Microbiotain IBD

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

REvIEwS

Host–microbiota interactions in
inflammatory bowel disease
Roberta Caruso1,2, Bernard C. Lo1,2 and Gabriel Núñez 1
*
Abstract | The mammalian intestine is colonized by trillions of microorganisms that have
co-evolved with the host in a symbiotic relationship. The presence of large numbers of symbionts
near the epithelial surface of the intestine poses an enormous challenge to the host because
it must avoid the activation of harmful inflammatory responses to the microorganisms while
preserving its ability to mount robust immune responses to invading pathogens. In patients with
inflammatory bowel disease, there is a breakdown of the multiple strategies that the immune
system has evolved to promote the separation between symbiotic microorganisms and the
intestinal epithelium and the effective killing of penetrant microorganisms, while suppressing
the activation of inappropriate T cell responses to resident microorganisms. Understanding the
complex interactions between intestinal microorganisms and the host may provide crucial insight
into the pathogenesis of inflammatory bowel disease as well as new avenues to prevent and treat
the disease.

Pathobionts
Inflammatory bowel disease (IBD), which includes often referred to as pathobionts because they can cause
Microorganisms that, under Crohn’s disease and ulcerative colitis, is a chronic and disease under certain conditions. The composition of
normal circumstances, live as relapsing inflammatory disorder of the intestine that the gut microbiota exhibits broad inter-​individual and
non-​harmful symbionts but can affects ~3 million people in the United States1. Although intra-​individual variability7,8 and is thought to be a cru-
induce pathology under certain
the pathogenesis of IBD is poorly understood, multiple cial determinant of host susceptibility to several diseases,
conditions, usually involving
environmental and/or genetic lines of evidence suggest that the disease is caused by including IBD9–11. A key feature of IBD is alteration of
alterations. a confluence of genetic and environmental factors that the composition of the gut microbiota (known as dysbi-
alter gut homeostasis, thereby triggering immune-​ osis)12; however, the precise role of dysbiosis in disease
mediated inflammation in genetically susceptible indi- remains poorly understood.
viduals2. Genetic studies have identified more than 200 The close proximity of a large number of symbiotic
loci that regulate IBD risk, most of which are associated microorganisms to the epithelial surface represents
with immunological pathways that regulate microbial a unique challenge to the mucosal immune system in
recognition and killing, immune responses to micro- that it must preserve the ability to mount an immune
organisms and the reinforcement of the intestinal bar- response against an invading pathogen while avoiding
rier3,4. Although the pathogenesis of both Crohn’s disease harmful inflammatory responses to the gut micro­
and ulcerative colitis involves intestinal inflammation, biota. In this Review, we discuss the mechanisms that
these two disorders differ in several features, including limit the inappropriate accumulation of particular
the association with specific susceptibility loci and the microorganisms near the epithelial surface and pro-
type of immune response and pathology associated with mote the clearance of penetrant organisms to minimize
disease2,4 (Box 1). the activation of harmful inflammatory responses
1
Department of Pathology
A key aspect of both forms of IBD pathogenesis is in the gut. We also discuss how IBD-​associated genetic
and Rogel Cancer Center, their link to the presence of symbiotic microorganisms mutations can disrupt these protective mechanisms,
the University of Michigan living in the intestinal tract. The resident symbionts which have been referred to as ‘mucosal firewalls’13.
Medical School, Ann Arbor, contribute to many host physiological processes, includ- In addition, the immune system has developed effec-
Michigan, USA.
ing digestive and metabolic functions, regulation of tive regulatory mechanisms to suppress the activ­
2
These authors contributed the epithelial barrier, development and regulation of the ation of inappropriate T cell responses to the resident
equally: Roberta Caruso,
Bernard C. Lo.
host immune system, and protection against patho­gen microbiota that can be compromised in IBD. We also
colonization5,6. Whereas the great majority of intes­ describe how IBD-​associated genetic defects can lead
*e-​mail:
gabriel.nunez@umich.edu tinal microorganisms live in a mutualistic relation- to pathobiont accumulation and penetration into the
https://doi.org/10.1038/ ship with the host, some symbiotic organisms, such as intestinal tissue, which further promotes dysbiosis and
s41577-019-0268-7 Mucispirillum schaedleri and Helicobacter species, are inflammation. This Review focuses on bacteria as these

Nature Reviews | Immunology


Reviews

Box 1 | Comparing Crohn’s disease and ulcerative colitis a formidable physical barrier and is also a scaffold for
antimicrobial peptides and immunoglobulins, which
Crohn’s disease and ulcerative colitis, the two major forms of inflammatory bowel prevent microorganisms from contacting the epithelial
disease in humans, are chronic relapsing inflammatory disorders. although these two surface18. The small intestine lacks a well-​demarcated
diseases have historically been studied together and share some clinical characteristics, inner mucus layer but it contains large numbers of
it is now clear that they represent two distinct pathophysiological entities. Crohn’s
Paneth cells, specialized intestinal cells that reside at
disease is characterized by segmental inflammation with sharp demarcation between
involved and uninvolved bowel segments (skip lesions). Pathologically, Crohn’s disease the base of the crypts of Lieberkühn and are rich in anti-
is characterized by transmural inflammation, narrow and penetrating ulcers, and the microbial molecules19. In response to bacterial stimula-
presence of non-​caseating granulomas in many patients. although lesions in Crohn’s tion, Paneth cells release their antimicrobial molecules,
disease may affect any site of the gastrointestinal tract, the terminal ileum is most including α-​defensins, into the intestinal lumen to limit
commonly affected and the earliest mucosal lesions in Crohn’s disease are often the accumulation of bacterial symbionts19,20. In addi-
located over Peyer’s patches. tion, intestinal epithelial cells (IECs) in the small intes-
unlike Crohn’s disease, ulcerative colitis is characterized by continuous inflammation tine secrete antimicrobial lectins, such as regenerating
that extends proximally from the rectum to a variable distance along the colon. islet-derived protein 3γ (REG3γ), that accumulate in the
inflammation in ulcerative colitis is superficial, limited to the mucosal layer and mucus layer and further promote the segregation of
characterized by the presence of neutrophils that infiltrate the lamina propria and the
the microbiota from the host21.
intestinal crypts, where they form micro-​abscesses (known as cryptic abscesses).
Depletion of goblet cells is also a common histological feature in ulcerative colitis2. Although the lamina propria is largely devoid of
Genome-​wide association studies have identified more than 200 susceptibility genes acute inflammatory cells, some neutrophils can transmi-
that are either shared by both diseases or specifically associated with one disease3. grate into the intestinal luminal side at steady state and
some of the Crohn’s disease-​specific genes are involved in immune responses against kill bacteria near the epithelial surface through several
bacteria, whereas genes associated with the risk of developing ulcerative colitis are mechanisms, including induction of an oxidative burst22.
mainly involved in T cell signalling and epithelial barrier function203,204. although The cytokine IL-22 also has a role in establishing
dysregulated intestinal inflammation is the hallmark of both Crohn’s disease and host–microbial mutualism by acting on epithelial cells
ulcerative colitis, mucosal inflammation associated with these two diseases is mediated to mediate barrier function and antimicrobial host
by different types of effector T cell. Whereas Crohn’s disease is mainly characterized by defence23. For example, secretion of IL-22 by group 3
an imbalance of effector T cells, predominantly T helper 1 (TH1) cells or TH17 cells, versus
innate lymphoid cells (ILC3s) is required for the contain-
regulatory T cells, ulcerative colitis has a cytokine profile associated with TH17 cells and
tH2 cells2,205,206. ment of commensal bacteria by inducing the expression
of antimicrobial peptides that prevent systemic dissem-
ination of Alcaligenes spp. bacteria24. Innate sources of
microorganisms have been best studied in the context of IL-22 are also important for controlling the proliferation
intestinal inflammation; however, the role of other com- of segmented filamentous bacteria (SFB) in the gut and
ponents of the intestinal microbiota, such as viruses and limiting T helper 17 (TH17) cell-​mediated colitis25. IL-22
fungi, in IBD is mentioned where appropriate. Finally, also promotes epithelial glycan fucosylation to support
we conclude the article by discussing how the microbiota the growth of mutualistic bacteria that are adapted to
can be targeted as a potential therapy for IBD. use fucose as a nutrient source26–28. Disruption of IL-22
signalling and fucosylation leads to greater susceptibil-
Mucosal firewalls ity to enteric infections and colitis, in part owing to the
The vast population of symbiotic microorganisms in the dysbiotic overgrowth of opportunistic pathobionts27,28.
Crypts of Lieberkühn intestine is separated from host tissues by a single-​cell The separation of intestinal bacteria from the epi-
Invaginations of the small layer of epithelium. To maintain its homeostatic relation- thelial surface is also promoted by IgA, which is pro-
intestine that contain epithelial ship with the intestinal microbiota, the host has evolved duced by plasma cells that reside in gut-​associated
stem cells and Paneth cells.
several strategies to minimize the contact between lymphoid tissues. Large amounts of polymeric IgA
Oxidative burst microorganisms and the epithelial surface and to limit are transcytosed across the epithelium into the lumen,
The rapid production of the presence of penetrant symbionts that could trigger where IgA has a role in maintaining barrier function
reactive oxygen species by unwanted inflammatory responses. by coating bacteria and binding to microbial antigens
phagocytes that can directly
and their toxins as well as in shaping the composition
kill bacteria.
Segregating symbionts from the epithelial surface. The of the microbiota through unresolved mechanisms29,30.
Segmented filamentous host restricts the access of potentially harmful micro­ IgA responses occur constitutively during homeosta-
bacteria organisms to the mucosal surface by several mecha- sis through T cell-​independent and T cell-​dependent
(SFB). A species of Clostridium-​ nisms, including mucus secretion and the release of processes that preferentially target distinct microbial
related bacteria that primarily
inhabits the terminal ileum of
antimicrobial proteins and immunoglobulins into the species29. IgA antibodies are typically polyreactive and
mice and promotes T helper intestinal lumen13,14 (Fig. 1). The mucus lining the gut bind with low affinity to microbial lipopolysaccharides,
17 cell development. epi­thelium is composed primarily of highly glycosylated DNA and flagellar antigens29. Microbial sensing through
proteins, including mucin 2 (MUC2), that are secre­ Toll-​like receptor (TLR) engagement can directly stim-
Endoplasmic reticulum
ted by goblet cells. The glycosylation of MUC2 protects ulate IgA production, although T cell-​independent IgA
stress
A consequence of dysregulated the protein from degradation by host and bacterial can also be induced independently of the microbiota by
protein processing in the proteases and promotes interaction with water to form endoplasmic reticulum stress in IECs to provide protection
endoplasmic reticulum that the mucus gel15. In the large intestine, two clearly distinct against intestinal inflammation31. In addition, T cell-​
initiates unfolded protein mucus layers are present: the outer layer, which contains intrinsic sensing of microbial signals through myeloid
response pathways; unresolved
endoplasmic reticulum stress is
large numbers of mucus-​dwelling bacteria, and the inner differentiation primary response protein 88 (MYD88)
associated with inflammatory layer near the epithelium, which is largely depleted of is important for homeostatic IgA responses to prevent
bowel disease. microorganisms16,17. The inner mucus layer provides dysbiosis and enteropathy32. Select members of the

www.nature.com/nri
Reviews

intestinal microbiota, which are heavily coated by IgA proximity to the epithelial surface, where they elicit an
during colitis, confer enhanced susceptibility to colitis antigen-​dependent, high-​affinity, T cell-​dependent IgA
when transferred to germ-​free animals, which indicates response33,34. In contrast to its role in protecting barrier
that preferential IgA binding during dysbiosis may iden- surfaces by microbial exclusion, IgA can be used by some
tify bacterial species that are relevant to IBD33. Whereas symbiotic species to stably colonize the intestine35. These
most commensals strongly elicit T cell-​independent IgA studies indicate that IgA can enforce a ‘healthy’ micro-
binding, ‘atypical’ bacteria, such as SFB, M. schaedleri, biota by promoting the colonization of beneficial com-
Prevotella spp. and Helicobacter sp. flexispira, can evade mensals but that, in states of dysbiosis, IgA responses are
T cell-​independent antibody responses to come in close induced against potentially colitic species29,30.

Small intestine Large intestine


Neutrophil
Mucus layer Intestinal
lumen Intestinal
microbiota

Goblet cell Detection


and killing

MUC2 IgG

Pathobiont
SIgA
M cell

Transcytosis IFNγ
Epithelium
AMPs,
FUT2
DC
TH1
α-Defensins Plasma cell
cell
TLR TFH cell
Peyer’s patch
Paneth IL-1β,
cell IL-6, IL-17,
IL-23 IL-22
TH17 cell

ILC3 Kupffer
cell
Lymphoid tissue Liver and systemic sites
Lamina propria

Preventing symbiont
Segregating symbionts from the epithelial surface Control at systemic sites
accumulation in the mucosa

Fig. 1 | mucosal firewalls. The host has evolved several strategies to mini- intestinal microorganisms, preventing their translocation across the epi­
mize the contact between intestinal microorganisms and the epithelial thelium. T follicular helper (TFH) cell-​dependent responses further contribute
surface and to limit the presence of penetrant symbionts that could trigger to homeostasis by supporting the production of high-​affinity IgA , which
unwanted inflammatory responses. Goblet cell secretion of glycoproteins, binds to select bacteria. Myeloid cell-​derived cytokines, including IL-1β, IL-6
including mucin 2 (MUC2), forms a mucus barrier that prevents micro­ and IL-23, promote T helper 17 (TH17) cell differentiation and group 3 innate
organisms in the intestinal lumen from making contact with the epithelium. lymphoid cell (ILC3) activation. IL-22 produced by ILC3s and TH17 cells fur-
In the small intestine, a loosely formed mucus layer coats the epi­thelium, ther reinforces the segregation of symbionts by inducing the expression of
whereas, in the large intestine, the inner mucus layer is largely devoid of antimicrobial peptides (AMPs), including regenerating islet-​derived protein
bacteria and mucus-​dwelling symbionts are restricted to the outer mucus 3γ (REG3γ). IL-22 also promotes fucosyltransferase 2 (FUT2)-mediated fuco-
layer. Paneth cells located in the crypts of the small intestine constitutively sylation of epithelial glycans to support microbial symbiosis and protect
express microbicidal α-​defensins. Microfold (M) cells are specialized epi­ against the invasion of potentially harmful bacteria. If bacteria are able to
thelial cells that overlay lymphoid tissues, such as Peyer’s patches, and facil- overcome these barriers, IL-17-stimulated neutrophils can eliminate patho-
itate antigen sampling. Dendritic cells (DCs) sample antigens in the Peyer’s bionts localized near the apical epithelial surface or those that have reached
patches delivered by M cells or possibly directly in the lumen through pro- the lamina propria. Microbial killing by tissue-​resident macrophages and
jections that penetrate the epithelial layer. Secretory IgA (SIgA) is produced DCs is further enhanced by TH1 cell-​derived interferon-​γ (IFNγ). The produc-
by plasma cells located in the Peyer’s patches and is secreted across the tion of bacteria-​specific IgG controls systemic infection by binding patho-
epithelium into the gut lumen by transcytosis. The majority of SIgA is pro- bionts to facilitate opsonization. In addition, penetrant microorganisms
duced following Toll-​like receptor (TLR) engagement on T cells and B cells are phagocytosed by Kupffer cells in the liver and splenic macrophages to
by microbial antigens and binds with low affinity to a broad fraction of control systemic dissemination.

Nature Reviews | Immunology


Reviews

Preventing symbiont accumulation in the mucosa. The to counter antimicrobial proteins produced by IECs.
protective mechanisms that promote the segregation For example, S. enterica can express genes involved in
of symbionts from the epithelium are not fool-​proof. lipopolysaccharide modifications that regulate resis­
Given the large numbers of microorganisms that reside tance to antimicrobial peptides as well as genes involved
near the epithelial surface, some bacteria can breach the in the sequestration, efflux and degradation of anti­
epithelial barrier under conditions of homeostasis36–38. microbial peptides52,53. Likewise, Listeria monocytogenes
To counter this situation, the host immune system has deacetylates N-​acetylglucosamine residues in peptido-
strategies to limit damaging inflammation in the mucosa glycan to evade the bacteriolytic activity of lysozyme,
and the dissemination of penetrant microorganisms to an enzyme produced by Paneth cells54. Unlike bacterial
systemic tissues (Fig. 1). These strategies include the symbionts, S. enterica produces virulence proteins to
killing of bacterial symbionts by specialized lamina evade lysosomal degradation in phagocytic cells55. Upon
propria macrophages that reside beneath the epithe- entry into enterocytes, S. flexneri ruptures the vacuolar
lium at high numbers, which engulf and kill penetrant membrane and escapes into the host cytosol, where it
microorganisms through several mechanisms, including evades autophagy-​mediated degradation, replicates and
the production of antimicrobial molecules and reactive spreads between IECs56. Within neutrophils, S. flexneri
oxygen species39. Under homeostatic conditions, these can inhibit reactive oxygen species-​mediated killing
resident macrophages are defective in microbial sens- by the expression of superoxide dismutases and cata-
ing and hence do not induce inflammatory responses, lases57. Thus, enteric pathogens have evolved multiple
a regulatory activity that is mediated through IL-10 mechanisms to evade mucosal firewalls. Perhaps more
stimulation40–42. Intestinal microorganisms can also be pertinent to IBD are the pathobionts that can trigger
engulfed by dendritic cells (DCs) in the lamina pro- disease in a susceptible host deficient for one or more
pria and transported to the mesenteric lymph nodes, components of the mucosal firewall.
where bacteria-​containing DCs induce protective IgA
and regulatory T (Treg) cells; these DCs do not reach sys- Breakdown of mucosal firewalls in IBD
temic secondary lymphoid structures, which limits their Breakdown of the homeostatic processes that reduce
systemic dissemination43. the contact between microorganisms and the epithelial
cell surface may increase susceptibility to IBD devel-
Control at systemic sites. Despite the presence of robust opment13,14. This notion is strengthened by the observ­
mucosal firewalls, rare intestinal microorganisms dis- ation that multiple IBD susceptibility genes encode
seminate systemically through the venous portal sys- proteins that function to limit the penetration of bacte-
tem or blood vessels (Fig. 1). These microorganisms rial symbionts into the mucosa or to promote bacterial
can be engulfed and killed by Kupffer cells in the liver killing (Fig. 2).
or macrophages in the spleen39,44. Symbiotic bacteria
can also induce homeostatic IgG responses through Segregating symbionts from the epithelial surface.
T cell-​dependent and T cell-​independent mechanisms. In mice, deficiency of MUC2 leads to abnormalities in
IgG antibodies can bind multiple symbionts, including the mucus layer that promote the close proximity
Proteobacteria and related pathogens, through the rec- of the microbiota to the intestinal epithelial surface and
ognition of highly conserved proteins, such as murein the development of spontaneous colitis58. The presence
lipoprotein, to limit the systemic dissemination of bac- of bacteria deep within crypts in close contact with the
teria from the intestine38. In addition, IgG2b and IgG3 epithelium is a common feature observed in patients
isotypes have similar reactivity to symbiotic species with ulcerative colitis58,59. Although aberrant expres-
that are bound by IgA and can be transferred to neo- sion of MUC2 has been described in patients with
nates in the breast milk; these maternal IgG isotypes, ulcerative colitis60,61, variants in mucin genes have not
together with IgA, bind the neonatal microbiota to limit been associated with IBD susceptibility. However, loss-​
aberrant commensal-​specific T cell-​mediated inflam- of-function variants in fucosyltransferase 2 (FUT2),
mation45. Systemic IgG responses to the microbiota are which encodes a protein that promotes mucosal barrier
enhanced in animals deficient in IgA or the MYD88 and function, are associated with increased susceptibility to
TIR domain-​containing adaptor-​inducing interferon-​β Crohn’s disease62,63.
(TRIF) signalling adaptors for TLR-​mediated sensing of Genetic variants of nucleotide-​binding oligomer-
bacteria46,47. This enhanced IgG response likely represents ization domain 2 (NOD2) were the first gene variants
a compensatory adaptation to the increased presence of to be associated with Crohn’s disease and remain the
symbionts in systemic tissues as the result of impaired strongest known genetic risk factors for disease devel-
containment and killing of penetrant symbiotic bacteria. opment64,65. NOD2 is an intracellular receptor that senses
peptidoglycan-​derived muramyl dipeptide and induces
Evasion by pathogens. Enteric pathogens have evolved immune responses against bacteria64. NOD2 is expressed
multiple strategies to disrupt and evade the mucosal in Paneth cells and may regulate their antimicrobial
firewalls that limit the penetration of bacterial sym- function66,67. However, Paneth cell ablation or deficiency
bionts into the mucosal tissues. For example, several in matrix metalloproteinase 7 (MMP7), which converts
pathogens, including Salmonella enterica, Shigella inactive pro-​α-defensins to bactericidal forms, does not
flexneri, Yersinia enterocolitica and Vibrio cholerae, lead to spontaneous inflammation in mice68. This sug-
produce mucin-​degrading enzymes48–51. In addition, gests that IBD pathogenesis requires additional genetic
enteric pathogens have evolved resistance mechanisms defects or the presence of particular pathobionts that are

www.nature.com/nri
Reviews

Small intestine Large intestine

Intestinal Mucus layer


lumen Intestinal
microbiota

Goblet cell

MUC2

Pathobiont
SIgA
M cell

Epithelium Macrophage AMPs,


FUT2
DC
AMPs
Peyer’s
patch
Impaired IL-6,
bacterial IL-1β,
Paneth recognition IL-23 Defective TH17 cell
cell • NOD2
IL-22 autophagy
Neutrophil • ATG16L1 TGFβ
IL-17 • IRGM
• NOD2
IL-10
Decreased secretion of ILC3 TH17 cell Defective
anti-microbial molecules bacterial killing
by Paneth cells Impaired ILC3-dependent • CYBB
• NOD2 and TH17 cell-dependent • CYBA TH1 cell Treg cell
• ATG16L1 immunity • NCF1
• XBP1 • TNFSF15 • NCF2
• LRRK2 • IL23R • NCF4 Defective
• RAC1 immunoregulation
• RAC2 • IL10R
Lamina propria

Fig. 2 | Breakdown of mucosal firewalls in inflammatory bowel disease. Mutations in susceptibility genes for
inflammatory bowel disease can impair the mucosal strategies that are used by the host to prevent harmful
microorganisms from gaining access to the intestinal lamina propria. In the small intestine, variants in nucleotide-​binding
oligomerization domain 2 (NOD2) and autophagy-​related genes (ATG16L1, IRGM, XBP1 and LRRK2) lead to decreased
secretion of Paneth cell-​derived antimicrobial peptides (AMPs). Polymorphisms of TNFSF15 and IL23R genes can affect
group 3 innate lymphoid cells (ILC3s) and T helper 17 (TH17) cells, which segregate bacterial symbionts from the mucosa
through the production of IL-22 and IL-17. Crohn’s disease-​associated loss-​of-function NOD2 mutations in phagocytes
such as macrophages and dendritic cells (DCs) impair bacterial recognition. Mutations in autophagy-​related genes
(ATG16L1, IRGM and NOD2) in intestinal epithelial cells may result in defective bacterial clearance. In the large intestine,
genetic variants in components or regulators of the phagosomal nicotinamide adenine dinucleotide phosphate (NADPH)
oxidase complex (CYBB, CYBA, NCF1, NCF2, NCF4, RAC1 and RAC2) impair the oxidative burst and production of reactive
oxygen species, leading to defective bacterial killing in phagocytes. Loss-​of-function mutations in IL10R alter intestinal
immune homeostasis, leading to dysregulated T cell responses. FUT2, fucosyltransferase 2; M, microfold; MUC2, mucin 2;
SIgA, secretory IgA; TGFβ, transforming growth factor-​β; Treg cell, regulatory T cell.

not found in the microbiota of the vast majority of mice autophagy pathways in IECs is associated with spontane-
housed under specific-​pathogen-free (SPF) conditions. ous Crohn’s disease-​like transmural ileitis71. Two Crohn’s
Mutations in the Crohn’s disease-​associated protein disease susceptibility genes, TNFSF15 and IL23R, regu-
autophagy-​related protein 16-like 1 (ATG16L1) may late ILC3s and TH17 cells, which have a crucial role in the
also contribute to ileal disease by impairing the exocy- containment of symbiotic microorganisms through
tosis of secretory granules within Paneth cells, an activ- the production of IL-17 and IL-22 (ref.72). However,
Unfolded protein response
(UPR). A group of intracellular ity that limits the penetration of bacterial symbionts69. further investigation is needed to understand how
signal transduction pathways Gene variants of the unfolded protein response (UPR) variants of these genes are linked to Crohn’s disease.
that facilitates the folding, transcription factor X-​box binding protein 1 (XBP1)
processing, export and have been associated with increased risk for Crohn’s Preventing symbiont accumulation in the mucosa.
degradation of proteins
derived from the endoplasmic
disease70. Specific epithelial deletion of XBP1 results in Killing of bacterial symbionts by specialized macro­
reticulum during stressed endoplasmic reticulum stress and structural defects phages in the lamina propria has a key role in limiting
conditions. in Paneth cells. Notably, impairment of both the UPR and damaging inflammation in the mucosa and preventing

Nature Reviews | Immunology


Reviews

Very early-​onset IBD


the dissemination of penetrant microorganisms. In the Studies using gnotobiotic mice have identified bacterial
A form of inflammatory bowel intestine, NOD2 is expressed by phagocytes, epithelial species that can induce an adaptive immune response
disease (IBD) diagnosed when cells and stromal cells as well as Paneth cells. Notably, during homeostasis. Perhaps the best studied example
symptoms manifest before the NOD2 deficiency together with deficiency of CYBB (also are the SFB, spore-​forming Gram-​p ositive bacteria
age of 6 years; it presents with
known as NOX2) — a component of the phagosomal related to Clostridium spp. that can adhere tightly to the
a severe disease course,
extensive colonic involvement, nicotinamide adenine dinucleotide phosphate (NADPH) epithelium of the terminal ileum in mice and promote
poor response to therapy and oxidase complex, which kills bacteria through the oxi- IL-17 expression by retinoic acid receptor-​related orphan
the frequent need for dative burst — triggers the bloom of M. schaedleri and receptor-​γt (RORγt+) CD4+ TH17 cells in the lamina pro-
abdominal surgery.
spontaneous Crohn’s disease-​like TH1 cell-​driven colitis pria86. The adhesion of SFB to intestinal epithelia trig-
in mice22. Because Crohn’s disease-​associated NOD2 var- gers the epithelial release of serum amyloid A, which
iants are loss-​of-function mutations, the resulting defec- conditions local DCs to secrete the TH17 cell-​promoting
tive bacterial sensing within phagocytic cells or intestinal cytokines IL-1β and IL-23; these cytokines also enhance
and/or stromal cells may promote luminal accumula- the production of IL-22 by gut-​resident ILC3s, which
tion and mucosal penetration of pathobionts, lead- reinforces the expression of serum amyloid A by epi-
ing to T cell-​mediated intestinal inflammation (Fig. 2). thelial cells87,88 (Fig. 3). Consequently, SFB colonization
However, whether NOD2-associated Crohn’s disease confers host protection against extracellular pathogens
is caused by the accumulation of specific pathobionts but is also linked with autoimmune disorders86,89. In
in humans requires further investigation. SFB-​colonized animals, the majority of lamina propria
NOD2 also has a role in autophagy, a pathway that TH17 cells express T cell receptors (TCRs) that recognize
mediates lysosomal degradation and clearance of intra- SFB antigens90.
cellular bacteria73. NOD2 recruits the Crohn’s disease-​ In addition to epithelial adherence, the ability of
associated protein ATG16L1 to the plasma membrane at symbiotic bacteria to reside in the mucus layer near the
sites of bacterial entry74, but Crohn’s disease-​associated intestinal epithelium may also be an important behav-
NOD2 variants have defective recruitment of ATG16L1 ioural feature to influence host immunity. For example,
and impaired bacteria-​induced intraepithelial auto- intestinal colonization by the mucus-​dwelling com-
phagy75. In addition to ATG16L1, immunity-​related mensal Akkermansia muciniphila can spontaneously
GTPase family M (IRGM) and leucine-​rich repeat kinase induce an adaptive immune response during homeo-
2 (LRRK2) also regulate the autophagy pathway and var- stasis in gnotobiotic mice with a simplified microbiota.
iants of these genes have been associated with Crohn’s A. muciniphila-​specific T cells primarily differentiate
disease risk76,77. As defects in autophagy impair the clear- into T follicular helper cells to mediate an IgG1 response
ance of intracellular bacteria78,79, it is conceivable that against the bacteria but the same TCR-​transgenic CD4+
mutations in autophagy-​associated genes could result T cells can adopt multiple cell fates under SPF condi-
in pathobiont penetration and inflammation in the gut. tions, which indicates that a complex microbiome can
A clear example of the link between bacterial killing affect the localization or behaviour of this symbiont91
and susceptibility to IBD is provided in patients with (Fig. 3). Similarly, M. schaedleri, another mucus-​dwelling
very early-​onset IBD80,81, which is associated with genetic symbiont, triggers specific IgG and IgA responses that
variants in components and regulators of the phagocyte protect neonates from colitis through antibody delivery
NADPH oxidase complex82,83. Similarly, up to 40% of to the intestinal lumen during breastfeeding22.
patients with chronic granulomatous disease, a primary
immunodeficiency disorder caused by loss-​of-function Limiting inappropriate inflammation. Treg cells orig-
mutations in the NADPH oxidase components, develop inate from two distinct ontogenic lineages: thymus-
Crohn’s disease-​like colitis84,85. Collectively, these obser-
derived Treg (tTreg) cells and peripherally derived Treg
vations suggest that defects in the killing of bacterial (pTreg) cells (Fig. 4a). Most pTreg cells develop in the colon
symbionts promote the development of Crohn’s disease. extrathymically in the presence of symbiotic bacteria
as the frequency of pTreg cells in the colon is markedly
Beneficial effects of symbionts decreased in germ-free mice92–94. Treg cells have a crucial
Symbiotic bacteria promote protective immunity by role in the preservation of tissue homeo­stasis by prevent-
reinforcing the mucosal firewalls that limit the pene- ing the induction of inappropriate T cell responses to
tration of symbionts while controlling aberrant T cell microbial antigens (Fig. 4a). For example, mice lacking
responses to microorganisms. Disruption of these Treg cells or factors that are important for their regula-
mutualistic host–symbiont interactions may promote tion or function, including IL-10, transforming growth
inappropriate immune responses against the dysbiotic factor-β (TGFβ) and αVβ8 integrin, develop spontane-
microbiota in patients with IBD. ous colitis, although the precise phenotype depends on
the animal facility95–98. Consistent with the animal data,
Inducing protective immune responses. The enteric young children with mutations in the IL-10 receptor
microbiota has a crucial role in modulating host immu- develop colonic Crohn’s disease99. Furthermore, mice and
nity to establish and maintain intestinal homeostasis. humans with mutations in forkhead box P3 (FOXP3),
Germ-​free animals have several impairments in these the transcription factor required for Treg cell develop-
processes, including defective formation of mucosa-​ ment, have autoimmune disorders, including colitis100,101.
associated lymphoid tissues, incomplete TH17 cell and A role for Treg cells in suppressing immune responses
Treg cell development, and reduced numbers of intraep- to sym­biotic bacteria is also supported by the observ­
ithelial CD8αβ T cells and IgA-​producing plasma cells5. ation that microbiota-​dependent colitis induced by the

www.nature.com/nri
Reviews

Small intestine Large intestine

Intestinal
microbiota

Akkermansia Symbionts
Symbionts muciniphila

SFB SIgA
AMPs

Intestinal IL-17,
epithelium IL-22
IgG
IL-1β, Neutrophil Plasma
IL-23 TFH cell
cell
TFH cell TLR
SAA
ILC3 TH17 cell
DC
Lamina propria

Fig. 3 | Beneficial effects of symbionts. Adhesion of segmented filamentous bacteria (SFB) to the intestinal epithelium
triggers the release of serum amyloid A (SAA), which acts on dendritic cells (DCs) of the lamina propria to stimulate the
secretion of cytokines, including IL-1β and IL-23, and induce retinoic acid receptor-​related orphan receptor-​γt (RORγt)+
T helper 17 (TH17) cell differentiation and group 3 innate lymphoid cell (ILC3) activation. IL-22 secretion by ILC3s
reinforces SAA production by the epithelium to augment TH17 cell-​mediated mucosal defences, including antimicrobial
peptide (AMP) secretion and neutrophil recruitment. Symbionts also promote intestinal homeostasis through IgA
responses induced by B cell-intrinsic and T cell-intrinsic microbial sensing by Toll-​like receptors (TLRs). Secreted IgA (SIgA)
contributes to barrier function by binding to intestinal microorganisms and preventing epithelial translocation. Similarly,
IgG production can occur directly by TLR engagement on B cells, and intestinal colonization by Akkermansia muciniphila
can induce homeostatic IgG production through antigen-specific T follicular helper (TFH) cell responses. In addition, IgG
contributes to the systemic control of pathobionts that penetrate the epithelial barrier (not shown).

transfer of CD45RBhiCD4+ T cells into lymphopenic IL-10-producing FOXP3+ pTreg cells and the B. fragilis-​
mice is ablated by the co-​transfer of Treg cells102. Although derived polysaccharide A is sufficient to reverse disease
the selective depletion of pTreg cells by deletion of the in experimental colitis105.
Foxp3 enhancer conserved non-​coding DNA sequence 1 Bacterial symbionts can promote the differentiation
(CNS1) in mice does not lead to the spontaneous multi­ of intestinal Treg cells by inducing the local production of
organ autoimmunity observed in complete Foxp3- TGFβ by epithelial cells and of retinoic acid by CD103+
null scurfy mice, type 2 immune pathologies can be DCs106–108 (Fig. 4b). Secretion of IL-10 and retinoic acid
observed in the gastrointestinal tract103. This immuno­ by DCs to support Treg cell proliferative expansion is
pathology of the mucosal barrier in pTreg cell-deficient further reinforced by granulocyte–macrophage colony-​
animals suggests that tTreg cells are sufficient to maintain stimulating factor (GM-​CSF) produced by ILC3s109.
tolerance to self-​antigens systemically, whereas pTreg cells Although different bacterial species can modulate
have a non-​redundant role in restraining inflammation intestinal Treg cell activity, the specificity of intestinal
in the intestine103. Treg cells for microbial antigens in the context of a com-
Colonic pTreg cells are profoundly influenced by local plex microbiota remains unclear. Bacteria-​derived short-​
antigens as their TCR repertoire is distinct from analo- chain fatty acids (SCFAs) produced by the fermentation
gous Treg cells of the peripheral lymph nodes and spleen of dietary sugars can restore the Treg cell population in
and can recognize antigens expressed by Clostridium and germ-​free animals and protect against T cell-​mediated
Parabacteroides spp., which further supports that bac- colitis, although the mechanism remains unclear110–112.
teria are important for intestinal Treg cell induction93 SCFAs inhibit histone deacetylase silencing of Foxp3
(Fig. 4b). A limited consortium of 17 human-derived gene expression, thus enhancing Treg cell differentiation
Clostridium spp., which can colonize the colonic mucus through epigenetic modifications111,112. A population of
layer near the epithelium, can induce Treg cells in germ-​ intestinal microbiota-​inducible RORγt+ Treg cells seems
free mice and can suppress intestinal inflammation to be crucial for suppressing gut immunopathology
when administered to mice with colitis104. Colonization mediated by TH1 cells and TH17 cells or by type 2 aller-
with the symbiont Bacteroides fragilis in germ-free gic responses113,114. In the T cell-​transfer model of coli-
animals can similarly induce the differentiation of tis, germ-​free mice colonized with an IBD-​associated

Nature Reviews | Immunology


Reviews

a b
Symbionts Bacteroides
fragilis (PSA)

Helicobacter
hepaticus

Clostridium spp.

SCFAs

TLR2
Macrophage
IL-10, IL-10, GM-CSF IL-10
TGFβ TGFβ
TH1 TH17
cell cell IL-1β IL-10,
Naive pTreg cell tTreg cell ILC3 TGFβ, Naive pTreg cell
T cell RA T cell

DC

CTLA4-mediated and/or Local symbiont antigens


TCR-mediated inhibition

Fig. 4 | Regulatory T cells support intestinal homeostasis. a | Peripherally derived regulatory T (pTreg) cells and thymus-​
derived Treg (tTreg) cells restrain aberrant inflammation in the intestine. Treg cells produce IL-10 and transforming growth
factor-​β (TGFβ) to suppress effector T helper 1 (TH1) cells and TH17 cells. In addition, myeloid cells such as macrophages
are an important target for IL-10-driven intestinal homeostasis. Intestinal Treg cells also restrain effector T cell responses
by the inhibition of antigen-​presenting cells such as dendritic cells (DCs) through cytotoxic T lymphocyte antigen 4
(CTLA4)-mediated or T cell receptor (TCR)-mediated processes. Self-​reactive tTreg cells may also contribute to antigen-​
specific immunosuppression through TCR cross-​reactivity with microbial antigens. b | Symbionts have a crucial role in the
peripheral education of pTreg cells through antigen-​dependent and antigen-​independent processes. Macrophage sensing
of bacteria leads to the secretion of granulocyte–macrophage colony-​stimulating factor (GM-​CSF) by group 3 innate
lymphoid cells (ILC3s) stimulated by IL-1β, which in turn enhances DC expression of retinoic acid (RA) and IL-10 to
support pTreg cell differentiation and proliferation in the colon. Moreover, symbionts can promote the expression of
TGFβ by epithelial cells and DCs in addition to RA and IL-10. Bacteroides fragilis can enhance Treg cell activity through
polysaccharide A (PSA) binding to Toll-​like receptor 2 (TLR2) expressed by Treg cells and DCs. Select bacterial species can
directly induce pTreg cell differentiation; these pTreg cells have TCR specificity for antigens expressed by Clostridium spp.
and Helicobacter spp., which indicates a role for symbiotic antigens in supporting Treg cell differentiation. Short-​chain fatty
acids (SCFAs) are fermentation by-​products produced by symbiotic bacteria, including Clostridium spp., that can promote
pTreg cell population expansion directly by epigenetic modification or through DCs via enhanced RA production.

microbiota have a selective bias towards the expansion of In addition, decreased numbers of SCFA-​producing
TH17 cell populations at the expense of RORγt+ Treg cells, micro­organisms, such as Clostridium spp., are observed
which accounts for disease severity and is predictive of in patients with IBD117. Microbiota-​derived SCFAs, in
human disease status115. particular butyrate, promote the development of Treg cells
and mucus production to downregulate inflammatory
Disruption of beneficial symbionts in IBD signalling pathways and to strengthen the epithelial
Genetic defects in patients with IBD often result in an barrier112,118. Thus, the loss of SCFA-​producing micro­
altered composition of the microbiota, further com- organisms may have detrimental effects in IBD. Another
promising the beneficial effects that some microorgan- example of disruption of the beneficial effects of sym­
isms exert on host immunity. A well-​described example bionts is the reduction of tryptophan metabolism, which
of a beneficial microbial species that is decreased in is a feature of the IBD microbiome119,120. The tryptophan
patients with ileal Crohn’s disease is Faecalibacterium metabolite indoleacrylic acid, which is produced by sev-
prausnitzii, a member of the Firmicutes phylum that eral Peptostreptococcus spp., promotes mucosal barrier
secretes anti-​inflammatory metabolites. A decreased function and reduces inflammatory responses119. The
abundance of F. prausnitzii in the ileum is associated symbiont Peptostreptococcus russellii increases goblet cell
with increased risk of postoperative recurrence of ileal numbers and mucin fucosylation, and mediates protec-
Crohn’s disease and endoscopic recurrence at 6 months116. tion from dextran sodium sulfate (DSS)-induced colitis119.

www.nature.com/nri
Reviews

Faecal stream diversion


Thus, the microbial imbalance associated with IBD may Dysbiosis in patients with IBD. The composition of the
A diverting terminal ileostomy contribute to disruption of the regulatory mechanisms microbiota in patients with IBD has been extensively
constructed proximally to an that suppress inflammation. studied132. 16S ribosomal RNA gene sequencing analysis of
ileocolonic anastomosis, stool and mucosal samples has revealed the presence of a
thereby excluding the
Breakdown of symbiosis in IBD dysbiotic microbiota, characterized by decreased com-
neoterminal ileum, the
anastomosis and the colon A large body of evidence suggests that the microbiota munity diversity and a shift in bacterial taxa, including
from the faecal intestinal has a crucial role in triggering IBD. Furthermore, genetic alterations in certain genera of the phylum Firmicutes
transit. defects linked to IBD together with environmental fac- and increased abundance of Enterobacteriaceae spe-
tors can induce the accumulation and penetration of cies117,133; these alterations are typically more pronounced
Tbx21−/−Rag2−/− mice
Immunocompromised mice
pathobionts into the intestinal tissue, which further in patients with Crohn’s disease than in patients with
that have no adaptive immune promote dysbiosis and inflammation. ulcerative colitis134. Some studies also showed changes
cells and only innate immune in Bacteroides spp., particularly in patients with Crohn’s
cells with a deficiency in T-​box The microbiota as a trigger of IBD. Although no path- disease135. Differences in the microbiota are also present
transcription factor 21
ogen or pathobiont has been consistently identified between patients with IBD and close relatives, includ-
(TBX21).
as being responsible for IBD, multiple lines of evi- ing twins136,137, which suggests that dysbiosis correlates
16S ribosomal RNA gene dence support an important role for the microbiota in with disease rather than with genetic factors. However,
sequencing driving intestinal inflammation2. For example, faecal longitudinal studies are needed to determine whether
A DNA amplification
stream diversion decreases or abolishes the inflammation dysbiosis precedes the onset of inflammation. The
technology for the study of a
gene conserved among
of ileal Crohn’s disease121. Furthermore, recurrence of imbalance in the microbial community towards a dys-
bacteria that is used for Crohn’s disease in the neoterminal ileum after curative biotic state is more pronounced in mucosal samples
species identification and ileal resection depends on exposure to luminal con- than in faecal samples collected at the time of diagnosis
taxonomic classification of tents122. Antibiotic treatment can induce remission in in untreated patients with Crohn’s disease138. Studies of
bacteria.
patients with active ulcerative colitis and Crohn’s disease mucosal samples also revealed a greater net loss of poten-
as well as prevent relapse in some patients with quiescent tially beneficial bacteria in Crohn’s disease compared
Crohn’s disease123,124. However, the interpretation of anti- with ulcerative colitis, the correlation of certain taxa,
biotic studies is difficult given the effects of these drugs including Enterobacteriaceae in Crohn’s disease and
on multiple bacterial species. Ruminococcus spp. in ulcerative colitis, with disease
Animal models further support a role for the gut activity, and an association of certain taxa with response
microbiota in the pathogenesis of IBD. For example, to TNF therapy in Crohn’s disease134. Metagenomic
oral transfer of the faecal microbiota from mice with sequencing of stool samples has shown that there is a
colitis into healthy animals is sufficient to trigger dis- marked separation between patients with Crohn’s dis-
ease125–127. Most importantly, genetically susceptible mice ease and those with non-​IBD, whereas patients with
develop colitis with a conventional microbiota but not ulcerative colitis were more heterogeneous with a gen-
under germ-​free conditions. For example, SPF mice, but eral trend towards loss of species diversity139. These stud-
not germ-​free mice, with null mutations in TCR genes ies also found a loss of metabolite diversity in the faeces
develop colitis128. Furthermore, colitis was not observed of patients with IBD that is comparable to the loss of
in germ-​f ree TCR-​deficient mice colonized with a species diversity139. Although metagenomic studies have
defined commensal community128, which suggests that been limited so far to the analysis of stool samples, owing
specific microorganisms are required to trigger disease. to the high levels of host-​derived DNA that are present
Similarly, the intestinal inflammation that develops in in mucosal biopsy samples, they have provided insight
Il2–/– mice, Il10–/– mice and rats transgenic for HLA-​B27 into the functional disruption of the microbiota in IBD.
and human β2-microglobulin is largely abrogated under Whereas most microbiome studies in IBD have
germ-​f ree conditions95,96,129,130. In addition, Tbx21 –/– focused on the bacterial contribution to disease patho-
Rag2–/– mice develop ulcerative colitis-​like spontaneous genesis, a few studies highlight the importance of other
inflammation that is markedly inhibited by antibiotic microorganisms in IBD. For example, there is increased
treatment125. In models of Crohn’s disease-​like ileitis, fungal diversity in colonic biopsy samples from patients
TNF∆ARE mice, which have a deletion in the tumour with Crohn’s disease compared with healthy individ­
necrosis factor (TNF) AU-​rich elements (ARE), develop uals140. Similarly, increased fungal diversity and increased
TNF-​driven transmural inflammation in the presence of prevalence of Candida albicans, Aspergillus clavatus and
the microbiota126, and SAMP1/Yit mice, a recombinant-​ Cryptococcus neoformans were observed in ileal mucosal
inbred line, develop ileitis that is attenuated under specimens and stool samples from patients with Crohn’s
germ-​free conditions131. Although studies of germ-​free disease141. Additional evidence has confirmed the pres-
animals suggest a crucial role for the microbiota in ence of fungal dysbiosis in IBD, including a decreased
IBD, the interpretation of these experiments is difficult proportion of Saccharomyces cerevisiae and an increase
given that multiple deficiencies are present in germ-​free of C. albicans compared with healthy subjects142. Notably,
mice. Studies with Nod2–/–Cybb–/– mice showed that they some studies correlated increased diversity of the fungal
develop Crohn’s disease-​like colitis when harbouring microbiota with disease severity and suggested that the
the microbiota of mice from Taconic Biosciences but Crohn’s disease environment favours fungi at the expense
not the microbiota of mice from the Jackson Laboratory22, of bacteria or that antibiotic treatment creates specific
which indicates that colitis requires the presence of niches for fungal expansion141–143. Intestinal fungi inter-
particular pathobionts to trigger disease in genetically act with the host immune receptor dectin 1, which sig-
susceptible mice. nals through the adaptor protein caspase recruitment

Nature Reviews | Immunology


Reviews

Adherent and invasive


domain-containing protein 9 (CARD9) to induce the susceptibility genes (Fig. 5). During late dysbiosis, intes-
Escherichia coli production of inflammatory molecules and TH17 cell tinal inflammation drives further changes in bacterial
E. coli strains that can adhere responses 144. CARD9 variants are associated with taxa, including the expansion of Proteobacteria popu-
to and invade intestinal increa­sed risk of developing IBD145. Similarly, Card9- lations (Fig. 5). Given that distinct members of the gut
epithelial cells.
null mice have altered intestinal fungal communities microbiota have beneficial effects on the host immune
and increased susceptibility to chemically induced system and the intestinal barrier119,154, depletion of cer-
colitis146. Furthermore, a polymorphism in the gene tain taxa may contribute to the exacerbation or persis-
encoding dectin 1 has been linked to medically refractory tence of intestinal inflammation. Blooms of particular
ulcerative colitis144. bacteria, such as adherent and invasive Escherichia coli that
The intestine also harbours a large community of accumulate in the inflamed mucosa of patients with IBD,
viruses that may have a role in IBD pathogenesis. For could further promote inflammation (Fig. 5). Adherent
example, infection with murine norovirus induces and invasive E. coli adhere to intestinal epithelia using
Paneth cell abnormalities in the presence of a mutation the common type 1 pili adhesin FimH, which recog-
of ATG16L1, a Crohn’s disease susceptibility gene147. nizes the carcinoembryonic antigen-​related cell adhesion
Furthermore, genetic variation in FUT2 has been impli- molecule 6 (CEACAM6) that is abnormally expressed
cated in susceptibility to both norovirus infection and in the ileum of patients with Crohn’s disease155. Well-​
Crohn’s disease62. Other viruses that may be relevant to designed longitudinal studies of mucosal samples from
IBD are bacteriophages. Metagenomic sequencing of the humans with increased IBD susceptibility and/or patients
enteric virome in patients with IBD revealed an expan- with IBD before clinical relapse are needed to understand
sion of Caudovirales bacteriophages compared with the role of dysbiosis in disease.
control individuals, which did not seem to be second-
ary to changes in bacterial populations148; however, this Adaptive immune responses to microbial antigens in
expansion was cohort specific and not confirmed across IBD. Increased T cell and antibody responses to micro-
validation cohorts148. Thus, more studies are needed to bial antigens are often detected in patients with IBD,
understand the role of fungi and viruses in human IBD. providing further evidence for a role of the microbiota
in disease pathogenesis. Patients with IBD produce large
Cause or consequence of IBD? Although alterations in amounts of IgG antibodies against symbiotic bacteria156
the microbiota can occur early in IBD and independently as well as having high serum reactivity against fungal
of therapy, there is no direct evidence for a causal role of and/or bacterial peptides and glycans compared with
dysbiosis in IBD pathogenesis. In mice, chemically indu­ healthy individuals157. Although antimicrobial antibod-
ced colitis and intestinal infection trigger robust changes ies can be used as markers of disease and diagnosis, the
in the composition of the microbiota, some of which role of such antibodies in IBD remains unclear. Given
are comparable to those observed in patients with IBD, that healthy mice and humans develop antibodies against
including expansion of Proteobacteria populations149,150. bacterial symbionts36,38,158, the presence of high titres of
Inflammation-induced increased oxygenation of the IBD-​associated antibodies may reflect enhanced adaptive
intestinal lumen and increased availability of nitrate immune responses or increased exposure to microbial
and host-derived electron acceptors can drive anaerobic antigens in the inflamed gut of patients with IBD.
respiration and blooming of Enterobacteriaceae149,151. In addition to humoral responses, dysregulated T cell
Thus, many of the microbiota alterations that are responses against the microbiota are observed in IBD.
observed in the intestinal mucosa and lumen of patients Early work showed that T cells or mononuclear cells from
with IBD are likely to be secondary to inflammation. the inflamed mucosa of patients with IBD have increased
In a genetic model of spontaneous Crohn’s disease, reactivity compared with cells from normal mucosa after
longitudinal analyses have revealed the population expan- stimulation with gut microbial antigens159,160. However,
sion of a single or limited number of pathobionts before CD4+ T cells reactive against bacterial symbionts can be
the onset of colitis and this dysbiotic microbiota is suffi- detected in the blood and mucosa of both patients with
cient to trigger disease in mice with a complex microbi- IBD and healthy individuals161,162. Microbiota-​responsive
ota22. Based on these observations, dysbiosis may proceed human T cells are mainly of a memory phenotype and
in two sequential stages. At an early stage, IBD-​associated have a diverse TCR Vβ repertoire, which is consistent
genetic and environmental factors may lead to the accu- with reactivity against a wide array of microbial anti-
mulation of disease-​causing pathobionts, which may gens162. In agreement with human data, T cells reactive
precede the development of clinical disease. Although against symbiotic bacteria and bacterial antigens can
the identity and number of pathobionts involved in also be detected in animal models of colitis163. Transfer
early dysbiosis are unknown, limited evidence in animal of CD4+ T cells reactive against commensal antigens into
models of IBD suggests that the genetic and metabolic lymphopenic animals is sufficient to trigger colitis164–166.
features of the bacteria may be important. For example, Similarly, monocolonization of Il10–/– germ-​free mice
M. schaedleri and Helicobacter hepaticus, two pathobionts with some commensal bacteria, including E. coli and
that can trigger spontaneous colitis in genetically suscep- Enterococcus faecalis or the pathobiont H. hepaticus,
tible mice, produce virulence factors and live near the elicits colitis167,168. Interestingly, H. hepaticus-​colonized
epithelium, although the role of these activities in induc- immunocompetent animals are healthy because the CD4+
ing colitis remains unclear22,152,153. Such bacterial features TH cells that express H. hepaticus-​specific TCRs predom-
may lower the threshold for local penetration when the inantly differentiate into RORγt+ pTreg cells and T folli-
mucosal firewalls are compromised by mutations in IBD cular helper cells169,170. However, under IL-10-deficient

www.nature.com/nri
Reviews

a Early dysbiosis b Late dysbiosis

Genetic factors Environmental Inflammation


factors

Decreased Loss of Pathobiont


microbial beneficial expansion
diversity symbionts

Decreased abundance Expansion of


of Firmicutes Proteobacteria

Firmicute
O2– + NO NO3– NO2–
Pathobiont
accumulation
and penetration Iron O2

SCFAs
O2

Neutrophil
TH1 cell
Pathobiont
Monocyte penetration
TH17 cell
DC Treg cell

Macrophage

Early inflammatory response Chronic inflammation

Fig. 5 | Dysbiosis in inflammatory bowel disease. The inflammatory bowel disease-​associated genetic defects, together
with environmental factors such as diet and antibiotic use, can lead to the accumulation and penetration of disease-​
causing pathobionts into the intestinal lamina propria (early dysbiosis), which may precede the development of clinically
overt disease. Inflammation may result in larger alterations of bacterial taxa, including the expansion of Proteobacteria
(late dysbiosis), through enhanced luminal oxygenation and increased availability of nitrate (NO3−), host-​derived oxygen
acceptors and iron in the inflamed gut environment. This late stage of dysbiosis is also characterized by an overall
decrease in microbial diversity with a loss of beneficial symbionts, which may result in increased mucosal adherence and
translocation of symbiotic microorganisms, thus triggering chronic inflammation. DC, dendritic cell; TH cell, T helper cell;
Treg cell, regulatory T cell; SCFAs, short-​chain fatty acids.

conditions, Helicobacter spp.-specific CD4+ T cells differ- Targeting the microbiota for therapy
entiate into pathogenic TH17 and TH1 cells169,170. Notably, Most current therapies for IBD, including steroids and
antigens expressed by symbionts that have limited access biologicals such as anti-​TNF or anti-​integrin therapies,
to the host are not sufficient to initiate T cell activation suppress the host immune system but do not directly
and colitis; these include Bacteroides spp., which prefer- target the microorganisms that cause or contribute to
entially reside in the intestinal lumen, and CBir1 flagellin-​ inflammation. Given that IBD therapies induce com-
expressing bacteria, which require a severe perturbation plete remission in less than 50% of patients, the devel-
in the mucosal barrier or selective impairment of IgA to opment of therapeutic approaches that target intestinal
trigger an antigen-​specific T cell response170–173. These microorganisms may provide a unique approach to treat
observations suggest that pathogenic CD4+ TH cells IBD. Our increased understanding of dysbiosis and the
recog­nizing pathobionts can develop under homeostatic immunomodulatory functions of select microorganisms
conditions and that the breakdown of Treg cell-​mediated has presented several innovative strategies for modify-
immunosuppression to these pathobionts is crucial for ing the intestinal microbiota to prevent or ameliorate
colitis development. However, the mechanism by which IBD (Fig. 6).
Treg cells act on effector T cells to inhibit bacteria-induced
inflammation in the gut remains unclear. Treg cells could Faecal microbiota transplantation. Features of dys­
inhibit effector T cells in an antigen-independent man- biosis in IBD and recurrent Clostridium difficile infec-
ner174 and/or act on antigen-presenting cells through tion include the loss of phylum-​level diversity and
their TCRs to suppress antigen-​specific effector T cells175. the overgrowth of facultative anaerobic bacteria of the

Nature Reviews | Immunology


Reviews

Faecal microbiota
Enterobacteriaceae family, including Proteobacteria5. efficacy are limited in cohort size and lack a placebo
transplantation Therefore, approaches aimed at correcting imbal- treatment group180,181. Further studies are needed to
(FMT). The transfer of the ances in the microbial community may be effective understand the mechanism by which FMT is effective
intestinal microbial community for the treatment of IBD. The high success rate of in some patients with ulcerative colitis and to develop
from a healthy individual to a
faecal microbiota transplantation (FMT) in resolving more effective protocols to treat IBD.
patient through infusion of
stool, typically by endoscopy. recurrent C. difficile infection by restoring gut micro-
bial community diversity has reinforced the potential Antibiotics and probiotics. Antibiotic usage in the treat-
of microbiota-​modulating therapies176. Although the ment of IBD remains limited as a result of conflicting
number of patients with IBD who have been treated findings but studies using broad-​spectrum antibiotic
with FMT is limited and the responses to treatment cocktails have shown that they may be efficacious in
have been variable, induction of clinical remission has cases of severe acute colitis and chronic ulcerative coli-
been achieved in about 30% of patients with ulcerative tis182. Meta-​analyses of randomized controlled trials also
colitis177,178. Pretreatment with antibiotics and the use indicate the benefits of antibiotics in the treatment of
of multiple faecal infusions seem to improve the effec- Crohn’s disease and ulcerative colitis183,184. However, the
tiveness of FMT in patients with ulcerative colitis177,178. use of antibiotics to modify the microbiota is limited
However, one double-​blinded, randomized, placebo-​ by the inability to selectively eliminate disease-​causing
controlled study did not observe any beneficial effect of bacteria without potentially affecting beneficial micro-
FMT in patients with ulcerative colitis179. In addition, organisms, which could lead to unexpected adverse
the effectiveness of FMT in patients with Crohn’s disease outcomes. In the future, once the metabolic pathways
remains unclear as current studies reporting potential of IBD-​causing microorganisms have been identified,

FMT Antibiotics Probiotics Prebiotics Drugs Synthetic Diet


microorganisms
Gut
lumen
Direct Direct Indirect Direct Direct Indirect Direct
• Metabolites Pathobiont Population Population Target Host Shapes
• Bacteriophages killing expansion of expansion of metabolic immune microbiota
• Beneficial beneficial Direct beneficial pathways modulation composition
microorganisms microorganisms Microcins microorgansims

Indirect Symbionts
Host Indirect
immune Host
stimulation immune
modulation
Pathobionts SCFAs
Mucus Indirect
layer MAMPs Bacterial
competition

Epithelial
barrier

TLR

IL-10
Macrophage Naive Treg cell
Lamina T cell
propria

TH17 cell
DC

Fig. 6 | Effects of microbiome-​based therapeutics in inflammatory bowel of microcins, small polypeptides with antimicrobial activity that directly
disease. Given the crucial role of the gut microbiota in the pathogenesis of suppress closely related Enterobacteriaceae. Supplementation with
inflammatory bowel disease, the development of therapeutic approaches prebiotics can foster the population expansion of beneficial symbionts that
that target intestinal microorganisms may provide a unique approach to in turn outcompete harmful bacteria. Targeting of metabolic pathways used
treat the disease. Faecal microbiota transplantation (FMT) may have direct by bacteria to promote intestinal inflammation may provide a new strategy
beneficial effects through the transfer of bacteria-​derived metabolites and to treat inflammatory bowel disease. Administration of synthetically
bacteriophages or by restoring beneficial microorganisms, or indirect engineered microorganisms may have beneficial effects on the host through
beneficial effects through host innate immune stimulation by bacterial modulation of the immune system, for example, by secretion of IL-10. Diet
components (for example, Toll-​like receptor (TLR) triggering by shapes the composition of the gut microbiota by promoting the growth of
microorganism-​associated molecular patterns (MAMPs)). Antibiotics may beneficial microorganisms or the depletion of pathobionts. Short-​chain fatty
have broad effects by directly killing pathobionts or indirectly promoting the acids (SCFAs) derived from dietary polysaccharides can also support the
population expansion of beneficial microorganisms. Administration of differentiation and expansion of intestinal regulatory T (Treg) cell populations.
probiotics, such as Escherichia coli Nissle 1917, can result in the secretion DC, dendritic cell; TH17 cell, T helper 17 cell.

www.nature.com/nri
Reviews

it may be possible to specifically target them with a new and polymeric formula diet that contains no solid food,
generation of antibiotics. is one of the few dietary interventions that has been
Alternatively, administering live microorganisms extensively studied in IBD197. EEN is as effective as cor-
may be advantageous in several ways. Once stable col- ticosteroids in inducing remission in paediatric patients
onization is achieved, beneficial microbial factors can with Crohn’s disease, but without the side effects that
be delivered continuously to the host and live bacteria are associated with corticosteroid therapy198–200. EEN
may trigger beneficial immune responses. The targeted rapidly alters the microbiota composition independently
modulation of the microbiota by administering probi- of other environmental factors and effectively reduces
otics, including Lactobacillus spp. or Bifidobacterium intestinal inflammation in paediatric patients with
spp., has been successful for some bowel disorders185. In Crohn’s disease 201. The mechanism by which EEN
addition, oral treatment with the probiotic E. coli Nissle induces remission in Crohn’s disease is unclear but it
1917 has similar efficacy to the standard treatment with may promote the growth of beneficial microorganisms
mesalazine in terms of maintaining disease remission or the depletion of pathobionts. In experimental DSS-​
in patients with ulcerative colitis186. Probiotic E. coli induced colitis, a survey of various refined diets identi-
Nissle 1917 can secrete microcins with antimicrobial fied the beneficial effects of psyllium fibre, whereas an
activity to suppress competing Enterobacteriaceae that increase in dietary proteins, including casein, increased
can potentially exacerbate intestinal inflammation187. faecal microbial density and exacerbated colitis sever-
However, probiotics typically have limited effects on the ity through alterations in intestinal permeability202.
overall composition of the microbiome as these bacte- Importantly, host susceptibility to colitis is highly
rial species are unable to persistently colonize healthy dependent on combinations of specific fibre or protein
adults188. Concurrent supplementation with prebiot- components that shape the microbiota, which indicates
ics can improve the engraftment efficiency of exoge- the importance of dietary composition in IBD202.
nous microorganisms in some indications, including
enhanced colonization resistance against C. difficile189. Future directions
Advances in synthetic biology have provided inno- In the past decade, there has been an enormous increase
vative approaches for the treatment and management in knowledge regarding microbiota–host interactions
of IBD (Fig. 6). Oral administration of Lactococcus lactis as well as the role of genetics and the immune system
engineered to express and secrete IL-10 was sufficient in IBD. However, several aspects of our understanding
to protect mice from colitis induced by DSS and IL-10 of IBD pathogenesis remain unclear.
deficiency190. In a small cohort of patients with Crohn’s A major deficiency is the lack of knowledge about the
disease, the recombinant IL-10-producing L. lactis strain identity of IBD-​causing pathobionts that trigger inflam-
was safely administered using a thymine-​limiting con- mation in genetically susceptible individuals. Multiple
tainment strategy but it induced only a small improve- species of pathobionts may cause disease and these may
ment in disease activity191. Owing to the inert properties vary according to the specific IBD susceptibility loci
of L. lactis, this probiotic has also been modified to of a patient. Therefore, studies using individuals with
express insulin growth factor 1, haem oxygenase 1 or identical or similar genetic defects may be important to
serine protease inhibitors; when orally administered to identify such microorganisms.
mice, these synthetic probiotics can alleviate the devel- There is a large intra-​species genetic diversity in the
opment of experimental colitis192–194. However, such symbionts that live in the mammalian intestine. Carefully
strategies were not successful in clinical trials, perhaps designed longitudinal studies coupled to metagenomic
owing to the inability of the engineered L. lactis strains analyses of mucosal samples and to new methodologies
to persistently colonize patients with IBD195. that can reveal intra-​species variation may be impor-
tant to understand the role of dysbiosis in disease. The
Targeting microbial metabolism. Precise targeting identification of IBD-​causing microorganisms will be
of the metabolic pathways that are used by harmful important for understanding disease pathogenesis, mon-
microorganisms to proliferate during dysbiosis has itoring patients for alterations of these pathobionts and
been effective in ameliorating colitis in mice. For exam- the rational development of new therapies.
ple, Proteobacteria take advantage of increased nitrogen Animal models of IBD have provided important
sources, including nitric oxide, produced in the inflamed insights into the roles of the microbiota and the immune
gut to bloom during IBD. Notably, targeting the molyb- system in the development of disease. However, with few
denum cofactor-​dependent enzymes that are required to exceptions, such models are not based on genetic defects
use nitric oxide for anaerobic respiration protects mice linked to human disease and do not fully recapitulate the
from DSS-​induced colitis by blunting the expansion of pathology of IBD. Furthermore, animal studies often rely
Enterobacteriaceae, including Proteobacteria196. Thus, on the use of gnotobiotic mice with a simplified micro-
the development of drugs that target metabolic pathways biota that does not reflect the complex microbial–host
used by harmful bacteria may provide a new strategy to interactions that occur in the intact gut. In addition,
treat IBD. mouse models do not incorporate the genetic and micro-
bial diversity of human populations or their environ-
Diet. Diet has an important role in shaping the compo- mental exposures. Thus, it will be important to develop
sition of the microbiota and can be modulated for the new models that are more relevant to human IBD.
management of IBD symptoms. Exclusive enteral nutri-
tion (EEN), which is a nutritionally complete elemental Published online xx xx xxxx

Nature Reviews | Immunology


Reviews

1. Kaplan, G. G. The global burden of IBD: from 2015 27. Goto, Y. et al. Innate lymphoid cells regulate intestinal protease of Vibrio cholerae that specifically cleaves
to 2025. Nat. Rev. Gastroenterol. Hepatol. 12, epithelial cell glycosylation. Science 345, 1254009 mucin glycoproteins. Microbiology 157, 516–525
720–727 (2015). (2014). (2011).
2. Xavier, R. J. & Podolsky, D. K. Unravelling the 28. Pham, T. A. N. et al. Epithelial IL-22RA1-mediated 52. Guo, L. et al. Lipid A acylation and bacterial resistance
pathogenesis of inflammatory bowel disease. Nature fucosylation promotes intestinal colonization against vertebrate antimicrobial peptides. Cell 95,
448, 427–434 (2007). resistance to an opportunistic pathogen. Cell Host 189–198 (1998).
3. Liu, J. Z. et al. Association analyses identify 38 Microbe 16, 504–516 (2014). 53. Matamouros, S. & Miller, S. I. S. Typhimurium strategies
susceptibility loci for inflammatory bowel disease 29. Bunker, J. J. & Bendelac, A. IgA responses to to resist killing by cationic antimicrobial peptides.
and highlight shared genetic risk across populations. microbiota. Immunity 49, 211–224 (2018). Biochim. Biophys. Acta 1848, 3021–3025 (2015).
Nat. Genet. 47, 979–986 (2015). 30. Kubinak, J. L. & Round, J. L. Do antibodies select a 54. Boneca, I. G. et al. A critical role for peptidoglycan
4. McGovern, D. P., Kugathasan, S. & Cho, J. H. Genetics healthy microbiota? Nat. Rev. Immunol. 16, 767–774 N-​deacetylation in Listeria evasion from the host
of inflammatory bowel diseases. Gastroenterology (2016). innate immune system. Proc. Natl Acad. Sci. USA
149, 1163–1176.e2 (2015). 31. Grootjans, J. et al. Epithelial endoplasmic reticulum 104, 997–1002 (2007).
5. Pickard, J. M., Zeng, M. Y., Caruso, R. & Nunez, G. stress orchestrates a protective IgA response. Science 55. D’Costa, V. M. et al. Salmonella disrupts host
Gut microbiota: role in pathogen colonization, immune 363, 993–998 (2019). endocytic trafficking by SopD2-mediated inhibition
responses, and inflammatory disease. Immunol. Rev. 32. Kubinak, J. L. et al. MyD88 signaling in T cells directs of Rab7. Cell Rep. 12, 1508–1518 (2015).
279, 70–89 (2017). IgA-​mediated control of the microbiota to promote 56. De Souza Santos, M. & Orth, K. The role of the
6. Round, J. L. & Mazmanian, S. K. The gut microbiota health. Cell Host Microbe 17, 153–163 (2015). type III secretion system in the intracellular lifestyle
shapes intestinal immune responses during health 33. Palm, N. W. et al. Immunoglobulin A coating identifies of enteric pathogens. Microbiol. Spectr. https://doi.
and disease. Nat. Rev. Immunol. 9, 313–323 (2009). colitogenic bacteria in inflammatory bowel disease. org/10.1128/microbiolspec.BAI-0008-2019 (2019).
7. Eckburg, P. B. et al. Diversity of the human intestinal Cell 158, 1000–1010 (2014). 57. Franzon, V. L., Arondel, J. & Sansonetti, P. J.
microbial flora. Science 308, 1635–1638 (2005). 34. Bunker, J. J. et al. Innate and adaptive humoral Contribution of superoxide dismutase and catalase
8. The Human Microbiome Project Consortium. responses coat distinct commensal bacteria with activities to Shigella flexneri pathogenesis. Infect.
Structure, function and diversity of the healthy human immunoglobulin A. Immunity 43, 541–553 (2015). Immun. 58, 529–535 (1990).
microbiome. Nature 486, 207–214 (2012). This study shows that T cell-independent IgA binds 58. Van der Sluis, M. et al. Muc2-deficient mice
9. Hooper, L. V., Littman, D. R. & Macpherson, A. J. commensal bacteria in the gut but that select spontaneously develop colitis, indicating that MUC2
Interactions between the microbiota and the immune species that evade T cell-independent IgA can is critical for colonic protection. Gastroenterology
system. Science 336, 1268–1273 (2012). localize near the intestinal mucosa, where they 131, 117–129 (2006).
10. Littman, D. R. & Pamer, E. G. Role of the commensal elicit an alternate T cell-dependent IgA response. 59. Johansson, M. E. et al. Bacteria penetrate the
microbiota in normal and pathogenic host immune 35. Donaldson, G. P. et al. Gut microbiota utilize normally impenetrable inner colon mucus layer in
responses. Cell Host Microbe 10, 311–323 (2011). immunoglobulin A for mucosal colonization. Science both murine colitis models and patients with
11. Lozupone, C. A., Stombaugh, J. I., Gordon, J. I., 360, 795–800 (2018). ulcerative colitis. Gut 63, 281–291 (2014).
Jansson, J. K. & Knight, R. Diversity, stability and 36. Haas, A. et al. Systemic antibody responses to gut 60. Van Klinken, B. J.-W., Van der Wal, J.-W. G.,
resilience of the human gut microbiota. Nature 489, commensal bacteria during chronic HIV-1 infection. Einerhand, A., Buller, H. & Dekker, J. Sulphation
220–230 (2012). Gut 60, 1506–1519 (2011). and secretion of the predominant secretory human
12. Honda, K. & Littman, D. R. The microbiome in 37. Sedman, P. C. et al. The prevalence of gut colonic mucin MUC2 in ulcerative colitis. Gut 44,
infectious disease and inflammation. Annu. Rev. translocation in humans. Gastroenterology 107, 387–393 (1999).
Immunol. 30, 759–795 (2012). 643–649 (1994). 61. Larsson, J. M. et al. Altered O-​glycosylation profile of
13. Macpherson, A. J., Slack, E., Geuking, M. B. & 38. Zeng, M. Y. et al. Gut microbiota-​induced MUC2 mucin occurs in active ulcerative colitis and is
McCoy, K. D. The mucosal firewalls against commensal immunoglobulin G controls systemic infection by associated with increased inflammation. Inflamm.
intestinal microbes. Semin. Immunopathol. 31, symbiotic bacteria and pathogens. Immunity 44, Bowel Dis. 17, 2299–2307 (2011).
145–149 (2009). 647–658 (2016). 62. McGovern, D. P. et al. Fucosyltransferase 2 (FUT2)
14. Belkaid, Y. & Hand, T. W. Role of the microbiota in This work shows that selective gut symbionts can non-​secretor status is associated with Crohn’s disease.
immunity and inflammation. Cell 157, 121–141 (2014). penetrate the mucosa and disseminate to induce Hum. Mol. Genet. 19, 3468–3476 (2010).
15. Arike, L., Holmen-​Larsson, J. & Hansson, G. C. IgG responses, which confer protection against 63. Tong, M. et al. Reprograming of gut microbiome
Intestinal Muc2 mucin O-​glycosylation is affected by systemic infection by enteric pathogens. energy metabolism by the FUT2 Crohn’s disease risk
microbiota and regulated by differential expression of 39. Kelsall, B. Recent progress in understanding the polymorphism. ISME J. 8, 2193–2206 (2014).
glycosyltranferases. Glycobiology 27, 318–328 phenotype and function of intestinal dendritic cells 64. Ogura, Y. et al. A frameshift mutation in NOD2
(2017). and macrophages. Mucosal Immunol. 1, 460–469 associated with susceptibility to Crohn’s disease.
16. Johansson, M. E. et al. The inner of the two Muc2 (2008). Nature 411, 603–606 (2001).
mucin-​dependent mucus layers in colon is devoid of 40. Smythies, L. E. et al. Human intestinal macrophages 65. Hugot, J. P. et al. Association of NOD2 leucine-​rich
bacteria. Proc. Natl Acad. Sci. USA 105, display profound inflammatory anergy despite avid repeat variants with susceptibility to Crohn’s disease.
15064–15069 (2008). phagocytic and bacteriocidal activity. J. Clin. Invest. Nature 411, 599–603 (2001).
17. Johansson, M. E., Larsson, J. M. & Hansson, G. C. 115, 66–75 (2005). 66. Ogura, Y. et al. Expression of NOD2 in Paneth cells:
The two mucus layers of colon are organized by the 41. Manicassamy, S. et al. Activation of β-​catenin in a possible link to Crohn’s ileitis. Gut 52, 1591–1597
MUC2 mucin, whereas the outer layer is a legislator of dendritic cells regulates immunity versus tolerance (2003).
host-​microbial interactions. Proc. Natl Acad. Sci. USA in the intestine. Science 329, 849–853 (2010). 67. Kobayashi, K. S. et al. Nod2-dependent regulation of
108 (Suppl. 1), 4659–4665 (2011). 42. Franchi, L. et al. NLRC4-driven production of IL-1β innate and adaptive immunity in the intestinal tract.
18. McGuckin, M. A., Linden, S. K., Sutton, P. & Florin, T. H. discriminates between pathogenic and commensal Science 307, 731–734 (2005).
Mucin dynamics and enteric pathogens. Nat. Rev. bacteria and promotes host intestinal defense. 68. Wilson, C. L. et al. Regulation of intestinal α-​defensin
Microbiol. 9, 265–278 (2011). Nat. Immunol. 13, 449–456 (2012). activation by the metalloproteinase matrilysin in
19. Ayabe, T. et al. Secretion of microbicidal α-​defensins 43. Macpherson, A. J. & Uhr, T. Induction of protective innate host defense. Science 286, 113–117 (1999).
by intestinal Paneth cells in response to bacteria. IgA by intestinal dendritic cells carrying commensal 69. Cadwell, K. et al. A key role for autophagy and the
Nat. Immunol. 1, 113–118 (2000). bacteria. Science 303, 1662–1665 (2004). autophagy gene Atg16l1 in mouse and human
20. Vaishnava, S., Behrendt, C. L., Ismail, A. S., Eckmann, L. 44. Balmer, M. L. et al. The liver may act as a firewall intestinal Paneth cells. Nature 456, 259–263 (2008).
& Hooper, L. V. Paneth cells directly sense gut mediating mutualism between the host and its gut 70. Kaser, A. et al. XBP1 links ER stress to intestinal
commensals and maintain homeostasis at the commensal microbiota. Sci. Transl Med. 6, 237ra266 inflammation and confers genetic risk for human
intestinal host-​microbial interface. Proc. Natl Acad. (2014). inflammatory bowel disease. Cell 134, 743–756
Sci. USA 105, 20858–20863 (2008). 45. Koch, M. A. et al. Maternal IgG and IgA antibodies (2008).
21. Vaishnava, S. et al. The antibacterial lectin RegIIIγ dampen mucosal T helper cell responses in early life. 71. Adolph, T. E. et al. Paneth cells as a site of origin for
promotes the spatial segregation of microbiota and Cell 165, 827–841 (2016). intestinal inflammation. Nature 503, 272–276
host in the intestine. Science 334, 255–258 (2011). 46. Macpherson, A. J. et al. A primitive T cell-​independent (2013).
22. Caruso, R. et al. A specific gene-​microbe interaction mechanism of intestinal mucosal IgA responses to 72. Siakavellas, S. I. & Bamias, G. Tumor necrosis factor-​
drives the development of Crohn’s disease-​like colitis commensal bacteria. Science 288, 2222–2226 like cytokine TL1A and its receptors DR3 and DcR3:
in mice. Sci. Immunol. 4, eaaw4341 (2019). (2000). important new factors in mucosal homeostasis and
This paper shows that a specific pathobiont can 47. Slack, E. et al. Innate and adaptive immunity inflammation. Inflamm. Bowel Dis. 21, 2441–2452
accumulate before the development of intestinal cooperate flexibly to maintain host-​microbiota (2015).
inflammation and trigger spontaneous Crohn’s mutualism. Science 325, 617–620 (2009). 73. Levine, B. & Kroemer, G. Autophagy in the
disease-like colitis in genetically susceptible mice. 48. Arabyan, N. et al. Salmonella degrades the host pathogenesis of disease. Cell 132, 27–42 (2008).
23. Sabat, R., Ouyang, W. & Wolk, K. Therapeutic glycocalyx leading to altered infection and glycan 74. Travassos, L. H. et al. Nod1 and Nod2 direct
opportunities of the IL-22–IL-22R1 system. Nat. Rev. remodeling. Sci. Rep. 6, 29525 (2016). autophagy by recruiting ATG16L1 to the plasma
Drug Discov. 13, 21–38 (2014). 49. Henderson, I. R., Czeczulin, J., Eslava, C., Noriega, F. membrane at the site of bacterial entry. Nat. Immunol.
24. Sonnenberg, G. F. et al. Innate lymphoid cells promote & Nataro, J. P. Characterization of Pic, a secreted 11, 55–62 (2010).
anatomical containment of lymphoid-​resident protease of Shigella flexneri and enteroaggregative 75. Homer, C. R., Richmond, A. L., Rebert, N. A.,
commensal bacteria. Science 336, 1321–1325 (2012). Escherichia coli. Infect. Immun. 67, 5587–5596 Achkar, J. P. & McDonald, C. ATG16L1 and NOD2
25. Qiu, J. et al. Group 3 innate lymphoid cells inhibit (1999). interact in an autophagy-​dependent antibacterial
T-cell-mediated intestinal inflammation through aryl 50. Mantle, M. & Rombough, C. Growth in and breakdown pathway implicated in Crohn’s disease pathogenesis.
hydrocarbon receptor signaling and regulation of of purified rabbit small intestinal mucin by Yersinia Gastroenterology 139, 1630–1641 (2010).
microflora. Immunity 39, 386–399 (2013). enterocolitica. Infect. Immun. 61, 4131–4138 76. Rioux, J. D. et al. Genome-​wide association study
26. Pickard, J. M. et al. Rapid fucosylation of intestinal (1993). identifies new susceptibility loci for Crohn disease
epithelium sustains host-​commensal symbiosis in 51. Szabady, R. L., Yanta, J. H., Halladin, D. K., and implicates autophagy in disease pathogenesis.
sickness. Nature 514, 638–641 (2014). Schofield, M. J. & Welch, R. A. TagA is a secreted Nat. Genet. 39, 596–604 (2007).

www.nature.com/nri
Reviews

77. Hampe, J. et al. A genome-​wide association scan 101. Brunkow, M. E. et al. Disruption of a new forkhead/ 126. Schaubeck, M. et al. Dysbiotic gut microbiota causes
of nonsynonymous SNPs identifies a susceptibility winged-​helix protein, scurfin, results in the fatal transmissible Crohn’s disease-​like ileitis independent
variant for Crohn disease in ATG16L1. Nat. Genet. 39, lymphoproliferative disorder of the scurfy mouse. of failure in antimicrobial defence. Gut 65, 225–237
207–211 (2007). Nat. Genet. 27, 68–73 (2001). (2016).
78. Lapaquette, P., Glasser, A. L., Huett, A., Xavier, R. J. 102. Mottet, C., Uhlig, H. H. & Powrie, F. Cutting edge: cure 127. Couturier-​Maillard, A. et al. NOD2-mediated dysbiosis
& Darfeuille-​Michaud, A. Crohn’s disease-​associated of colitis by CD4+CD25+ regulatory T cells. J. Immunol. predisposes mice to transmissible colitis and
adherent-​invasive E. coli are selectively favoured by 170, 3939–3943 (2003). colorectal cancer. J. Clin. Invest. 123, 700–711
impaired autophagy to replicate intracellularly. 103. Josefowicz, S. Z. et al. Extrathymically generated (2013).
Cell. Microbiol. 12, 99–113 (2010). regulatory T cells control mucosal TH2 inflammation. 128. Dianda, L. et al. T cell receptor-​alpha beta-​deficient
79. Brest, P. et al. A synonymous variant in IRGM alters Nature 482, 395–399 (2012). mice fail to develop colitis in the absence of a
a binding site for miR-196 and causes deregulation 104. Atarashi, K. et al. Induction of colonic regulatory microbial environment. Am. J. Pathol. 150, 91–97
of IRGM-​dependent xenophagy in Crohn’s disease. T cells by indigenous Clostridium species. Science (1997).
Nat. Genet. 43, 242–245 (2011). 331, 337–341 (2011). 129. Taurog, J. D. et al. The germfree state prevents
80. Kelsen, J. R., Baldassano, R. N., Artis, D. & 105. Round, J. L. & Mazmanian, S. K. Inducible Foxp3+ development of gut and joint inflammatory disease
Sonnenberg, G. F. Maintaining intestinal health: the regulatory T-​cell development by a commensal in HLA-​B27 transgenic rats. J. Exp. Med. 180,
genetics and immunology of very early onset bacterium of the intestinal microbiota. Proc. Natl 2359–2364 (1994).
inflammatory bowel disease. Cell. Mol. Gastroenterol. Acad. Sci. USA 107, 12204–12209 (2010). 130. Rath, H. C. et al. Normal luminal bacteria, especially
Hepatol. 1, 462–476 (2015). 106. Atarashi, K. et al. Treg induction by a rationally Bacteroides species, mediate chronic colitis, gastritis,
81. Uhlig, H. H. et al. The diagnostic approach to selected mixture of Clostridia strains from the human and arthritis in HLA-​B27/human beta2 microglobulin
monogenic very early onset inflammatory bowel microbiota. Nature 500, 232–236 (2013). transgenic rats. J. Clin. Invest. 98, 945–953 (1996).
disease. Gastroenterology 147, 990–1007.e3 107. Benson, M. J., Pino-​Lagos, K., Rosemblatt, M. & 131. Barnes, S. L. et al. Resistin-​like molecule β (RELMβ/
(2014). Noelle, R. J. All-​trans retinoic acid mediates enhanced FIZZ2) is highly expressed in the ileum of SAMP1/YitFc
82. Dhillon, S. S. et al. Variants in nicotinamide adenine T reg cell growth, differentiation, and gut homing in mice and is associated with initiation of ileitis.
dinucleotide phosphate oxidase complex components the face of high levels of co-​stimulation. J. Exp. Med. J. Immunol. 179, 7012–7020 (2007).
determine susceptibility to very early onset 204, 1765–1774 (2007). 132. Dalal, S. R. & Chang, E. B. The microbial basis of
inflammatory bowel disease. Gastroenterology 147, 108. Mucida, D. et al. Retinoic acid can directly promote inflammatory bowel diseases. J. Clin. Invest. 124,
680–689.e2 (2014). TGF-β-mediated Foxp3+ Treg cell conversion of naive 4190–4196 (2014).
83. Muise, A. M. et al. NADPH oxidase complex and IBD T cells. Immunity 30, 471–472; author reply 133. Swidsinski, A., Weber, J., Loening-​Baucke, V.,
candidate gene studies: identification of a rare variant 472–473 (2009). Hale, L. P. & Lochs, H. Spatial organization and
in NCF2 that results in reduced binding to RAC2. 109. Mortha, A. et al. Microbiota-​dependent crosstalk composition of the mucosal flora in patients with
Gut 61, 1028–1035 (2012). between macrophages and ILC3 promotes intestinal inflammatory bowel disease. J. Clin. Microbiol. 43,
84. Schappi, M. G. et al. The nature of colitis in chronic homeostasis. Science 343, 1249288 (2014). 3380–3389 (2005).
granulomatous disease. J. Pediatr. Gastroenterol. 110. Smith, P. M. et al. The microbial metabolites, 134. Yilmaz, B. et al. Microbial network disturbances in
Nutr. 36, 623–631 (2003). short-​chain fatty acids, regulate colonic Treg cell relapsing refractory Crohn’s disease. Nat. Med. 25,
85. Marciano, B. E. et al. Gastrointestinal involvement homeostasis. Science 341, 569–573 (2013). 323–336 (2019).
in chronic granulomatous disease. Pediatrics 114, 111. Arpaia, N. et al. Metabolites produced by commensal This work shows that, in patients with Crohn’s
462–468 (2004). bacteria promote peripheral regulatory T-​cell disease, microbiota networks are related to long-​
86. Ivanov, I. I. et al. Induction of intestinal Th17 cells by generation. Nature 504, 451–455 (2013). term disease type and severity, the extent and
segmented filamentous bacteria. Cell 139, 485–498 112. Furusawa, Y. et al. Commensal microbe-​derived localization of the inflamed segments, and
(2009). butyrate induces the differentiation of colonic responsiveness to therapy.
87. Atarashi, K. et al. Th17 cell induction by adhesion regulatory T cells. Nature 504, 446–450 (2013). 135. Gophna, U., Sommerfeld, K., Gophna, S., Doolittle, W. F.
of microbes to intestinal epithelial cells. Cell 163, 113. Sefik, E. et al. Mucosal immunology. Individual & Veldhuyzen van Zanten, S. J. O. Differences between
367–380 (2015). intestinal symbionts induce a distinct population of tissue-​associated intestinal microfloras of patients
88. Sano, T. et al. An IL-23R/IL-22 circuit regulates RORγ+ regulatory T cells. Science 349, 993–997 with Crohn’s disease and ulcerative colitis. J. Clin.
epithelial serum amyloid A to promote local effector (2015). Microbiol. 44, 4136–4141 (2006).
Th17 responses. Cell 164, 324 (2016). 114. Ohnmacht, C. et al. Mucosal immunology. The 136. Willing, B. P. et al. A pyrosequencing study in twins
89. Gaboriau-​Routhiau, V. et al. The key role of segmented microbiota regulates type 2 immunity through RORγt+ shows that gastrointestinal microbial profiles vary with
filamentous bacteria in the coordinated maturation of T cells. Science 349, 989–993 (2015). inflammatory bowel disease phenotypes.
gut helper T cell responses. Immunity 31, 677–689 115. Britton, G. J. et al. Microbiotas from humans with Gastroenterology 139, 1844–1854.e1 (2010).
(2009). inflammatory bowel disease alter the balance of gut 137. Lepage, P. et al. Twin study indicates loss of interaction
90. Yang, Y. et al. Focused specificity of intestinal TH17 Th17 and RORγt+ regulatory T cells and exacerbate between microbiota and mucosa of patients with
cells towards commensal bacterial antigens. Nature colitis in mice. Immunity 50, 212–224.e4 (2019). ulcerative colitis. Gastroenterology 141, 227–236
510, 152–156 (2014). 116. Sokol, H. et al. Faecalibacterium prausnitzii is an (2011).
91. Ansaldo, E. et al. Akkermansia muciniphila induces anti-​inflammatory commensal bacterium identified 138. Gevers, D. et al. The treatment-​naive microbiome in
intestinal adaptive immune responses during by gut microbiota analysis of Crohn disease patients. new-​onset Crohn’s disease. Cell Host Microbe 15,
homeostasis. Science 364, 1179–1184 (2019). Proc. Natl Acad. Sci. USA 105, 16731–16736 382–392 (2014).
This paper shows that A. muciniphila-​specific T cells (2008). 139. Franzosa, E. A. et al. Gut microbiome structure and
differentiate into T follicular helper cells to 117. Frank, D. N. et al. Molecular-​phylogenetic metabolic activity in inflammatory bowel disease.
generate homeostatic IgG1 in gnotobiotic animals characterization of microbial community imbalances Nat. Microbiol. 4, 293–305 (2019).
but can also adopt multiple TH cell lineages in mice in human inflammatory bowel diseases. Proc. Natl This paper reports metagenomic and metabolomic
with a complex microbiota. Acad. Sci. USA 104, 13780–13785 (2007). profiling of cross-​sectional stool samples from IBD
92. Weiss, J. M. et al. Neuropilin 1 is expressed on 118. Gaudier, E. et al. Butyrate specifically modulates MUC cohorts to identify associations between intestinal
thymus-​derived natural regulatory T cells, but not gene expression in intestinal epithelial goblet cells bacteria and metabolites that can predict IBD
mucosa-​generated induced Foxp3+ T reg cells. J. Exp. deprived of glucose. Am. J. Physiol. Gastrointest. status.
Med. 209, 1723–1742 (2012). Liver Physiol. 287, G1168–G1174 (2004). 140. Ott, S. J. et al. Fungi and inflammatory bowel
93. Lathrop, S. K. et al. Peripheral education of the 119. Wlodarska, M. et al. Indoleacrylic acid produced by diseases: alterations of composition and diversity.
immune system by colonic commensal microbiota. commensal Peptostreptococcus species suppresses Scand. J. Gastroenterol. 43, 831–841 (2008).
Nature 478, 250–254 (2011). inflammation. Cell Host Microbe 22, 25–37.e6 141. Li, Q. et al. Dysbiosis of gut fungal microbiota is
94. Geuking, M. B. et al. Intestinal bacterial colonization (2017). associated with mucosal inflammation in Crohn’s
induces mutualistic regulatory T cell responses. 120. Lamas, B. et al. CARD9 impacts colitis by altering disease. J. Clin. Gastroenterol. 48, 513–523 (2014).
Immunity 34, 794–806 (2011). gut microbiota metabolism of tryptophan into aryl 142. Sokol, H. et al. Fungal microbiota dysbiosis in IBD.
95. Kuhn, R., Lohler, J., Rennick, D., Rajewsky, K. & hydrocarbon receptor ligands. Nat. Med. 22, 598–605 Gut 66, 1039–1048 (2017).
Muller, W. Interleukin-10-deficient mice develop (2016). 143. Lewis, J. D. et al. Inflammation, antibiotics, and diet as
chronic enterocolitis. Cell 75, 263–274 (1993). 121. Rutgeerts, P. et al. Effect of faecal stream diversion environmental stressors of the gut microbiome in
96. Sellon, R. K. et al. Resident enteric bacteria are on recurrence of Crohn’s disease in the neoterminal pediatric Crohn’s disease. Cell Host Microbe 18,
necessary for development of spontaneous colitis and ileum. Lancet 338, 771–774 (1991). 489–500 (2015).
immune system activation in interleukin-10-deficient 122. D’Haens, G. R. et al. Early lesions of recurrent Crohn’s 144. Iliev, I. D. et al. Interactions between commensal fungi
mice. Infect. Immun. 66, 5224–5231 (1998). disease caused by infusion of intestinal contents in and the C-​type lectin receptor Dectin-1 influence
97. Li, M. O., Wan, Y. Y., Sanjabi, S., Robertson, A. K. & excluded ileum. Gastroenterology 114, 262–267 colitis. Science 336, 1314–1317 (2012).
Flavell, R. A. Transforming growth factor-​β regulation (1998). 145. McGovern, D. P. et al. Genome-​wide association
of immune responses. Annu. Rev. Immunol. 24, 123. Thia, K. T. et al. Ciprofloxacin or metronidazole for identifies multiple ulcerative colitis susceptibility loci.
99–146 (2006). the treatment of perianal fistulas in patients with Nat. Genet. 42, 332–337 (2010).
98. Travis, M. A. et al. Loss of integrin αvβ8 on dendritic Crohn’s disease: a randomized, double-​blind, 146. Sokol, H. et al. Card9 mediates intestinal epithelial
cells causes autoimmunity and colitis in mice. Nature placebo-controlled pilot study. Inflamm. Bowel Dis. cell restitution, T-​helper 17 responses, and control of
449, 361–365 (2007). 15, 17–24 (2009). bacterial infection in mice. Gastroenterology 145,
99. Glocker, E. O. et al. Inflammatory bowel disease and 124. Prantera, C. et al. Rifaximin-​extended intestinal 591–601.e3 (2013).
mutations affecting the interleukin-10 receptor. release induces remission in patients with moderately 147. Cadwell, K. et al. Virus-​plus-susceptibility gene
N. Engl. J. Med. 361, 2033–2045 (2009). active Crohn’s disease. Gastroenterology 142, interaction determines Crohn’s disease gene Atg16L1
100. Bennett, C. L. et al. The immune dysregulation, 473–481.e4 (2012). phenotypes in intestine. Cell 141, 1135–1145 (2010).
polyendocrinopathy, enteropathy, X-​linked syndrome 125. Garrett, W. S. et al. Communicable ulcerative colitis 148. Norman, J. M. et al. Disease-​specific alterations in
(IPEX) is caused by mutations of FOXP3. Nat. Genet. induced by T-​bet deficiency in the innate immune the enteric virome in inflammatory bowel disease.
27, 20–21 (2001). system. Cell 131, 33–45 (2007). Cell 160, 447–460 (2015).

Nature Reviews | Immunology


Reviews

149. Hughes, E. R. et al. Microbial respiration and formate 169. Xu, M. et al. c-​MAF-dependent regulatory T cells 192. Liu, S., Li, Y., Deng, B. & Xu, Z. Recombinant
oxidation as metabolic signatures of inflammation-​ mediate immunological tolerance to a gut pathobiont. Lactococcus lactis expressing porcine insulin-​like
associated dysbiosis. Cell Host Microbe 21, 208–219 Nature 554, 373–377 (2018). growth factor I ameliorates DSS-​induced colitis in
(2017). 170. Chai, J. N. et al. Helicobacter species are potent mice. BMC Biotechnol. 16, 25 (2016).
150. Lupp, C. et al. Host-​mediated inflammation disrupts drivers of colonic T cell responses in homeostasis and 193. Shigemori, S. et al. Oral delivery of Lactococcus lactis
the intestinal microbiota and promotes the overgrowth inflammation. Sci. Immunol. 2, aal5068 (2017). that secretes bioactive heme oxygenase-1 alleviates
of Enterobacteriaceae. Cell Host Microbe 2, 119–129 References 169 and 170 show that Helicobacter development of acute colitis in mice. Microb. Cell Fact.
(2007). species drive inducible Treg cell differentiation in the 14, 189 (2015).
151. Litvak, Y., Byndloss, M. X., Tsolis, R. M. & Baumler, A. J. colon during homeostasis but the same bacteria 194. Bermudez-​Humaran, L. G. et al. Serine protease
Dysbiotic proteobacteria expansion: a microbial also induce pathobiont-​specific colitic CD4+ TH cells inhibitors protect better than IL-10 and TGF-​β anti-​
signature of epithelial dysfunction. Curr. Opin. in the absence of Treg cells. inflammatory cytokines against mouse colitis when
Microbiol. 39, 1–6 (2017). 171. Chiaranunt, P., Tometich, J. T., Ji, J. & Hand, T. W. T cell delivered by recombinant lactococci. Microb. Cell Fact.
152. Loy, A. et al. Lifestyle and horizontal gene transfer-​ proliferation and colitis are initiated by defined 14, 26 (2015).
mediated evolution of Mucispirillum schaedleri, a core intestinal microbes. J. Immunol. 201, 243–250 (2018). 195. Wang, X., Wong, K., Ouyang, W. & Rutz, S. Targeting
member of the murine gut microbiota. mSystems 2, 172. Hand, T. W. et al. Acute gastrointestinal infection IL-10 family cytokines for the treatment of human
e00171–16 (2017). induces long-​lived microbiota-​specific T cell responses. diseases. Cold Spring Harb. Perspect. Biol. 11,
153. Fox, J. G., Ge, Z., Whary, M. T., Erdman, S. E. & Science 337, 1553–1556 (2012). a028548 (2019).
Horwitz, B. H. Helicobacter hepaticus infection in 173. Cong, Y., Feng, T., Fujihashi, K., Schoeb, T. R. & 196. Zhu, W. et al. Precision editing of the gut
mice: models for understanding lower bowel Elson, C. O. A dominant, coordinated T regulatory microbiota ameliorates colitis. Nature 553, 208–211
inflammation and cancer. Mucosal Immunol. 4, cell-IgA response to the intestinal microbiota. Proc. Natl (2018).
22–30 (2011). Acad. Sci. USA 106, 19256–19261 (2009). 197. Lee, D. et al. Diet in the pathogenesis and treatment
154. Faith, J. J., Ahern, P. P., Ridaura, V. K., Cheng, J. & 174. Josefowicz, S. Z., Lu, L. F. & Rudensky, A. Y. Regulatory of inflammatory bowel diseases. Gastroenterology
Gordon, J. I. Identifying gut microbe-​host phenotype T cells: mechanisms of differentiation and function. 148, 1087–1106 (2015).
relationships using combinatorial communities in Annu. Rev. Immunol. 30, 531–564 (2012). 198. Lee, D. et al. Comparative effectiveness of nutritional
gnotobiotic mice. Sci. Transl Med. 6, 220ra211 175. Liu, Z. et al. Immune homeostasis enforced by and biological therapy in North American children with
(2014). co-localized effector and regulatory T cells. Nature active Crohn’s disease. Inflamm. Bowel Dis. 21,
155. Barnich, N. et al. CEACAM6 acts as a receptor for 528, 225–230 (2015). 1786–1793 (2015).
adherent-​invasive E. coli, supporting ileal mucosa 176. van Nood, E. et al. Duodenal infusion of donor feces 199. Borrelli, O. et al. Polymeric diet alone versus
colonization in Crohn disease. J. Clin. Invest. 117, for recurrent Clostridium difficile. N. Engl. J. Med. corticosteroids in the treatment of active pediatric
1566–1574 (2007). 368, 407–415 (2013). Crohn’s disease: a randomized controlled open-​label
156. Macpherson, A., Khoo, U. Y., Forgacs, I., Philpott-​ 177. Paramsothy, S. et al. Faecal microbiota transplantation trial. Clin. Gastroenterol. Hepatol. 4, 744–753
Howard, J. & Bjarnason, I. Mucosal antibodies in for inflammatory bowel disease: a systematic review (2006).
inflammatory bowel disease are directed against and meta-​analysis. J. Crohns Colitis 11, 1180–1199 200. Grover, Z., Muir, R. & Lewindon, P. Exclusive enteral
intestinal bacteria. Gut 38, 365–375 (1996). (2017). nutrition induces early clinical, mucosal and
157. Ferrante, M. et al. New serological markers in 178. Levy, A. N. & Allegretti, J. R. Insights into the role transmural remission in paediatric Crohn’s disease.
inflammatory bowel disease are associated with of fecal microbiota transplantation for the treatment of J. Gastroenterol. 49, 638–645 (2014).
complicated disease behaviour. Gut 56, 1394–1403 inflammatory bowel disease. Therap. Adv. Gastroenterol. 201. Lewis, J. D. et al. Inflammation, antibiotics, and diet
(2007). 12, 1756284819836893 (2019). as environmental stressors of the gut microbiome in
158. Benckert, J. et al. The majority of intestinal IgA+ and 179. Rossen, N. G. et al. Findings from a randomized pediatric Crohn’s disease. Cell Host Microbe 22, 247
IgG+ plasmablasts in the human gut are antigen-​ controlled trial of fecal transplantation for patients (2017).
specific. J. Clin. Invest. 121, 1946–1955 (2011). with ulcerative colitis. Gastroenterology 149, 202. Llewellyn, S. R. et al. Interactions between diet and
159. Pirzer, U., Schonhaar, A., Fleischer, B., Hermann, E. 110–118.e4 (2015). the intestinal microbiota alter intestinal permeability
& Meyer zum Buschenfelde, K. H. Reactivity of 180. Suskind, D. L. et al. Fecal microbial transplant effect on and colitis severity in mice. Gastroenterology 154,
infiltrating T lymphocytes with microbial antigens in clinical outcomes and fecal microbiome in active Crohn’s 1037–1046.e2 (2018).
Crohn’s disease. Lancet 338, 1238–1239 (1991). disease. Inflamm. Bowel Dis. 21, 556–563 (2015). 203. Anderson, C. A. et al. Meta-​analysis identifies 29
160. Duchmann, R. et al. Tolerance exists towards resident 181. Vaughn, B. P. et al. Increased intestinal microbial additional ulcerative colitis risk loci, increasing the
intestinal flora but is broken in active inflammatory diversity following fecal microbiota transplant for number of confirmed associations to 47. Nat. Genet.
bowel disease (IBD). Clin. Exp. Immunol. 102, active Crohn’s disease. Inflamm. Bowel Dis. 22, 43, 246–252 (2011).
448–455 (1995). 2182–2190 (2016). 204. Franke, A. et al. Genome-​wide meta-​analysis
161. Duchmann, R. et al. T cell specificity and cross 182. Ledder, O. & Turner, D. Antibiotics in IBD: still a role in increases to 71 the number of confirmed Crohn’s
reactivity towards enterobacteria, Bacteroides, the biological era? Inflamm. Bowel Dis. 24, disease susceptibility loci. Nat. Genet. 42, 1118–1125
Bifidobacterium, and antigens from resident intestinal 1676–1688 (2018). (2010).
flora in humans. Gut 44, 812–818 (1999). 183. Khan, K. J. et al. Antibiotic therapy in inflammatory 205. Bouma, G. & Strober, W. The immunological and
162. Hegazy, A. N. et al. Circulating and tissue-​resident bowel disease: a systematic review and meta-​analysis. genetic basis of inflammatory bowel disease. Nat. Rev.
CD4+ T cells with reactivity to intestinal microbiota are Am. J. Gastroenterol. 106, 661–673 (2011). Immunol. 3, 521–533 (2003).
abundant in healthy individuals and function is altered 184. Wang, S. L., Wang, Z. R. & Yang, C. Q. Meta-​analysis 206. Baumgart, D. C. & Sandborn, W. J. Crohn’s disease.
during inflammation. Gastroenterology 153, of broad-​spectrum antibiotic therapy in patients with Lancet 380, 1590–1605 (2012).
1320–1337.e16 (2017). active inflammatory bowel disease. Exp. Ther. Med. 4,
This study reports the presence of microbiota-​ 1051–1056 (2012). Acknowledgements
reactive memory CD4+ T cells with a diverse TCR Vβ 185. Ford, A. C. et al. Efficacy of prebiotics, probiotics, and The authors apologize to colleagues whose work was not
repertoire in the blood and intestinal mucosa of synbiotics in irritable bowel syndrome and chronic cited or was cited through other review articles because of
both patients with IBD and healthy individuals. idiopathic constipation: systematic review and meta-​ space limitations. The authors thank G. Chen for critical
163. Cong, Y. et al. CD4+ T cells reactive to enteric bacterial analysis. Am. J. Gastroenterol. 109, 1547–1561 review of the manuscript. Work in G.N.’s laboratory is sup-
antigens in spontaneously colitic C3H/HeJBir mice: (2014). ported by US National Institutes of Health grants. R.C. is
increased T helper cell type 1 response and ability to 186. Kruis, W. et al. Maintaining remission of ulcerative supported by a Career Developments Award from the Crohn’s
transfer disease. J. Exp. Med. 187, 855–864 (1998). colitis with the probiotic Escherichia coli Nissle 1917 and Colitis Foundation. B.C.L. is supported by a Canadian
164. Iqbal, N. et al. T helper 1 and T helper 2 cells are is as effective as with standard mesalazine. Gut 53, Institutes of Health Research (CIHR) Fellowship.
pathogenic in an antigen-​specific model of colitis. 1617–1623 (2004).
J. Exp. Med. 195, 71–84 (2002). 187. Sassone-​Corsi, M. et al. Microcins mediate Author contributions
165. Kullberg, M. C. et al. Induction of colitis by a CD4+ competition among Enterobacteriaceae in the The authors contributed equally to all aspects of the article.
T cell clone specific for a bacterial epitope. Proc. Natl inflamed gut. Nature 540, 280–283 (2016).
Acad. Sci. USA 100, 15830–15835 (2003). 188. Kristensen, N. B. et al. Alterations in fecal microbiota Competing interests
166. Lodes, M. J. et al. Bacterial flagellin is a dominant composition by probiotic supplementation in healthy The authors declare no competing interests.
antigen in Crohn disease. J. Clin. Invest. 113, adults: a systematic review of randomized controlled
1296–1306 (2004). trials. Genome Med. 8, 52 (2016). Peer review information
167. Kim, S. C. et al. Variable phenotypes of enterocolitis in 189. Mills, J. P., Rao, K. & Young, V. B. Probiotics for Nature Reviews Immunology thanks J. Faith, J. Round and
interleukin 10-deficient mice monoassociated with two prevention of Clostridium difficile infection. Curr. Opin. the other, anonymous, reviewer(s) for their contribution to the
different commensal bacteria. Gastroenterology 128, Gastroenterol. 34, 3–10 (2018). peer review of this work.
891–906 (2005). 190. Steidler, L. et al. Treatment of murine colitis by
168. Kullberg, M. C. et al. Helicobacter hepaticus triggers Lactococcus lactis secreting interleukin-10. Science Publisher’s note
colitis in specific-​pathogen-free interleukin-10 (IL-10)- 289, 1352–1355 (2000). Springer Nature remains neutral with regard to jurisdictional
deficient mice through an IL-12- and gamma 191. Braat, H. et al. A phase I trial with transgenic bacteria claims in published maps and institutional affiliations.
interferon-​dependent mechanism. Infect. Immun. 66, expressing interleukin-10 in Crohn’s disease. Clin.
5157–5166 (1998). Gastroenterol. Hepatol. 4, 754–759 (2006). © Springer Nature Limited 2020

www.nature.com/nri

You might also like