Sie sind auf Seite 1von 10

PersPecTives

bacteria are then followed by hundreds


OPINION
of environmentally acquired species,
which differ between individuals but
The immune system and the gut mainly belong to two bacterial phylotypes,
Firmicutes spp. and Bacteroidetes spp.2,4.
microbiota: friends or foes? A growing number of studies support
the view that eukaryotic hosts and their
symbionts have co-evolved towards mutu-
Nadine Cerf-Bensussan and Valrie Gaboriau-Routhiau alistic interactions that are based on the
nutritional benefits that each partner gains
Abstract | The mammalian intestine is home to a complex community of trillions of from the association5 (BOX 1). However,
bacteria that are engaged in a dynamic interaction with the host immune system. the huge collection of bacteria at the gut
Determining the principles that govern hostmicrobiota relationships is the focus surface is also a major threat to host integ-
of intense research. Here, we describe how the intestinal microbiota is able to rity and has driven the selection of highly
influence the balance between pro-inflammatory and regulatory responses and flexible defence mechanisms, which enable
eukaryotic hosts to cope with their micro-
shape the hosts immune system. We suggest that improving our understanding of
bial environment and compensate for their
the intestinal microbiota has therapeutic implications, not only for intestinal less rapid genetic adaptation.
immunopathologies but also for systemic immune diseases.
The gut immune system. Recent reviews
Initially, all microorganisms were viewed as the gut might influence the development have highlighted how the microbiota elicits
pathogens that cause and propagate infectious and functions of the immune system has innate and adaptive immune mechanisms
diseases and, as a field, immunology was built become a major focus of interest. A prevalent that cooperate to protect the host and main-
around the paradigm that the host immune theory, derived from hypotheses that were tain intestinal homeostasis6,7. Epithelial cells
system should recognize and eliminate these first postulated by Metchnikoff a century are a central component of the immune
intruders (non-self) while tolerating self- ago, proposes that individual members of system of the gut. In a similar manner to
molecules to preserve homeostasis. However, the microbiota might influence the balance immune cells, epithelial cells express recep-
the persistent association of animal and plant between pro-inflammatory and regulatory tors for microbial-associated molecular
species with obligate and facultative symbionts host responses and that alterations in the patterns (MAMPs). These receptors activate
now shows that both bacteria and their composition of the microbiota (a process that signalling cascades that finely tune epithelial
eukaryote hosts benefit from their coopera- is known as dysbiosis) could jeopardize host cell production of antimicrobial products
tive relationships. These benefits suggest that immune responses and promote the devel- and chemokines, depending on the signals
co-evolution has selected mechanisms that opment of various inflammatory disorders. that are delivered by the microbiota (FIG. 1).
promote and maintain associations between Here, we discuss the principles that govern Thus, gut epithelial cells form a potent and
bacteria and eukaryotes. In humans, trillions the interactions between the intestinal micro- inducible physico-chemical barrier, which
of bacteria are distributed in complex and biota and the host immune system, both in limits microbial growth and access to the
site-specific communities on the skin health and in disease. Moreover, we stress gut surface. They can also recruit leukocytes
and at mucosal surfaces, and the largest how the complexity of the gut ecological sys- to complement their barrier function or to
community is found in the distal gut. As these tem and the reciprocal nature of the regula- participate in the activation of gut adap-
bacteria encode hundreds of genes that are tion of the immune system and of microbial tive immune responses. In mammals, the
absent in the human genome1, the idea has community structures must be considered development of gut-associated lymphoid
emerged that together with our microbiota, before one can draw any conclusions about tissues (GALTs) is initiated before birth by
we form superorganisms in which energy and the role of the microbiota in disease and a genetic programme8. However, GALT
metabolites can be exchanged2 and homeo- propose therapeutic interventions. maturation and the recruitment of IgA-
stasis is maintained by the immune system3. secreting plasma cells and activated T cells
Therefore, a new paradigm proposes that the The hostmicrobiota interaction in the gut to mucosal sites only occurs after birth and
immune system has evolved to accommodate The intestine is an open ecological system is strictly dependent on microbiota-derived
colonization by symbiotic bacterial commu- that is colonized immediately after birth signals; these signals influence the crosstalk
nities of increasing complexity while retaining by a microbial population that reaches between epithelial cells and gut dendritic
the capacity to fight pathogens. an impressive density of 1012 bacteria per cells (DCs), thereby modulating the nature
The gastrointestinal tract is the primary gram of luminal content in the distal gut. and intensity of intestinal B and T cell
site of interactions between the host and the Colonization is initiated by maternally responses7,9 (FIG. 2). In immunocompetent
microbiota. How bacterial colonization of acquired bacteria during birth; these mice, intestinal colonization stimulates

nATuRE REvIEws | Immunology voLuME 10 | o CToBER 2010 | 735

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

Box 1 | mutualistic relationships between hosts and their intestinal microbiota Anti-inflammatory roles of the microbiota.
Current results indicate that a trade-off is
The human intestine harbours an estimated 100 trillion bacteria, 7080% of which cannot yet be established between the host immune sys-
cultured. Each individual is thought to host several hundred species of bacteria from only 7 to 9 tem and the bulk of the microbiota, so that
phylotypes; these are mainly Gram-positive Firmicutes spp. (most notably Clostridium spp.,
in a healthy individual, intestinal coloniza-
Enterococcus spp. and Lactobacillus spp.) and Gram-negative Bacteroidetes spp. Recent
tion stimulates host production of micro-
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 bicidal peptides11 and secretory IgA, which
encodes a core of redundant bacterial genes that are likely to be needed to resist stressful in turn contain the microbiota within the
conditions in the host intestine63 and to harvest nutrients that are necessary for bacterial intestinal lumen and neutralize MAMPs12.
growth2,4. Competition between bacteria with distinct metabolic requirements might explain the These mechanisms protect the host from
massive and rapid shifts in the structure of the intestinal microbial community that are provoked the systemic translocation of bacteria or
by changes in host diet64. In addition to the genes that are necessary for microbial adaptation to bacterial products and from the outburst of
the host environment, the gut microbiome encodes multiple biosynthetic pathways that are pro-inflammatory cascades in intestinal epi-
predicted to greatly increase the hosts capacity to metabolize glycans and xenobiotics and to thelial and innate cells13. Conversely, the resi-
synthesize vitamins2,4. Moreover, studies in gnotobiotic mice have shown the broad influence of
dent bacteria also benefit from the symbiotic
the gut microbiota on host physiology. Intestinal colonizaton induces a spectrum of intestinal
relationship and can thrive in the mucus,
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 thus minimizing destruction by host-derived
signalling pathways that are involved remain largely elusive, but recent observations suggest inflammatory mediators. Hosts and bacteria
that overlapping mechanisms have been selected during hostmicrobiota co-evolution that have evolved additional strategies to main-
simultaneously control host metabolic and innate immune responses to the microbiota. In mice, tain friendly relationships. Thus, signalling
inactivation of Toll-like receptor 5 (TLR5), which is a receptor for bacterial flagellin that has an cascades that occur downstream of Toll-like
established role in host innate immune responses, results in severe obesity and profound receptors (TLRs) can be desensitized by
alterations in the microbiota structure68. Furthermore, peroxisome proliferator-activated continuous exposure to lipopolysaccharide
receptor- (PPAR), which is a transcription factor that has a central role in glucidolipidic (LPs)14 or can be attenuated by other soluble
metabolism, can control the production of microbicidal peptides by colonocytes and serves
mediators that are produced by the micro-
as a feedback mechanism for the activation of nuclear factor-B (NF-B) in enterocytes69.
biota (FIG. 1). Furthermore, some microbiota-
derived soluble products can promote the
the production of secretory IgA, the dif- regulatory immune responses, and to establish functions of TReg cells15,16.
ferentiation of effector T helper 1 (TH1), whether the composition of the microbiota The mechanism that maintains this
TH2 and TH17 cells, and the development of can influence the development of inflamma- friendly relationship has been elucidated
regulatory T (TReg) cells10. tory diseases in and beyond the gut. Before in the case of Bacteroides fragilis, which is a
It is increasingly clear how these adap- considering the possible role of the microbiota common culturable member of the micro-
tive immune elements cooperate with innate in disease, we will first highlight how the dif- biota15. This bacterium possesses an unusual
immune cells to strengthen the gut barrier ferent colonization strategies of individual capsular polysaccharide A (PsA) that is able
and protect the host from invading patho- members of the microbiota can influence the to drive the differentiation of interleukin-10
gens. An outstanding issue now is to define development and function of the gut immune (IL-10)-secreting TReg cells. Colonization by
how individual members of the microbiota system and show that, ultimately, it is the host a wild-type B. fragilis, but not by a mutant
or microbiota-derived products can affect immune system that determines whether a strain that lacks PsA, protected mice from
the balance between pro-inflammatory and bacterium is a friend or a foe. the severe experimental colitis that is induced

glossary
Ankylosing enthesopathy IgE-associated allergies mucosa. Together with mesenteric lymph nodes, they form
An inflammatory autoimmune disease of the joints Type 1 hypersensitivity reactions that are mediated by the inductive compartment for intestinal immune responses.
that naturally occurs in mice on a C57BL/10 genetic IgE, which induces mast cell activation and degranulation.
background; the disease is similar to human ankylosing Such immune reactions are seen in asthma, allergic rhinitis, Proteobacteria
spondylitis. The pathology is characterized by the systemic anaphylaxis and food allergies. Gram-negative microorganisms that colonize very distinct
proliferation of cartilage and connective tissue, which environments and are the second largest group of bacteria
culminates in ankylosis of the joints. Obligate and facultative symbionts on earth. Proteobacteria that colonize the intestine include
Obligate microbial symbionts need to colonize a host commensal, pathogenic and opportunistic species, such as
Germinal centres to develop and multiply, unlike facultative microbial Salmonella, Shigella and Helicobacter spp. and Escherichia
Highly specialized and dynamic microenvironments that symbionts, which can also develop outside a host. coli strains. In healthy adults, proteobacteria represent less
are located in secondary lymphoid tissues and give rise than 1% of the enteric microbiota, but they are a major
to secondary B cell follicles during an immune response. Pathobionts cause of intestinal and extraintestinal diseases.
Germinal centres are the main sites of B cell proliferation Microbial symbionts that can cause defined disease in
and differentiation, which leads to the generation of predisposed hosts following changes in the gastrointestinal Type VI secretion system
memory B cells and plasma cells that produce high-affinity environment. (T6SS). Like T3SS and T4SS, T6SS is a multi-subunit
antibodies. complex that acts like a needle and syringe to
Microbiome translocate bacterial products across the
Gnotobiotic mice The whole genome of all of the microorganisms that double-membrane of Gram-negative bacteria into
Germ-free mice are born and raised in sterile isolators colonize a specific environment. the cytoplasm of eukaryotic cells.
and are devoid of colonization by any microorganisms,
but after they have been experimentally colonized by Peyers patches Xenobiotics
known bacteria, they are said to be gnotobiotic. They Collections of lymphoid follicles that are located in the Chemical compounds that are foreign to a living organism
are kept in isolators to control their bacterial status. intestinal mucosa and are particularly abundant in the ileal and that can be toxic, even at low concentrations.

736 | o CToBER 2010 | voLuME 10 www.nature.com/reviews/immunol

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

by Helicobacter hepaticus15,17. Furthermore,


administration of PsA reduced the sever- PAMP
ity of disease in a model of trinitrobenzene
sulphonic acid (TnBs)-induced colitis17.
Bacterium
Therefore, B. fragilis may represent a proto-
type peace-keeper strain. Yet the outcome
of hostmicrobiota interactions cannot be
predicted from only the bacterium itself, and
a bacterium that is beneficial for an immuno-
TLR
competent host can become a dangerous foe Intestinal lumen
when the immune system is weakened. This
is illustrated by the fact that B. fragilis causes Intestinal
MYD88 epithelial cell
severe sepsis in immunocompromised hosts.
Expression of IRAK1
Likewise, normally harmless members of the decreased by LPS IRAK1
ROS induced by
microbiota can initiate intestinal inflamma- from microbiota microbiota inhibit
tion in individuals who cannot mount effi- ubiquitin ligases
cient intestinal humoral responses18,19 and in IB ubiquitylated and
individuals with impaired intestinal immuno- IB targeted to the proteasome
regulation, most notably in those who lack a for degradation
p50 p65
functional IL-10 signalling pathway 20,21.
NF-B PPAR induced in
Promotion of effector immune responses. Nuclear translocation response to microbiota
p65 PPAR LPS diverts NF-B
of NF-B
Although certain members of the microbiota from nucleus
have adopted peace-keeper activities to colo-
nize the intestine, others are, undoubtedly,
PPAR upregulates
endowed with pro-inflammatory properties. p50 p65 PPAR colonic -defensins
one such group, which has recently attracted to sustain gut barrier
much attention, is segmented filamentous
bacteria (sFB). These unculturable species NF-B transcribes
settle in the rodent intestine at the time of pro-inflammatory Pathway impaired
cytokines and in patients with
weaning and stimulate the postnatal matu- chemokines and Crohns disease
ration of immune responses in the mouse defensins
gut. Mice that are colonized by a microbiota
that lacks sFB have weaker IgA antibody
Figure 1 | modulation of intestinal epithelial cell pro-inflammatory responses by the microbiota.
responses22 and much poorer intestinal in a similar manner to immune cells, epithelial cells detect microbes throughNature
pattern-recognition recep-
Reviews | Immunology
T cell responses compared with mice that tors, including Toll-like receptors (TLrs). Upon TLr ligation, adaptor proteins, such as myeloid differen-
are colonized with sFB. notably, mice that are tiation primary-response protein 88 (MYD88), are recruited and activate signalling cascades, notably
colonized by an sFB-deficient microbiota the nuclear factor-B (NF-B) pathway, which stimulates the transcription of antimicrobial proteins,
lack mucosal TH17 cells10,23. Furthermore, pro-inflammatory cytokines and chemokines. in resting cells, NF-B is sequestered in the cytoplasm by
these animals cannot control colonization by its inhibitor iB. Following TLr activation, iB is phosphorylated, ubiquitylated and degraded by the
the invasive pathogen Citrobacter rodentium, proteasome, which allows nuclear translocation of NF-B and transcription of NF-B target genes. This
which suggests that microbiota-induced pathway can be modulated by microbiota-derived factors, preventing excessive and potentially delete-
immune responses participate in the bar- 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-B cascade, is downregulated by
rier function of the flora23. This hypothesis
microbiota-derived lipopolysaccharide (LPs)14. The polyubiquitylation and degradation of iB can be
is also supported by recent work that shows inhibited by commensal bacteria, which inhibit a common ubiquitin ligase by inducing reactive oxygen
that the destruction of the microbiota fol- species (rOs)70. Peroxisome proliferator- activated receptor- (PPAr), which is induced in response to
lowing treatment with antibiotics can jeop- TLr4 activation by LPs71, can also divert NF-B from the nucleus72. checkpoints that are controlled by
ardize innate immune responses in the gut the microbiota are indicated by T bars. interestingly, PPAr positively controls the expression of the
and promote colonization by pathogens24. colonic microbicidal peptide defensin 1 (rEF. 69) and thus can simultaneously sustain the gut barrier
A striking feature of sFB is their strong and prevent excessive inflammation. This mechanism may be impaired in a subset of patients with
adherence to the surface epithelium of the colonic crohns disease69. PAMP, pathogen-associated molecular pattern.
ileum and the Peyers patches shortly after
weaning 25. This is in contrast with most other
members of the microbiota, which remain signalling pathways in epithelial cells and mouse intestine, but it also benefits the host
entrapped within the mucus and have little DCs, resulting in robust innate and adap- by strengthening the gut barrier. strikingly,
or no physical contact with host epithelium25. tive immune responses in the intestine10,23. on the basis of morphological studies, sFB
This attachment, which is perhaps necessary such behaviour is characteristic of bona fide have been detected in all species studied from
to initiate the replication of sFB, is likely to pathogens, which use host inflammatory arthropods to mammals, including humans,
facilitate the sampling and presentation of responses to eliminate the resident flora and and closely related 16s rRnA sequences have
sFB antigens to T cells by DCs in the Peyers to colonize the remaining niches26,27. This been found in chickens, trout and rodents25,28.
patches and to stimulate pro-inflammatory attachment might enable sFB to settle in the Therefore, it is tempting to speculate that this

nATuRE REvIEws | Immunology voLuME 10 | o CToBER 2010 | 737

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

a
M cell
Intestinal lumen
Bacterium

MHC
Peptide
Intestinal epithelium
TCR
CX3CR1- Naive T cell
expressing Transcytosed bacteria are
phagocyte CD103+ DCs acquire Peyers patch
bacterial antigens acquired by DCs, which activate
Direct sampling of adaptive immune responses
Lamina propria bacteria in lumen?
Antigen transfer?
Peyers patch and CD103+ DCs
CD103+ DC migrate to MLNs and initiate
adaptive immune responses

b c
IgA molecules are B. fragilis
transcytosed by polymeric SFB
IgA receptors and retain
bacteria in the mucus Microbiota-
derived products
activate TLRs

TSLP, B. fragilis capsular ATP Bacterial ATP and


TLR bacteria-induced
TGF, polysaccharide A SAA
Mucus layer retinoic SAA activate DCs
acid
SFB induce
TReg cell IL-12-producing
induction DCs by unknown
Dimeric Epithelial Inflammatory mechansims
BAFF,
secretory cell DCs
APRIL
IgA FOXP3 +
Tolerogenic DC IL-1, IL-6, IL-12
BAFF and APRIL TReg cell
promote T cell- IL-23
TReg cells maintain
dependent and tolerance to food
T cell-independent and commensals
IgA class-switching
IgA+ plasma cell
TH17 cell TH1 cell
TH1 and TH17 cells sustain intestinal barrier
by recruiting macrophages and neutrophils
and inducing antibacterial defensins

Figure 2 | | modulation of adaptive immune responses in the gut by the secretory igA molecules form immune complexes with bacteria, which are
microbiota. a | intestinal adaptive immune responses can be initiated in then retained in the mucus. c | cD103+ Dcs are conditioned by epithelial
Peyers patches or in mesenteric lymph nodes (MLNs). Activated T and B cells cell-derived factors, such as thymic stromal lymphopoietin (TsLP), transform-
subsequently leave these lymphoid tissues and home to the intestinal lamina ing growth factor- (TGF) and retinoic acid, to acquire a tolerogenic pheno-
propria via the bloodstream. Bacteria are mainly sampled by Peyers patch type; these Dcs can promote the induction of forkhead box P3 (FOXP3)+
dendritic cells (Dcs) after transcytosis across the specialized epithelium that regulatory T (Treg) cells77. Bacteria-derived products, such as the capsular
overlays these lymphoid organs. it has also been suggested that a population polysaccharide A from Bacteroides fragilis, can further
Naturepromote
Reviews the induction
| Immunology
of cX3c-chemokine receptor 1 (cX3cr1)+ lamina propria cells with both Dc- of interleukin-10 (iL-10)-producing Treg cells through a TLr2-dependent
and macrophage-like characteristics can send dendrites into the intestinal mechanism15,17. However, some commensal bacteria can stimulate the dif-
lumen and directly capture bacteria. Their role in antigen presentation ferentiation of inflammatory mucosal T cells. T helper 17 (TH17) cell differen-
remains controversial73,74, but they may pass antigens to lamina propria tiation can be promoted by bacteria-derived ATP, which activates a subset of
cD103+ Dcs, which can migrate to the MLNs and present antigens to T cells74. Dcs that produce iL-1, iL-6 and iL-23 (rEF. 78), or by serum amyloid A pro-
b | Microbiota-derived products activate Toll-like receptors (TLrs) that are tein (sAA), which is an acute phase protein that is produced in response to
expressed by intestinal epithelial cells, which leads to the production of segmented filamentous bacteria (sFB)23. sFB can also drive the expansion of
B cell-activating factor (BAFF) and a proliferation-inducing ligand (APriL); mucosal TH1 cell populations11, presumably by inducing iL-12 production by
these cytokines promote both T cell-dependent and T cell-independent igA Dcs. in addition, inflammatory Dcs might stimulate the conversion of
class-switching responses in the intestine75,76. Plasma cells produce dimeric Treg cells into TH17 and/or TH1 cells in the lamina propria (not shown)79. TH1
igA molecules that are transcytosed into the intestinal lumen by the epithe- and TH17 cells maintain the intestinal barrier by recruiting and activating
lial polymeric ig receptor, the expression of which is upregulated by the macrophages and neutrophils that eliminate penetrating bacteria. iL-22 that
microbiota. The extracellular part of this receptor remains associated with is produced by a subset of TH17 cells can also promote the production of
igA following release into the lumen and forms secretory igA. These antibacterial defensins by epithelial cells. M cell, microfold cell.

738 | o CToBER 2010 | voLuME 10 www.nature.com/reviews/immunol

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

unusual symbiont has a particularly impor- Intestinal colonization by SFB


tant role in shaping the gut immune system
across evolution. However, again, the out-
come of sFBhost interactions depends on
the immune status of the host; sFB has been
associated with the development of intestinal
inflammation29 or arthritis30 in mice with
impaired immunoregulation and can also Effects on intestinal compartment Effects on peripheral compartment
aggravate experimental autoimmune enceph- Stimulation of innate immune Increased germinal centre Enhanced
alomyelitis31 (FIG. 3). Interestingly, sFB alone responses e.g. Reg III/ production formation in the spleen sensitization
were not able to induce intestinal inflamma- Stimulation of CD4+ T cell responses e.g. Autoantibody-secreting following
TH1, TH2, TH17 and TReg cells IgG plasma cells challenge with
tion in immunodeficient mice and instead Induction of IgA responses Circulating immune MOG peptide
synergized with a pathogen-free flora. This Recruitment and activation of IELs complexes
finding highlights how interactions in the
microbiota community can influence host
immune responses, thereby adding an Physiological inflammation Intestinal inflammation
strengthens gut barrier in in SCID mice transferred Arthritis in Increased
additional level of complexity. immunocompetent hosts with effector T cells K/BxN mice severity of EAE

The case of pathobionts. A particular sub- 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
set of bacteria further exemplifies how the Nature Reviews | Immunology
mother microbiota, sFB develop strong interactions with the ileal mucosa and in immunocompe-
behaviour of the microbiota is dependent on
tent mice, the bacteria can largely recapitulate the inducing effects of the whole microbiota on
the immune status of the host. Although these the postnatal maturation of the gut immune system. sFB induce the production of reg iii/ micro-
bacteria, which are known as pathobionts, bicidal peptides10,23,80, which protect against colonizing pathogens24. Additionally, sFB simultane-
colonize the gastrointestinal tract of many ously activate strong secretory igA responses22, induce the recruitment and activation of cytotoxic
individuals asymptomatically, they also have intraepithelial lymphocytes (ieLs)81 and drive various T cell responses, including a robust T helper 17
the potential to cause disease. A recent study (TH17) cell response11,23. in immunocompetent mice, sFB-induced pro-inflammatory and regulatory
showed that Helicobacter hepaticus, which responses balance each other, which results in physiological inflammation that strengthens the
is a member of the epsilon subgroup of gut barrier. By contrast, colonization by sFB promotes the development of colitis in severe com-
proteobacteria, uses its type VI secretion system 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
(T6ss) to regulate bacterial colonization and
gut. sFB promote arthritis in autoimmune non-obese diabetic (NOD) mice that express a trans-
inhibit host innate and adaptive immune
genic T cell receptor (Tcr) that is specific for a self peptide (known as K/BxN mice), an effect
responses, thereby actively maintaining ascribed to the induction of TH17 cells 30. sFB also enhance the severity of myelin oligodendrocyte
symbiotic relationships with immunocom- glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (eAe). These aggravating
petent hosts32. However, this bacterium can effects may reflect the strong adjuvant properties of sFB. Treg cells, regulatory T cells.
cause severe typhocolitis in Il10/ mice or
in the severe combined immunodeficient
(sCID) transfer model of colitis. These with potent acid-suppressive properties33. patients with Crohns disease than in healthy
findings highlight the central role of host other recently characterized pathobionts controls, which suggests that these bacteria
adaptive regulatory responses in main- include enterotoxigenic B. fragilis (ETBF), can initiate disease36. Yet to induce intestinal
taining symbiotic relationships with the which can stimulate colonic inflammation inflammation, the prototype E. coli LF82
microbiota32. and tumorigenesis in predisposed multiple strain first needs to bind to an epithelial
In humans, the most classical example intestinal neoplasia (MIn) mice34, and some cell-expressed receptor. This receptor
of a pathobiont is Helicobacter pylori, which Escherichia coli strains, which can promote is absent in the normal ileal mucosa but is
uses various mechanisms to dampen host gut inflammation in patients with Crohns upregulated by interferon- (IFn) and
immune responses and persist in the stom- disease35,36. TnF that are produced during intestinal
ach. Gastric colonization by this bacterium inflammation35,37. An alternative hypo-
remains asymptomatic in most individu- Intestinal dysbiosis and IBDs thesis suggests that the reduced frequency
als and has even been suggested to protect Inflammatory bowel diseases (IBDs) are of a Firmicutes species, Faecalibacterium
against the development of oesophageal thought to arise owing to a combination of prausnitzii, in the intestinal microbiota is
carcinomas owing to downmodulation of genetic and environmental factors that result a causative factor of Crohns disease16. This
gastric acid secretion. Yet this bacterium in dysregulated immune responses to the gut strain releases an unidentified soluble factor,
is the major cause of gastritis and gastric microbiota and the subsequent development which inhibits pro-inflammatory epithelial
cancers. The circumstances that lead to this of gut inflammation20. Compelling evidence cell responses in vitro and attenuates
bacterium becoming a serious health con- from a variety of studies has shown dysbiosis inflammation in a mouse model of colitis16.
cern are not completely understood but are in patients with IBD compared with healthy other studies, however, suggest that
thought to include the selection of specifi- controls, and this suggests a causative role for more global changes in the composition
cally aggressive strains and/or host predis- dysbiosis in gut inflammation. of the microbiota are associated with IBD,
posing factors, notably polymorphisms in several scenarios can be considered. such as abnormal adherence of bacteria to
gene promoters that increase the production Pro-inflammatory bacteria, such as entero- the gut mucosa, reduced bacterial diversity,
of tumour necrosis factor (TnF) or IL-1, invasive Escherichia coli strains, are more decreased levels of resident Firmicutes spp.
which is a pro-inflammatory cytokine frequently seen in the ileal mucosa of and/or Bacteroides spp. and an overgrowth

nATuRE REvIEws | Immunology voLuME 10 | o CToBER 2010 | 739

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

of proteobacteria3840. strikingly, comparable also seen in Il10/ mice after, but not before, inability of DCs to properly regulate TnF
changes in the microbiota have been seen the onset of intestinal inflammation, which production results in a severe and highly
in mice in which intestinal inflammation further highlights the profound influence of penetrating colitis. Intestinal inflammation
was induced by either an invasive pathogen the host immune response on the structure spontaneously progresses to colonic dyspla-
or by the injection of transgenic T cells of the microbial community 27. sia and rectal adenocarcinoma; therefore,
that attacked the gut epithelium27. In these Together, these observations underscore disease progression in this model is similar
two distinct models, host inflammation the confounding role of host-predisposing to that seen in human IBD41,42. TRuC colitis
suppressed the growth of Firmicutes and factors and the difficulty in assigning a can be prevented by eradicating the micro-
Bacteroides spp., allowing proteobacteria, causative role for dysbiosis in IBD. However, biota with broad-spectrum antibiotics.
which are apparently more resistant to host- experimental evidence suggests that intes- Moreover, in a situation that recalls rare
derived microbicidal factors, to outcompete tinal inflammation can select for bacterial reports of intrafamilial transmission of IBD
these normally dominant resident bacteria27. species with colitogenic properties. In T-bet/ in humans, colitis can be transmitted from
notably, outgrowth of proteobacteria was Rag2/ ulcerative colitis (TRuC) mice, the TRuC mice to wild-type mice in both cross-
fostering and co-housing experiments41.
Furthermore, the microbiota in TRuC
a b Altered gut environment mice exhibits complex changes, including
Healthy gut environment
Antibiotics, diet, hygiene,
pollutants, virus? selective enrichment of two proteobacteria,
Proteus mirabilis and Klebsiella pneumonia.
These two bacteria are not sufficient to
Dysbiosis
Gut
Physiological microbiota induce colitis in gnotobiotic TRuC mice;
lumen Decrease in peace-keeping bacteria however, they can colonize the intestine
Peace-keeping and increase in pathobionts
of wild-type mice and, in concert with a
bacterium Pathobiont pathogen-free microbiota, induce colonic
Mucus inflammation43. These data highlight how
the host immune response can shape the
Healthy
epithelial microbiota and eventually lead to the selec-
barrier tion of aggressive bacteria, which not only
Damaged epithelial survive in the inflamed gut but also promote
barrier, increased inflammation.
bacterial adherence
and penetration what are the therapeutic implications
Pathological inflammation of these findings? At the very least, they
IgA+ TH17 cell explain the difficulties in establishing and
TH1 cell plasma cell TReg cell Altered host immune system maintaining remission in patients with IBD
Lamina Genetics using drugs that target host inflammatory
propria Physiological inflammation components or the microbiota without also
Severe monogenic Immune
immunodeficiency gene variants correcting host-predisposing factors. They
IL-10R mutations, NOD2, ATG16L1, highlight the importance of identifying
CVID IL-23R, IRGM
such predisposing factors and designing
Environment more specific therapies for IBD. They also
suggest the need for combined approaches
Stress, diet, infections, vaccine?
that can restore local ecological conditions
Figure 4 | Schematic representation of hostmicrobiota interactions in the healthy and and correct dysbiosis to reinstate balanced
inflamed gut. a | in healthy hosts, an efficient immune barrier contains the microbiota in the gut hostmicrobiota interactions in the long
lumen and feedback mechanisms avoid excessive activation of host immune responses. Peace- term (FIG. 4).
Nature Reviews | Immunology
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
Microbiota and systemic immunity
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 so far we have focused on the roles of the
patients with common variable immunodeficiency (cviD)19) or a key regulatory pathway (for exam- microbiota in intestinal immunity, both
ple, loss-of-function mutations that affect the interleukin-10 receptor (iL-10r)21) spontaneously in health and disease; however, growing
develop intestinal inflammation when exposed to the microbiota. in more common forms of inflam- evidence suggests that the intestinal micro-
matory bowel disease (iBD), a complex genetic background results in more subtle alterations of gut biota can also have an important impact on
immune responses that may weaken the gut barrier and/or impair immunoregulation82. in these indi- the development of the peripheral immune
viduals, lifestyle changes or medical practices (for example, stress, diet, hygiene, smoking, antibiotics, system. Moreover, dysbiosis has been
vaccines or appendectomy) may promote the onset of gut inflammation by affecting the immune implicated in the development of extra-
balance and/or the gut microbiota82. intestinal inflammation results in increased bacterial adherence, intestinal immune-mediated diseases. It will
epithelial damage and increased entry of bacteria into the intestinal lamina propria, thus sustaining
be crucial to determine the extent to which
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 the impact of the microbiota depends
of peace-keeping species16, which results in even more severe and uncontrolled inflammation. These on the host immune status, whether indi-
pathobionts might become sufficiently aggressive to also cause disease in immunocompetent indi- vidual bacterial species can exert distinctive
viduals43. ATG16L1, autophagy-related 16-like 1; irGM, immunity-related GTPase family M; sciD, roles and, ultimately, whether the observed
severe combined immunodeficient; TH cell, T helper cell; Treg cell, regulatory T cell. dysbiosis has a causative role in disease.

740 | o CToBER 2010 | voLuME 10 www.nature.com/reviews/immunol

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

Effects on the peripheral immune system. postnatal pre- and probiotic supplementa- development of an autoimmune ankylosing
The exact impact of the gut microbiota on tion can reduce the incidence of eczema enthesopathy, disease does not develop
the hosts peripheral immune system is con- and food-specific IgE in a subset of high- under germ-free conditions. Furthermore,
troversial. Two groups have convincingly risk children who are born by caesarean disease develops in mice that are colonized
shown that specific immune responses to section51. This protective effect was not with a mixture of culturable anaerobes
the intestinal microbiota are largely confined seen in vaginally delivered children and but not in mice that are colonized with
to the intestinal lymphoid compartment in was transient, as it was significant at Lactobacillus or Staphylococcus spp.59. This
immunocompetent mice with an efficient 2 years of age but not by 5 years of age. finding suggests a role for a specific com-
gut barrier 13,44,45. However, studies compar- The results of such interventions remain ponent (or components) of the microbiota
ing mice that were raised in conventional conflicting and a causative link between dys- in disease progression. A triggering role
or germ-free conditions highlight the biosis, which is induced by changes in life- for the microbiota, specifically for sFB, was
importance of the intestinal microbiota for style or recent medical practices, and allergy also seen in the autoimmune arthritis that
the development of the peripheral immune remains difficult to establish51,52. Moreover, develops in K/Bxn mice30. In this model,
system in immunocompetent hosts. notably, experimental studies that support the role an uncontrolled TH17 cell response that is
the spleens of germ-free mice contain fewer of the microbiota in the development of induced by sFB stimulated the production
and smaller germinal centres46 and decreased allergic diseases are still scarce (TABLE 1). one of autoantibodies and led to the deposi-
numbers of memory CD4+ T cells, and study has reported increased development tion of immune complexes in the joints
cytokine production by these T cells shows a of allergic airway disease in mice that were (FIG. 3). The microbiota also promoted
TH2-type profile47. treated with a short course of oral antibiot- disease in another IL-17-dependent model
The mechanisms that underlie the ics53. Another interesting study showed that of arthritis, which develops in mice that
stimulatory effects of the microbiota on the TLR4 activation by microbiota-derived lack the IL-1 receptor antagonist 60. Finally,
peripheral immune system of immuno- LPs was necessary to prevent anaphylaxis the microbiota and, to a lesser degree, sFB
competent hosts are not well understood. after oral immunization with the peanut- enhanced the severity of experimental
Recent work suggests that soluble factors derived allergen Arah 1. TLR4-deficient or autoimmune encephalomyelitis31. The
that are produced by the microbiota can antibiotic-treated mice showed an increased aggravating role of the microbiota in these
translocate from the gut to the bloodstream TH2-type skewing of cytokine responses models is consistent with its central role in
and activate innate immune cells. For compared with control mice. Conversely, the induction of TH17 cell responses.
example, the opsonophagocytic activity of activation of TLR9 by oral administration of By contrast, the microbiota was shown
neutrophils is primed by microbiota-derived CpG oligodeoxynucleotides could abrogate to have a protective role in collagen-
peptidoglycan, which results in enhanced allergic symptoms and correct the TH1/TH2 induced arthritis61 and to prevent diabetes
protection against pneumococcal sepsis48. imbalance54. However, additional work is development in myeloid differentiation
Moreover, monocolonization with B. fragilis needed to delineate whether and how the primary-response protein 88 (MYD88)-
corrects systemic T cell deficiencies and composition of the microbiota might deficient non-obese diabetic (noD) mice62,
the TH1/TH2 imbalance of germ-free mice influence the onset of allergy. as in both models disease was more severe
due to the stimulation of DC IL-12 produc- if animals were housed in germ-free condi-
tion by the bacteriums unusual capsular Autoimmunity and the microbiota. The tions. It remains unclear why a protective
polysaccharide47. possibility that the intestinal microbiota is role for the microbiota during the develop-
involved in the development of systemic ment of diabetes is seen in the presence
Allergy and dysbiosis. The hygiene autoimmunity has recently attracted grow- of impaired TLR signalling. The complete
hypothesis, which has been revised ing attention. Changes in the composition of lack of TLR signalling in Myd88/ Trif
several times since its initial formula- the gut flora have been reported in patients (TIR-domain-containing adaptor protein
tion by strachan in 1989, stipulates that in the early phases of rheumatoid arthritis55 inducing IFn)/ mice has been associ-
decreased exposure to infectious agents, when compared with a control group with ated with abnormal bacterial translocation
as well as changes in the intestinal micro- fibromyalgia. However, such studies cannot into the spleen and activation of systemic
biota during infancy, might alter immune establish a link between dysbiosis and the adaptive responses. It will be interesting to
regulatory networks and account for the development of disease. Therefore, the role evaluate whether comparable mechanisms
dramatic increase in the incidence of of the microbiota has been more directly operate in Myd88/ noD mice and par-
allergic diseases that has been observed in addressed by comparing the onset and/or ticipate in the protection against diabetes.
developed countries49. several studies have severity of experimental autoimmune dis- Interestingly, the caecal microbiota of
reported differences in the composition eases in germ-free mice with disease onset Myd88/ noD mice differed significantly
of the faecal microbiota of infants who and/or severity in mice that have been from that of noD mice and could attenuate
develop an allergic disease and those who colonized by a diverse microbiota. the development of type 1 diabetes when
do not. notably, a decreased frequency In several models of autoimmunity, transferred to the newborn progeny of
of Lactobacillus and Bifidobacterium spp. the progression of disease was compa- germ-free noD mice62; this suggests that
has been suggested to precede the onset rable between both sets of mice5658. In one or more species that confer protection
of allergy 50, and prophylactic approaches, other models, contradictory roles for the against disease might have been selected
which are based on the administration of microbiota have been reported (TABLE 1). for in the noD mice with impaired
probiotics to mothers and newborns at The microbiota was shown to trigger TLR signalling.
high risk for IgE-associated allergies, have disease in several mouse models of auto- Taken together, these data provide
been initiated. A recent study reports immune arthritis. For example, in a mouse novel examples of the complex interplay
that 1 month of prenatal and 6 months of strain that is prone to the spontaneous that exists between the host immune

nATuRE REvIEws | Immunology voLuME 10 | o CToBER 2010 | 741

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

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 allergy Weanling c3H mice or Oral cocktail of antibiotics for 3 weeks induction of anaphylactic 54
to peanut allergen c57BL/6 mice symptoms, increased production of
ige and iL-13
Tlr4 mutant or Tlr4/ mice None
ige-mediated allergic BALB/c and c57BL/6 mice Oral cefoperazone (cefobid, Pfizer; increase in pulmonary eosinophils 49,53
airway disease induced cefazone, Pharco B international) and enhanced synthesis of ige, iL-5
by Aspergillus fumigatus for 5 days and a single oral gavage of and iL-13
spores or ovalbumin Candida albicans
Autoimmunity
systemic lupus MrL/lpr mice Germ-free mice No change in autoimmunity 58
erythematosus
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 specific No disease
pathogen-free flora
spontaneous ankylosing Male B10.Br mice Germ-free mice No disease 59
enthesopathy
colonization with probiotic No disease
Lactobacillus spp.
colonization with a mixture of Disease restored
Bacteroides, Enterococcus, Veillonella
and Staphylococcus spp.
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 segmented Disease restored
filamentous bacteria
experimental c57BL/6J mice Germ-free mice Weak severity 31
autoimmune
encephalomyelitis specific pathogen-free flora Maximal severity
colonization with segmented intermediate severity
filamentous bacteria
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. *Aire
regulates the transcription of peripheral autoantigens in medullary thymic epithelial cells and is necessary for thymic negative selection; AiD is central for
class-switch recombination and somatic hypermutation in B cells; NOD mice provide a polygenic model of type 1 diabetes; ||Mice with a transgenic T cell receptor
against a self peptide that is derived from glucose-6-phosphate isomerase.

system and the microbiota, and the dif- Conclusion also indicates the possible contribution of
ficulties in attempting to delineate the An increasing number of studies are pro- the intestinal microbiota to immunological
respective roles of host predisposing gressively unravelling the fascinating inter- diseases outside the gut. However, from the
factors and specific bacterial species. To actions that occur between eukaryotes and microbial perspective, the host is simply a
determine the therapeutic implications their bacterial symbionts. It is now clear complex environment and the distinction
of these observations, further research is that the intestinal microbiota profoundly between health and disease is important
needed to elucidate how the microbiota influences host metabolic and immune only as far as it affects microbial fitness.
is able to influence peripheral immune pathways and participates in human health The challenge that lies ahead is to determine
responses and how such microbiota- and disease. Compelling evidence shows a when changes in our microbiota are the
driven responses can interfere with the pivotal role for the microbiota in the devel- primary cause of a disease and when these
immunological mechanisms that underlie opment of many gastrointestinal diseases, changes merely reflect the enormous capacity
a given autoimmune disease. from inflammation to cancer. Recent work of bacteria for rapid and continuous genetic

742 | o CToBER 2010 | voLuME 10 www.nature.com/reviews/immunol

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

adaptation to new ecological conditions. An 19. Malamut, G. et al. The enteropathy associated with 43. Garrett, W. et al. Enterobacteriaceae act in concert
common variable immunodeficiency: the delineated with the gut microbiota to induce spontaneous and
in-depth understanding of the principles frontiers with celiac disease. Am. J. Gastroenterol. maternally-transmitted colitis. Cell Host Microbe
and mechanisms that underlie microbial 15 Jun 2010 (doi:10.1038/ajg.2010.214). (in the press).
20. Bouma, G. & Strober, W. The immunological and 44. Konrad, A., Cong, Y., Duck, W., Borlaza, R. &
community structures and hostsymbiont genetic basis of inflammatory bowel disease. Nature Elson, C. O. Tight mucosal compartmentation of the
relationships will be pivotal for the develop- Rev. Immunol. 3, 521533 (2003). murine immune response to antigens of the enteric
21. Glocker, E. O. et al. Inflammatory bowel disease and microbiota. Gastroenterology 130, 20502059
ment of therapeutic approaches that manip- mutations affecting the interleukin-10 receptor. (2006).
ulate the intestinal microbiota to maintain N. Engl. J. Med. 361, 20332045 (2009). 45. Slack, E. et al. Innate and adaptive immunity
22. Talham, G. L., Jiang, H. Q., Bos, N. A. & Cebra, J. J. cooperate flexibly to maintain host-microbiota
human health. Segmented filamentous bacteria are potent stimuli of mutualism. Science 325, 617620 (2009).
a physiologically normal state of the murine gut 46. Bauer, H., Horowitz, R. E., Levenson, S. M. &
Nadine Cerf-Bensussan and Valrie Gaboriau-Routhiau mucosal immune system. Infect. Immun. 67, Popper, H. The response of the lymphatic tissue
are at the Institut National de la Sant et de la 19922000 (1999). to the microbial flora. Studies on germfree mice.
Recherche Mdicale (INSERM) U989, Universit Paris 23. Ivanov, I. I. et al. Induction of intestinal Th17 cells by Am. J. Pathol. 42, 471483 (1963).
segmented filamentous bacteria. Cell 139, 485498 47. Mazmanian, S. K., Liu, C. H., Tzianabos, A. O. &
Descartes, 156 rue de Vaugirard, 75730 Paris
(2009). Kasper, D. L. An immunomodulatory molecule of
Cedex 15, France; and the Institut National de la 24. Brandl, K. et al. Vancomycin-resistant enterococci symbiotic bacteria directs maturation of the host
Recherche Agronomique (INRA) UM1319, Domaine de exploit antibiotic-induced innate immune deficits. immune system. Cell 122, 107118 (2005).
Vilvert, 78350 Jouy-en-Josas, France. Nature 455, 804807 (2008). 48. Clarke, T. B. et al. Recognition of peptidoglycan from
Correspondence to N.C.-B. 25. Klaasen, H. L. et al. Intestinal, segmented, filamentous the microbiota by Nod1 enhances systemic innate
bacteria in a wide range of vertebrate species. immunity. Nature Med. 16, 228231 (2010).
e-mail: nadine.cerf-bensussan@inserm.fr
Lab. Anim. 27, 141150 (1993). 49. Noverr, M. C. & Huffnagle, G. B. The microflora
doi:10.1038/nri2850 26. Lupp, C. et al. Host-mediated inflammation disrupts hypothesis of allergic diseases. Clin. Exp. Allergy 35,
the intestinal microbiota and promotes the overgrowth 15111520 (2005).
of Enterobacteriaceae. Cell Host Microbe 2, 119129 50. Sjogren, Y. M., Jenmalm, M. C., Bottcher, M. F.,
1. Costello, E. K. et al. Bacterial community variation in (2007). Bjorksten, B. & Sverremark-Ekstrom, E. Altered early
human body habitats across space and time. Science 27. Stecher, B. et al. Salmonella enterica serovar infant gut microbiota in children developing allergy up
326, 16941697 (2009). typhimurium exploits inflammation to compete with to 5 years of age. Clin. Exp. Allergy 39, 518526
2. Gill, S. R. et al. Metagenomic analysis of the human the intestinal microbiota. PLoS Biol. 5, 21772189 (2009).
distal gut microbiome. Science 312, 13551359 (2007). 51. Kuitunen, M. et al. Probiotics prevent IgE-associated
(2006). 28. Snel, J. et al. Comparison of 16S rRNA sequences of allergy until age 5 years in cesarean-delivered children
3. Eberl, G. A new vision of immunity: homeostasis of the segmented filamentous bacteria isolated from mice, but not in the total cohort. J. Allergy Clin. Immunol.
superorganism. Mucosal Immunol. 5 May 2010 rats, and chickens and proposal of Candidatus 123, 335341 (2009).
(doi:10.1038/mi.2010.20). Arthromitus. Int. J. Syst. Bacteriol. 45, 780782 52. Penders, J., Stobberingh, E. E., van den Brandt, P. A.
4. Qin, J. et al. A human gut microbial gene catalogue (1995). & Thijs, C. The role of the intestinal microbiota in the
established by metagenomic sequencing. Nature 464, 29. Stepankova, R. et al. Segmented filamentous bacteria development of atopic disorders. Allergy 62,
5965 (2010). in a defined bacterial cocktail induce intestinal 12231236 (2007).
5. Moran, N. A., McCutcheon, J. P. & Nakabachi, A. inflammation in SCID mice reconstituted with 53. Noverr, M. C., Falkowski, N. R., McDonald, R. A.,
Genomics and evolution of heritable bacterial CD45RBhigh CD4+ T cells. Inflamm. Bowel Dis. 13, McKenzie, A. N. & Huffnagle, G. B. Development of
symbionts. Annu. Rev. Genet. 42, 165190 (2008). 12021211 (2007). allergic airway disease in mice following antibiotic
6. Garrett, W. S., Gordon, J. I. & Glimcher, L. H. 30. Wu, H.-S. et al. Gut-residing segmented filamentous therapy and fungal microbiota increase: role of host
Homeostasis and inflammation in the intestine. bacteria drive autoimmune arthritis via T helper 17 genetics, antigen, and interleukin-13. Infect. Immun.
Cell 140, 859870 (2010). cells. Immunity 32, 815827 (2010). 73, 3038 (2005).
7. Hooper, L. V. & Macpherson, A. J. Immune 31. Lee, Y. K., Menezes, J. S., Umesaki, Y. & 54. Bashir, M. E., Louie, S., Shi, H. N. & Nagler-Anderson, C.
adaptations that maintain homeostasis with the Mazmanian, S. K. Microbes and health sackler Toll-like receptor 4 signaling by intestinal microbes
intestinal microbiota. Nature Rev. Immunol. 10, colloquium: proinflammatory T-cell responses to gut influences susceptibility to food allergy. J. Immunol.
159169 (2010). microbiota promote experimental autoimmune 172, 69786987 (2004).
8. Eberl, G. & Lochner, M. The development of encephalomyelitis. Proc. Natl Acad. Sci. USA 28 Jul 55. Vaahtovuo, J., Munukka, E., Korkeamaki, M.,
intestinal lymphoid tissues at the interface of self 2010 (doi:10.1073/pnas.10000.82107). Luukkainen, R. & Toivanen, P. Fecal microbiota in early
and microbiota. Mucosal Immunol. 2, 478485 32. Chow, J. & Mazmanian, S. K. A pathobiont of the rheumatoid arthritis. J. Rheumatol. 35, 15001505
(2009). microbiota balances host colonization and intestinal (2008).
9. Rescigno, M. & Di Sabatino, A. Dendritic cells in inflammation. Cell Host Microbe 7, 265276 (2010). 56. Gray, D. H., Gavanescu, I., Benoist, C. & Mathis, D.
intestinal homeostasis and disease. J. Clin. Invest. 33. Polk, D. B. & Peek, R. M. Jr. Helicobacter pylori: Danger-free autoimmune disease in Aire-deficient
119, 24412450 (2009). gastric cancer and beyond. Nature Rev. Cancer 10, mice. Proc. Natl Acad. Sci. USA 104, 1819318198
10. Gaboriau-Routhiau, V. et al. The key role of segmented 403414 (2010). (2007).
filamentous bacteria in the coordinated maturation of 34. Wu, S. et al. A human colonic commensal promotes 57. Hase, K. et al. Activation-induced cytidine deaminase
gut helper T cell responses. Immunity 31, 677689 colon tumorigenesis via activation of T helper type 17 deficiency causes organ-specific autoimmune disease.
(2009). T cell responses. Nature Med. 15, 10161022 PLoS ONE 3, e3033 (2008).
11. Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L. (2009). 58. Maldonado, M. A. et al. The role of environmental
& Hooper, L. V. Paneth cells directly sense gut 35. Carvalho, F. A. et al. Crohns disease adherent- antigens in the spontaneous development of
commensals and maintain homeostasis at the invasive Escherichia coli colonize and induce strong autoimmunity in MRL-lpr mice. J. Immunol. 162,
intestinal host-microbial interface. Proc. Natl Acad. gut inflammation in transgenic mice expressing 63226330 (1999).
Sci. USA 105, 2085820863 (2008). human CEACAM. J. Exp. Med. 206, 21792189 59. Sinkorova, Z., Capkova, J., Niederlova, J.,
12. Peterson, D. A., McNulty, N. P., Guruge, J. L. & (2009). Stepankova, R. & Sinkora, J. Commensal intestinal
Gordon, J. I. IgA response to symbiotic bacteria as a 36. Darfeuille-Michaud, A. et al. High prevalence of bacterial strains trigger ankylosing enthesopathy of
mediator of gut homeostasis. Cell Host Microbe 2, adherent-invasive Escherichia coli associated with ileal the ankle in inbred B10.BR (H-2k) male mice. Hum.
328339 (2007). mucosa in Crohns disease. Gastroenterology 127, Immunol. 69, 845850 (2008).
13. Macpherson, A. J. & Uhr, T. Induction of protective 412421 (2004). 60. Abdollahi-Roodsaz, S. et al. Stimulation of TLR2 and
IgA by intestinal dendritic cells carrying commensal 37. Barnich, N. et al. CEACAM6 acts as a receptor for TLR4 differentially skews the balance of T cells in a
bacteria. Science 303, 16621665 (2004). adherent-invasive E. coli, supporting ileal mucosa mouse model of arthritis. J. Clin. Invest. 118,
14. Lotz, M. et al. Postnatal acquisition of endotoxin colonization in Crohn disease. J. Clin. Invest. 117, 205216 (2008).
tolerance in intestinal epithelial cells. J. Exp. Med. 15661574 (2007). 61. Breban, M. A., Moreau, M. C., Fournier, C.,
203, 973984 (2006). 38. Frank, D. N. et al. Molecular-phylogenetic Ducluzeau, R. & Kahn, M. F. Influence of the bacterial
15. Mazmanian, S. K., Round, J. L. & Kasper, D. L. characterization of microbial community imbalances in flora on collagen-induced arthritis in susceptible and
A microbial symbiosis factor prevents intestinal human inflammatory bowel diseases. Proc. Natl Acad. resistant strains of rats. Clin. Exp. Rheumatol. 11,
inflammatory disease. Nature 453, 620625 Sci. USA 104, 1378013785 (2007). 6164 (1993).
(2008). 39. Sokol, H. et al. Specificities of the fecal microbiota in 62. Wen, L. et al. Innate immunity and intestinal
16. Sokol, H. et al. Faecalibacterium prausnitzii is an inflammatory bowel disease. Inflamm. Bowel Dis. 12, microbiota in the development of Type 1 diabetes.
anti-inflammatory commensal bacterium identified 106111 (2006). Nature 455, 11091113 (2008).
by gut microbiota analysis of Crohn disease patients. 40. Swidsinski, A., Weber, J., Loening-Baucke, V., Hale, L. P. 63. Giraud, A. et al. Dissecting the genetic components
Proc. Natl Acad. Sci. USA 105, 1673116736 & Lochs, H. Spatial organization and composition of of adaptation of Escherichia coli to the mouse gut.
(2008). the mucosal flora in patients with inflammatory bowel PLoS Genet. 4, e2 (2008).
17. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ disease. J. Clin. Microbiol. 43, 33803389 (2005). 64. Turnbaugh, P. J., Backhed, F., Fulton, L. & Gordon, J. I.
regulatory T-cell development by a commensal 41. Garrett, W. S. et al. Communicable ulcerative colitis Diet-induced obesity is linked to marked but reversible
bacterium of the intestinal microbiota. Proc. Natl induced by T-bet deficiency in the innate immune alterations in the mouse distal gut microbiome.
Acad. Sci. USA 107, 1220412209 (2010). system. Cell 131, 3345 (2007). Cell Host Microbe 3, 213223 (2008).
18. Fagarasan, S. et al. Critical roles of activation-induced 42. Garrett, W. S. et al. Colitis-associated colorectal cancer 65. Backhed, F. et al. The gut microbiota as an
cytidine deaminase in the homeostasis of gut flora. driven by T-bet deficiency in dendritic cells. Cancer Cell environmental factor that regulates fat storage.
Science 298, 14241427 (2002). 16, 208219 (2009). Proc. Natl Acad. Sci. USA 101, 1571815723 (2004).

nATuRE REvIEws | Immunology voLuME 10 | o CToBER 2010 | 743

2010 Macmillan Publishers Limited. All rights reserved


PersPectives

66. Samuel, B. S. et al. Effects of the gut microbiota on extension into the small bowel lumen in response to indigenous intestinal bacteria that activate
host adiposity are modulated by the short-chain epithelial cell TLR engagement. J. Exp. Med. 203, intraepithelial lymphocytes and induce MHC class II
fatty-acid binding G protein-coupled receptor, Gpr41. 28412852 (2006). molecules and fucosyl asialo GM1 glycolipids on the
Proc. Natl Acad. Sci. USA 105, 1676716772 74. Schulz, O. et al. Intestinal CD103+, but not CX3CR1+, small intestinal epithelial cells in the ex-germ-free
(2008). antigen sampling cells migrate in lymph and serve mouse. Microbiol. Immunol. 39, 555562 (1995).
67. Turnbaugh, P. J. et al. The human microbiome project. classical dendritic cell functions. J. Exp. Med. 206, 82. Mayer, L. Evolving paradigms in the pathogenesis of
Nature 449, 804810 (2007). 31013114 (2009). IBD. J. Gastroenterol. 45, 916 (2010).
68. Vijay-Kumar, M. et al. Metabolic syndrome and altered 75. Tsuji, M. et al. Preferential generation of follicular B
gut microbiota in mice lacking Toll-like receptor 5. helper T cells from Foxp3+ T cells in gut Peyers Acknowledgements
Science 328, 228231 (2010). patches. Science 323, 14881492 (2009). The authors thank W. Garrett for sharing unpublished data.
69. Peyrin-Biroulet, L. et al. Peroxisome proliferator- 76. Cerutti, A. & Rescigno, M. The biology of intestinal Their work is supported by grants from the Institut National
activated receptor activation is required for immunoglobulin A responses. Immunity 28, 740750 de la Sant et de la Recherche Mdicale (INSERM), the
maintenance of innate antimicrobial immunity in the (2008). Institut National de la Recherche Agronomique (INRA) and
colon. Proc. Natl Acad. Sci. USA 107, 87728777 77. Coombes, J. L. & Powrie, F. Dendritic cells in intestinal the Agence Nationale de la Recherche and Fondation
(2010). immune regulation. Nature Rev. Immunol. 8, Princesse Grace. The authors are partners of the European
70. Kumar, A. et al. Commensal bacteria modulate 435446 (2008). Community networks Cross-Talk (contract number
cullin-dependent signaling via generation of reactive 78. Atarashi, K. et al. ATP drives lamina propria TH17 cell PITN-GA-2008-215553) and Tornado (FP7 222720).
oxygen species. EMBO J. 26, 44574466 (2007). differentiation. Nature 455, 808812 (2008).
71. Dubuquoy, L. et al. Impaired expression of peroxisome 79. Zhou, L., Chong, M. M. & Littman, D. R. Plasticity of Competing interests statement
proliferator-activated receptor in ulcerative colitis. CD4+ T cell lineage differentiation. Immunity 30, The authors declare no competing financial interests.
Gastroenterology 124, 12651276 (2003). 646655 (2009).
72. Kelly, D. et al. Commensal anaerobic gut bacteria 80. Keilbaugh, S. A. et al. Activation of RegIII/ and
attenuate inflammation by regulating nuclear- interferon expression in the intestinal tract of SCID FURTHER INFORMATION
cytoplasmic shuttling of PPAR- and RelA. Nature mice: an innate response to bacterial colonisation of Nadine cerf-Bensussans homepage: http://www.
Immunol. 5, 104112 (2004). the gut. Gut 54, 623629 (2005). fondationimagine.org/Nadine-cerf-Bensussan_uk.php
73. Chieppa, M., Rescigno, M., Huang, A. Y. & 81. Umesaki, Y., Okada, Y., Matsumoto, S., Imaoka, A. & All lInkS ARE ActIvE In thE onlInE PdF
Germain, R. N. Dynamic imaging of dendritic cell Setoyama, H. Segmented filamentous bacteria are

744 | o CToBER 2010 | voLuME 10 www.nature.com/reviews/immunol

2010 Macmillan Publishers Limited. All rights reserved

Das könnte Ihnen auch gefallen