International Immunology International Immunology Advance Access published May 28, 2014

Journal:

INTIMM-14-0071

r Fo

Manuscript ID:

International Immunology

Manuscript Type: Date Submitted by the Author:

Complete List of Authors:

27-Apr-2014

Suzuki, Keiichiro; Kyoto University, Graduate School of Medicine Nakajima, Akira; Kyoto University, Graduate School of Medicine

Pe

Keywords:

Invited Review

IgA, gut microbiota, homeostasis

er ew

vi

Re

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

New aspects of IgA synthesis in the gut

Page 1 of 24

International Immunology

New aspects of IgA synthesis in the gut

Keiichiro Suzuki1 and Akira Nakajima1 1

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Sakyo-ku, Kyoto 606-8501, Japan,

Corresponding author: Keiichiro Suzuki

r Fo

Email: [email protected] Fax: +81-75-753-9500 Tel: +81-75-753-9526

Pe

Running title: Regulation of gut homeostasis by IgA

er

Kyewords: IgA, gut microbiota, homeostasis

ew

vi

Re

The number of Figures: two Figures

1

International Immunology

Page 2 of 24

Abstract In mammals, the gastrointestinal tract is colonized by extremely dense and diverse bacterial communities that are beneficial for health. Maintenance of complexity and

when disrupted, the microbial community attacks the host’s tissues and cause inflammatory reactions. Our immune system provides the necessary mechanisms to maintain the homeostatic balance between microbial communities and the host. The

r Fo

immunoglobulin A (IgA) plays crucial roles in regulation of host-bacterial interactions in the gut. IgA is the most abundant immunoglobulin (Ig) isotype in our body, mostly produced by the IgA plasma cells residing in the lamina propria of the

Pe

small and large intestine. Although it was well known that IgA provides protection against pathogens, only recently it became clear that IgA plays critical roles in

er

regulation of bacterial communities in the gut in steady-state conditions. Here we

Re

summarize recent progress in our understandings for the various mechanisms of IgA synthesis in multiple anatomical sites and discuss how IgA limits bacterial access to

ew

vi

the internal milieu of the host.

2

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

proper localization and distribution of gut bacteria is of prime importance, because

Page 3 of 24

International Immunology

Introduction More than a half-century has passed since IgA was found as the most predominant class of antibody in mucosal secretions (1, 2). The observation that IgA but not IgG is

mucosal immune system. Pioneering works done by John Cebra and coworkers provided the cellular basis for the origin of IgA producing cells and the evidence for the involvement of gut bacteria in maturation of the immune system (3, 4). By using

r Fo

the transfer model in rabbit, they showed that Peyer’s patch (PP) cells were much more efficient than peripheral lymph node (pLN) cells to repopulate in small intestine of the transferred hosts (3). In addition, their experiments with germ free mice showed

Pe

that germinal centers (GCs) formation in PPs and subsequent IgA secretion efficiently occurred only in response to the bacterial colonization in the intestine (4). These and

er

many other subsequent studies led to the concept that the precursors of IgA plasma

Re

cells are generated in PP GCs by T cell-dependent mechanisms triggered by commensal bacteria. However, recent observations have revealed that the sites and

vi

mechanisms for the induction of IgA+ cells are multi-layered (5); IgA+ cells are also induced in isolated lymphoid follicles (ILFs) and in non-organized lamina propria

ew

(LP). In these anatomical sites, T cells are dispensable for the generation of IgA+ cells (5).

Although the IgAs generated by different mechanisms may play different roles in distinct contexts, the primary function of IgA is to maintain homeostasis at the mucosal surface. IgA was originally recognized as an important factor to protect the host from mucosal pathogens. More recently, we begin to appreciate that IgA plays an important role in steady-state conditions in the gut (6). Technical advances in the analysis of complex microbial community rapidly expanded our knowledge about the

3

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

secreted across the mucosal surfaces attracted attentions for the uniqueness of

International Immunology

Page 4 of 24

IgA-dependent mechanisms for the maintenance of microbial community in the gut. In this review, we discuss the critical role of IgA for the maintenance of “healthy” gut microbiota and emphasize the mechanisms responsible for IgA synthesis in the gut .

The gut harbors more than 1000 species of bacteria that outnumber our own cells by a factor of 10. Since most of the normal gut bacteria are uncultured, the studies

r Fo

based on culture-dependent experiments did not give an accurate view of their composition. A more precise characterization became possible only recently with the development of culture-independent molecular approaches based on bacterial

Pe

sequencing, (including the largely used sequencing of bacterial 16S rRNA genes) (7, 8). Such technical development allowed us to evaluate in detail the composition of

er

microbial communities, and to monitor how they change with conditions of the hosts.

Re

One powerful driving force for shaping gut microbiota appears to be dietary habit of the hosts (9-12) (Fig1). Comparative analysis revealed that humans harbor a

vi

representative gut microbiota of omnivorous primates that is clearly distinct from the microbial community of carnivores and herbivores (9). Within different human

ew

populations, European children taking typical “western” diet have clearly distinctive bacterial community from rural African children who mainly take plant-based diet, suggesting that long-term dietary intake may have a significant effect on the composition of the gut microbiota (10). Even short-term dietary interventions can quickly change gut microbial community. For example, changing the diet from high fiber diet to high fat diet drastically altered within several days the bacterial composition and their metabolic activity such as the production of short chain fatty acids (SCFAs) in the gut (11, 12). These changes seem to deeply impact on the host’s

4

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

1. IgA-independent regulation of gut microbiota

Page 5 of 24

International Immunology

immune system, including the production of pro-inflammatory cytokines such as IFNγ, IL1-β, IL-22, IL-23, and possibly the maintenance of regulatory T cells (Tregs) (1318).

inflammatory responses at bay provides another layer of microbiota regulation (Fig1). For example, NLRP6 inflammasome pathway in intestinal epithelial cells inhibits the development of colitis by IL-18 dependent regulation of some colitogenic

r Fo

commensals such as Prevotellaceae and TM7, or by clearing pathogens through mucus secretions by goblet cells (discussed by Flavell in this issue) (19, 20). Another example is that the expression of T-bet in innate immune system prevents the

Pe

expansion of Klebsiella pneumoniae and Proteus mirabilis probably through the function of dendritic cells, thus avoiding inflammatory responses in the colon (21).

er

IL-22 produced by innate lymphoid cells was also reported to be involved in

Re

regulation of the gut microbiota such as segmented filamentous bacteria (SFB), and avoiding the systemic dissemination of Alcaligenes species (discussed by Sonnenberg

vi

in this issue) (15, 22). Thus, the innate immune system by regulating the local

commensal microbiota.

ew

cytokine milieu contributes to modulation of “pathogenic” stimulus derived from

2. Regulation of gut microbiota by antigen specific IgA Acquired immune system has a non-redundant role in regulation of host-microbial interactions at mucosal barrier. Thus, production and secretion of IgA play an important role for the regulation of gut microbiota that cannot be achieved by the diet or the innate immune system (Fig1). The first evidence that secretory IgA play critical roles in regulating gut microbiota came from the studies of activation-induced

5

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

In addition to diet, the innate immune system by keeping the undesirable

International Immunology

Page 6 of 24

cytidine deaminase (AID)-deficient mice, which lack the key enzyme required for class-switch recombination (CSR) and somatic hypermutation (SHM) of the Ig genes (23-25). In the absence of IgA, culturable bacteria in AID-/- small intestine expanded

also changed, with SFB aberrantly expanding in all segments of the small intestine (25). The expansion of SFB led to over-activation of not only the gut immune system, but also the whole body immune system. When IgA-producing cells were

r Fo

reconstituted in AID-/- mice upon parabiosis with normal mice, the amount and composition of gut microbiota were normalized (25). Although SFB expansion was reported to induce mostly differentiation of T cells into Th17 cells (26), the

Pe

composition of Th17 cells in the gut LP of AID-/- mice was equivalent to those of normal mice, suggesting that the over-activated phenotype of AID-deficiency is not

er

due to the activation of Th17 cells (our unpublished observation).

Re

Since AID is required both for CSR and SHM, the studies with AID-/- mice did not dissect whether the simple presence/absence or the quality of IgA is important for the

vi

regulation of gut bacterial communities and in general for the body homeostasis. The study with AIDG23S, a mutant from AID that has normal CSR but impaired SHM

ew

activity, addressed this issue (27). The knock-in mice expressing AIDG23S had the normal concentration of serum and fecal IgA (as well as other Ig isotypes), but had a limited diversity of the Ig repertoire due SHM defect. However, similar to AID-/mice, AIDG23S mice showed expansion of culturable bacteria in their small intestine, and systemic over-activation with induction of GCs in peripheral lymphoid tissues (27). These results clearly indicated that SHM and selection of IgAs are needed for specific recognition of the bacterial epitope to regulate their amount and composition of microbial community in the gut.

6

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

more than 100 fold (24). Moreover, the composition of uncultured gut microbiota was

Page 7 of 24

International Immunology

The diversification of IgA repertoire by SHM occurs mostly in the PP GCs and the formation of GCs depends on the T cells help. Most of the T cells present in the GCs are CD4+ T cells generally known as follicular B helper T cells, of TFH cells. TFH cells

sustain GC reactions. Interestingly, TFH cells also express high amount of the inhibitory receptor programmed cell death-1 (PD-1), a molecule that also regulate the several aspects of GC biology. Thus, the absence of PD-1 led to considerable

r Fo

expansion of PP TFH cells with different properties (i.e producing more IFN-γ and less IL-21) that caused impaired selection of GC B cells. Thus, PD-1-/- mice had reduced affinity maturation of IgA-producing plasma cells, and less IgA binding capacity to

Pe

gut bacteria (28). Associated with reduced IgA coating of bacteria, PD-1-/- mice had a marked reduction in the number of “healthy” gut bacteria, such as Bifidobacterium. In

er

contrast, the number of Enterobacteriaceae was significantly increased in PD-1-/-

Re

mice. Like in AID-deficient or AIDG23S mice, the gut dysbiosis induced overactivation of the systemic immune system, which led to T and B cell hyperplasia in

vi

PD-1-/- mice. The systemic activation could be rescued by antibiotic treatment (28).

ew

These observations clearly indicate that the T cell-dependent selection of bacteriaspecific B cells in PP GCs is critical for regulating symbiotic relationships with the gut microbiota leading to homeostasis.

IgA not only coat the bacteria, but can modulate bacterial epitopes and even change the bacterial metabolism. By using mono-colonization system, it was demonstrated that Bacteroides thetaiotaomicron elicits less pro-inflammatory signals in the small intestine in the presence of a specific gut IgA that recognize capsular polysaccharide CPS4 (29). The bacteria evaded anti-CPS4 IgA response by inducing alternative expression of another capsular locus, CPS5. Thus, not only that IgA is required to

7

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

are characterized by expression of co-stimulatory molecules and cytokines required to

International Immunology

Page 8 of 24

regulate the amount and composition of gut microbiota, but that the adaptive immune system drives diversification of bacterial surface structures by exposing their antigenic epitope(s) to appropriately selected IgA.

Although it is clear that the T cell-dependent selection of antigen specific B cells in PP GCs is critical to regulate gut microbiota, significant numbers of IgA+ cells are

r Fo

generated outside of GCs even in the absence of T cells (30). Many of these cells originate from B1 cells that reside in the peritoneal cavity. This was shown in lethally irradiated T cell-deficient mice, reconstituted with allotype-marked B1 cells together

Pe

with host-derived bone marrow cells (31). Many of B1-derived IgA+ cells do not pass through T cell-dependent selection in GCs. However, IgA secreted from these cells

er

can abundantly coat the surface of gut bacteria even with the germ line-encoded VH

Re

genes (32). Secretory IgA may naturally bind on bacteria through their heavy glycosylation on secretory components or on the heavy chains (33, 34). By coating in

vi

abundance the bacterial surfaces, innate IgA produced by B1-derived plasma cells may play an important role for preventing systemic dissemination of commensal

ew

bacteria (Fig1). Indeed, T cell-deficient mice that produce B1-derived gut IgA do not have serum IgA or IgG with the same specificities (31). By contrast, mice with IgA deficiency (IgA-/- mice and alymphoplasia aly/aly mice) have serum IgGs that are specific for commensal bacteria produced with the help of T cells (31). Coating of gut bacteria by non-selected “natural” IgA appears to protect neonatal mucosa from abrupt encounter with commensals. Pups exchanging experiments showed that SFB colonization and subsequent generation of IgA+ cells in the small intestine occurs earlier in the pups nursed by SCID females that lack milk Ig (35).

8

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

3. Innate function of IgA

Page 9 of 24

International Immunology

Similarly, normal pups nursed by JH-/- mother that lack mature B cells and Ig had early induction of serum IgA (36). Milk Ig and pups-derived IgA prevented the penetration of bacteria through the intestinal epithelium even without diversification

nitrophenol-specific IgA produced in young quasi-monoclonal mice was sufficient to limit the penetration of commensal Enterobacter cloacae (36). Non-specific IgA coating on bacteria is also functionally important in adult gut. This

r Fo

is supported by the following observations. First, the invasion of Salmonella typhimurium to epithelial cells was increased in polymeric immunoglobulin receptor deficient (pIgR-/-) mice, which lack IgA secretion (37). Secretory IgA in naive mice

Pe

inhibited this process without developing high-affinity antibodies that recognize surface epitopes of S. typhimurium. Second, pIgR-/- mice showed exacerbation of

er

chemical colitis induced by exposure to dextran sodium sulfate (DSS) depend on the

Re

presence of intestinal microbiota (38). Third, similar to neonatal mice, the presence of secretory IgA limits the translocation of aerobic bacteria from intestinal lumen to

vi

mesenteric lymph nodes (mLNs) (39). Intestinal DCs carry living luminal bacteria to mLN and prevent further dissemination into the systemic immune compartment

ew

through MyD88-dependent Toll-like receptor (TLR) signaling. These DCs have capacity to induce IgA production in the gut (39-41). Thus, it seems that the presence of intestinal IgA and bacterial access to DCs are tightly correlated each other, and may be considered as the first and the second gateway respectively to prevent bacterial penetration into the deeper tissues (Fig1).

4. Impact of gut microbiota and other environmental factors on IgA synthesis 4-1. Generation of IgA+ cells in PP GCs

9

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

of the antibody repertoire. This was shown by the observation that the non-mutated

International Immunology

Page 10 of 24

One of the most striking response to the colonization of commensal microbiota is the formation of PP GCs (4). In conventional mice, GCs are continuously found in PPs because of constant stimulation by gut bacteria. Interestingly, GCs in PPs but not in

receptors, given that B cells are activated by bacteria through a TLR signaling pathway (42). GC B cells in PPs are also unique in the sense that they undergo CSR almost

r Fo

exclusively to IgA. We found that the stimulation through bacterial and food-derived factors on stromal cells in PP GCs, i.e. follicular dendritic cells (FDCs) play an important role for the CSR to IgA (43). FDCs express TLRs and retinoic acid

Pe

receptors (RARs) and stimulation through these receptors by bacterial and foodderived products induce large amounts of active TGF-β1 and BAFF (Fig2). Indeed,

er

conditioned media from PP FDCs supports more efficiently the generation of IgA+ B

Re

cells and IgA plasmablasts than pLN FDCs by mechanisms that involves the TGF-β1 activation and BAFF production (43). In addition, PP FDCs produce enhanced level

vi

of CXCL13, by which FDCs may contribute to the augmentation of recruitment and

ew

motility of GC B cells in PPs. A recent report of in vivo ablation of FDCs supports this idea; FDC depletion resulted in the reduced level of Cxcl13 transcripts, disorganized the follicular structures and reduced B cell motility (44). The correct positioning of activated CXCR5-expressing T cells also requires CXCL13 production by FDCs. Priming of naive T cells by DCs at the T-B border induce upregulation of CXCR5 and downregulation of CCR7 on T cells, and those T cells are directed to Bcell follicles via the gradients of CXCL13, and mature into TFH cells (45). Interestingly, TFH cells in PPs can be generated from Foxp3+ T cells with IgA helper properties (46) (Fig2). Upon the adoptive transfer into T cell- deficient mice, Foxp3+

10

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

pLNs can be induced in the absence of antigen-specific recognition by B cells

Page 11 of 24

International Immunology

T cells lose their Foxp3 expression, covert into TFH cells and efficiently induce GCs in the PPs and IgA plasma cells in the LP. The conversion of Foxp3+ cells into TFH cells is a gut-specific phenomenon, because this conversion was observed only in PPs

Thus, in PPs there seems to be a link between the Foxp3+ T cells, GCs and IgA+ cells. This is likely due to the unique microenvironment generated by the presence of bacteria and food-derived components, and also the products secreted by activated

r Fo

epithelial cells, DCs, B/T cells and stromal cells in the gut milieu (Fig2).

4-2. Generation of IgA+ cells outside of PP GCs

Pe

Some of IgA+ cells are also generated in LP and ILFs, albeit less efficient that in the PP GCs (5). Pre-switched IgM+ B cells are found either scattered in the villi or

er

forming aggregates called isolated lymphoid follicles (ILFs). The development of

Re

ILFs depends on the RORγt-expressing lymphoid tissue inducer (LTi) cells and their interaction via LT-LTβR with local stromal cells (47, 48). The ILF or LP B cells

ew

(Fig2).

vi

undergo CSR to IgA in situ upon their stimulation by commensal bacteria (47-49)

In LP and ILFs, B cells are directly activated by DCs that have access to luminal bacteria via extended dendrites through the tight junction of the epithelium, or through the transport of bacteria by microfold cells (M cells) (50, 51). Similar to PPs, the epithelium covering ILFs and intestinal villi differentiate into glycoprotein 2 (GP2)-expressing M cells through the stimulation of stromal-derived TNF superfamily member receptor activator of NF-κB ligand (RANKL) and transcription factor Spi-B (discussed by Ohno in this issue) (52-54).

11

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

and partially in mLNs but not in spleen even with the repeated immunizations (46).

International Immunology

Page 12 of 24

Upon activation by bacterial antigens, the DCs present in the LP or ILFs are sufficient to induce CSR to IgA even in the absence of T cells. Two major subsets of DCs are present in LP: CX3CR1+ DCs and CD103+ DCs (55). CX3CR1+ DCs are

possibly IL-10 (56, 57). CD103+ DCs also help in situ CSR probably through the TLR5-dependent production of retinoic acid (58). In addition, DCs in LP or ILFs produce large amount of TNF-α, and stimulate local stromal cells to release the active

r Fo

form of TGF-β1 via the effect of matrix metalloproteinase 9 (MMP9) and MMP13 (48). Interestingly, a subset of CD11c+ DCs called Tip-DCs and also a fraction of IgA+ plasma cells were found to be crucial for IgA induction by producing iNOS and

Pe

TNF-α in the gut (56, 59). Thus, interactions among bacteria, DCs, stromal cells and IgA-plasma cells likely generate a positive feedback loop to induce IgA+ cells in the

Re

5. Conclusion

er

local environment (Fig2).

vi

It is now clear that IgA is critical for the maintenance of a diversified microbial

ew

community in the gut. Accumulating evidence show that IgA producing cells are generated by multiple pathways, in organized and non-organized follicular structures, by T-dependent and T-independent mechanisms. However, the functional differences between IgAs generated in different anatomical sites by different mechanisms are still unclear. Also unknown are the mechanisms by which the antigen-specific IgAs regulate the amount and composition of microbial community, or how exactly the innate IgA inhibits host-bacterial interactions in the gut. Further analyses are required to understand the complex and dynamic interactions among IgA, bacteria and immune

12

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

resident cells that may regulate CSR to IgA through their production of BAFF and

Page 13 of 24

International Immunology

cells in the gut, which should pave the way to develop new strategies for the treatment of gut dysbiosis leading to inflammatory diseases or autoimmunity.

We thank to Sidonia Fagarasan for discussions and critically reading of the manuscript. This work was supported in part by the Special Coordination Funds for

r Fo

Promoting Science and Technology of the Japanese Government and in part by Astellas Pharma Inc. in the Formation of Innovation Center for Fusion of Advanced Technologies Program. Also supported in part by Grants-in-Aid for Scientific

Pe

Research from the Japanese Ministry of Education, Culture, Sports, Science and Technology, and Japan Society for the Promotion of Science to K.S.

er ew

vi

Re 13

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

ACKNOWLEDGMENTS

International Immunology

Page 14 of 24

REFERENCES 1

Chodirker W.B., and Tomasi T.B., Jr. 1963. Gamma-Globulins: Quantitative

Relationships in Human Serum and Nonvascular Fluids. Science. 142:1080-1081. Crabbe P.A., Carbonara A.O., and Heremans J.F. 1965. The Normal Human

Intestinal Mucosa as a Major Source of Plasma Cells Containing Gamma-aImmunoglobulin. Lab Invest. 14:235-248. 3

Craig S.W., and Cebra J.J. 1971. Peyer's patches: an enriched source of

4

r Fo

precursors for IgA-producing immunocytes in the rabbit. J Exp Med. 134:188-200. Cebra J.J. 1999. Influences of microbiota on intestinal immune system

development. Am J Clin Nutr. 69:1046S-1051S.

Pe

5

Fagarasan S., Kawamoto S., Kanagawa O., and Suzuki K. 2010. Adaptive

immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis.

Kato L.M., Kawamoto S., Maruya M., and Fagarasan S. 2014. Gut TFH and

Re

6

er

Annu Rev Immunol. 28:243-273.

IgA: key players for regulation of bacterial communities and immune homeostasis.

7

vi

Immunol Cell Biol. 92:49-56.

Backhed F., Ley R.E., Sonnenburg J.L., Peterson D.A., and Gordon J.I. 2005.

ew

Host-bacterial mutualism in the human intestine. Science. 307:1915-1920. 8

Ley R.E., Peterson D.A., and Gordon J.I. 2006. Ecological and evolutionary

forces shaping microbial diversity in the human intestine. Cell. 124:837-848. 9

Ley R.E., Hamady M., Lozupone C., et al. 2008. Evolution of mammals and

their gut microbes. Science. 320:1647-1651. 10

De Filippo C., Cavalieri D., Di Paola M., et al. 2010. Impact of diet in shaping

gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 107:14691-14696.

14

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

2

Page 15 of 24

International Immunology

11

Turnbaugh P.J., Ridaura V.K., Faith J.J., Rey F.E., Knight R., and Gordon J.I.

2009. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 1:6ra14. David L.A., Maurice C.F., Carmody R.N., et al. 2014. Diet rapidly and

reproducibly alters the human gut microbiome. Nature. 505:559-563. 13

Devkota S., Wang Y., Musch M.W., et al. 2012. Dietary-fat-induced

taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature.

14

r Fo

487:104-108.

Yoshimoto S., Loo T.M., Atarashi K., et al. 2013. Obesity-induced gut

microbial metabolite promotes liver cancer through senescence secretome. Nature.

15

Pe

499:97-101.

Upadhyay V., Poroyko V., Kim T.J., et al. 2012. Lymphotoxin regulates

er

commensal responses to enable diet-induced obesity. Nat Immunol. 13:947-953. Smith P.M., Howitt M.R., Panikov N., et al. 2013. The microbial metabolites,

Re

16

short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 341:569-573. Arpaia N., Campbell C., Fan X., et al. 2013. Metabolites produced by

vi

17

commensal bacteria promote peripheral regulatory T-cell generation. Nature.

18

ew

504:451-455.

Furusawa Y., Obata Y., Fukuda S., et al. 2013. Commensal microbe-derived

butyrate induces the differentiation of colonic regulatory T cells. Nature. 19

Elinav E., Strowig T., Kau A.L., et al. 2011. NLRP6 inflammasome regulates

colonic microbial ecology and risk for colitis. Cell. 145:745-757. 20

Wlodarska M., Thaiss C.A., Nowarski R., et al. 2014. NLRP6 Inflammasome

Orchestrates the Colonic Host-Microbial Interface by Regulating Goblet Cell Mucus Secretion. Cell. 156:1045-1059.

15

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

12

International Immunology

21

Page 16 of 24

Garrett W.S., Gallini C.A., Yatsunenko T., et al. 2010. Enterobacteriaceae act

in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe. 8:292-300. Sonnenberg G.F., Monticelli L.A., Alenghat T., et al. 2012. Innate lymphoid

cells promote anatomical containment of lymphoid-resident commensal bacteria. Science. 336:1321-1325. 23

Muramatsu M., Kinoshita K., Fagarasan S., Yamada S., Shinkai Y., and Honjo

r Fo

T. 2000. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell. 102:553-563. 24

Fagarasan S., Muramatsu M., Suzuki K., Nagaoka H., Hiai H., and Honjo T.

Pe

2002. Critical roles of activation-induced cytidine deaminase (AID) in the homeostasis of gut flora. Science. 298:1424-1427.

er

25

Suzuki K., Meek B., Doi Y., et al. 2004. Aberrant expansion of segmented

26

Re

filamentous bacteria in IgA-deficient gut. Proc Natl Acad Sci U S A. 101:1981-1986. Ivanov, II, Atarashi K., Manel N., et al. 2009. Induction of intestinal Th17

27

vi

cells by segmented filamentous bacteria. Cell. 139:485-498.

Wei M., Shinkura R., Doi Y., Maruya M., Fagarasan S., and Honjo T. 2011.

ew

Mice carrying a knock-in mutation of Aicda resulting in a defect in somatic hypermutation have impaired gut homeostasis and compromised mucosal defense. Nat Immunol. 12:264-270. 28

Kawamoto S., Tran T.H., Maruya M., et al. 2012. The inhibitory receptor PD-

1 regulates IgA selection and bacterial composition in the gut. Science. 336:485-489. 29

Peterson D.A., McNulty N.P., Guruge J.L., and Gordon J.I. 2007. IgA

response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe. 2:328-339.

16

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

22

Page 17 of 24

International Immunology

30

Suzuki K., Maruya M., Kawamoto S., and Fagarasan S. 2010. Roles of B-1

and B-2 cells in innate and acquired IgA-mediated immunity. Immunol Rev. 237:180190. Macpherson A.J., Gatto D., Sainsbury E., Harriman G.R., Hengartner H., and

Zinkernagel R.M. 2000. A primitive T cell-independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science. 288:2222-2226. 32

Bos N.A., Bun J.C., Popma S.H., et al. 1996. Monoclonal immunoglobulin A

r Fo

derived from peritoneal B cells is encoded by both germ line and somatically mutated VH genes and is reactive with commensal bacteria. Infect Immun. 64:616-623. 33

Royle L., Roos A., Harvey D.J., et al. 2003. Secretory IgA N- and O-glycans

Pe

provide a link between the innate and adaptive immune systems. J Biol Chem. 278:20140-20153.

er

34

Mathias A., and Corthesy B. 2011. Recognition of gram-positive intestinal

Re

bacteria by hybridoma- and colostrum-derived secretory immunoglobulin A is mediated by carbohydrates. J Biol Chem. 286:17239-17247. Jiang H.Q., Bos N.A., and Cebra J.J. 2001. Timing, localization, and

vi

35

persistence of colonization by segmented filamentous bacteria in the neonatal mouse

ew

gut depend on immune status of mothers and pups. Infect Immun. 69:3611-3617. 36

Harris N.L., Spoerri I., Schopfer J.F., et al. 2006. Mechanisms of neonatal

mucosal antibody protection. J Immunol. 177:6256-6262. 37

Wijburg O.L., Uren T.K., Simpfendorfer K., Johansen F.E., Brandtzaeg P.,

and Strugnell R.A. 2006. Innate secretory antibodies protect against natural Salmonella typhimurium infection. J Exp Med. 203:21-26. 38

Reikvam D.H., Derrien M., Islam R., et al. 2012. Epithelial-microbial

crosstalk in polymeric Ig receptor deficient mice. Eur J Immunol. 42:2959-2970.

17

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

31

International Immunology

39

Page 18 of 24

Macpherson A.J., and Uhr T. 2004. Induction of protective IgA by intestinal

dendritic cells carrying commensal bacteria. Science. 303:1662-1665. 40

Slack E., Hapfelmeier S., Stecher B., et al. 2009. Innate and adaptive

325:617-620. 41

Diehl G.E., Longman R.S., Zhang J.X., et al. 2013. Microbiota restricts

trafficking of bacteria to mesenteric lymph nodes by CX(3)CR1(hi) cells. Nature.

42

r Fo

494:116-120.

Casola S., Otipoby K.L., Alimzhanov M., et al. 2004. B cell receptor signal

strength determines B cell fate. Nat Immunol. 5:317-327.

Pe

43

Suzuki K., Maruya M., Kawamoto S., et al. 2010. The sensing of

environmental stimuli by follicular dendritic cells promotes immunoglobulin A

er

generation in the gut. Immunity. 33:71-83.

Wang X., Cho B., Suzuki K., et al. 2011. Follicular dendritic cells help

Re

44

establish follicle identity and promote B cell retention in germinal centers. J Exp Med.

45

vi

208:2497-2510.

Haynes N.M., Allen C.D., Lesley R., Ansel K.M., Killeen N., and Cyster J.G.

ew

2007. Role of CXCR5 and CCR7 in follicular Th cell positioning and appearance of a programmed cell death gene-1high germinal center-associated subpopulation. J Immunol. 179:5099-5108. 46

Tsuji M., Komatsu N., Kawamoto S., et al. 2009. Preferential generation of

follicular B helper T cells from Foxp3+ T cells in gut Peyer's patches. Science. 323:1488-1492.

18

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

immunity cooperate flexibly to maintain host-microbiota mutualism. Science.

Page 19 of 24

International Immunology

47

Suzuki K., Meek B., Doi Y., Honjo T., and Fagarasan S. 2005. Two

distinctive pathways for recruitment of naive and primed IgM+ B cells to the gut lamina propria. Proc Natl Acad Sci U S A. 102:2482-2486. Tsuji M., Suzuki K., Kitamura H., et al. 2008. Requirement for lymphoid

tissue-inducer cells in isolated follicle formation and T cell-independent immunoglobulin A generation in the gut. Immunity. 29:261-271. 49

Fagarasan S., Kinoshita K., Muramatsu M., Ikuta K., and Honjo T. 2001. In

r Fo

situ class switching and differentiation to IgA-producing cells in the gut lamina propria. Nature. 413:639-643. 50

Rescigno M., Urbano M., Valzasina B., et al. 2001. Dendritic cells express

Pe

tight junction proteins and penetrate gut epithelial monolayers to sample bacteria. Nat Immunol. 2:361-367.

er

51

Jang M.H., Kweon M.N., Iwatani K., et al. 2004. Intestinal villous M cells: an

Re

antigen entry site in the mucosal epithelium. Proc Natl Acad Sci U S A. 101:61106115.

Hase K., Kawano K., Nochi T., et al. 2009. Uptake through glycoprotein 2 of

vi

52

FimH(+) bacteria by M cells initiates mucosal immune response. Nature. 462:226-

ew

230. 53

Knoop K.A., Kumar N., Butler B.R., et al. 2009. RANKL is necessary and

sufficient to initiate development of antigen-sampling M cells in the intestinal epithelium. J Immunol. 183:5738-5747. 54

Kanaya T., Hase K., Takahashi D., et al. 2012. The Ets transcription factor

Spi-B is essential for the differentiation of intestinal microfold cells. Nat Immunol. 13:729-736.

19

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

48

International Immunology

55

Page 20 of 24

Varol C., Zigmond E., and Jung S. 2010. Securing the immune tightrope:

mononuclear phagocytes in the intestinal lamina propria. Nat Rev Immunol. 10:415426. Tezuka H., Abe Y., Iwata M., et al. 2007. Regulation of IgA production by

naturally occurring TNF/iNOS-producing dendritic cells. Nature. 448:929-933. 57

Murai M., Turovskaya O., Kim G., et al. 2009. Interleukin 10 acts on

regulatory T cells to maintain expression of the transcription factor Foxp3 and

58

r Fo

suppressive function in mice with colitis. Nat Immunol. 10:1178-1184. Uematsu S., Fujimoto K., Jang M.H., et al. 2008. Regulation of humoral and

cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor

Pe

5. Nat Immunol. 9:769-776. 59

Fritz J.H., Rojas O.L., Simard N., et al. 2012. Acquisition of a multifunctional

er

IgA+ plasma cell phenotype in the gut. Nature. 481:199-203. He B., Xu W., Santini P.A., et al. 2007. Intestinal bacteria trigger T cell-

Re

60

independent immunoglobulin A(2) class switching by inducing epithelial-cell

61

vi

secretion of the cytokine APRIL. Immunity. 26:812-826.

Chu V.T., Beller A., Rausch S., et al. 2014. Eosinophils promote generation

ew

and maintenance of immunoglobulin-a-expressing plasma cells and contribute to gut immune homeostasis. Immunity. 40:582-593.

20

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

56

Page 21 of 24

International Immunology

Figure legends Figure 1. Regulation of gut microbiota and its feed-back to the host. There are at least three

different dietary habits (i.e. high fat and high sugar “western” diet versus high fiber and low fat “traditional” diet ) results in different bacterial community. Second, innate cytokines produced by epithelial cells (IL-18), innate lymphoid cells (ILCs; IL-22)

r Fo

and dendritic cells (DCs; TNF-α) play an important role to regulate some of bacteria species that may induce diseases such as colitis. Although the detailed mechanisms how these cytokines change the bacterial composition is unclear, the regulation of

Pe

epithelial-derived mucus and/or Paneth cells-derived anti-microbial peptides might be involved in this process. Third, secretory IgA that specifically recognize bacterial

er

epitopes are crucial for regulating the bacterial composition, by inhibiting their

Re

expansion and facilitating diversification, thus shaping “healthy” bacterial community in the gut. Significant amounts of secretory IgA bind on bacterial surfaces even in the

vi

absence of specificities to bacterial antigens. These are “innate IgAs” –mostly

ew

generated by B1 cells- that inhibit the attachment/penetration of bacteria to the epithelial cells, and thus prevent translocation of bacteria to the systemic immune compartment. Most of bacteria-specific IgAs are generated in germinal centers (GCs) of Peyer’s patches (PPs) with the help of TFH cells.

Figure 2. T cell-dependent and T cell-independent induction of IgA+ cells in the gut. In PPs, DCs located under specialized M cells take up bacteria and then activate T cells. The activated T cells express CXCR5 and migrate into B cell follicles along with CXCL13

21

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

major factors that contribute to shaping the commensal communities in the gut. First,

International Immunology

Page 22 of 24

gradient produced by FDCs. Some TFH cells are derived from Foxp3+ T cells that lose the Foxp3 expression. Upon activation and interaction with T cells, GC B cells express AID and undergo CSR and SHM. In the GC of PPs, B cells preferentially

FDCs producing TGF-β1 and BAFF upon their stimulation of TLRs and RARs. Unlike in the PPs, induction of IgA+ cells in ILFs and LP does not necessarily require help from T cells. Recruitment of IgM+ B cells from bone marrow (BM) or peritoneal

r Fo

cavity (PEC) into LP depends on the stromal cells (SCs) expressing functional LTβR. Local B cells are directly activated by DCs that take up luminal bacteria via the extended dendrites through tight junction, or via the M cells present on the epithelium

Pe

that covers the villi and ILFs. TNF-α is abundantly produced by iNOS-expressing

er

Tip DCs and a subset of IgA+ plasma cells. Upon TNF-α stimulation, SCs produce active form of TGF-β1 via the effect of matrix metalloproteinase (MMPs), which

Re

together with APRIL produced by ECs (and perhaps gut eosinophils) induce class switching of activated B cells from IgM to IgA(60, 61). IgA+ B cells differentiate into

vi

antibody-producing plasma cells upon the stimulation of IL-6, IL-10, BAFF and

ew

APRIL produced by SCs, DCs or epithelial cells.

22

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

switch their isotype from IgM to IgA, due to help of TFH cells producing IL-21 and

International Immunology Page 23 of 24

er Pe

ew vi Re

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

r Fo

209x148mm (300 x 300 DPI)

er Pe

ew vi Re

Downloaded from http://intimm.oxfordjournals.org/ at Polytechnic University. Bern Dibner Library of Science and Technology on September 2, 2014

r Fo

209x148mm (300 x 300 DPI)

Page 24 of 24 International Immunology

New aspects of IgA synthesis in the gut.

In mammals, the gastrointestinal tract is colonized by extremely dense and diverse bacterial communities that are beneficial for health. Maintenance o...
640KB Sizes 2 Downloads 3 Views