cells with both DC-

    and macrophage-like characteristics can send dendrites into the intestinal

    lumen and directly capture bacteria. Their role in antigen presentation

    remains controversial73,74, but they may pass antigens to lamina propria

    CD103+ DCs, which can migrate to the MLNs and present antigens to T cells74.

    b | Microbiota-derived products activate Toll-like receptors (TLRs) that are

    expressed by intestinal epithelial cells, which leads to the production of

    B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL);

    these cytokines promote both T cell-dependent and T cell-independent IgA

    class-switching responses in the intestine75,76. Plasma cells produce dimeric

    IgA molecules that are transcytosed into the intestinal lumen by the epithe-

    lial polymeric Ig receptor, the expression of which is upregulated by the

    microbiota. The extracellular part of this receptor remains associated with

    IgA following release into the lumen and forms secretory IgA. These

    secretory IgA molecules form immune complexes with bacteria, which are

    then retained in the mucus. c | CD103+ DCs are ‘conditioned’ by epithelial

    cell-derived factors, such as thymic stromal lymphopoietin (TSLP), transform-

    ing growth factor-β (TGFβ) and retinoic acid, to acquire a tolerogenic pheno-type; these DCs can promote the induction of forkhead box P3 (FOXP3) + 

    regulatory T (TReg

    ) cells77. Bacteria-derived products, such as the capsular

    polysaccharide A from Bacteroides fragilis, can further promote the induction

    of interleukin-10 (IL-10)-producing TReg cells through a TLR2-dependentmechanism15,17. However, some commensal bacteria can stimulate the dif-

    ferentiation of inflammatory mucosal T cells. T helper 17 (TH17) cell differen-

    tiation can be promoted by bacteria-derived ATP, which activates a subset of

    DCs that produce IL-1β, IL-6 and IL-23 (REF. 78), or by serum amyloid A pro-tein (SAA), which is an acute phase protein that is produced in response to

    segmented filamentous bacteria (SFB)23. SFB can also drive the expansion of

    mucosal TH1 cell populations11, presumably by inducing IL-12 production by

    DCs. In addition, inflammatory DCs might stimulate the conversion of

    TReg

     cells into TH17 and/or T

    H1 cells in the lamina propria (not shown)79. T

    H1

    and TH17 cells maintain the intestinal barrier by recruiting and activating

    macrophages and neutrophils that eliminate penetrating bacteria. IL-22 that

    is produced by a subset of TH17 cells can also promote the production of

    antibacterial defensins by epithelial cells. M cell, microfold cell.

    P E R S P E C T I V E S

    738 | OCTOBER 2010 | VOLUME 10 www.nature.com/reviews/immunol

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    5/10

      | 

    Intestinal colonization by SFB

    Effects on intestinal compartmentEffects on peripheral compartment

    Arthritis inK/BxN mice

    Increasedseverity of EAE

    Physiological inflammationstrengthens gut barrier inimmunocompetent hosts

    Intestinal inflammationin SCID mice transferredwith effector T cells

    • Stimulation of innate immuneresponses e.g. Reg IIIβ /γ production

    • Stimulation of CD4+ T cell responses e.g.↑ TH1, TH2, TH17 and TReg cells

    • Induction of IgA responses• Recruitment and activation of IELs

    • Increased germinal centreformation in the spleen

      ↑ Autoantibody-secretingIgG plasma cells

      ↑ Circulating immunecomplexes

    • Enhancedsensitizationfollowingchallenge withMOG peptide

    unusual symbiont has a particularly impor-tant role in shaping the gut immune systemacross evolution. However, again, the out-come of SFB–host interactions depends onthe immune status of the host; SFB has beenassociated with the development of intestinalinflammation29 or arthritis30 in mice withimpaired immunoregulation and can alsoaggravate experimental autoimmune enceph-alomyelitis31 (FIG. 3). Interestingly, SFB alonewere not able to induce intestinal inflamma-tion in immunodeficient mice and insteadsynergized with a pathogen-free flora. Thisfinding highlights how interactions in themicrobiota community can influence hostimmune responses, thereby adding anadditional level of complexity.

    The case of pathobionts. A particular sub-set of bacteria further exemplifies how thebehaviour of the microbiota is dependent on

    the immune status of the host. Although thesebacteria, which are known as pathobionts,colonize the gastrointestinal tract of manyindividuals asymptomatically, they also havethe potential to cause disease. A recent studyshowed that Helicobacter hepaticus, whichis a member of the epsilon subgroup ofproteobacteria, uses its type VI secretion system (T6SS) to regulate bacterial colonization andinhibit host innate and adaptive immuneresponses, thereby actively maintainingsymbiotic relationships with immunocom-petent hosts32. However, this bacterium cancause severe typhocolitis in Il10–/– mice orin the severe combined immunodeficient(SCID) transfer model of colitis. Thesefindings highlight the central role of hostadaptive regulatory responses in main-taining symbiotic relationships with themicrobiota32.

    In humans, the most classical exampleof a pathobiont is Helicobacter pylori, whichuses various mechanisms to dampen hostimmune responses and persist in the stom-ach. Gastric colonization by this bacteriumremains asymptomatic in most individu-als and has even been suggested to protect

    against the development of oesophagealcarcinomas owing to downmodulation ofgastric acid secretion. Yet this bacteriumis the major cause of gastritis and gastriccancers. The circumstances that lead to thisbacterium becoming a serious health con-cern are not completely understood but arethought to include the selection of specifi-cally aggressive strains and/or host predis-posing factors, notably polymorphisms ingene promoters that increase the productionof tumour necrosis factor (TNF) or IL-1β,which is a pro-inflammatory cytokine

    with potent acid-suppressive properties33.Other recently characterized pathobiontsinclude enterotoxigenic B. fragilis (ETBF),which can stimulate colonic inflammationand tumorigenesis in predisposed multipleintestinal neoplasia (MIN) mice34, and someEscherichia coli strains, which can promotegut inflammation in patients with Crohn’sdisease35,36.

    Intestinal dysbiosis and IBDs

    Inflammatory bowel diseases (IBDs) are

    thought to arise owing to a combination ofgenetic and environmental factors that resultin dysregulated immune responses to the gutmicrobiota and the subsequent developmentof gut inflammation20. Compelling evidencefrom a variety of studies has shown dysbiosisin patients with IBD compared with healthycontrols, and this suggests a causative role fordysbiosis in gut inflammation.

    Several scenarios can be considered.Pro-inflammatory bacteria, such as entero-invasive Escherichia coli strains, are morefrequently seen in the ileal mucosa of

    patients with Crohn’s disease than in healthycontrols, which suggests that these bacteriacan initiate disease36. Yet to induce intestinalinflammation, the prototype E. coli LF82strain first needs to bind to an epithelialcell-expressed receptor. This receptoris absent in the normal ileal mucosa but isupregulated by interferon-γ (IFNγ) andTNF that are produced during intestinalinflammation35,37. An alternative hypo-thesis suggests that the reduced frequencyof a Firmicutes species, Faecalibacterium

     prausnitzii, in the intestinal microbiota isa causative factor of Crohn’s disease16. Thisstrain releases an unidentified soluble factor,which inhibits pro-inflammatory epithelialcell responses in vitro and attenuatesinflammation in a mouse model of colitis16.

    Other studies, however, suggest thatmore global changes in the compositionof the microbiota are associated with IBD,such as abnormal adherence of bacteria tothe gut mucosa, reduced bacterial diversity,decreased levels of resident Firmicutes spp.and/or Bacteroides spp. and an overgrowth

    Figure 3 | Effects of SFB colonization on the immune system. Segmented filamentous bacteria

    (SFB) are spore-forming bacteria that are related to the genus Clostridium28. Inherited from the

    mother microbiota, SFB develop strong interactions with the ileal mucosa and in immunocompe-

    tent mice, the bacteria can largely recapitulate the inducing effects of the whole microbiota on

    the postnatal maturation of the gut immune system. SFB induce the production of Reg IIIβ/γ micro-bicidal peptides10,23,80, which protect against colonizing pathogens24. Additionally, SFB simultane-

    ously activate strong secretory IgA responses22, induce the recruitment and activation of cytotoxic

    intraepithelial lymphocytes (IELs)81 and drive various T cell responses, including a robust T helper 17

    (TH17) cell response11,23. In immunocompetent mice, SFB-induced pro-inflammatory and regulatory

    responses balance each other, which results in physiological inflammation that strengthens the

    gut barrier. By contrast, colonization by SFB promotes the development of colitis in severe com-

    bined immunodeficient (SCID) mice that have been reconstituted with effector T cells29. Intestinal

    colonization by SFB can also promote the development of inflammatory diseases outside of the

    gut. SFB promote arthritis in autoimmune non-obese diabetic (NOD) mice that express a trans-

    genic T cell receptor (TCR) that is specific for a self peptide (known as K/BxN mice), an effect

    ascribed to the induction of TH17 cells 30. SFB also enhance the severity of myelin oligodendrocyte

    glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). These aggravating

    effects may reflect the strong adjuvant properties of SFB. TReg

     cells, regulatory T cells.

    P E R S P E C T I V E S

    NATURE REVIEWS | IMMUNOLOGY VOLUME 10 | OCTOBER 2010 | 739

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    6/10

      | 

    a b

    Physiological microbiota

    Pathobiont

    Damaged epithelialbarrier, increasedbacterial adherenceand penetration

    ‘Peace-keeping’bacterium

    Mucus

    Laminapropria

    Healthyepithelial

    barrier

    Physiological inflammation

    Pathological inflammation

    TH17 cell

    TReg cellTH1 cell

    IgA+

    plasma cell

    Gutlumen

    Healthy gut environmentAltered gut environmentAntibiotics, diet, hygiene,pollutants, virus?

    Dysbiosis

    Decrease in ‘peace-keeping’ bacteriaand increase in pathobionts

    Severe monogenicimmunodeficiencyIL-10R mutations,

    CVID

    Immunegene variantsNOD2, ATG16L1,

    IL-23R, IRGM

    Genetics

    Environment

    Stress, diet, infections, vaccine?

    Altered host immune system

    of proteobacteria38–40. Strikingly, comparablechanges in the microbiota have been seenin mice in which intestinal inflammationwas induced by either an invasive pathogenor by the injection of transgenic T cellsthat attacked the gut epithelium27. In thesetwo distinct models, host inflammationsuppressed the growth of Firmicutes andBacteroides spp., allowing proteobacteria,which are apparently more resistant to host-derived microbicidal factors, to outcompetethese normally dominant resident bacteria27.Notably, outgrowth of proteobacteria was

    also seen in Il10–/– mice after, but not before,the onset of intestinal inflammation, whichfurther highlights the profound influence ofthe host immune response on the structureof the microbial community 27.

    Together, these observations underscorethe confounding role of host-predisposingfactors and the difficulty in assigning acausative role for dysbiosis in IBD. However,experimental evidence suggests that intes-tinal inflammation can select for bacterialspecies with colitogenic properties. In T -bet –/– Rag2–/– ulcerative colitis (TRUC) mice, the

    inability of DCs to properly regulate TNFproduction results in a severe and highlypenetrating colitis. Intestinal inflammationspontaneously progresses to colonic dyspla-sia and rectal adenocarcinoma; therefore,disease progression in this model is similarto that seen in human IBD41,42. TRUC colitiscan be prevented by eradicating the micro-biota with broad-spectrum antibiotics.Moreover, in a situation that recalls rarereports of intrafamilial transmission of IBDin humans, colitis can be transmitted fromTRUC mice to wild-type mice in both cross-fostering and co-housing experiments41.Furthermore, the microbiota in TRUCmice exhibits complex changes, includingselective enrichment of two proteobacteria,Proteus mirabilis and Klebsiella pneumonia.These two bacteria are not sufficient toinduce colitis in gnotobiotic TRUC mice;however, they can colonize the intestine

    of wild-type mice and, in concert with apathogen-free microbiota, induce colonicinflammation43. These data highlight howthe host immune response can shape themicrobiota and eventually lead to the selec-tion of aggressive bacteria, which not onlysurvive in the inf lamed gut but also promoteinflammation.

    What are the therapeutic implicationsof these findings? At the very least, theyexplain the difficulties in establishing andmaintaining remission in patients with IBDusing drugs that target host inflammatorycomponents or the microbiota without alsocorrecting host-predisposing factors. Theyhighlight the importance of identifyingsuch predisposing factors and designingmore specific therapies for IBD. They alsosuggest the need for combined approachesthat can restore local ecological conditionsand correct dysbiosis to reinstate balancedhost–microbiota interactions in the longterm (FIG. 4).

    Microbiota and systemic immunity

    So far we have focused on the roles of themicrobiota in intestinal immunity, both

    in health and disease; however, growingevidence suggests that the intestinal micro-biota can also have an important impact onthe development of the peripheral immunesystem. Moreover, dysbiosis has beenimplicated in the development of extra-intestinal immune-mediated diseases. It willbe crucial to determine the extent to whichthe impact of the microbiota dependson the host immune status, whether indi-

     vidual bacterial species can exert distinctiveroles and, ultimately, whether the observeddysbiosis has a causative role in disease.

    Figure 4 | Schematic representation of host–microbiota interactions in the healthy and

    inflamed gut. a | In healthy hosts, an efficient immune barrier contains the microbiota in the gut

    lumen and feedback mechanisms avoid excessive activation of host immune responses. ‘Peace-

    keeping’ bacteria that release anti-inflammatory products participate in the tuning of host responses

    towards tolerance and help to prevent the pro-inflammatory effects of any pathobionts that are

    present in the microbiota, thus maintaining intestinal homeostasis. b | Immunodeficient patients,

    who lack an important component of the gut barrier (for example, secretory immunoglobulins in

    patients with common variable immunodeficiency (CVID)19) or a key regulatory pathway (for exam-

    ple, loss-of-function mutations that affect the interleukin-10 receptor (IL-10R)21) spontaneouslydevelop intestinal inflammation when exposed to the microbiota. In more common forms of inflam-

    matory bowel disease (IBD), a complex genetic background results in more subtle alterations of gut

    immune responses that may weaken the gut barrier and/or impair immunoregulation82. In these indi-

    viduals, lifestyle changes or medical practices (for example, stress, diet, hygiene, smoking, antibiotics,

    vaccines or appendectomy) may promote the onset of gut inflammation by affecting the immune

    balance and/or the gut microbiota82. Intestinal inflammation results in increased bacterial adherence,

    epithelial damage and increased entry of bacteria into the intestinal lamina propria, thus sustaining

    a vicious inflammatory circle. Moreover, inflammation can favour the selection of aggressive patho-

    bionts, which are more resistant to host-derived microbicidal mediators26,27, and reduce the number

    of peace-keeping species16, which results in even more severe and uncontrolled inflammation. These

    pathobionts might become sufficiently aggressive to also cause disease in immunocompetent indi-

    viduals43. ATG16L1, autophagy-related 16-like 1; IRGM, immunity-related GTPase family M; SCID,

    severe combined immunodeficient; TH cell, T helper cell; T

    Reg cell, regulatory T cell.

    P E R S P E C T I V E S

    740 | OCTOBER 2010 | VOLUME 10 www.nature.com/reviews/immunol

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    7/10

    Effects on the peripheral immune system. The exact impact of the gut microbiota onthe host’s peripheral immune system is con-troversial. Two groups have convincinglyshown that specific immune responses tothe intestinal microbiota are largely confinedto the intestinal lymphoid compartment inimmunocompetent mice with an efficientgut barrier13,44,45. However, studies compar-ing mice that were raised in conventionalor germ-free conditions highlight theimportance of the intestinal microbiota forthe development of the peripheral immunesystem in immunocompetent hosts. Notably,the spleens of germ-free mice contain fewerand smaller germinal centres46 and decreasednumbers of memory CD4+ T cells, andcytokine production by these T cells shows aT

    H2-type profile47.The mechanisms that underlie the

    stimulatory effects of the microbiota on the

    peripheral immune system of immuno-competent hosts are not well understood.Recent work suggests that soluble factorsthat are produced by the microbiota cantranslocate from the gut to the bloodstreamand activate innate immune cells. Forexample, the opsonophagocytic activity ofneutrophils is primed by microbiota-derivedpeptidoglycan, which results in enhancedprotection against pneumococcal sepsis48.Moreover, monocolonization with B. fragilis corrects systemic T cell deficiencies andthe T

    H1/T

    H2 imbalance of germ-free mice

    due to the stimulation of DC IL-12 produc-tion by the bacterium’s unusual capsularpolysaccharide47.

     Allerg y and dysbiosis. The ‘hygienehypothesis’, which has been revisedseveral times since its initial formula-tion by Strachan in 1989, stipulates thatdecreased exposure to infectious agents,as well as changes in the intestinal micro-biota during infancy, might alter immuneregulatory networks and account for thedramatic increase in the incidence ofallergic diseases that has been observed in

    developed countries49. Several studies havereported differences in the compositionof the faecal microbiota of infants whodevelop an allergic disease and those whodo not. Notably, a decreased frequencyof Lactobacillus and Bifidobacterium spp.has been suggested to precede the onsetof allergy 50, and prophylactic approaches,which are based on the administration ofprobiotics to mothers and newborns athigh risk for IgE-associated allergies, havebeen initiated. A recent study reportsthat 1 month of prenatal and 6 months of

    postnatal pre- and probiotic supplementa-tion can reduce the incidence of eczemaand food-specific IgE in a subset of high-risk children who are born by caesareansection51. This protective effect was notseen in vaginally delivered children andwas transient, as it was significant at2 years of age but not by 5 years of age.

    The results of such interventions remainconflicting and a causative link between dys-biosis, which is induced by changes in life-style or recent medical practices, and allergyremains difficult to establish51,52. Moreover,experimental studies that support the roleof the microbiota in the development ofallergic diseases are still scarce (TABLE 1). Onestudy has reported increased developmentof allergic airway disease in mice that weretreated with a short course of oral antibiot-ics53. Another interesting study showed thatTLR4 activation by microbiota-derived

    LPS was necessary to prevent anaphylaxisafter oral immunization with the peanut-derived allergen Arah 1. TLR4-deficient orantibiotic-treated mice showed an increasedT

    H2-type skewing of cytokine responses

    compared with control mice. Conversely,activation of TLR9 by oral administration ofCpG oligodeoxynucleotides could abrogateallergic symptoms and correct the T

    H1/T

    H2

    imbalance54. However, additional work isneeded to delineate whether and how thecomposition of the microbiota mightinfluence the onset of allergy.

     Autoimmunity and the microbiota. Thepossibility that the intestinal microbiota isinvolved in the development of systemicautoimmunity has recently attracted grow-ing attention. Changes in the composition ofthe gut flora have been reported in patientsin the early phases of rheumatoid arthritis55 when compared with a control group withfibromyalgia. However, such studies cannotestablish a link between dysbiosis and thedevelopment of disease. Therefore, the roleof the microbiota has been more directlyaddressed by comparing the onset and/or

    severity of experimental autoimmune dis-eases in germ-free mice with disease onsetand/or severity in mice that have beencolonized by a diverse microbiota.

    In several models of autoimmunity,the progression of disease was compa-rable between both sets of mice56–58. Inother models, contradictory roles for themicrobiota have been reported (TABLE 1).The microbiota was shown to triggerdisease in several mouse models of auto-immune arthritis. For example, in a mousestrain that is prone to the spontaneous

    development of an autoimmune ankylosingenthesopathy , disease does not developunder germ-free conditions. Furthermore,disease develops in mice that are colonizedwith a mixture of culturable anaerobesbut not in mice that are colonized withLactobacillus or Staphylococcus spp.59. Thisfinding suggests a role for a specific com-ponent (or components) of the microbiotain disease progression. A triggering rolefor the microbiota, specifically for SFB, wasalso seen in the autoimmune arthritis thatdevelops in K/BxN mice30. In this model,an uncontrolled T

    H17 cell response that is

    induced by SFB stimulated the productionof autoantibodies and led to the deposi-tion of immune complexes in the joints(FIG. 3). The microbiota also promoteddisease in another IL-17-dependent modelof arthritis, which develops in mice thatlack the IL-1 receptor antagonist60. Finally,

    the microbiota and, to a lesser degree, SFBenhanced the severity of experimentalautoimmune encephalomyelitis31. Theaggravating role of the microbiota in thesemodels is consistent with its central role inthe induction of T

    H17 cell responses.

    By contrast, the microbiota was shownto have a protective role in collagen-induced arthritis61 and to prevent diabetesdevelopment in myeloid differentiationprimary-response protein 88 (MYD88)-deficient non-obese diabetic (NOD) mice62,as in both models disease was more severeif animals were housed in germ-free condi-tions. It remains unclear why a protectiverole for the microbiota during the develop-ment of diabetes is seen in the presenceof impaired TLR signalling. The completelack of TLR signalling in Myd88–/– Trif(TIR-domain-containing adaptor proteininducing IFNβ)–/– mice has been associ-ated with abnormal bacterial translocationinto the spleen and activation of systemicadaptive responses. It will be interesting toevaluate whether comparable mechanismsoperate in Myd88–/– NOD mice and par-ticipate in the protection against diabetes.

    Interestingly, the caecal microbiota of Myd88–/– NOD mice differed significantlyfrom that of NOD mice and could attenuatethe development of type 1 diabetes whentransferred to the newborn progeny ofgerm-free NOD mice62; this suggests thatone or more species that confer protectionagainst disease might have been selectedfor in the NOD mice with impairedTLR signalling.

    Taken together, these data providenovel examples of the complex interplaythat exists between the host immune

    P E R S P E C T I V E S

    NATURE REVIEWS | IMMUNOLOGY VOLUME 10 | OCTOBER 2010 | 741

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    8/10

    also indicates the possible contribution ofthe intestinal microbiota to immunologicaldiseases outside the gut. However, from themicrobial perspective, the host is simply acomplex environment and the distinctionbetween health and disease is importantonly as far as it affects microbial fitness.The challenge that lies ahead is to determinewhen changes in our microbiota are theprimary cause of a disease and when thesechanges merely reflect the enormous capacityof bacteria for rapid and continuous genetic

    system and the microbiota, and the dif-ficulties in attempting to delineate therespective roles of host predisposingfactors and specific bacterial species. Todetermine the therapeutic implicationsof these observations, further research isneeded to elucidate how the microbiotais able to influence peripheral immuneresponses and how such microbiota-driven responses can interfere with theimmunological mechanisms that underliea given autoimmune disease.

    Conclusion

    An increasing number of studies are pro-gressively unravelling the fascinating inter-actions that occur between eukaryotes andtheir bacterial symbionts. It is now clearthat the intestinal microbiota profoundlyinfluences host metabolic and immunepathways and participates in human healthand disease. Compelling evidence shows apivotal role for the microbiota in the devel-opment of many gastrointestinal diseases,from inflammation to cancer. Recent work

    Table 1 | Effect of the microbiota on systemic immune-mediated diseases

    Model Animal strain Protocol to alter microbiota Observed effects Refs

     Allergy 

    IgE-mediated food allergyto peanut allergen

    Weanling C3H mice orC57BL/6 mice

    Oral cocktail of antibiotics for 3 weeks Induction of anaphylacticsymptoms, increased production ofIgE and IL-13

    54

    Tlr4 mutant or Tlr4–/– mice None

    IgE-mediated allergicairway disease inducedby Aspergillus fumigatus spores or ovalbumin

    BALB/c and C57BL/6 mice Oral cefoperazone (Cefobid, Pfizer;Cefazone, Pharco B International)for 5 days and a single oral gavage ofCandida albicans

    Increase in pulmonary eosinophilsand enhanced synthesis of IgE, IL-5and IL-13

    49,53

     Autoimmunity 

    Systemic lupuserythematosus

    MRL/lpr mice  Germ-free mice No change in autoimmunity 58

    APECED   Aire–/– mice* Germ-free mice No change in autoimmunity 56

    Spontaneous gastritis   Aid–/– mice‡ Germ-free mice No change in autoimmunity 57

    Collagenous arthritis Fischer rats (resistant) Germ-free rats Enhanced humoral responses 61

    Dark Agouti rats (sensitive) Germ-free rats Increased severity

    Type 1 diabetes NOD mice§ x Myd88 –/–mice Germ-free mice Increased incidence and severity 62

    NOD Myd88–/–mice Colonization with specificpathogen-free flora

    No disease

    Spontaneous ankylosingenthesopathy

    Male B10.BR mice Germ-free mice No disease 59

    Colonization with probioticLactobacillus spp.

    No disease

    Colonization with a mixture ofBacteroides, Enterococcus, Veillonella and Staphylococcus spp.

    Disease restored

    Spontaneous arthritis   Il1Ra–/– BALB/c mice Germ-free mice No disease 60

    Colonization with Lactobacillus bifidus Disease restored

    Il1Ra–/–  Tlr4–/– mice None Same disease incidence,decreased severity

    Il1Ra–/–  Tlr2–/– mice None Increased severity

    Autoimmune arthritis KRN-C57BL/6||  NOD mice Germ-free mice No disease 30

    Colonization with segmentedfilamentous bacteria

    Disease restored

    Experimentalautoimmuneencephalomyelitis

    C57BL/6J mice Germ-free mice Weak severity 31

    Specific pathogen-free flora Maximal severity

    Colonization with segmentedfilamentous bacteria

    Intermediate severity

     Aid, activation-induced cytidine deaminase; Aire, autoimmune regulator; APECED, autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy; IL,interleukin; Il1Ra, IL-1 receptor antagonist; Myd88, myeloid differentiation primary-response protein 88; NOD, non-obese diabetic; Tlr , Toll-like receptor. *AIREregulates the transcription of peripheral autoantigens in medullary thymic epithelial cells and is necessary for thymic negative selection; ‡AID is central forclass-switch recombination and somatic hypermutation in B cells; §NOD mice provide a polygenic model of type 1 diabetes; ||Mice with a transgenic T cell receptoragainst a self peptide that is derived from glucose-6-phosphate isomerase.

    P E R S P E C T I V E S

    742 | OCTOBER 2010 | VOLUME 10 www.nature.com/reviews/immunol

    © 20 Macmillan Publishers Limited. All rights reserved10

  • 8/16/2019 Nature the Immune System and the Gut

    9/10

    19. Malamut, G. et al. The enteropathy associated with

    common variable immunodeficiency: the delineated

    frontiers with celiac disease. Am. J. Gastroenterol. 

    15 Jun 2010 (doi:10.1038/ajg.2010.214).

    20. Bouma, G. & Strober, W. The immunological and

    genetic basis of inflammatory bowel disease. Nature

    Rev. Immunol. 3, 521–533 (2003).

    21. Glocker, E. O. et al. Inflammatory bowel disease and

    mutations affecting the interleukin-10 receptor.

    N. Engl. J. Med. 361, 2033–2045 (2009).

    22. Talham, G. L., Jiang, H. Q., Bos, N. A. & Cebra, J. J.

    Segmented filamentous bacteria are potent stimuli ofa physiologically normal state of the murine gut

    mucosal immune system. Infect. Immun. 67,

    1992–2000 (1999).

    23. Ivanov, I. I. et al. Induction of intestinal Th17 cells by

    segmented filamentous bacteria. Cell  139, 485–498

    (2009).

    24. Brandl, K. et al. Vancomycin-resistant enterococci

    exploit antibiotic-induced innate immune deficits.

    Nature 455, 804–807 (2008).

    25. Klaasen, H. L. et al. Intestinal, segmented, filamentous

    bacteria in a wide range of vertebrate species.

    Lab. Anim. 27, 141–150 (1993).

    26. Lupp, C. et al. Host-mediated inflammation disrupts

    the intestinal microbiota and promotes the overgrowth

    of Enterobacteriaceae . Cell Host Microbe 2, 119–129

    (2007).

    27. Stecher, B. et al. Salmonella enterica serovar

    typhimurium exploits inflammation to compete with

    the intestinal microbiota. PLoS Biol. 5, 2177–2189

    (2007).

    28. Snel, J. et al. Comparison of 16S rRNA sequences of

    segmented filamentous bacteria isolated from mice,

    rats, and chickens and proposal of “Candidatus

     Arthromitus”. Int. J. Syst. Bacteriol. 45, 780–782

    (1995).

    29. Stepankova, R. et al. Segmented filamentous bacteria

    in a defined bacterial cocktail induce intestinal

    inflammation in SCID mice reconstituted with

    CD45RBhigh CD4+ T cells. Inflamm. Bowel Dis. 13,

    1202–1211 (2007).

    30. Wu, H.-S. et al. Gut-residing segmented filamentous

    bacteria drive autoimmune arthritis via T helper 17

    cells. Immunity 32, 815–827 (2010).

    31. Lee, Y. K., Menezes, J. S., Umesaki, Y. &

    Mazmanian, S. K. Microbes and health sackler

    colloquium: proinflammatory T-cell responses to gut

    microbiota promote experimental autoimmune

    encephalomyelitis. Proc. Natl Acad. Sci. USA 28 Jul

    2010 (doi:10.1073/pnas.10000.82107).

    32. Chow, J. & Mazmanian, S. K. A pathobiont of the

    microbiota balances host colonization and intestinalinflammation. Cell Host Microbe 7, 265–276 (2010).

    33. Polk, D. B. & Peek, R. M. Jr. Helicobacter pylori :

    gastric cancer and beyond. Nature Rev. Cancer  10,

    403–414 (2010).

    34. 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).

    35. Carvalho, F. A. et al. Crohn’s disease adherent-

    invasive Escherichia coli  colonize and induce strong

    gut inflammation in transgenic mice expressing

    human CEACAM. J. Exp. Med. 206, 2179–2189

    (2009).

    36. Darfeuille-Michaud, A. et al. High prevalence of

    adherent-invasive Escherichia coli  associated with ileal

    mucosa in Crohn’s disease. Gastroenterology 127,

    412–421 (2004).

    37. Barnich, N. et al. CEACAM6 acts as a receptor for

    adherent-invasive E. coli , supporting ileal mucosa

    colonization in Crohn disease. J. Clin. Invest. 117,1566–1574 (2007).

    38. Frank, D. N. et al. Molecular-phylogenetic

    characterization of microbial community imbalances in

    human inflammatory bowel diseases. Proc. Natl Acad.

    Sci. USA 104, 13780–13785 (2007).

    39. Sokol, H. et al. Specificities of the fecal microbiota in

    inflammatory bowel disease. Inflamm. Bowel Dis. 12,

    106–111 (2006).

    40. Swidsinski, A., Weber, J., Loening-Baucke, V., Hale, L. P.

    & Lochs, H. Spatial organization and composition of

    the mucosal flora in patients with inflammatory bowel

    disease. J. Clin. Microbiol. 43, 3380–3389 (2005).

    41. Garrett, W. S. et al. Communicable ulcerative colitis

    induced by T-bet deficiency in the innate immune

    system. Cell  131, 33–45 (2007).

    42. Garrett, W. S. et al. Colitis-associated colorectal cancer

    driven by T-bet deficiency in dendritic cells. Cancer Cell  

    16, 208–219 (2009).

    43. Garrett, W. et al. Enterobacteriaceae  act in concert

    with the gut microbiota to induce spontaneous and

    maternally-transmitted colitis. Cell Host Microbe 

    (in the press).

    44. Konrad, A., Cong, Y., Duck, W., Borlaza, R. &

    Elson, C. O. Tight mucosal compartmentation of the

    murine immune response to antigens of the enteric

    microbiota. Gastroenterology 130, 2050–2059

    (2006).

    45. Slack, E. et al. Innate and adaptive immunity

    cooperate flexibly to maintain host-microbiota

    mutualism. Science 325, 617–620 (2009).46. Bauer, H., Horowitz, R. E., Levenson, S. M. &

    Popper, H. The response of the lymphatic tissue

    to the microbial flora. Studies on germfree mice.

     Am. J. Pathol. 42, 471–483 (1963).

    47. 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).

    48. Clarke, T. B. et al. Recognition of peptidoglycan from

    the microbiota by Nod1 enhances systemic innate

    immunity. Nature Med. 16, 228–231 (2010).

    49. Noverr, M. C. & Huffnagle, G. B. The ‘microflora

    hypothesis’ of allergic diseases. Clin. Exp. Allergy 35,

    1511–1520 (2005).

    50. Sjogren, Y. M., Jenmalm, M. C., Bottcher, M. F.,

    Bjorksten, B. & Sverremark-Ekstrom, E. Altered early

    infant gut microbiota in children developing allergy up

    to 5 years of age. Clin. Exp. Allergy 39, 518–526

    (2009).

    51. Kuitunen, M. et al. Probiotics prevent IgE-associated

    allergy until age 5 years in cesarean-delivered children

    but not in the total cohort. J. Allergy Clin. Immunol. 

    123, 335–341 (2009).

    52. Penders, J., Stobberingh, E. E., van den Brandt, P. A.

    & Thijs, C. The role of the intestinal microbiota in the

    development of atopic disorders. Allergy 62,

    1223–1236 (2007).

    53. Noverr, M. C., Falkowski, N. R., McDonald, R. A.,

    McKenzie, A. N. & Huffnagle, G. B. Development of

    allergic airway disease in mice following antibiotic

    therapy and fungal microbiota increase: role of host

    genetics, antigen, and interleukin-13. Infect. Immun. 

    73, 30–38 (2005).

    54. 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).

    55.  Vaahtovuo, J., Munukka, E., Korkeamaki, M.,

    Luukkainen, R. & Toivanen, P. Fecal microbiota in early

    rheumatoid arthritis. J. Rheumatol. 35, 1500–1505

    (2008).56. Gray, D. H., Gavanescu, I., Benoist, C. & Mathis, D.

    Danger-free autoimmune disease in Aire-deficient

    mice. Proc. Natl Acad. Sci. USA 104, 18193–18198

    (2007).

    57. Hase, K. et al. Activation-induced cytidine deaminase

    deficiency causes organ-specific autoimmune disease.

    PLoS ONE  3, e3033 (2008).

    58. Maldonado, M. A. et al. The role of environmental

    antigens in the spontaneous development of

    autoimmunity in MRL-lpr mice. J. Immunol. 162,

    6322–6330 (1999).

    59. Sinkorova, Z., Capkova, J., Niederlova, J.,

    Stepankova, R. & Sinkora, J. Commensal intestinal

    bacterial strains trigger ankylosing enthesopathy of

    the ankle in inbred B10.BR (H-2k) male mice. Hum.

    Immunol. 69, 845–850 (2008).

    60.  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).61. Breban, M. A., Moreau, M. C., Fournier, C.,

    Ducluzeau, R. & Kahn, M. F. Influence of the bacterial

    flora on collagen-induced arthritis in susceptible and

    resistant strains of rats. Clin. Exp. Rheumatol. 11,

    61–64 (1993).

    62. Wen, L. et al. Innate immunity and intestinal

    microbiota in the development of Type 1 diabetes.

    Nature 455, 1109–1113 (2008).

    63. Giraud, A. et al. Dissecting the genetic components

    of adaptation of Escherichia coli  to the mouse gut.

    PLoS Genet. 4, e2 (2008).

    64. Turnbaugh, P. J., Backhed, F., Fulton, L. & Gordon, J. I.

    Diet-induced obesity is linked to marked but reversible

    alterations in the mouse distal gut microbiome.

    Cell Host Microbe 3, 213–223 (2008).

    65. Backhed, F. et al. The gut microbiota as an

    environmental factor that regulates fat storage.

    Proc. Natl Acad. Sci. USA 101, 15718–15723 (2004).

    adaptation to new ecological conditions. Anin-depth understanding of the principlesand mechanisms that underlie microbialcommunity structures and host–symbiontrelationships will be pivotal for the develop-ment of therapeutic approaches that manip-ulate the intestinal microbiota to maintainhuman health.

    Nadine Cerf-Bensussan and Valérie Gaboriau-Routhiau

    are at the Institut National de la Santé et de la

    Recherche Médicale (INSERM) U989, Université Paris

    Descartes, 156 rue de Vaugirard, 75730 Paris

    Cedex 15, France; and the Institut National de la

    Recherche Agronomique (INRA) UM1319, Domaine de

    Vilvert, 78350 Jouy-en-Josas, France.

    Correspondence to N.C.-B.

    e-mail: [email protected] 

    doi:10.1038/nri2850

    1. Costello, E. K. et al. Bacterial community variation in

    human body habitats across space and time. Science 

    326, 1694–1697 (2009).

    2. Gill, S. R. et al. Metagenomic analysis of the human

    distal gut microbiome. Science 312, 1355–1359

    (2006).3. Eberl, G. A new vision of immunity: homeostasis of the

    superorganism. Mucosal Immunol. 5 May 2010

    (doi:10.1038/mi.2010.20).

    4. Qin, J. et al. A human gut microbial gene catalogue

    established by metagenomic sequencing. Nature 464,

    59–65 (2010).

    5. Moran, N. A., McCutcheon, J. P. & Nakabachi, A.

    Genomics and evolution of heritable bacterial

    symbionts. Annu. Rev. Genet. 42, 165–190 (2008).

    6. Garrett, W. S., Gordon, J. I. & Glimcher, L. H.

    Homeostasis and inflammation in the intestine.

    Cell 140, 859–870 (2010).

    7. Hooper, L. V. & Macpherson, A. J. Immune

    adaptations that maintain homeostasis with the

    intestinal microbiota. Nature Rev. Immunol. 10,

    159–169 (2010).

    8. Eberl, G. & Lochner, M. The development of

    intestinal lymphoid tissues at the interface of self

    and microbiota. Mucosal Immunol. 2, 478–485

    (2009).9. Rescigno, M. & Di Sabatino, A. Dendritic cells in

    intestinal homeostasis and disease. J. Clin. Invest. 

    119, 2441–2450 (2009).

    10. 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).

    11.  Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L.

    & Hooper, L. V. Paneth cells directly sense gut

    commensals and maintain homeostasis at the

    intestinal host-microbial interface. Proc. Natl Acad.

    Sci. USA 105, 20858–20863 (2008).

    12. 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).

    13. Macpherson, A. J. & Uhr, T. Induction of protective

    IgA by intestinal dendritic cells carrying commensal

    bacteria. Science 303, 1662–1665 (2004).

    14. Lotz, M. et al. Postnatal acquisition of endotoxintolerance in intestinal epithelial cells. J. Exp. Med. 203, 973–984 (2006).

    15. Mazmanian, S. K., Round, J. L. & Kasper, D. L.

     A microbial symbiosis factor prevents intestinal

    inflammatory disease. Nature 453, 620–625

    (2008).

    16. 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).

    17. 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).

    18. Fagarasan, S. et al. Critical roles of activation-induced

    cytidine deaminase in the homeostasis of gut flora.

    Science 298, 1424–1427 (2002).

    P E R S P E C T I V E S

    NATURE REVIEWS | IMMUNOLOGY VOLUME 10 | OCTOBER 2010 | 743

    © 20 Macmillan Publishers Limited. All rights reserved10

    mailto:[email protected]:[email protected]

  • 8/16/2019 Nature the Immune System and the Gut

    10/10

    66. Samuel, B. S. et al. Effects of the gut microbiota on

    host adiposity are modulated by the short-chain

    fatty-acid binding G protein-coupled receptor, Gpr41.

    Proc. Natl Acad. Sci. USA 105, 16767–16772

    (2008).

    67. Turnbaugh, P. J. et al. The human microbiome project.

    Nature 449, 804–810 (2007).

    68.  Vijay-Kumar, M.et al. Metabolic syndrome and altered

    gut microbiota in mice lacking Toll-like receptor 5.

    Science 328, 228–231 (2010).

    69. Peyrin-Biroulet, L. et al. Peroxisome proliferator-

    activated receptor γ activation is required formaintenance of innate antimicrobial immunity in thecolon. Proc. Natl Acad. Sci. USA 107, 8772–8777

    (2010).

    70. Kumar, A. et al. Commensal bacteria modulate

    cullin-dependent signaling via generation of reactive

    oxygen species. EMBO J. 26, 4457–4466 (2007).

    71. Dubuquoy, L. et al. Impaired expression of peroxisome

    proliferator-activated receptor γ in ulcerative colitis.Gastroenterology 124, 1265–1276 (2003).

    72. Kelly, D. et al. Commensal anaerobic gut bacteria

    attenuate inflammation by regulating nuclear-

    cytoplasmic shuttling of PPAR-γ and RelA. NatureImmunol. 5, 104–112 (2004).

    73. Chieppa, M., Rescigno, M., Huang, A. Y. &

    Germain, R. N. Dynamic imaging of dendritic cell

    extension into the small bowel lumen in response to

    epithelial cell TLR engagement. J. Exp. Med. 203,

    2841–2852 (2006).

    74. Schulz, O. et al. Intestinal CD103+, but not CX3CR1+,

    antigen sampling cells migrate in lymph and serve

    classical dendritic cell functions. J. Exp. Med. 206,

    3101–3114 (2009).

    75. Tsuji, M. et al. Preferential generation of follicular B

    helper T cells from Foxp3+ T cells in gut Peyer’s

    patches. Science 323, 1488–1492 (2009).

    76. Cerutti, A. & Rescigno, M. The biology of intestinal

    immunoglobulin A responses. Immunity 28, 740–750(2008).

    77. Coombes, J. L. & Powrie, F. Dendritic cells in intestinal

    immune regulation. Nature Rev. Immunol. 8,

    435–446 (2008).

    78.  Atarashi, K. et al. ATP drives lamina propria TH17 cell

    differentiation. Nature 455, 808–812 (2008).

    79. Zhou, L., Chong, M. M. & Littman, D. R. Plasticity of

    CD4+ T cell lineage differentiation. Immunity 30,

    646–655 (2009).

    80. Keilbaugh, S. A. et al. Activation of RegIIIβ/γ andinterferon γ expression in the intestinal tract of SCIDmice: an innate response to bacterial colonisation of

    the gut. Gut  54, 623–629 (2005).

    81. Umesaki, Y., Okada, Y., Matsumoto, S., Imaoka, A. &

    Setoyama, H. Segmented filamentous bacteria are

    indigenous intestinal bacteria that activate

    intraepithelial lymphocytes and induce MHC class II

    molecules and fucosyl asialo GM1 glycolipids on the

    small intestinal epithelial cells in the ex-germ-free

    mouse. Microbiol. Immunol. 39, 555–562 (1995).

    82. Mayer, L. Evolving paradigms in the pathogenesis of

    IBD. J. Gastroenterol. 45, 9–16 (2010).

     AcknowledgementsThe authors thank W. Garrett for sharing unpublished data.

    Their work is supported by grants from the Institut National

    de la Santé et de la Recherche Médicale (INSERM), theInstitut National de la Recherche Agronomique (INRA) and

    the Agence Nationale de la Recherche and Fondation

    Princesse Grace. The authors are partners of the European

    Community networks Cross-Talk (contract number

    PITN-GA-2008-215553) and Tornado (FP7 222720).

    Competing interests statementThe authors declare no competing financial interests.

    FURTHER INFORMATIONNadine Cerf-Bensussan’s homepage: http://www.

    fondationimagine.org/Nadine-Cerf-Bensussan_uk.php

    ALL LINKS ARE ACTIVE IN THE ONLINE PDF

    P E R S P E C T I V E S

    744 | OCTOBER 2010 | VOLUME 10 www.nature.com/reviews/immunol

    http://www.fondationimagine.org/Nadine-Cerf-Bensussan_uk.phphttp://www.fondationimagine.org/Nadine-Cerf-Bensussan_uk.phphttp://www.fondationimagine.org/Nadine-Cerf-Bensussan_uk.phphttp://www.fondationimagine.org/Nadine-Cerf-Bensussan_uk.php