Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Role of the Microbiota in Immune Development

Qing Zhao and Charles O Elson, University of Alabama at Birmingham, Birmingham, AL, USA
Ó 2016 Elsevier Ltd. All rights reserved.

Glossary
Altered Schaedler flora (ASF) A group of eight defined recognized by CD8þ T cells, while class II MHC molecules
cultivable bacteria from normal mice gut microbiota that is are recognized by CD4þ T cells.
used to colonize germ-free mice and to study the effect of Mesenteric lymph node (MLN) Draining lymph node of
a limited but defined set of gut commensal bacteria in vivo. intestinal mucosa that locates in the mesentery.
Aryl hydrocarbon receptor (AhR) A cytosolic transcription Microbe-associated molecular patterns (MAMPs) A series
factor involved in the differentiation of regulatory T cell, of conserved molecular motifs such as bacterial
Th17 cell, type 3 innate lymphoid cell, and intraepithelial lipopolysaccharide and flagellin that are associated with
cells. After its binding of dietary or microbial products, it microbes. They can be recognized by pattern recognition
translocates into the nucleus and initiates downstream receptors in both animals and plants.
gene expression. Microfold cell (M cell) A specialized epithelial cell that
C-type lectin A large family of carbohydrate-binding lacks microvilli but has shorter and broader microfolds on
receptors. Antimicrobial peptide RegIIIg is a C-type lectin. the apical cell surface. In the small intestine, it locates
Defensin A type of antimicrobial peptides produced by above Peyer’s patches and can transport antigens from
neutrophils and epithelial cells. Defensins can disrupt intestinal lumen to cells in the Peyer’s patches.
microbial cell membrane integrity thus resulting in Mutualism An ecological relationship that two species live
microbial killing. together and benefit from each other’s activities.
Hygiene hypothesis Hypothesis that the lack of exposure Paneth cell A specialized epithelial cell in the small
to symbiotic microbes and pathogens in early childhood intestine that produces a number of antimicrobial
predisposes individuals to increased allergic and peptides/proteins.
autoimmune diseases later on in life. Pattern recognition receptor (PRR) Germ line–encoded
Inflammasomes Protein complexes belonging to the innate proteins that can recognize microbe-associated molecular
immune system to sense damage-associated molecular patterns and induce signaling cascade in innate immune
patterns. They can activate inflammatory caspases and trigger responses.
the cleavage of proinflammatory IL-1 family cytokines into Peyer’s patch (PP) Isolated secondary lymphoid organs
their active forms. Subtypes of inflammasomes are named that locate under the small intestinal epithelium at the side
after the protein that initiates the signaling cascade, including opposite to mesenteric attachment. It is composed of a dome
nucleotide-binding oligomerization domain receptor (NLR) area, B cell follicles and interfollicular T cell areas, and
or AIM2-like receptor (ALR) protein. functions as an important inductive site of IgA responses.
Inflammatory bowel disease (IBD) Diseases of immune- Recombination-activating gene (RAG) Encodes enzymes
mediated intestinal inflammation caused by environment that are important for rearrangement and recombination of
and genetic predisposition. Crohn’s disease and ulcerative immunoglobulin and T cell receptor genes. The lack of either
colitis are two main types of IBD. RAG1 or RAG2 will lead to T cell and B cell deficiencies.
Lamina propria (LP) A layer of loose connective tissue Serum amyloid A (SAA) One of a group of highly
beneath the epithelium lining the mucosal tissues of the conserved proteins associated with high-density
body. It contains a rich vascular, nerve, and lymphatic lipoproteins in the serum. The level of SAA proteins
network and various types of immune cells, providing drastically increases during inflammation and participate
nutritional and immunological support at the mucosal in the induction of immune responses.
surface. Severe combined immunodeficiency (SCID) mice A
Major histocompatibility complex (MHC) A set of cell mouse model broadly used in studies of the immune
surface molecules that can bind peptide fragments and system and tissue transplants. These mice lack mature T and
display them on antigen-presenting cells for recognition by B cells due to a recessive mutation of a gene important for
corresponding T cells. Class I MHC molecules can be DNA repair.

Abstract

A well-functioning immune system is required for proper protection from infections and diseases. Vertebrates have coevolved
a comprehensive immune network with the numerous microbes colonizing every surface of their body, particularly in the
digestive tract. As such, the immune system has to be trained to discriminate pathogens from symbiotic microbes as well as
self from nonself. Commensal microbiota in the gut plays a critical role in this development. Investigations have found

Encyclopedia of Immunobiology, Volume 5 http://dx.doi.org/10.1016/B978-0-12-374279-7.19019-8 109


110 Physiology of the Immune System j Role of the Microbiota in Immune Development

correlations of gut microbiota with the development and maturation of a number of components of innate and adaptive
immunity. Disruption of this microbiota-host dialog may lead to immune disorders such as allergy and immune-mediated
inflammatory diseases.

Introduction microbial containment upon pathogen challenge, and increased


risk of immune-mediated inflammatory diseases (Dimmitt et al.,
The complex community of commensal, symbiotic, and path- 2010; Hill et al., 2010; Fukuda et al., 2011). In humans, antibiotic
ogenic microorganisms colonizing the body surface of all verte- treatment during infancy has resulted in increased risk of allergy,
brates is termed the ‘microbiota,’ with the largest mass residing asthma, and inflammatory bowel disease (IBD) later in life (Hviid
in the lower intestine. Normally, a healthy human intestine et al., 2011; Kronman et al., 2012; Munyaka et al., 2014).
harbors 1014 microbes – 10 times greater than the number of Thus microbiota colonization and concomitant immune devel-
cells in the human body (Gill et al., 2006). These organisms opment during neonatal life and infancy are crucial for a properly
include mostly bacteria, but also fungi, archaea, and viruses, functioning immune system. In the sections that follow, we will
which interact with the mucosal surface and play a variety of discuss immune system development and maturation, much of
physiological roles, such as facilitating host food digestion which occurs during this early-life exposure to the microbiota.
via degradation of plant polysaccharide and fibers, producing
vitamins and energy, promoting the development of the intes-
tinal epithelium, the gut vasculature, and the enteric nervous Intestinal Lymphoid Tissue Development
system (Duerkop et al., 2009). Recently, there are also studies
correlating altered gut microbiota composition with cognitive Lymphoid tissue inducer (LTi) cells, which are generated prena-
disorders such as depression, anxiety, and autism (Schmidt, tally in the fetal liver due to the expression of transcription factor
2015). But most importantly, a complete set of balanced gut Rorgt and Id2 (Eberl et al., 2004), are fundamental for the early
microbiota helps with the development and regulation of development of lymph nodes (LNs), Peyer’s patches (PPs), cryp-
a healthy, fully functioning mucosal and systemic immune topatches (CPs), and isolated lymphoid follicles (ILFs) (Eberl and
system. In particular to the gut, the microbiota influences the Littman, 2004). LNs and PPs develop in the fetus in utero, in the
structural development of lymphoid tissues, epithelial barrier absence of microbial stimulation under the influence of a number
function, and the development of the innate and adaptive of factors (Table 1). However, microbial signals are required for
immune systems, assisting with the reciprocal challenges that the postnatal development and maturation of PPs and mesenteric
the intimate contact of gut microbiota and the mucosal surface lymph nodes (MLNs) (Pollard and Sharon, 1970; Round and
brings – intestinal infections and autoimmune diseases Mazmanian, 2009). The development and maturation of micro-
(Rakoff-Nahoum et al., 2004; Macpherson and Harris, 2004). fold cell (M cell), a specialized epithelial cell localized on top of
Microbiota composition is greatly diversified among individ- PPs to facilitate antigen translocation, are also influenced by
uals, but generally similar within the same species. Depending bacterial colonization (Randall and Mebius, 2014).
on the delivery method of the baby (vaginally or with cesarean The formation of CPs and ILFs happens postnatally. CPs are
section), the founder bacteria of the gut will be different. mainly composed of clusters of LTi cells in the LP at the base of
Commonly, the gut microbiota composition of unweaned intestinal villi. In mice, over 1500 CPs can be detected about
infants is structured with fewer microbial species and shows 2 weeks after birth, even in GF mice without microbial coloniza-
greater variations between individuals (Kurokawa et al., 2007). tion (Kanamori et al., 1996). Upon microbe-associated molec-
The intestinal ecosystem undergoes a vast change when infants ular pattern (MAMP) recognition through Toll-like receptors
start eating solid food: more microbial species appear due to (TLRs), stromal cells become activated by LTi cells through lym-
the expansion of nutritional niches and a rapidly developing photoxin-b receptor signaling, and recruit DCs and B cells into
immune system, and tends to be relatively stable at the end of the CPs to form ILFs (Tsuji et al., 2008). Recognition of gram-
early childhood (Dominguez-Bello et al., 2011). In adult gut, negative bacterial peptidoglycan through nucleotide oligomeri-
Bacteroidetes and Firmicutes phyla become predominant. zation domain 1 (NOD1) expressed in intestinal epithelial cells
Although highly variable between individuals, the assemblage (IECs) induces CCL20 and b-defensin 3 and recruits B cells to the
of microbial species shows a functional similarity in the gut LTi-DC clusters to form ILFs (Bouskra et al., 2008). ILFs are
regardless of age and sex (Human Microbiome Project, 2012). dynamic reservoirs for the induction of secretory IgA (sIgA) via
The understanding of how intestinal microbiota shape the both T-dependent and -independent pathways (Figure 1; Tsuji
gut immune system is largely established via comparing germ- et al., 2008; Knoop and Newberry, 2012; Maynard et al., 2012).
free (GF) mice or microbial-reduced mice with conventional Yet another type of intestinal lymphoid tissue is tertiary
mice raised under specific pathogen free (SPF) conditions. lymphoid tissue (tLT), which contains B cell follicles and T cell
Lack of microbial antigen stimulation results in underdevelop- areas and is generated in the LP during chronic inflammation
ment of the mucosal as well as the systemic immune system, (Aloisi and Pujol-Borrell, 2006) but is absent in healthy tissues.
including impaired development of lymphoid organs, decreased Contrary to PPs, CPs, and ILFs; the development of tLTs is not
recruitment of lymphocytes, unbalanced lymphocyte differentia- dependent on Rorgt, because Rorgt-deficient mice can form
tion, and immune cell malfunction. These homeostatic immune them in the intestine in the presence of a gut microbiota. These
system defects in GF and antibiotic-treated mice can tLTs along with increased serum IgG help contain microbes in
subsequently lead to altered immune responses, impaired Rorgt/ mice at steady state in compensation for the lack of
Physiology of the Immune System j Role of the Microbiota in Immune Development 111

Table 1 Requirements for the development of intestinal lymphoid tissues

Lymphoid tissue Rorgt Id2 LTab-LTbR Chemokines and cytokines Age References

MLNs þ þ þ TRANCE Prenatal De Togni et al. (1994), Yokota et al. (1999),


Eberl et al. (2004), and Kim et al. (2000)
Peyer’s patches þ þ þ IL-7, TNF, CXCL13, CCL19, Prenatal De Togni et al. (1994), Yokota et al. (1999),
CCL20, CCL21 Eberl et al. (2004), and Eberl and Lochner (2009)
Cryptopatches þ ? þ IL-7 Postnatal Kanamori et al. (1996)
ILFs þ ? þ CXCL13 and CCL20 needed Postnatal Tsuji et al. (2008), Bouskra et al. (2008),
for small intestine and Baptista et al. (2013)
tLTs  ? LTab-LTbR and CCL19, CCL21, CXCL12, Postnatal Aloisi and Pujol-Borrell (2006) and Lochner
TNFSF14-LTbR CXCL13 et al. (2011)

MLNs, mesenteric lymph nodes; ILFs, isolated lymphoid follicles; tLTs, tertiary lymphoid tissues; Rorgt, retinoid acid receptor–related orphan receptor gt; Id2, inhibitor of DNA
binding 2; LTab, lymphotoxin-ab; LTbR, lymphotoxin-b receptor; TRANCE, tumor necrosis factor–related activation-induced cytokine; TNF, tumor necrosis factor; CXCL13,
chemokine (CeXeC motif) ligand 13; CCL9, chemokine (CeC motif) ligand 9; TNFSF14, tumor necrosis factor superfamily member 14; þ, required.

(a) Prenatal (b) Postnatal


Dendritic cell
Bacteria
Antimicrobial
Intraepithelial peptides
Lumen lymphocyte
Dendritic cell
MAMPs Dimeric IgA
Goblet cell
M cell IgA+ cell
Epithelium
T cell

B cell
Lamina
propria
Peyer’s patch
Cryptopatch
Paneth cell
Peyer’s patch
LTi cell Mature isolated
Mesenteric lymphoid follicle
lymph node

Figure 1 The development of intestinal lymphoid tissues. (a) Prenatally, secondary lymphoid tissues (Peyer’s patches and mesenteric lymph nodes)
and cryptopatches develop by the spatiotemporal recruitment of lymphoid tissue inducer (LTi) cells to sites of the developing intestine and supporting
neurovascular structures. This, in turn, stimulates the recruitment of dendritic cells (DCs), T cells, and B cells in preparation for the immune response to
the microbiota. Intraepithelial lymphocytes seed the epithelium before birth. (b) Postnatally, bacteria colonize the neonatal intestine immediately, initiating
multiple events that affect the development or functional maturation of the mucosa- and gut-associated lymphoid tissues. Shown from left to right:
microbe-associated molecular patterns (MAMPs) sensed by pattern recognition receptors on intestinal epithelial cells and DCs adjacent to cryptopatches
stimulate the further recruitment of B cells and T cells, causing the cryptopatches to develop into mature isolated lymphoid follicles. The isolated
lymphoid follicles release IgA-producing plasma cells – which are formed through T cell–dependent and –independent interactions – into the lamina
propria. Microbes also cross the epithelium and enter Peyer’s patches through M cells, from which they are endocytosed by DCs in the subepithelial
dome. Antigen-loaded DCs in the Peyer’s patch interact with local lymphocytes to induce T cell differentiation and T cell–dependent B cell maturation in
the germinal centre to induce the development of IgA-producing plasma cells that home to the lamina propria, where they release dimeric IgA for trans-
port into the intestinal lumen. Dendritic-cell-mediated luminal sampling of microbial products or transcytosis of bacteria across the epithelium results in
antigen loading of lamina propria dendritic cells, which then migrate through the afferent lymphatics vessels (not shown) to a draining mesenteric lymph
node to induce differentiation of effector T cells that traffic to the lamina propria. Shown on the far right, sensing of MAMPs stimulates the proliferation
of intestinal epithelial cells in crypts, resulting in their increased depth and, in the small intestine, increased density of Paneth cells. This sensing also
arms the intestinal epithelial cells for release of antimicrobial peptides (figure and legend from Maynard et al., 2012).

other lymphoid organs. However, when colonic injury was the host, while at the same time transporting and absorbing
induced with chemicals in Rorgt/ mice, such tLTs contributed dietary substances for the host’s needs. The epithelial surface
to exaggerated disease (Lochner et al., 2011). is constantly shedding but rapidly self-renewing by specialized
stem cells that reside at the base of the intestinal crypt. Five
Intestinal Barrier Function and Innate Immune types of epithelial cells – Paneth cells, goblet cells, M cells,
System Development absorptive enterocytes, and enteroendocrine cells, can all
perform immune functions that either facilitate the physical
Microbiota and IECs
clearance of infiltrating microbes and/or orchestrate innate
Gut epithelium is the most critical physical and physiological and adaptive immune machinery to maintain intestinal
frontline barrier that separates the intestinal microbiota from homeostasis.
112 Physiology of the Immune System j Role of the Microbiota in Immune Development

IECs express a series of pattern recognition receptors (PRRs) to also be induced by monocolonization with Bacteroides thetaio-
sense microbial antigens, referred to as MAMPs. TLRs are a family taomicron (Hooper et al., 2003). Another example is the expres-
of cell surface PRRs, which are indispensible for sensing intestinal sion of C-type lectin RegIIIg, which primarily targets gram-
microbiota to control IEC homeostasis and protect from epithe- positive bacteria in the intestine, and requires microbial sensing
lial injury (Rakoff-Nahoum et al., 2004). Shortly after birth, through TLR and MyD88 (Vaishnava et al., 2011).
IECs become activated due to the stimulation of TLR4 by lipo- Goblet cells contribute to immune defense by the production
polysaccharide (LPS), which is a major component of the outer of highly glycosylated mucin proteins that form a viscous layer
membrane of gram-negative bacteria. This spontaneous activa- outside the epithelial surface to restrict the penetration of resident
tion is microbial signal driven, since it is not seen in cesarean microbes. The thickness of the mucus layer correlates with bacte-
section–delivered mice or TLR4-deficient mice. Within hours of rial load in the intestine (Petersson et al., 2011). MUC2/ mice
initial microbial exposure, the posttranscriptional downregula- develop colitis spontaneously (Van der Sluis et al., 2006), and
tion of a protein essential for TLR4 signaling, interleukin-1 (IL- following Citrobacter rodentium infection, MUC2 deficiency results
1) receptor–associated kinase 1, leads to the lifelong loss of in exaggerated colonic pathology due to impaired clearance and
responsiveness of IECs to LPS stimulation. This programmed increased translocation of this intestinal pathogen (Bergstrom
negative-regulatory mechanism helps the host adapt to the rapid et al., 2010). GF mice have fewer numbers and smaller-sized
microbial colonization of the intestine (Lotz et al., 2006). IEC goblet cells in the epithelium (Kandori et al., 1996), and corre-
expression of TLR3 also shows an age-dependent increase due spondingly the inner mucus layer is substantially diminished
to microbial colonization (Pott et al., 2012). compared to conventionally reared mice. These defects can be
Intestinal antimicrobial peptides/proteins (AMPs) are restored with LPS or peptidoglycan exposure (Petersson et al.,
a group of natural antibiotics produced by multiple types of cells 2011), indicating the importance of microbial stimulation on
lining the gut epithelium, but primarily by intestinal enterocytes goblet cell development and mucin production (Figure 2).
and Paneth cells. AMPs are predominantly present in the mucus Furthermore, microbiota are involved in the regulation of
layer above the epithelial surface, either directly killing or inac- an array of other barrier pathways. For example, epithelial
tivating bacteria and thus creating a relatively sterile zone that cell expression of polymeric immunoglobulin receptors
greatly reduces the direct contact of microbes and the epithelium (PIgR), which can bind IgA and IgM produced in the LP and
(Gallo and Hooper, 2012). Microbial signals are involved in the facilitate their transport across the epithelium into the intes-
regulation of the expression and secretion of most AMPs other tinal lumen, is strongly induced by microbial colonization
than a-defensins. One example is Paneth cell production of (Hooper et al., 2001; Hapfelmeier et al., 2010). Microbiota
angiogenin-4 which rapidly increases to adult levels during also induce the glycosylation of IECs by stimulating group 3
weaning due to microbial colonization; this upregulation can innate lymphoid cell (see below) production of IL-22 and

Small intestine Large intestine

Intestinal
lumen

~50μm 50-200μm

Epithelium

Paneth Goblet Enterocytes


cell cell

AMPs Outer loose mucus


sIgA Inner mucus

Commensal bacteria

Figure 2 IECs contribute to intestinal barrier functions. Intestinal epithelial cells secrete mucin and a series of antimicrobial peptides (AMPs) to
create a spatial segregation of the commensal microbiota and the epithelium. MUC2 mucin is the major mucin protein in the intestine. It forms
a loose, nonadherent single mucus layer in the small intestine, and a two-layered mucus complex in the large intestine where the inner layer is
compact and firmly attached to the epithelium. The inner layer is 50–200 mm thick and hardly permeable by commensal bacteria. It converts to the
outer loose mucus layer at the luminal border, where commensal bacteria are densely colonized. In addition to mucin proteins, enterocytes and Pan-
eth cells (present only in the small intestine) can secrete AMPs to separate the microbiota from the epithelial surface. In the small intestine where the
inner mucus layer is absent, AMPs can form a  50 mm barrier to limit direct association of luminal bacteria with the epithelium.
Physiology of the Immune System j Role of the Microbiota in Immune Development 113

lymphotoxin (Goto et al., 2014a). This benefits certain micro- Sonnenberg et al., 2013; Robinette et al., 2015). ILCs are
biota species that can use epithelial glycans as a nutritional lymphocytes that lack antigen receptors but respond rapidly
substrate and thus maintains colonization (Hooper et al., to microbiota and cytokine signals. They develop from
1999). a common precursor in an IL-7Ra- and Id2-dependent
manner, and differentiate into three groups depending on
the tissue and the environmental stimuli. In the gut, the major
Microbiota and Innate Cells
type is Rorgtþ group 3 ILCs, which resemble their adaptive
IECs constantly interact with the innate cells present in the intes- counterpart Th17 cells by the production of IL-17A and
tinal lamina propria (LP) at steady state. Innate cells such as IL-22. Microbial induction of IL-22 production by ILC3 can
dendritic cells (DCs), macrophages, and innate lymphoid cells subsequently stimulate epithelial AMP and mucin expression,
(ILCs), are responsible for antigen uptake and presentation, as well as epithelial fucosylation (Satoh-Takayama et al.,
degradation of cell debris and pathogens, as well as secreting 2008; Sonnenberg et al., 2011).
cytokines and chemokines in response to MAMP recognition, It has been controversial whether microbiota plays a role on
linking microbiota, the epithelium, and adaptive immune the generation of ILC3s. Studies show that the absolute number
system all together to form a homeostatic system maintaining of ILC3 subset NKp46þ cells is decreased in GF mice and can be
mutualism. Intestinal microbiota plays an important role in restored with bacterial colonization (Satoh-Takayama et al.,
the regulation of innate cell phenotype and functions. 2008; Sanos et al., 2009). However, others have found no
DCs are derived from a common progenitor in the bone difference of either total or NKp46þ ILC3 numbers in GF,
marrow, and migrate directly via blood or afferent lymph into antibiotic-treated, or SPF mice. Also, microbiota can suppress
the intestine to develop into resident DCs and migratory DCs, ILC3 production of IL-22 by inducing epithelial cell IL-25
respectively (Persson et al., 2013). Colonization of bacteria trig- production, consistent with a negative regulatory loop for
gers the recruitment of LP CD103þ DCs to the epithelium for ILC3 cells at steady state (Sawa et al., 2011).
antigen uptake and subsequent presentation to T cells in the ILC3s can produce GM-CSF in response to microbiota-
MLNs (Farache et al., 2013). Commonly, TLR recognition induced IL-1b secretion by intestinal macrophages. Impaired
induces DC production of IL-12 and subsequently promotes GM-CSF production can lead to altered cell functions of mono-
Th1 cell differentiation. However, the abundance of TGFb and nuclear phagocytes and Treg cells, and therefore disturbed gut
retinoic acid (RA) in the gut environment facilitates the differen- homeostasis (Mortha et al., 2014). A proper balance of this
tiation of tolerogenic DCs, which produce IL-10 instead of IL-12 macrophage-ILC3 loop is needed because excessive IL-1b
and promote the differentiation of regulatory T (Treg) cells, there- during Helicobacter hepaticus infection promotes an IL-17A
fore contributing to the maintenance of gut homeostasis (Stein- response in both CD4þ T cells and ILCs, leading to an exagger-
man et al., 2003; Coombes et al., 2007; Cording et al., 2014). ated colonic inflammation (Coccia et al., 2012).
Macrophages have phenotypic overlap with DCs but distinct
functions. Both resident and inflammatory microphages in the
gut are derived from circulating Ly6Chi monocytes (Bain et al., Microbiota and the Development of Adaptive Immune
2013), and their recruitment is CCR2-dependent in mice (Bain System
et al., 2014). Although highly phagocytic and bactericidal, human
mucosal macrophages exhibit nonresponsiveness to a broad Due to the active interactions with intestinal microbiota and
range of microbial antigens and inflammatory stimuli at steady the mucosal immune system, the majority of lymphocytes
state (Smythies et al., 2005; Lotz et al., 2006), exerting immune reside in the healthy intestine. In fact, based on observations
surveillance functions at mucosal borders as well as contributing from immune-deficient mouse models such as severe
to immune tolerance. Systemic macrophages isolated from combined immunodeficiency mice and recombination-
antibiotic-treated adult mice show defective antiviral-related activating gene-1/2 (RAG-1/2) knock out mice that have no
gene expression, impaired type I and II IFN production, weakened mature T and B cells, innate immune pathways are sufficient
antiviral functions, and increased susceptibility to influenza infec- to provide host protection in response to commensal micro-
tion (Abt et al., 2012), indicating the role of microbiota in main- biota stimulation. However, the vulnerability of such mice to
taining macrophage function systemically as well as mucosally. pathogens demonstrates the importance of the adaptive
Microbiota also regulate the homeostasis of systemic immune system. Similar to the innate components of the gut
neutrophils via NOD1 recognition of microbial peptidoglycan mucosa, the development and maturation of gut adaptive
(Clarke et al., 2010). Neutrophils are normally not present in immune system are actively driven by the microbiota as well.
the gut, but are rapidly recruited via chemokines in response
of intestinal pathogens and inflammation. Lacking the priming
Microbiota and T cells
by intestinal microbial signals, GF and antibiotic-treated mice
develop reduced numbers of bone marrow and circulating Distinct subsets of T cells populate different parts of the intes-
neutrophils, as well as impaired killing functions against path- tinal wall – intraepithelial lymphocytes (IELs) are mainly
ogens (Deshmukh et al., 2014). composed of CD8þ T cells, while CD4þ T cells are the major
During recent years, an important cell type belonging to cell type in the LP. The recruitment and function of gut T cells
the innate immune system, termed ILCs, have been are microbiota driven, because GF or antibiotic-treated mice
discovered and are beginning to be characterized (Colonna, have reduced numbers of IELs and LP CD4þ T cells in the
2009; Spits and Di Santo, 2011; Spits and Cupedo, gut, as well as altered immune functions (Ichinohe et al.,
2012; Sonnenberg and Artis, 2012; Walker et al., 2013; 2011; Chung et al., 2012). Interestingly, although exposed to
114 Physiology of the Immune System j Role of the Microbiota in Immune Development

rapidly colonizing microbiota, LP CD4þ T cells remain imma- (SFB) appear to be potent stimulators of the Th17 subset (Iva-
ture throughout the postnatal period (Torow et al., 2015). nov et al., 2008, 2009). SFB is a gram-positive, spore-forming
Microbial recognition through nucleotide oligomerization anaerobic organism that is classified as a unique member of
domain 2 (NOD2) plays a critical role in IEL homeostasis the Clostridiales, which in the vegetative state forms typical
(Jiang et al., 2013). Functionally, unlike peripheral CD8 T cells, long filaments. SFB is an auxotroph and interacts closely
gut IELs freshly isolated from conventional mice are constitu- with the epithelium and PPs of the terminal ileum. Coloniza-
tively cytolytic. However, IELs isolated from GF mice have tion of SFB induces a moderate upregulation of a broad range
substantially reduced expression of Thy-1 and have almost no of proinflammatory cytokines with mechanisms still not
lytic function, revealing the crucial role of microbiota on IEL known, although recently it is postulated that TLR2 recogni-
activation (Lefrancois and Goodman, 1989). tion might be involved in this process (Gaboriau-Routhiau
et al., 2009; Schnupf et al., 2015). SFB specifically induces
Effector T Cell Development the differentiation of endogenous Th17 cells in the small
Lack of microbial exposure in early life leads to a skewed intestine by MHC-II-dependent antigen presentation through
LP T cell development toward Th2 phenotype (Mazmanian intestinal DCs (Goto et al., 2014b) and stimulation of epithe-
et al., 2005; Cahenzli et al., 2013), most likely due to the lial production of serum amyloid A (SAA) (Ivanov et al.,
impaired development of other CD4þ T cell subsets including 2009). The function of Th17 cells in the gut remains contro-
Th1, Th17, and Treg cells. Helminth infection greatly induces versial. Th17 cells help maintain gut homeostasis under steady
proliferation and activation of gut Th2 cells as well as Treg cells, state conditions and provide host protection against certain
which in turn inhibit the differentiation of Th1 cells, thus gut pathogens by facilitating epithelial production of AMPs,
pig worm ingestion has been proposed as a therapeutic B cell production of IgA, and upregulation of PIgR. However,
intervention for inflammatory bowel disorders (Elliott and Th17 cells have been implicated in the pathogenesis of IBD
Weinstock, 2012). and other autoimmune diseases. During colitis, Th17 cells
The normal adult mouse LP shows a Th1/Th17 dominant differentiate into an IL-17AþIFNgþ double-positive CD4 T
effector phenotype. Both Th1 and Th17 cells contribute to cell phenotype, and then into IFNgþ single positive ex-Th17,
normal intestinal homeostasis but can also mediate inflamma- Th1 cells. The latter cell has recently been identified as the
tion in IBD and other autoimmune diseases by their cytokine effector cell driving experimental colitis (Harbour et al.,
production and their ability to recruit neutrophils (Strober 2015). Interestingly, regarding antigen specificity, the majority
and Fuss, 2011; Feng et al., 2011; Neurath, 2014; Harbour of SFB-induced endogenous Th17 cells in the gut react
et al., 2015). Several microorganisms have been shown to strongly to SFB antigen stimulation, suggesting that SFB
participate in the induction of Th1 cells mucosally and system- does not work as an adjuvant in inducing Th17 cells to other
ically (Hsieh et al., 1993; Mazmanian et al., 2005; Jakobsson commensals (Yang et al., 2014). How SFB-reactive Th17 cells
et al., 2014). Although pathogenic during inflammation, gut migrate to organs other than the gut and contribute to autoim-
Th1 cells exert important immune protective functions against mune diseases remain to be elucidated (Wu et al., 2010).
enteric pathogens such as Listeria monocytogenes and interact
with the innate immune system to maintain gut homeostasis. Regulatory T Cell Development
It is shown that Th1-associated cytokine IFNg, induced in The differentiation of naïve T cells in the gut occurs in the PPs
wild-type mice by bacterial colonization, contributes to the and MLNs. When CD103þ DCs sample intestinal antigens and
inhibition of innate IL-23 expression, which can function as migrate to the draining MLNs, they present the processed
a pathogenic factor of intestinal inflammation (Sheikh et al., peptides to activate the naïve T cells (Pabst and Mowat,
2010). IFNg also participates in the upregulation of major 2012). Intestinal CD103þ DCs express high levels of aldh1a2,
histocompatibility complex (MHC)-II molecules on IECs which can convert the vitamin A metabolite retinal into RA.
(Thelemann et al., 2014) and in the control of T cell expansion RA together with TGFb favors Treg development in the gut
in lymphopenic mice (Do et al., 2014), contributing to preven- (Coombes et al., 2007; Sun et al., 2007). Gut Treg cells are
tion of intestinal inflammation. required for the establishment of oral tolerance and are respon-
Th17 cells are dependent on transcription factors Rorgt sible for suppression of effector T cell responses and the resolu-
and aryl hydrocarbon receptor (AhR) for their development tion of intestinal inflammation (Hadis et al., 2011). They are
(Ivanov et al., 2006; Yang et al., 2008b), and secrete IL-17A, also indispensable for the maintenance of gut immune homeo-
IL-17F, and IL-22 as their main cytokines. Th17 cells share stasis with the microbiota. The pathogenesis of IBD is proposed
the developmental requirement of microbiota-driven TGFb to be uncontrolled immune responses to commensal antigens
with induced Tregs (Mangan et al., 2006; Qin et al., 2009; in the gut in a genetically susceptible host (Maloy and Powrie,
Lee et al., 2012), but in vitro Th17 cell differentiation also 2011). A number of current experimental mouse models are
needs IL-6. The differentiation of intestinal Th17 cells is due to genetic targeting of the Treg pathway. Treg cells perform
dependent on microbial signals, in that GF animals rarely their immunosuppressive function with mechanisms such as
have them (Ivanov et al., 2008, 2009). It is shown that at the production of anti-inflammatory cytokines IL-10, TGFb,
steady state, the induction of intestinal Th17 cells needs and IL-35 and consumption of IL-2; they can also inhibit
microbiota-dependent IL-1b, but not IL-6 (Shaw et al., Ca2þ signaling and subsequently inhibit the activation of
2012). In addition, other microbiota-induced factors, such NF-kB and NFAT pathways in conventional T cells (Schmidt
as ATP (Atarashi et al., 2008) and IL-23, are shown to help et al., 2012). A number of studies have shown that Treg cells
with intestinal Th17 cell development or maintenance, respec- stimulate the production of sIgA to promote host-microbiota
tively. Among the microbiota, segmented filamentous bacteria mutualism (Kawamoto et al., 2014). One possible pathway is
Physiology of the Immune System j Role of the Microbiota in Immune Development 115

Clostridium
SFB IV and XIVa B. fragilis ASF

Intestinal
lumen

Epithelium
IEL

• SAA
• DC activation • SCFA PSA TLR activation
• Bacteria-derived • GPR43 activation
Lamina ATP
propria

Th17 Treg
RORgt+ Foxp3+

T
Naïve

Figure 3 Microbiota induction of intestinal Treg/Th17 cell differentiation. Commensal bacteria such as Clostridium cluster IV and XIVa, Bacteroides
fragilis and altered Schaedler flora (ASF) promote the differentiation and expansion of Treg cells in the intestine through various mechanisms,
whereas microbial-derived ATP and segmented filamentous bacteria (SFB) stimulate the induction of intestinal Th17 cells. Th17 and Treg cells are
both plastic and can convert into each other and into other T cell subsets under certain conditions.

that Treg cells can downregulate Foxp3 expression in the PPs Treg cells into large intestinal LP and contribute to gut homeo-
and convert into follicular helper T (Tfh) cells that facilitate B stasis (Kim et al., 2013). In addition, microbiota and their
cell synthesis of IgA (Tsuji et al., 2009). Treg cells also provide metabolic products impact the epigenetic regulation of Treg
a survival signal for IgAþ B cells in the LP. Depletion of CD25þ cells as well (Furusawa et al., 2013; Obata et al., 2014).
Treg cells results in decreased IgAþ B cells in the LP and corre- Microbiota-driven Treg cells are functionally protective in gut
spondingly reduced total and commensal-specific sIgA levels in inflammation and in allergic disease models.
the gut (Cong et al., 2009). LP T cells show high potential of plasticity due to the
Microbiota play an important role in the differentiation and complexity of the gut environment. The most reported plas-
expansion of Treg cells. Due to the high density of microbes ticity occurs between Treg/Th17 and Th17/Th1 conversions
and microbial metabolic products in the large intestine, it (Yang et al., 2008a; Lee et al., 2009; Zhou et al., 2009; Brown
becomes a primary location for the induction of peripherally et al., 2015; Gagliani et al., 2015; Harbour et al., 2015; Liu
derived Treg cells and the expansion of thymus-derived Treg et al., 2015). Treg and Th17 cells can convert into Tfh cells in
cells. There are a number of different microbiota products the PPs and facilitate high-affinity intestinal IgA production
that can induce Treg cells. For example, the immunomodula- (Tsuji et al., 2009; Hirota et al., 2013). Whether microbial
tory molecule PSA derived from commensal bacteria Bacteroides signals are involved in plasticity still remains to be investigated.
fragilis drives the induction of colonic Treg cells and IL-10
expression in a TLR2-dependent manner (Round and
Microbiota and B Cells
Mazmanian, 2010; Round et al., 2011). Colonization of GF
mice with 46 strains of Clostridium that primarily belong to IgA, as a noninflammatory immunoglobulin, does not possess
IV and XIVa clusters specifically induced IL-10þCTLA4þHelios the ability to bind common Fc receptors and activate the
Tregs and TGFb production in the colon (Atarashi et al., 2011). complement system, and represents a unique mechanism of
Colonization of GF mice with altered Schaedler flora (ASF) mucosal immune defense. It is produced in the largest quanti-
stimulated both Heliosþ and Helios Treg populations, which ties of any antibody isotypes (Macpherson et al., 2008), and
is shown to be dependent on TLR-mediated sensing (Geuking can be made through T-independent and -dependent manners
et al., 2011). Most recently, several groups showed that gut by B1 and B2 B cells, respectively (Fujihashi et al., 1996;
bacterial fermentation products, short-chain fatty acids, exclu- Macpherson et al., 2000). After being produced by plasma cells
sively induce Treg cells, but not other T cell subsets, in the colon in the LP, IgA dimers are transcytosed by PIgR expressed on
in a G protein–coupled receptor 43 (GPR43)-dependent epithelial cells from the LP into the intestinal lumen, where
manner (Figure 3; Arpaia et al., 2013; Furusawa et al., 2013; they bind to the surface of commensal bacteria and limit
Smith et al., 2013). GPR43 deficiency leads to increased inflam- them from penetrating the epithelium. Microbial colonization
matory responses in different disease models (Maslowski et al., predominantly drives the production of intestinal IgA, because
2009). Gut microbiota also regulate the expression of another GF mice have substantially impaired IgA production in the
G protein receptor – GPR15, which can promote homing of intestinal mucosa whereas serum IgA levels are less affected
116 Physiology of the Immune System j Role of the Microbiota in Immune Development

(Macpherson et al., 2008). Commensal bacteria such as Morga- the cleanliness of environment and the broad use of antibiotics
nella morganii or SFB can strongly stimulate germinal center (Bach, 2002; Noverr and Huffnagle, 2004; Okada et al., 2010),
reactions and IgA secreting B cells in the PPs (Shroff et al., consistent with the concept of the hygiene hypothesis, which
1995). SFB can also induce de novo IgA production in tLTs in was first brought up by Strachan in the late 1980s (Strachan,
the LP in mice lacking PPs and ILFs (Klaasen et al., 1993; 1989). The vast environmental transition from the sterile
Lecuyer et al., 2014). Microbiota can also contribute to IgA uterus to the outside world full of microbes greatly stimulates
catabolism, as commensal bacteria belonging to the Sutterella immune development and maturation, making the early child-
genus can degrade secretory component and IgA in the intes- hood an important period for the establishment of a well-
tinal lumen, leading to exaggerated intestinal damage upon functioning immune system. The human data linking altered
chemical challenge, and this pattern is transmissible from early-life microbial exposure with impaired immune develop-
mother to offspring (Moon et al., 2015). ment and long-term susceptibility of autoimmune and allergic
B cell differentiation and function is regulated by gut micro- diseases demonstrate the significance of proper microbiota
biota. For example, in response to microbial stimulation at colonization during this transitional period (Lundell et al.,
steady state, mucosal B cells can produce tumor necrosis factor 2011, 2012; Olszak et al., 2012; Cahenzli et al., 2013; Munyaka
(TNF) and inducible nitric oxide synthase (iNOS) for optimal et al., 2014). As we progress in the application of comprehen-
intestinal IgA production and maintenance of microbiota sive metagenomic and metatranscriptomic analysis, a better
diversity (Fritz et al., 2012). During inflammation, B cells can understanding of how microbiota impact immune develop-
acquire a regulatory phenotype and start to produce IL-10; ment will provide us with new insights in immune disease
microbiota-driven IL-1b and IL-6 production is required to prevention and treatment.
promote this process (Rosser et al., 2014). In the absence of
diversified microbial exposure, neonatal mice develop more See also: Allergy: The Hygiene Hypothesis of Allergy and
IgE switching B cells at the mucosal sites and correspondingly Asthma. Anatomy and Microanatomy of the Immune System:
higher serum IgE levels, in a CD4 T cell– and IL-4-dependent Tertiary Lymphoid Tissues. Cells of the Innate Immune System:
manner, leading to increased surface-bound IgE on mast cells Dendritic Cells and Dendritic Cell Subsets; Macrophage
and exaggerated oral-induced allergic reaction with ovalbumin Activation and Polarization; Neutrophils – Role in Innate
(Cahenzli et al., 2013). MyD88-dependent basophil activation Immunity. Cytokines and Their Receptors: Inflammasomes.
is also involved in this process (Hill et al., 2012). Development of T Cells and Innate Lymphoid Cells:
Development of Regulatory T Cells in the Thymus; Innate
Lymphoid Cells Type 3. Immune Deficiency: Combined T Cell
Microbiota and Immunological Diseases and B Cell Deficiency – SCID Forms: TBþ; Severe
Combined Immune Deficiency with Absence of B and T
With the profound impact on the development of gut immune Lymphocytes (TBNKþSCIDs): The Key Function of V(D)J
system, an unbalanced composition of gut microbiota or ‘dys- Recombination for Lymphocyte Development. Immunity to
biosis’ can lead to disturbed immune responses and has been Bacterial, Parasitic and Fungal Infections: The Hygiene
linked to a variety of autoimmune and allergic diseases in Hypothesis and Immunity to Parasitic Helminths. Molecular
recent years (Berer et al., 2011; Horai et al., 2015). One Aspects of Innate Immunity: The Mucins. Structure and
example is depletion of transcription factor T-bet in Rag/ Function of Diversifying Receptors: Structure and Function of
mice resulting in the development of spontaneous colitis, IgA. T Cell Activation: Th17 and Th22 Cells; Treg Cells.
which can be treated with broad-spectrum antibiotics, indi-
cating a microbiota-driven pathology. Cohousing of colitic
Tbet/Rag/ with wild-type mice results in colitis develop-
ment in the latter as well, suggesting that pathogenic micro- References
biota is sufficient to induce inflammation in the normal gut
Abt, M.C., Osborne, L.C., Monticelli, L.A., et al., 2012. Commensal bacteria calibrate
(Garrett et al., 2007). Similarly, perturbation of inflammasome
the activation threshold of innate antiviral immunity. Immunity 37, 158–170.
NLRP6 or IL-22 pathway also causes a shift in gut microbiota Aloisi, F., Pujol-Borrell, R., 2006. Lymphoid neogenesis in chronic inflammatory
and subsequently leads to either colonic inflammation or diseases. Nat. Rev. Immunol. 6, 205–217.
a greater risk to develop colonic inflammation, respectively Arpaia, N., Campbell, C., Fan, X., et al., 2013. Metabolites produced by commensal
(Elinav et al., 2011; Zenewicz et al., 2013). bacteria promote peripheral regulatory T-cell generation. Nature 504, 451–455.
Atarashi, K., Nishimura, J., Shima, T., et al., 2008. ATP drives lamina propria T(H)17
In accordance with findings in animal studies, human IBD cell differentiation. Nature 455, 808–812.
has been associated to dysbiosis. A subset of patients with IBD Atarashi, K., Tanoue, T., Shima, T., et al., 2011. Induction of colonic regulatory T cells
develops an altered gut microbiota with greatly increased Proteo- by indigenous Clostridium species. Science 331, 337–341.
bacteria and reduced Bacteroidetes and Firmicutes (Frank et al., Bach, J.F., 2002. The effect of infections on susceptibility to autoimmune and allergic
diseases. N. Engl. J. Med. 347, 911–920.
2007; Gevers et al., 2014). Children living in developing coun-
Bain, C.C., Scott, C.L., Uronen-Hansson, H., et al., 2013. Resident and pro-
tries with greater intestinal bacterial diversity have lower frequen- inflammatory macrophages in the colon represent alternative context-dependent
cies of inflammatory disorders in adulthood including IBD. Also, fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 6, 498–510.
early-life exposure to antibiotics has been shown highly associ- Bain, C.C., Bravo-Blas, A., Scott, C.L., et al., 2014. Constant replenishment from
ated with IBD development later in life (De Filippo et al., circulating monocytes maintains the macrophage pool in the intestine of adult
mice. Nat. Immunol. 15, 929–937.
2010; Hviid et al., 2011; Kronman et al., 2012; Lin et al., 2013). Baptista, A.P., Olivier, B.J., Goverse, G., et al., 2013. Colonic patch and colonic SILT
The increasing incidence of autoimmune and allergic development are independent and differentially regulated events. Mucosal Immu-
diseases in industrialized countries occurs concurrently with nol. 6, 511–521.
Physiology of the Immune System j Role of the Microbiota in Immune Development 117

Berer, K., Mues, M., Koutrolos, M., et al., 2011. Commensal microbiota and myelin Fritz, J.H., Rojas, O.L., Simard, N., et al., 2012. Acquisition of a multifunctional IgAþ
autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541. plasma cell phenotype in the gut. Nature 481, 199–203.
Bergstrom, K.S., Kissoon-Singh, V., Gibson, D.L., et al., 2010. Muc2 protects against Fujihashi, K., Mcghee, J.R., Kweon, M.N., et al., 1996. Gamma/delta T cell-deficient
lethal infectious colitis by disassociating pathogenic and commensal bacteria from mice have impaired mucosal immunoglobulin A responses. J. Exp. Med. 183,
the colonic mucosa. PLoS Pathog. 6, e1000902. 1929–1935.
Bouskra, D., Brezillon, C., Berard, M., et al., 2008. Lymphoid tissue genesis induced by Fukuda, S., Toh, H., Hase, K., et al., 2011. Bifidobacteria can protect from entero-
commensals through NOD1 regulates intestinal homeostasis. Nature 456, 507–510. pathogenic infection through production of acetate. Nature 469, 543–547.
Brown, C.C., Esterhazy, D., Sarde, A., et al., 2015. Retinoic acid is essential for Th1 cell Furusawa, Y., Obata, Y., Fukuda, S., et al., 2013. Commensal microbe-derived
lineage stability and prevents transition to a Th17 cell program. Immunity 42, 499–511. butyrate induces the differentiation of colonic regulatory T cells. Nature 504,
Cahenzli, J., Koller, Y., Wyss, M., Geuking, M.B., Mccoy, K.D., 2013. Intestinal 446–450.
microbial diversity during early-life colonization shapes long-term IgE levels. Cell Gaboriau-Routhiau, V., Rakotobe, S., Lecuyer, E., et al., 2009. The key role of
Host Microbe 14, 559–570. segmented filamentous bacteria in the coordinated maturation of gut helper T cell
Chung, H., Pamp, S.J., Hill, J.A., et al., 2012. Gut immune maturation depends on responses. Immunity 31, 677–689.
colonization with a host-specific microbiota. Cell 149, 1578–1593. Gagliani, N., Vesely, M.C., Iseppon, A., et al., 2015. Th17 cells transdifferentiate into
Clarke, T.B., Davis, K.M., Lysenko, E.S., et al., 2010. Recognition of peptidoglycan regulatory T cells during resolution of inflammation. Nature 523, 221–225.
from the microbiota by Nod1 enhances systemic innate immunity. Nat. Med. 16, Gallo, R.L., Hooper, L.V., 2012. Epithelial antimicrobial defence of the skin and
228–231. intestine. Nat. Rev. Immunol. 12, 503–516.
Coccia, M., Harrison, O.J., Schiering, C., et al., 2012. IL-1beta mediates chronic Garrett, W.S., Lord, G.M., Punit, S., et al., 2007. Communicable ulcerative colitis
intestinal inflammation by promoting the accumulation of IL-17A secreting innate induced by T-bet deficiency in the innate immune system. Cell 131, 33–45.
lymphoid cells and CD4(þ) Th17 cells. J. Exp. Med. 209, 1595–1609. Geuking, M.B., Cahenzli, J., Lawson, M.A., et al., 2011. Intestinal bacterial coloni-
Colonna, M., 2009. Interleukin-22-producing natural killer cells and lymphoid tissue zation induces mutualistic regulatory T cell responses. Immunity 34, 794–806.
inducer-like cells in mucosal immunity. Immunity 31, 15–23. Gevers, D., Kugathasan, S., Denson, L.A., et al., 2014. The treatment-naïve micro-
Cong, Y., Feng, T., Fujihashi, K., Schoeb, T.R., Elson, C.O., 2009. A dominant, biome in new-onset Crohn’s disease. Cell Host Microbe 15, 382–392.
coordinated T regulatory cell-IgA response to the intestinal microbiota. Proc. Natl. Gill, S.R., Pop, M., Deboy, R.T., et al., 2006. Metagenomic analysis of the human
Acad. Sci. U.S.A. 106, 19256–19261. distal gut microbiome. Science 312, 1355–1359.
Coombes, J.L., Siddiqui, K.R., Arancibia-Carcamo, C.V., et al., 2007. A functionally Goto, Y., Obata, T., Kunisawa, J., et al., 2014a. Innate lymphoid cells regulate
specialized population of mucosal CD103þ DCs induces Foxp3þ regulatory T cells intestinal epithelial cell glycosylation. Science 345, 1254009.
via a TGF-beta and retinoic acid-dependent mechanism. J. Exp. Med. 204, Goto, Y., Panea, C., Nakato, G., et al., 2014b. Segmented filamentous bacteria
1757–1764. antigens presented by intestinal dendritic cells drive mucosal Th17 cell differen-
Cording, S., Wahl, B., Kulkarni, D., et al., 2014. The intestinal micro-environment tiation. Immunity 40, 594–607.
imprints stromal cells to promote efficient Treg induction in gut-draining lymph Hadis, U., Wahl, B., Schulz, O., et al., 2011. Intestinal tolerance requires gut homing
nodes. Mucosal Immunol. 7, 359–368. and expansion of FoxP3þ regulatory T cells in the lamina propria. Immunity 34,
De Filippo, C., Cavalieri, D., Di Paola, M., et al., 2010. Impact of diet in shaping gut 237–246.
microbiota revealed by a comparative study in children from Europe and rural Hapfelmeier, S., Lawson, M.A., Slack, E., et al., 2010. Reversible microbial coloni-
Africa. Proc. Natl. Acad. Sci. U.S.A. 107, 14691–14696. zation of germ-free mice reveals the dynamics of IgA immune responses. Science
De Togni, P., Goellner, J., Ruddle, N.H., et al., 1994. Abnormal development of 328, 1705–1709.
peripheral lymphoid organs in mice deficient in lymphotoxin. Science 264, Harbour, S.N., Maynard, C.L., Zindl, C.L., Schoeb, T.R., Weaver, C.T., 2015.
703–707. Th17 cells give rise to Th1 cells that are required for the pathogenesis of colitis.
Deshmukh, H.S., Liu, Y., Menkiti, O.R., et al., 2014. The microbiota regulates Proc. Natl. Acad. Sci. U.S.A. 112, 7061–7066.
neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in Hill, D.A., Hoffmann, C., Abt, M.C., et al., 2010. Metagenomic analyses reveal
neonatal mice. Nat. Med. 20, 524–530. antibiotic-induced temporal and spatial changes in intestinal microbiota with
Dimmitt, R.A., Staley, E.M., Chuang, G., et al., 2010. Role of postnatal acquisition of associated alterations in immune cell homeostasis. Mucosal Immunol. 3,
the intestinal microbiome in the early development of immune function. J. Pediatr. 148–158.
Gastroenterol. Nutr. 51, 262–273. Hill, D.A., Siracusa, M.C., Abt, M.C., et al., 2012. Commensal bacteria-derived signals
Do, J.S., Asosingh, K., Baldwin 3rd, W.M., Min, B., 2014. Cutting edge: IFN-gammaR regulate basophil hematopoiesis and allergic inflammation. Nat. Med. 18,
signaling in non-T cell targets regulates T cell-mediated intestinal inflammation 538–546.
through multiple mechanisms. J. Immunol. 192, 2537–2541. Hirota, K., Turner, J.E., Villa, M., et al., 2013. Plasticity of Th17 cells in Peyer’s
Dominguez-Bello, M.G., Blaser, M.J., Ley, R.E., Knight, R., 2011. Development of the patches is responsible for the induction of T cell-dependent IgA responses. Nat.
human gastrointestinal microbiota and insights from high-throughput sequencing. Immunol. 14, 372–379.
Gastroenterology 140, 1713–1719. Hooper, L.V., Stappenbeck, T.S., Hong, C.V., Gordon, J.I., 2003. Angiogenins: a new
Duerkop, B.A., Vaishnava, S., Hooper, L.V., 2009. Immune responses to the micro- class of microbicidal proteins involved in innate immunity. Nat. Immunol. 4,
biota at the intestinal mucosal surface. Immunity 31, 368–376. 269–273.
Eberl, G., Littman, D.R., 2004. Thymic origin of intestinal alphabeta T cells revealed by Hooper, L.V., Xu, J., Falk, P.G., Midtvedt, T., Gordon, J.I., 1999. A molecular sensor
fate mapping of RORgammatþ cells. Science 305, 248–251. that allows a gut commensal to control its nutrient foundation in a competitive
Eberl, G., Lochner, M., 2009. The development of intestinal lymphoid tissues at the ecosystem. Proc. Natl. Acad. Sci. U.S.A. 96, 9833–9838.
interface of self and microbiota. Mucosal Immunol. 2, 478–485. Hooper, L.V., Wong, M.H., Thelin, A., et al., 2001. Molecular analysis of commensal
Eberl, G., Marmon, S., Sunshine, M.J., et al., 2004. An essential function for the host-microbial relationships in the intestine. Science 291, 881–884.
nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer Horai, R., Zarate-Blades, C.R., Dillenburg-Pilla, P., et al., 2015. Microbiota-dependent
cells. Nat. Immunol. 5, 64–73. activation of an autoreactive T cell receptor provokes autoimmunity in an immu-
Elinav, E., Strowig, T., Kau, A.L., et al., 2011. NLRP6 inflammasome regulates colonic nologically privileged site. Immunity 43, 343–353.
microbial ecology and risk for colitis. Cell 145, 745–757. Hsieh, C.S., Macatonia, S.E., Tripp, C.S., Wolf, S.F., O’garra, A., 1993. Development
Elliott, D.E., Weinstock, J.V., 2012. Helminth-host immunological interactions: of TH1 CD4þ T cells through IL-12 produced by Listeria-induced macrophages.
prevention and control of immune-mediated diseases. Ann. N.Y. Acad. Sci. 1247, Science 260, 547–549.
83–96. Human Microbiome Project Consortium, 2012. Structure, function and diversity of the
Farache, J., Koren, I., Milo, I., et al., 2013. Luminal bacteria recruit CD103þ dendritic healthy human microbiome. Nature 486, 207–214.
cells into the intestinal epithelium to sample bacterial antigens for presentation. Hviid, A., Svanstrom, H., Frisch, M., 2011. Antibiotic use and inflammatory bowel
Immunity 38, 581–595. diseases in childhood. Gut 60, 49–54.
Feng, T., Qin, H., Wang, L., et al., 2011. Th17 cells induce colitis and promote Th1 Ichinohe, T., Pang, I.K., Kumamoto, Y., et al., 2011. Microbiota regulates immune
cell responses through IL-17 induction of innate IL-12 and IL-23 production. defense against respiratory tract influenza A virus infection. Proc. Natl. Acad. Sci.
J. Immunol. 186, 6313–6318. U.S.A. 108, 5354–5359.
Frank, D.N., St Amand, A.L., Feldman, R.A., et al., 2007. Molecular-phylogenetic Ivanov, I.I., Mckenzie, B.S., Zhou, L., et al., 2006. The orphan nuclear receptor
characterization of microbial community imbalances in human inflammatory bowel RORgammat directs the differentiation program of proinflammatory IL-17þ T helper
diseases. Proc. Natl. Acad. Sci. U.S.A. 104, 13780–13785. cells. Cell 126, 1121–1133.
118 Physiology of the Immune System j Role of the Microbiota in Immune Development

Ivanov, I.I., Frutos Rde, L., Manel, N., et al., 2008. Specific microbiota direct the Maynard, C.L., Elson, C.O., Hatton, R.D., Weaver, C.T., 2012. Reciprocal interactions
differentiation of IL-17-producing T-helper cells in the mucosa of the small of the intestinal microbiota and immune system. Nature 489, 231–241.
intestine. Cell Host Microbe 4, 337–349. Mazmanian, S.K., Liu, C.H., Tzianabos, A.O., Kasper, D.L., 2005. An immunomodu-
Ivanov, I.I., Atarashi, K., Manel, N., et al., 2009. Induction of intestinal Th17 cells by latory molecule of symbiotic bacteria directs maturation of the host immune
segmented filamentous bacteria. Cell 139, 485–498. system. Cell 122, 107–118.
Jakobsson, H.E., Abrahamsson, T.R., Jenmalm, M.C., et al., 2014. Decreased gut Moon, C., Baldridge, M.T., Wallace, M.A., et al., 2015. Vertically transmitted faecal
microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 IgA levels determine extra-chromosomal phenotypic variation. Nature 521,
responses in infants delivered by caesarean section. Gut 63, 559–566. 90–93.
Jiang, W., Wang, X., Zeng, B., et al., 2013. Recognition of gut microbiota by NOD2 is Mortha, A., Chudnovskiy, A., Hashimoto, D., et al., 2014. Microbiota-dependent
essential for the homeostasis of intestinal intraepithelial lymphocytes. J. Exp. Med. crosstalk between macrophages and ILC3 promotes intestinal homeostasis.
210, 2465–2476. Science 343, 1249288.
Kanamori, Y., Ishimaru, K., Nanno, M., et al., 1996. Identification of novel lymphoid Munyaka, P.M., Khafipour, E., Ghia, J.E., 2014. External influence of early childhood
tissues in murine intestinal mucosa where clusters of c-kitþ IL-7Rþ Thy1þ establishment of gut microbiota and subsequent health implications. Front. Pediatr.
lympho-hemopoietic progenitors develop. J. Exp. Med. 184, 1449–1459. 2, 109.
Kandori, H., Hirayama, K., Takeda, M., Doi, K., 1996. Histochemical, lectin- Neurath, M.F., 2014. Cytokines in inflammatory bowel disease. Nat. Rev. Immunol. 14,
histochemical and morphometrical characteristics of intestinal goblet cells of 329–342.
germfree and conventional mice. Exp. Anim. 45, 155–160. Noverr, M.C., Huffnagle, G.B., 2004. Does the microbiota regulate immune responses
Kawamoto, S., Maruya, M., Kato, L.M., et al., 2014. Foxp3(þ) T cells regulate outside the gut? Trends Microbiol. 12, 562–568.
immunoglobulin a selection and facilitate diversification of bacterial species Obata, Y., Furusawa, Y., Endo, T.A., et al., 2014. The epigenetic regulator Uhrf1
responsible for immune homeostasis. Immunity 41, 152–165. facilitates the proliferation and maturation of colonic regulatory T cells. Nat.
Kim, D., Mebius, R.E., Macmicking, J.D., et al., 2000. Regulation of peripheral lymph Immunol. 15, 571–579.
node genesis by the tumor necrosis factor family member TRANCE. J. Exp. Med. Okada, H., Kuhn, C., Feillet, H., Bach, J.F., 2010. The ʻhygiene hypothesisʼ for
192, 1467–1478. autoimmune and allergic diseases: an update. Clin. Exp. Immunol. 160, 1–9.
Kim, S.V., Xiang, W.V., Kwak, C., et al., 2013. GPR15-mediated homing controls Olszak, T., An, D., Zeissig, S., et al., 2012. Microbial exposure during early life has
immune homeostasis in the large intestine mucosa. Science 340, 1456–1459. persistent effects on natural killer T cell function. Science 336, 489–493.
Klaasen, H.L., Van Der Heijden, P.J., Stok, W., et al., 1993. Apathogenic, intestinal, Pabst, O., Mowat, A.M., 2012. Oral tolerance to food protein. Mucosal Immunol. 5,
segmented, filamentous bacteria stimulate the mucosal immune system of mice. 232–239.
Infect. Immun. 61, 303–306. Persson, E.K., Scott, C.L., Mowat, A.M., Agace, W.W., 2013. Dendritic cell subsets in
Knoop, K.A., Newberry, R.D., 2012. Isolated lymphoid follicles are dynamic reservoirs the intestinal lamina propria: ontogeny and function. Eur. J. Immunol. 43,
for the induction of intestinal IgA. Front. Immunol. 3, 84. 3098–3107.
Kronman, M.P., Zaoutis, T.E., Haynes, K., Feng, R., Coffin, S.E., 2012. Antibiotic Petersson, J., Schreiber, O., Hansson, G.C., et al., 2011. Importance and regulation of
exposure and IBD development among children: a population-based cohort study. the colonic mucus barrier in a mouse model of colitis. Am. J. Physiol. Gastrointest.
Pediatrics 130, e794–e803. Liver Physiol. 300, G327–G333.
Kurokawa, K., Itoh, T., Kuwahara, T., et al., 2007. Comparative metagenomics revealed Pollard, M., Sharon, N., 1970. Responses of the Peyer’s patches in germ-free mice to
commonly enriched gene sets in human gut microbiomes. DNA Res. 14, 169–181. antigenic stimulation. Infect. Immun. 2, 96–100.
Lecuyer, E., Rakotobe, S., Lengline-Garnier, H., et al., 2014. Segmented filamentous Pott, J., Stockinger, S., Torow, N., et al., 2012. Age-dependent TLR3 expression of the
bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and intestinal epithelium contributes to rotavirus susceptibility. PLoS Pathog. 8,
specific T helper 17 cell responses. Immunity 40, 608–620. e1002670.
Lee, Y., Awasthi, A., Yosef, N., et al., 2012. Induction and molecular signature of Qin, H., Wang, L., Feng, T., et al., 2009. TGF-beta promotes Th17 cell development
pathogenic TH17 cells. Nat. Immunol. 13, 991–999. through inhibition of SOCS3. J. Immunol. 183, 97–105.
Lee, Y.K., Turner, H., Maynard, C.L., et al., 2009. Late developmental plasticity in the Rakoff-Nahoum, S., Paglino, J., Eslami-Varzaneh, F., Edberg, S., Medzhitov, R., 2004.
T helper 17 lineage. Immunity 30, 92–107. Recognition of commensal microflora by toll-like receptors is required for intestinal
Lefrancois, L., Goodman, T., 1989. In vivo modulation of cytolytic activity and Thy-1 homeostasis. Cell 118, 229–241.
expression in TCR-gamma deltaþ intraepithelial lymphocytes. Science 243, Randall, T.D., Mebius, R.E., 2014. The development and function of mucosal lymphoid
1716–1718. tissues: a balancing act with micro-organisms. Mucosal Immunol. 7, 455–466.
Lin, A., Bik, E.M., Costello, E.K., et al., 2013. Distinct distal gut microbiome diversity Robinette, M.L., Fuchs, A., Cortez, V.S., et al., 2015. Transcriptional programs define
and composition in healthy children from Bangladesh and the United States. PLoS molecular characteristics of innate lymphoid cell classes and subsets. Nat.
One 8, e53838. Immunol. 16, 306–317.
Liu, H.P., Cao, A.T., Feng, T., et al., 2015. TGF-beta converts Th1 cells into Th17 cells Rosser, E.C., Oleinika, K., Tonon, S., et al., 2014. Regulatory B cells are induced by
through stimulation of Runx1 expression. Eur. J. Immunol. 45, 1010–1018. gut microbiota-driven interleukin-1beta and interleukin-6 production. Nat. Med. 20,
Lochner, M., Ohnmacht, C., Presley, L., et al., 2011. Microbiota-induced tertiary 1334–1339.
lymphoid tissues aggravate inflammatory disease in the absence of RORgamma t Round, J.L., Lee, S.M., Li, J., et al., 2011. The Toll-like receptor 2 pathway estab-
and LTi cells. J. Exp. Med. 208, 125–134. lishes colonization by a commensal of the human microbiota. Science 332,
Lotz, M., Gutle, D., Walther, S., et al., 2006. Postnatal acquisition of endotoxin 974–977.
tolerance in intestinal epithelial cells. J. Exp. Med. 203, 973–984. Round, J.L., Mazmanian, S.K., 2009. The gut microbiota shapes intestinal immune
Lundell, A.C., Bjornsson, V., Ljung, A., et al., 2012. Infant B cell memory differentiation responses during health and disease. Nat. Rev. Immunol. 9, 313–323.
and early gut bacterial colonization. J. Immunol. 188, 4315–4322. Round, J.L., Mazmanian, S.K., 2010. Inducible Foxp3þ regulatory T-cell development
Lundell, A.C., Rabe, H., Quiding-Jarbrink, M., et al., 2011. Development of gut-homing by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci.
receptors on circulating B cells during infancy. Clin. Immunol. 138, 97–106. U.S.A. 107, 12204–12209.
Macpherson, A.J., Gatto, D., Sainsbury, E., et al., 2000. A primitive T cell-independent Sanos, S.L., Bui, V.L., Mortha, A., et al., 2009. RORgammat and commensal
mechanism of intestinal mucosal IgA responses to commensal bacteria. Science microflora are required for the differentiation of mucosal interleukin 22-producing
288, 2222–2226. NKp46þ cells. Nat. Immunol. 10, 83–91.
Macpherson, A.J., Harris, N.L., 2004. Interactions between commensal intestinal Satoh-Takayama, N., Vosshenrich, C.A., Lesjean-Pottier, S., et al., 2008. Microbial
bacteria and the immune system. Nat. Rev. Immunol. 4, 478–485. flora drives interleukin 22 production in intestinal NKp46þ cells that provide innate
Macpherson, A.J., Mccoy, K.D., Johansen, F.E., Brandtzaeg, P., 2008. The immune mucosal immune defense. Immunity 29, 958–970.
geography of IgA induction and function. Mucosal Immunol. 1, 11–22. Sawa, S., Lochner, M., Satoh-Takayama, N., et al., 2011. RORgammatþ innate
Maloy, K.J., Powrie, F., 2011. Intestinal homeostasis and its breakdown in inflam- lymphoid cells regulate intestinal homeostasis by integrating negative signals from
matory bowel disease. Nature 474, 298–306. the symbiotic microbiota. Nat. Immunol. 12, 320–326.
Mangan, P.R., Harrington, L.E., O’quinn, D.B., et al., 2006. Transforming growth Schmidt, C., 2015. Mental health: thinking from the gut. Nature 518, S12–S15.
factor-beta induces development of the T(H)17 lineage. Nature 441, 231–234. Schmidt, A., Oberle, N., Krammer, P.H., 2012. Molecular mechanisms of Treg-
Maslowski, K.M., Vieira, A.T., Ng, A., et al., 2009. Regulation of inflammatory mediated T cell suppression. Front. Immunol. 3, 51.
responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, Schnupf, P., Gaboriau-Routhiau, V., Gros, M., et al., 2015. Growth and host interaction
1282–1286. of mouse segmented filamentous bacteria in vitro. Nature 520, 99–103.
Physiology of the Immune System j Role of the Microbiota in Immune Development 119

Shaw, M.H., Kamada, N., Kim, Y.G., Nunez, G., 2012. Microbiota-induced IL-1beta, Tsuji, M., Suzuki, K., Kitamura, H., et al., 2008. Requirement for lymphoid tissue-
but not IL-6, is critical for the development of steady-state TH17 cells in the inducer cells in isolated follicle formation and T cell-independent immunoglobulin
intestine. J. Exp. Med. 209, 251–258. A generation in the gut. Immunity 29, 261–271.
Sheikh, S.Z., Matsuoka, K., Kobayashi, T., et al., 2010. Cutting edge: IFN-gamma is Vaishnava, S., Yamamoto, M., Severson, K.M., et al., 2011. The antibacterial lectin
a negative regulator of IL-23 in murine macrophages and experimental colitis. RegIIIgamma promotes the spatial segregation of microbiota and host in the
J. Immunol. 184, 4069–4073. intestine. Science 334, 255–258.
Shroff, K.E., Meslin, K., Cebra, J.J., 1995. Commensal enteric bacteria engender Van der Sluis, M., De Koning, B.A., De Bruijn, A.C., et al., 2006. Muc2-deficient mice
a self-limiting humoral mucosal immune-response while permanently colonizing the spontaneously develop colitis, indicating that MUC2 is critical for colonic protection.
gut. Infect. Immun. 63, 3904–3913. Gastroenterology 131, 117–129.
Smith, P.M., Howitt, M.R., Panikov, N., et al., 2013. The microbial metabolites, Walker, J.A., Barlow, J.L., Mckenzie, A.N., 2013. Innate lymphoid cells – how did we
short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, miss them? Nat. Rev. Immunol. 13, 75–87.
569–573. Wu, H.J., Ivanov, I.I., Darce, J., et al., 2010. Gut-residing segmented filamentous
Smythies, L.E., Sellers, M., Clements, R.H., et al., 2005. Human intestinal macro- bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32, 815–827.
phages display profound inflammatory anergy despite avid phagocytic and bac- Yang, X.O., Nurieva, R., Martinez, G.J., et al., 2008a. Molecular antagonism and
teriocidal activity. J. Clin. Invest. 115, 66–75. plasticity of regulatory and inflammatory T cell programs. Immunity 29, 44–56.
Sonnenberg, G.F., Artis, D., 2012. Innate lymphoid cell interactions with microbiota: Yang, X.O., Pappu, B.P., Nurieva, R., et al., 2008b. T helper 17 lineage differentiation
implications for intestinal health and disease. Immunity 37, 601–610. is programmed by orphan nuclear receptors ROR alpha and ROR gamma. Immunity
Sonnenberg, G.F., Mjosberg, J., Spits, H., Artis, D., 2013. SnapShot: innate lymphoid 28, 29–39.
cells. Immunity 39, 622–622. e1. Yang, Y., Torchinsky, M.B., Gobert, M., et al., 2014. Focused specificity of intestinal
Sonnenberg, G.F., Monticelli, L.A., Elloso, M.M., Fouser, L.A., Artis, D., 2011. CD4(þ) TH17 cells towards commensal bacterial antigens. Nature 510, 152–156.
lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, Yokota, Y., Mansouri, A., Mori, S., et al., 1999. Development of peripheral lymphoid
122–134. organs and natural killer cells depends on the helix-loop-helix inhibitor Id2. Nature
Spits, H., Cupedo, T., 2012. Innate lymphoid cells: emerging insights in 397, 702–706.
development, lineage relationships, and function. Annu. Rev. Immunol. 30, Zenewicz, L.A., Yin, X., Wang, G., et al., 2013. IL-22 deficiency alters colonic
647–675. microbiota to be transmissible and colitogenic. J. Immunol. 190, 5306–5312.
Spits, H., Di Santo, J.P., 2011. The expanding family of innate lymphoid cells: Zhou, L., Chong, M.M., Littman, D.R., 2009. Plasticity of CD4þ T cell lineage
regulators and effectors of immunity and tissue remodeling. Nat. Immunol. 12, differentiation. Immunity 30, 646–655.
21–27.
Steinman, R.M., Hawiger, D., Nussenzweig, M.C., 2003. Tolerogenic dendritic cells.
Annu. Rev. Immunol. 21, 685–711. Further Reading
Strachan, D.P., 1989. Hay fever, hygiene, and household size. BMJ 299,
1259–1260.
Abreu, M.T., 2010. Toll-like receptor signalling in the intestinal epithelium: how
Strober, W., Fuss, I.J., 2011. Proinflammatory cytokines in the pathogenesis of
bacterial recognition shapes intestinal function. Nat. Rev. Immunol. 10, 131–144.
inflammatory bowel diseases. Gastroenterology 140, 1756–1767.
Hooper, L.V., Littman, D.R., Macpherson, A.J., 2012. Interactions between the
Sun, C.M., Hall, J.A., Blank, R.B., et al., 2007. Small intestine lamina propria dendritic
microbiota and the immune system. Science 336, 1268–1273.
cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J. Exp.
Kamada, N., Seo, S.U., Chen, G.Y., Nunez, G., 2013. Role of the gut microbiota in
Med. 204, 1775–1785.
immunity and inflammatory disease. Nat. Rev. Immunol. 13, 321–335.
Thelemann, C., Eren, R.O., Coutaz, M., et al., 2014. Interferon-gamma induces
Mowat, A.M., Agace, W.W., 2014. Regional specialization within the intestinal immune
expression of MHC class II on intestinal epithelial cells and protects mice from
system. Nat. Rev. Immunol. 14, 667–685.
colitis. PLoS One 9, e86844.
Peterson, L.W., Artis, D., 2014. Intestinal epithelial cells: regulators of barrier function
Torow, N., Yu, K., Hassani, K., et al., 2015. Active suppression of intestinal CD4(þ)
and immune homeostasis. Nat. Rev. Immunol. 14, 141–153.
TCRalphabeta(þ) T-lymphocyte maturation during the postnatal period. Nat.
Saleh, M., Elson, C.O., 2011. Experimental inflammatory bowel disease: insights into
Commun. 6, 7725.
the host-microbiota dialog. Immunity 34, 293–302.
Tsuji, M., Komatsu, N., Kawamoto, S., et al., 2009. Preferential generation of follicular
Underhill, D.M., Iliev, I.D., 2014. The mycobiota: interactions between commensal
B helper T cells from Foxp3þ T cells in gut Peyer’s patches. Science 323,
fungi and the host immune system. Nat. Rev. Immunol. 14, 405–416.
1488–1492.

You might also like