Journal List > Immune Netw > v.15(1) > 1033455

Ko and Chang: Regulation of Intestinal Immune System by Dendritic Cells

Abstract

Innate immune cells survey antigenic materials beneath our body surfaces and provide a front-line response to internal and external danger signals. Dendritic cells (DCs), a subset of innate immune cells, are critical sentinels that perform multiple roles in immune responses, from acting as principal modulators to priming an adaptive immune response through antigen-specific signaling. In the gut, DCs meet exogenous, non-harmful food antigens as well as vast commensal microbes under steady-state conditions. In other instances, they must combat pathogenic microbes to prevent infections. In this review, we focus on the function of intestinal DCs in maintaining intestinal immune homeostasis. Specifically, we describe how intestinal DCs affect IgA production from B cells and influence the generation of unique subsets of T cell.

Abbreviations

DC

dendritic cell

sIgA

secretory IgA

SI

small intestine

FcRn

neonatal Fc receptors

iNOS

inducible nitric oxide synthase

Tip DC

TNF-α/iNOS producing DC

pDC

plasmacytoid DC

Treg

regulatory T cell

GAP

goblet cell associated antigen passage

MLN

mesenteric lymph node

RALDH2

retinaldehyde dehydrogenase type 2

RA

retinoic acid

ILF

isolated lymphoid follicle

SFB

segmented filamentous bacteria

APRIL

a proliferation-inducing ligand

BAFF

B cell-activating factor belonging to the TNF family

CSR

class-switch DNA recombination

INTRODUCTION

Our body is covered with tight physical barriers of skin and mucosal tissues. Mucosal surfaces are constantly exposed to the external environment, which includes commensal microbes and exogenous antigens. When pathogenic microbes breach the surface barrier, surveillance systems beneath sense the trespassers and send an alarm to defense headquarters. Mucosal immune tissues comprise lymphoid organs associated with the gastro-intestinal tract (e.g., intestine, oral cavity and pharynx), respiratory tract, and urogenital tract, as well as the glands associated with these tissues, such as the salivary glands and lacrimal glands (1). The lactating breast is also a mucosal immune tissue. Mucosal immunity can maintain peaceful body surface by generating secretory IgA (sIgA) from B cells as well as priming specific T cell immunity. The intestine, especially, harbors an enormous community of commensal microorganisms that may contribute to host defense by enforcing the host's barrier function (2) or by competing against other microorganisms metabolically (3,4). Dendritic cells (DCs) or other phagocytic cells continuously survey the mucosal environment by using innate pattern recognition receptors and sample antigens prior to integrate adaptive immune system. These cells also can adjust suppressive regulation to innocuous antigens by inducing Tregs and keep distance to commensals by producing sIgA. Moreover, these cells protect against pathogenic invasion by generating various kinds of helper T (TH) and CD8+ T cells as well as helping to produce sIgA antibodies. Here, we provide an overview of the gut immune response, focusing on unique functional features of intestinal DCs and other phagocytic cells.

INTESTINAL DCs AND MACROPHAGE SUBSETS

Lamina propria DCs in the small intestine (SI) haves been well studied as one of the intestinal DC subsets. CD11c+ major histocompatibility (MHC) class II+ cells in the gut comprise DCs as well as phagocytic macrophages. Genuine DCs are a CD11chigh MHC class IIhigh population, whereas macrophages are a CD11clow MHC class IIlow population (5). Lamina propria phagocytic cells in the gut have different origins and functions (6). CD103-expressing DCs are widely present in non-lymphoid tissues. CD103+ DC subsets are differentiated by Flt3 ligand-dependent manner whereas CX3CR1-expressing phagocytic cells are dependent on CSF-1R (6). Peripheral CD103+CD11b- DCs are developmentally dependent on Batf3 and are related to CD8α+ conventional DCs (7). DC migration is tightly controlled by the expression of CCR7, and it can be largely classified as non-migratory and migratory (8,9). Non-migratory DCs are generally tissue-resident macrophage-like cells. Migratory DCs travel into draining lymph nodes with sampled antigen and can be infiltrated under inflammation. DC and phagocytic cells in the gut and their functions therein are listed in Table I. In the gut, CD103+ CD11b+ DCs has been well reported by the function to induce lymphocytes. Gut CD103+ DCs comprise two major subsets, CD103+CD11b+ and CD103+CD11b- DCs (10). CD103+ CD11b- DCs are the dominant population of CD103+ DC in the Peyer's patches and colon lamina propria (11). In contrast, CD103+CD11b+ DCs are the major DC subset in the SI lamina propria (12). In addition, recent reports regarding resident CX3CR1+ phagocytic cells are increasing. TNF-α/iNOS-producing DCs (Tip DCs) were initially reported in the spleen, where they released large amounts of nitric oxide (NO) after recognizing commensal bacteria through toll-like receptors (TLRs) (13). Several TLR-expressing DCs are reported to induce IgA production. Gut plasmacytoid DCs (pDCs) can induce IgA production and repress inflammation. The detailed function of each subset will be discussed later.

REGULATORY T CELLS AND INTESTINAL DCs

The SI shows a strong propensity for immune suppression in response to exogenous antigens. The presentation of innocuous food antigens by antigen-presenting cells induces oral tolerance by generation of inducible regulatory T (Treg) cells (14). CD103+CD11b+ DCs can sample soluble antigens from the intestinal lumen through a process termed goblet cell-associated antigen passages (GAPs) (15) (Fig. 1). These DCs can also patrol among enterocytes while extending dendrites toward the lumen (16). These intraepithelial CD103+ DCs are recruited to sample bacterial antigens for presentation. In the SI lamina propria as well as mesenteric lymph node (MLN), these DCs express retinaldehyde dehydrogenase type 2 (RALDH2) which can convert retinal to retinoic acid (RA), a metabolic derivative of vitamin A found in food. This DC subset can induce regulatory Foxp3+CD4+ T cells dependent on RA and TGF-β (17,18,19); Alternatively, CD11b+ F4/80+CD11c- macrophages in the lamina propria has been reported as more potent inducers of Treg cells than DCs (20). CX3CR1+ phagocytic cells in the lamina propria support expansion of the Foxp3+CD4+ Treg cell population by producing IL-10 to harness immune tolerance (21). CX3CR1+ phagocytic cells can capture Salmonella by extending dendrites across epithelium in a CX3CR1-dependent manner (22). Antigens captured by CX3CR1+ phagocytic cells can be transferred through gap junctions to CD103+ DCs in the lamina propria to establish oral tolerance (23). In addition to luminal antigen, lamina propria CX3CR1+ cells facilitate the surveillance of circulatory antigens from blood vessels (24). These cells fail to prime naïve CD4+ T cells; however, cross-presentation by these cells can induce priming of and differentiation into CD8+ T cells that express IL-10, IL-13, and IL-9. These CD8+ T cells can suppress pathogen-specific CD4+ T cell activation through IL-10 (24). Finally, these CD8+ T cells act as a regulatory CD8αβ +TCRαβ + T cell population in the epithelium. CX3CR1+ cells regulate colonic IL-22 producing group 3 innate lymphoid cells (ILC3) to promote mucosal healing and maintain barrier integrity (25). Therefore, CD103+ DCs and CX3CR1+ phagocytic cells can generate two distinct regulatory T cell subsets by different mechanisms to maintain gut immune homeostasis at steady state (Fig. 1). pDCs may mediate anti-inflammatory responses following TLR2-polysaccharide A signaling (26) and Type I interferon supports Treg function and regulates colitis (27,28).

T CELL DIFFERENTIATION AND INTESTINAL DCs

After DCs sample antigen in the lamina propria, they can present antigenic epitope to naïve T cells in the isolated lymphoid follicles (ILF) or draining MLN. Distinct DCs subset instructs T cell differentiation (Fig. 2). CD103+CD11b+ DCs, one of the major DC subset in the SI lamina propria, are primarily a migratory population that responds to CCR7 expression and can be infiltrated under inflammatory conditions (29,30). Segmented filamentous bacteria (SFB), murine commensal bacteria, are shown to be sufficient for TH17 differentiation (31). MHC II-dependent presentation of SFB antigens by intestinal DCs is crucial for TH17 cell induction (32). Most TH17 cells recognize antigenic repertoires derived from SFB (33). CD103+ CD11b+ DCs produce IL-6 with TLR stimuli which enable to induce TH17 cell differentiation (34). Several studies suggest the possibility that CD103+CD11b+ DCs might interact with SFB and link signals to induce TH17. CD103+ CD11b+ DCs can express high amounts of IL-23 following TLR5 stimuli and then drove IL-22-dependent RegIIIγ production from Paneth cells (35). TLR5+ DCs promote the differentiation of antigen-specific TH17 and TH1 cells following stimulation by flagellin, a TLR5 ligand (36). CD103+CD11b-CD8α+ DCs expressing TLR3, TLR7, and TLR9 can produce IL-6 and IL-12p40 following stimulation of the respective TLR ligands (37). These DCs induce a TH1 response and cytotoxic T lymphocytes (CTL). CX3CR1+ phagocytic cells contribute to intestinal clearance of intracellular bacteria. While their function under conditions of inflammation or infection remains unclear, their suppressive functions are well described at steady state.

SECRETORY IgA AND INTESTINAL DCs

A unique feature of the mucosal immune system is local production of sIgA from plasma cells differentiated from B cells. IgA class switching generally occurs in gut-associated lymphoid tissues including Peyer's patches, MLNs, and ILFs within the lamina propria. SFB stimulates the postnatal development of ILFs and tertiary lymphoid tissue in the SI lamina propria, which can substitute for Peyer's patches as inductive sites for intestinal IgA (38). Intestinal IgA coating identifies inflammatory commensals that drive intestinal disease like inflammatory bowel disease (33,39). The sIgA within mucus establishes distance with commensals and forms a barrier between invading and commensal microorganisms. Intestinal DCs support IgA isotype class-switching and differentiation into IgA-secreting plasma cells, either together with the assistance of TH cells or in a T cell-independent manner by expressing B cell-activating factors (BAFFs) and a proliferation-inducing ligand (APRIL) (Fig. 3). Intestinal pDCs, in particular, have been shown to induce IgA production by expressing these factors (40). When commensal bacteria are recognized through TLRs, Tip DCs release large amounts of NO which enhances IgA class-switch DNA recombination (CSR) in B cells by inducing DC expression of BAFF and APRIL (13). Further, RA can be converted by RALDH2 from dietary vitamin A in DCs, and DCs expressing RALDH2 can also induce IgA CSR (41). Lamina propria CD103+CD11b+ DCs, Tip DCs, and TLR5+ DCs express RALDH2 and produce RA, which in turn can be used for IgA production together with TGF-β (13,36,42). Langerin-expressing DCs in the MLNs that emerge following trans-cutaneous vaccination can also induce RA-dependent antigen-specific IgA production in the SI (43). Moreover, RA confers gut homing signature such as α4β7 integrin and CCR9 on lymphocytes (42,44). Therefore, RA is essential to maintaining the intestinal immune environment (45,46). In normal lamina propria, large numbers of eosinophils are present,accounting for approximately 5% of all lamina propria leukocytes (47). Recently, eosinophils have been found to promote the generation and maintenance of IgA-producing plasma cells by inducing B cell activating factors (48) and supporting the function of CD103+ DCs (49).

CONCLUSION AND FUTURE PERSPECTIVE

In this review, we focused on the integral role of intestinal DCs in shaping the unique intestinal immunity. The advent of advanced experimental techniques for surveying mucosal tissues and analyzing metagenomic data of commensals, along with the wide availability of germ-free mice has facilitated a growing understanding of this unique mucosal immune environment. Diverse microbiota can drive microbe-dependent CD4 effector T-cell programs. For example, Clostridium strains provide a rich environment of TGF-β and induce Foxp3+ Treg in the colon (50,51), and SFB induce the generation of TH17 cell by IL-6 production from DCs (32). Some pathogens (e.g., Listeria spp.) specifically induce TH1 cells (33). Thus, microbial signals may induce polarizing cytokine secretion from DCs, other innate cells or stromal cells. Assembling combined information, DCs may coordinate to establish gut immune system.

Figures and Tables

Figure 1

Regulatory T cells induced by intestinal DCs. Intestinal DCs can take up antigen indirectly through M cell-dependent (1), Goblet cell-dependent (2), and neonatal Fc receptor (FcRn)-dependent (3), and apoptosis-dependent manner (4). Alternatively, intestinal DCs can sample luminal antigen using intraepithelial dendrites (5). CX3CR1+ phagocytes facilitate the surveillance of circulatory antigens (6). Under steady-state conditions, CD103+ DCs induce Foxp3+CD4+ Tregs using retinoic acid by delivering luminal innocuous antigen. CX3CR1+ phagocytes can induce CD8+ Tregs to both luminal and circulatory antigens. These cells can expand the Foxp3+CD4+ Treg populations by IL-10 secretion in the intestinal lamina propria. Plasmacytoid DCs can produce IL-10 in response to TLR2.

in-15-1-g001
Figure 2

Helper T cell induced by intestinal DCs. CD103+CD11b+ DCs and TLR5+ DCs induce TH17 cells. TLR5+ DCs and CD103+CD8α+ DCs can induce TH1 cells by means of TLR signaling. CD103+CD8α+ DCs can also induce CTL. Induced helper T cell and CD8+ T cells confer host defense and further inflammation. Intraepithelial CD103+ DCs and CX3CR1+ phagocytes can sample pathogenic bacteria by extending long dendrites across the epithelium to directly defend against bacterial infection. CD103+CD11b+ DCs produce IL-23 and IL-22 to promote anti-microbial peptide production from Paneth cells.

in-15-1-g002
Figure 3

Intestinal DCs support secretory IgA generation. Gut CD103+CD11b+ DCs, Tip DCs, and TLR5+ DCs express RALDH2 that is converted into retinoic acid from dietary vitamin A and can be used for IgA production. Gut pDCs and Tip DCs induce IgA generation from B cells by expressing BAFF and APRIL. Eosinophils promote IgA production by expressing BAFF and APRIL or support the function of CD103+ DCs.

in-15-1-g003
Table I

Representative subsets of DCs and phagocytes in the intestine

in-15-1-i001
Name Phenotype Characteristic features Functions References
CD103+ CD103+ CCR7 expression : migration into LN CD4+Foxp3+ Treg generation (17, 18, 19, 51)
CD11b+ DCs CD11b+ RALDH2 expression : RA production IgA class switching (41)
Imprinting of lymphocyte gut homing by expression of CCR9 (43, 52)
Antigen uptake by extending long dendrite or goblet cell associated antigen passage (GAP) (28)
TLR stimulation: IL-6 production TH17 generation (33)
TLR5 stimulation: IL-23, IL-22 production RegIIIγ induction (34)
CD103+ CD103+ Expression of TLR3, TLR7, and TLR9 TH1 response and CTL activity (36)
CD11b- DCs CD11b- Production of IL-6 and IL-12p40
CD8αα+
CX3CR1+ cells CX3CR1+ No CCR7 expression: tissue-resident Bacteria clearance (22)
F4/80+ Antigen uptake by extending long dendrite from luminal antigen and bacteria Generation of regulatory CD8+ (23)
CD11b+ Uptake of circulatory antigen T cells (CD8αβ+TCRαβ+)
IL-10 production Treg expansion (21)
IL-22 induction by ILC3 Enhanced barrier integrity (24)
Tip DCs TNF-α+iNOS+ TGF-β IgA production (13)
CD11b+ APRIL and BAFF production
TLR5+ DCs TLR5+D11chi IL-6 production Differentiation of TH17 and TH1 cells (35)
CD11bhiF4/80+ RALDH2 expression : RA production Generation of IgA-producing cells
CD103+ Expression of TLR5 and TLR9
pDCs CD11cintB220+ Type I IFN receptor expression T cell-independent IgA production (39)
mPDCA1+ APRIL and BAFF production
IL-10 induction by CD4+ T cells Immune suppression (25)

Abbreviations

DC

dendritic cell

sIgA

secretory IgA

SI

small intestine

FcRn

neonatal Fc receptors

iNOS

inducible nitric oxide synthase

Tip DC

TNF-α/iNOS producing DC

pDC

plasmacytoid DC

Treg

regulatory T cell

GAP

goblet cell associated antigen passage

MLN

mesenteric lymph node

RALDH2

retinaldehyde dehydrogenase type 2

RA

retinoic acid

ILF

isolated lymphoid follicle

SFB

segmented filamentous bacteria

APRIL

a proliferation-inducing ligand

BAFF

B cell-activating factor belonging to the TNF family

CSR

class-switch DNA recombination

ACKNOWLEDGEMENTS

This work was supported by the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT and future Planning (NRF-2014R1A1A1002129, NRF-2014R1A2A2A 01002576).

Notes

The authors have no financial conflict of interest.

References

1. Murphy K, Travers P, Walport M, Janeway C. Janeway's immunobiology. New York: Garland Science;p. 466–468.
2. Ciorba MA, Riehl TE, Rao MS, Moon C, Ee X, Nava GM, Walker MR, Marinshaw JM, Stappenbeck TS, Stenson WF. Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2-dependent manner. Gut. 2012; 61:829–838.
crossref
3. Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, Tobe T, Clarke JM, Topping DL, Suzuki T, Taylor TD, Itoh K, Kikuchi J, Morita H, Hattori M, Ohno H. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011; 469:543–547.
crossref
4. Pickard JM, Maurice CF, Kinnebrew MA, Abt MC, Schenten D, Golovkina TV, Bogatyrev SR, Ismagilov RF, Pamer EG, Turnbaugh PJ, Chervonsky AV. Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature. 2014; 514:638–641.
crossref
5. Denning TL, Norris BA, Medina-Contreras O, Manicassamy S, Geem D, Madan R, Karp CL, Pulendran B. Functional specializations of intestinal dendritic cell and macrophage subsets that control Th17 and regulatory T cell responses are dependent on the T cell/APC ratio, source of mouse strain, and regional localization. J Immunol. 2011; 187:733–747.
crossref
6. Varol C, Vallon-Eberhard A, Elinav E, Aychek T, Shapira Y, Luche H, Fehling HJ, Hardt WD, Shakhar G, Jung S. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 2009; 31:502–512.
crossref
7. Edelson BT, KC W, Juang R, Kohyama M, Benoit LA, Klekotka PA, Moon C, Albring JC, Ise W, Michael DG, Bhattacharya D, Stappenbeck TS, Holtzman MJ, Sung SS, Murphy TL, Hildner K, Murphy KM. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells. J Exp Med. 2010; 207:823–836.
crossref
8. Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I, Wolf E, Lipp M. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 1999; 99:23–33.
crossref
9. Jang MH, Sougawa N, Tanaka T, Hirata T, Hiroi T, Tohya K, Guo Z, Umemoto E, Ebisuno Y, Yang BG, Seoh JY, Lipp M, Kiyono H, Miyasaka M. CCR7 is critically important for migration of dendritic cells in intestinal lamina propria to mesenteric lymph nodes. J Immunol. 2006; 176:803–810.
crossref
10. Persson EK, Jaensson E, Agace WW. The diverse ontogeny and function of murine small intestinal dendritic cell/macrophage subsets. Immunobiology. 2010; 215:692–697.
crossref
11. Helft J, Ginhoux F, Bogunovic M, Merad M. Origin and functional heterogeneity of non-lymphoid tissue dendritic cells in mice. Immunol Rev. 2010; 234:55–75.
crossref
12. Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, Liu K, Jakubzick C, Ingersoll MA, Leboeuf M, Stanley ER, Nussenzweig M, Lira SA, Randolph GJ, Merad M. Origin of the lamina propria dendritic cell network. Immunity. 2009; 31:513–525.
crossref
13. Tezuka H, Abe Y, Iwata M, Takeuchi H, Ishikawa H, Matsushita M, Shiohara T, Akira S, Ohteki T. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature. 2007; 448:929–933.
crossref
14. Mucida D, Kutchukhidze N, Erazo A, Russo M, Lafaille JJ, Curotto de Lafaille MA. Oral tolerance in the absence of naturally occurring Tregs. J Clin Invest. 2005; 115:1923–1933.
crossref
15. McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, Newberry RD, Miller MJ. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012; 483:345–349.
crossref
16. Farache J, Koren I, Milo I, Gurevich I, Kim KW, Zigmond E, Furtado GC, Lira SA, Shakhar G. Luminal bacteria recruit CD103+ dendritic cells into the intestinal epithelium to sample bacterial antigens for presentation. Immunity. 2013; 38:581–595.
crossref
17. Coombes JL, Siddiqui KR, rancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, Powrie F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007; 204:1757–1764.
crossref
18. Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, Belkaid Y. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 Treg cells via retinoic acid. J Exp Med. 2007; 204:1775–1785.
crossref
19. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, Cheroutre H. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007; 317:256–260.
crossref
20. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007; 8:1086–1094.
crossref
21. Hadis U, Wahl B, Schulz O, Hardtke-Wolenski M, Schippers A, Wagner N, Muller W, Sparwasser T, Forster R, Pabst O. Intestinal tolerance requires gut homing and expansion of Foxp3+ regulatory T cells in the lamina propria. Immunity. 2011; 34:237–246.
crossref
22. Niess JH, Brand S, Gu X, Landsman L, Jung S, McCormick BA, Vyas JM, Boes M, Ploegh HL, Fox JG, Littman DR, Reinecker HC. CX3CR1-mediated dendritic cell access to the intestinal lumen and bacterial clearance. Science. 2005; 307:254–258.
crossref
23. Mazzini E, Massimiliano L, Penna G, Rescigno M. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1(+) macrophages to CD103(+) dendritic cells. Immunity. 2014; 40:248–261.
crossref
24. Chang SY, Song JH, Guleng B, Cotoner CA, Arihiro S, Zhao Y, Chiang HS, O'Keeffe M, Liao G, Karp CL, Kweon MN, Sharpe AH, Bhan A, Terhorst C, Reinecker HC. Circulatory antigen processing by mucosal dendritic cells controls CD8(+) T cell activation. Immunity. 2013; 38:153–165.
crossref
25. Longman RS, Diehl GE, Victorio DA, Huh JR, Galan C, Miraldi ER, Swaminath A, Bonneau R, Scherl EJ, Littman DR. CX(3)CR1(+) mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. J Exp Med. 2014; 211:1571–1583.
crossref
26. Dasgupta S, Erturk-Hasdemir D, Ochoa-Reparaz J, Reinecker HC, Kasper DL. Plasmacytoid dendritic cells mediate anti-inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms. Cell Host Microbe. 2014; 15:413–423.
crossref
27. Lee SE, Li X, Kim JC, Lee J, Gonzalez-Navajas JM, Hong SH, Park IK, Rhee JH, Raz E. Type I interferons maintain Foxp3 expression and T-regulatory cell functions under inflammatory conditions in mice. Gastroenterology. 2012; 143:145–154.
crossref
28. Kole A, He J, Rivollier A, Silveira DD, Kitamura K, Maloy KJ, Kelsall BL. Type I IFNs regulate effector and regulatory T cell accumulation and anti-inflammatory cytokine production during T cell-mediated colitis. J Immunol. 2013; 191:2771–2779.
crossref
29. Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, Pabst O. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med. 2009; 206:3101–3114.
crossref
30. Jaensson E, Uronen-Hansson H, Pabst O, Eksteen B, Tian J, Coombes JL, Berg PL, Davidsson T, Powrie F, Johansson-Lindbom B, Agace WW. Small intestinal CD103+ dendritic cells display unique functional properties that are conserved between mice and humans. J Exp Med. 2008; 205:2139–2149.
crossref
31. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009; 139:485–498.
crossref
32. Goto Y, Panea C, Nakato G, Cebula A, Lee C, Diez MG, Laufer TM, Ignatowicz L, Ivanov II. Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation. Immunity. 2014; 40:594–607.
crossref
33. Yang Y, Torchinsky MB, Gobert M, Xiong H, Xu M, Linehan JL, Alonzo F, Ng C, Chen A, Lin X, Sczesnak A, Liao JJ, Torres VJ, Jenkins MK, Lafaille JJ, Littman DR. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature. 2014; 510:152–156.
crossref
34. Persson EK, Uronen-Hansson H, Semmrich M, Rivollier A, Hagerbrand K, Marsal J, Gudjonsson S, Hakansson U, Reizis B, Kotarsky K, Agace WW. IRF4 transcription-factor-dependent CD103(+)CD11b(+) dendritic cells drive mucosal T helper 17 cell differentiation. Immunity. 2013; 38:958–969.
crossref
35. Kinnebrew MA, Buffie CG, Diehl GE, Zenewicz LA, Leiner I, Hohl TM, Flavell RA, Littman DR, Pamer EG. Interleukin 23 production by intestinal CD103(+)CD11b(+) dendritic cells in response to bacterial flagellin enhances mucosal innate immune defense. Immunity. 2012; 36:276–287.
crossref
36. Uematsu S, Fujimoto K, Jang MH, Yang BG, Jung YJ, Nishiyama M, Sato S, Tsujimura T, Yamamoto M, Yokota Y, Kiyono H, Miyasaka M, Ishii KJ, Akira S. Regulation of humoral and cellular gut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat Immunol. 2008; 9:769–776.
crossref
37. Fujimoto K, Karuppuchamy T, Takemura N, Shimohigoshi M, Machida T, Haseda Y, Aoshi T, Ishii KJ, Akira S, Uematsu S. A new subset of CD103+CD8alpha+ dendritic cells in the small intestine expresses TLR3, TLR7, and TLR9 and induces Th1 response and CTL activity. J Immunol. 2011; 186:6287–6295.
crossref
38. Lecuyer E, Rakotobe S, Lengline-Garnier H, Lebreton C, Picard M, Juste C, Fritzen R, Eberl G, McCoy KD, Macpherson AJ, Reynaud CA, Cerf-Bensussan N, Gaboriau-Routhiau V. Segmented filamentous bacterium uses secondary and tertiary lymphoid tissues to induce gut IgA and specific T helper 17 cell responses. Immunity. 2014; 40:608–620.
crossref
39. Palm NW, de Zoete MR, Cullen TW, Barry NA, Stefanowski J, Hao L, Degnan PH, Hu J, Peter I, Zhang W, Ruggiero E, Cho JH, Goodman AL, Flavell RA. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell. 2014; 158:1000–1010.
crossref
40. Tezuka H, Abe Y, Asano J, Sato T, Liu J, Iwata M, Ohteki T. Prominent role for plasmacytoid dendritic cells in mucosal T cell-independent IgA induction. Immunity. 2011; 34:247–257.
crossref
41. Molenaar R, Knippenberg M, Goverse G, Olivier BJ, de Vos AF, O'Toole T, Mebius RE. Expression of retinaldehyde dehydrogenase enzymes in mucosal dendritic cells and gut-draining lymph node stromal cells is controlled by dietary vitamin A. J Immunol. 2011; 186:1934–1942.
crossref
42. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, Otipoby KL, Yokota A, Takeuchi H, Ricciardi-Castagnoli P, Rajewsky K, Adams DH, von Andrian UH. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science. 2006; 314:1157–1160.
crossref
43. Chang SY, Cha HR, Igarashi O, Rennert PD, Kissenpfennig A, Malissen B, Nanno M, Kiyono H, Kweon MN. Cutting edge: Langerin+ dendritic cells in the mesenteric lymph node set the stage for skin and gut immune system cross-talk. J Immunol. 2008; 180:4361–4365.
crossref
44. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 2004; 21:527–538.
crossref
45. Chang SY, Cha HR, Chang JH, Ko HJ, Yang H, Malissen B, Iwata M, Kweon MN. Lack of retinoic acid leads to increased langerin-expressing dendritic cells in gut-associated lymphoid tissues. Gastroenterology. 2010; 138:1468–1478. e6
crossref
46. Chang SY, Kweon MN. Langerin-expressing dendritic cells in gut-associated lymphoid tissues. Immunol Rev. 2010; 234:233–246.
crossref
47. Sutherland DB, Fagarasan S. Gut reactions: Eosinophils add another string to their bow. Immunity. 2014; 40:455–457.
crossref
48. Chu VT, Beller A, Rausch S, Strandmark J, Zanker M, Arbach O, Kruglov A, Berek C. Eosinophils promote generation and maintenance of immunoglobulin-A-expressing plasma cells and contribute to gut immune homeostasis. Immunity. 2014; 40:582–593.
crossref
49. Chu DK, Jimenez-Saiz R, Verschoor CP, Walker TD, Goncharova S, Llop-Guevara A, Shen P, Gordon ME, Barra NG, Bassett JD, Kong J, Fattouh R, McCoy KD, Bowdish DM, Erjefalt JS, Pabst O, Humbles AA, Kolbeck R, Waserman S, Jordana M. Indigenous enteric eosinophils control DCs to initiate a primary Th2 immune response in vivo. J Exp Med. 2014; 211:1657–1672.
crossref
50. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, Cheng G, Yamasaki S, Saito T, Ohba Y, Taniguchi T, Takeda K, Hori S, Ivanov II, Umesaki Y, Itoh K, Honda K. Induction of colonic regulatory T cells by indigenous Clostridium species. Science. 2011; 331:337–341.
crossref
51. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B, Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013; 500:232–236.
crossref
52. Benson MJ, Pino-Lagos K, Rosemblatt M, Noelle RJ. All-trans retinoic acid mediates enhanced Treg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation1. J Exp Med. 2007; 204:1765–1774.
crossref
53. Mora JR, Bono MR, Manjunath N, Weninger W, Cavanagh LL, Rosemblatt M, Von Andrian UH. Selective imprinting of gut-homing T cells by Peyer's patch dendritic cells. Nature. 2003; 424:88–93.
crossref
TOOLS
ORCID iDs

Sun-Young Chang
https://orcid.org/http://orcid.org/0000-0001-7336-9245

Similar articles