Skip to main content

MINI REVIEW article

Front. Immunol., 04 July 2022
Sec. T Cell Biology
This article is part of the Research Topic Revisit the Roles of Regulatory T cells in Infection View all 6 articles

Reciprocal Interactions Between Regulatory T Cells and Intestinal Epithelial Cells

  • 1Sun-Yat Sen University, School of Medicine, Guangzhou, China
  • 2Experimental Immunology Branch, National Cancer Institute, National Institute of Health (NIH), Bethesda, MD, United States

It has been well established that Foxp3+ regulatory T cells (Treg cells) play a crucial role for immune repression and tolerance, protecting the body from autoimmunity and inflammation. Previous studies indicate that intestinal Treg cells are one specialized population of Treg cells, distinct from those in other organ compartments, both functionally and phenotypically. Specific external and internal signals, particularly the presence of microbiota, shape these Treg cells to better cooperate with the gut ecosystem, controlling intestinal physiology. The integrity of intestinal epithelial barrier represents a key feature of gut immune tolerance, which can be regulated by multiple factors. Emerging evidence suggests that bidirectional interactions between gut epithelium and resident T cells significantly contribute to intestinal barrier function. Understanding how Treg cells regulate intestinal barrier integrity provides insights into immune tolerance-mediated mucosal homeostasis, which can further illuminate potential therapeutic strategies for treating inflammatory bowel disease and colon cancer.

Introduction

Regulatory T cells (Treg cells) are a specialized T cell subset which play a critical role in controlling immune homeostasis and peripheral tolerance (1, 2). Intestinal Treg cells mainly develop and differentiate in the thymus as thymic Treg (tTreg) cells, or can be induced in the periphery as peripheral Treg (pTreg) cells (3, 4). tTreg cells are generated after self-antigen recognition by T cell receptor in the thymus while pTreg cells are derived by non-self-antigen from naïve T cells. While these two types of Treg cells show complementary functions and different genetic signatures, they both express master transcription factor Foxp3 (5, 6). The function of Foxp3+ Treg cells for gut physiology has been documented in patients with immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome who lost the Treg cells (7). These patients exhibit symptoms of spontaneous inflammation in multiple organs, with most severe disorders on the mucosal surfaces, including the gastrointestinal (GI) tract (8). Plus, Foxp3 deficient mice (Scurfy) as well display severe autoimmunity in the gut (5, 9). These findings indicate that Treg cells are crucial for the intestinal immune tolerance. Considering the distinct antigen repertoires, intestinal pTreg cells are mainly responsible for immune tolerance against environmental insults, whereas tTreg cells protect the tissue from autoreactive responses.

The intestinal epithelium represents the largest interface which protects the body from potential danger while sensing external milieu. The monolayer of intestinal epithelial cells (IECs) form a physical barrier to segregate external environment from the intestinal tissues. Given the constant challenges and insults from dietary and microbial antigens, the integrity of intestinal epithelium barrier is a key feature of gut homeostasis (10). In addition to immune suppression, newly emerging evidence suggests that intestinal Treg cells also exert function for epithelium tissue repair and mucosal barrier maintenance (11). Hence, to elucidate how Treg-IEC crosstalk participates in gut physiology and pathophysiology is essential for the comprehension of tissue adaptation of Treg cells in the intestinal microenvironment.

In this Review, we will summarize and discuss the current understanding of how mutualism between Treg cells and IECs contribute to GI physiology and immune tolerance.

Gut Treg Subsets

In general, tTreg cells infiltrating in lamina propria (LP) inductive site origin from those tTreg cells propagating in peripheral blood, while pTreg cells accumulating in LP inductive site are mainly comprised by locally differentiated naïve T cells (12). The surface homing molecules CCR7 and CD62L direct tTreg and naïve T cells migrate into gut-associated lymphoid tissue (GALT) or gut-draining mesenteric lymph nodes (mLN). In these lymphoid compartments, tTreg cells expand when expose to unknown signals (13) and a substantial proportion of the naïve T cells differentiate into pTreg cells. Thereafter tTreg and pTreg cells migrate into LP effective site facilitated by α4β7 integrin and CCR9 signaling (14, 15). Unlike tTreg cells, pTreg cells expand inside of LP after exposed to commensal and dietary antigens (13). With an exception of the common pTreg homing route, there remain some pTreg cells found in LP, differentiated by TGF-β and retinoic acid (RA) producing eosinophils (16). In concert with freshly infiltrated tTreg and pTreg cells, there is also a subset of memory Treg cells resident in LP expand and exert immune suppression functions when induced in the gut. These CD103 expressing memory Treg cells are generated in a previous induction event and quiescent to exhibit tissue resident feature to join in the Treg pool to maintain mucosal homeostasis (17).

Although tTreg and pTreg cells are both able to exert immunosuppression function in the gut, they function independently and synergistically to maintain mucosal tolerance. tTreg cells and pTreg cells have different TCR repertoires, and thus response to different antigens. tTreg cells normally recognize self-antigen, and therefore response to those exposed antigens expressed by IECs, particularly under certain intestinal perturbations such as sterile injuries (18). pTreg cells normally recognize alien-antigens such as dietary metabolites and microbe-antigens and expand at the induction site (19, 20). In addition, strong TCR affinity facilitates the generation of a small portion of cross-react Treg cells with not fully elucidated reasons (18), including self-antigen responding pTreg cells (21) and foreign antigen responding tTreg cells (22). The variety of TCR repertoires covered by tTreg cells and pTreg cells are both required in regulating intestinal immune responses. It has been shown that adoptive transfer of tTreg cells alone is not sufficient to fully rescue Foxp3 deficiency during murine model of colitis, unless Foxp3CD4+ T cells are co-transferred, suggesting that both tTreg and pTreg cells are required for optimal protection during intestinal inflammation (23, 24). These findings shed light on developing Treg transfer therapy for potential treatment of human IBD patients.

IEC-Mediated Intestinal Treg Cells Induction and Function

IEC-Expressed MHC-II Independent Intestinal Treg Cells

Different studies have shown that intestinal Treg cells can be controlled via both IEC independent and dependent manners. Interaction between IEC and dendritic cell (DC) facilitates generation of tolerogenic DC via TGF-β and RA, which promotes intestinal Treg cells differentiation and restrains inflammation of colitis (25). Meanwhile, IECs are known to secret exosomes to the extracellular environment, which induce the tolerogenic properties to DCs for the generation of Treg cells in the gut (26). Additionally, other IEC-derived factors such as cytokines are as well known to modulate Treg cells differentiation and function. For instance, IEC-derived IL-18 modulates effector T cell differentiation in the gut which indirectly influence Treg function (27). Another study indicates that during intestinal tumorigenesis, IECs promotes specific subset of KLRG1+GATA3+ Treg cells accumulation mediated via IL-33 (28).

IEC-Expressed MHC-II Dependent Intestinal Treg Cells

Complement to the DC studies, intestinal Treg cells can also be directly induced by MHC class II (MHC-II) on IECs. It has been shown that both human and mouse IECs express MHC-II molecules (2932). IECs single cell survey identifies the expression of MHC-II on IECs (33), suggesting that IECs function as non-conventional APCs (34). The induction of MHC-II on IECs has been demonstrated to be IFN-γ−dependent (3539). It has been reported that IEC-derived MHC-II is sufficient to induce effector CD4+ T cells activation in GvHD model (37). Several studies have implicate that IECs preferentially promote suppressive Treg cell responses (38). Loss of MHC-II on IECs results in elevated levels of colitis associated with reduced Treg cells (34, 38). The expression of antigens by IECs leads to the proliferation of antigen-specific Treg cells in the intestine, which is further shown to be MHC-II-dependent (40). Moreover, intestinal mononuclear phagocytes (MNPs) have been reported to acquire MHC-II from IECs, subsequently assisting the generation of Treg cells (41). However, contradictory data show that MHC-II molecules are dispensable for T cell activation during murine colitis (42), raising the possibility that IEC-mediated T cell activation is context-dependent. Additional to IEC-mediated Treg cell expansion, recent study demonstrates that intestinal Treg cells are converted into CD4+Foxp3 IELs to control intestinal inflammation, indicating the critical role of IECs in controlling environmental adaptation of Treg cells in the gut (43). IECs from small intestine also provide a unique IL-2 independent milieu for the maintenance and survival of Treg cells (44). Altogether, the microenvironment of epithelium calibrates cellular and functional properties of Treg cells to cope with dynamic change in the gut.

Microbiota-Derived Intestinal Treg Cells

IECs are critical for microbial-mediated T cell differentiation and accumulation. It has been long established that segmented filamentous bacteria (SFB) promote intestinal Th17 cell differentiation which requires the direct adhesion of SFB on epithelium (4547). The SFB-IEC interaction leads to the production of serum amyloid A (SAA) from IECs which is critical for Th17 cell differentiation (48). Loss of such interaction compromises the induction of Th17 cells, indicating IEC plays a role of a key mediator in T cell responses to microbes (48). Similarly, gut Treg cells have also been shown to be induced from naïve T cells by antigens derived from commensal bacteria, which are known as inducible Treg cells (49). It has been reported that commensal bacteria such as Clostridium species and B. fragilis are able to induce peripheral Treg cells via IEC dependent or independent manners (50, 51). The Clostridia colonize the mucus layer without direct adhesion to IECs. The colonization of Clostridium species is found to impact on IECs for the production of TGF-β and indoleamine 2,3-dioxygenase (IDO), which could contribute to the induction of colonic Treg cells (52, 53). More importantly, de novo generation of intestinal Treg cells may require synergistic effects with different Clostridia species, given the fact that a single species is insufficient in polarizing Treg cells (54). Additional to TGF-β and IDO-derived from IECs, Clostridia may also induce Treg cell generation via producing short chain fatty acids (SCFAs) by diffusing through the epithelium to LP (5558). Moreover, gut bacteria also generate secondary bile acids which can modulate the balance of Th17 and pTreg cells for intestinal immune homeostasis. (59). While Clostridia species are known to regulate Treg cells via IEC dependent manners, other microbiota species including Lactobacilli and Bifidobacteria, can also induce and activate colonic Treg cells by IEC independent manners (50, 6062). It is now commonly recognized that microbiota modulates T cell differentiation and function in the gut for intestinal physiology (63). Given that Clostridia and Bacteroides species are two prominent members of the mammalian gut microbiota, such microbiota-mediated Treg cell regulation could be one machinery for the maintenance of gut homeostasis. Recent study further elucidates that mucosa-associated fungi also modulates gut Th17 responses for intestinal barrier function (64). Specifically, both Candida albicans and Staphylococcus aureus are identified to be strong inducers of human Th17 responses (65, 66). These findings implicate that T cell differentiation and function could be regulated by a diverse community of bacteria, viruses, protozoa, and fungi within the GI tract (67). Given close proximity of gut microbiota and epithelium barrier, IECs play a critical role in bridging the crosstalk between different microbes and hosts for immune regulation in the gut. The precise mechanisms of how IECs collaborate with different microbial for immune tolerance is still under investigation, including Treg cells generation and function.

Diet-Derived Intestinal Treg Cells

Dietary components largely influence the development and function of intestinal Treg cells, which can be mediated by IEC-dependent and -independent manners. Dietary antigens are known to directly induce RORγt+ pTreg cells which are essential for the induction of oral tolerance (19, 68). Dietary vitamin A-derived retinoic acid regulates the differentiation and accumulation of Treg cells, which exerts both pro- and anti-inflammatory functions (69, 70). The metabolite of vitamin D3, 1,25-dihyroxyvitamin D3 can promote Treg cell differentiation (71). Interaction between vitamin B9 and its receptor (folic acid receptor 4) on Treg cells facilitates colonic Treg cell survival (72), protecting the mice from colitis (73). Additionally, vitamin C transporter was found to highly expressed on Treg cells. Vitamin C treatment leads to impaired suppressive function of tTreg cells, whereas it promotes pTreg cell generation both in vitro and in vivo (74). High salt diet (HSD) has also been reported to promote pathogenic Th17 responses via SGK1-Foxo1 signaling pathway while dampening Treg cell function, enhancing the susceptibility of autoimmunity and inflammation (7578). Moreover, it has been suggested that HSD modulates gut microbial responses for proinflammatory T cells generation in human (79). And clinical study shows that dietary sodium intake positively correlates with the severity of autoimmunity (80). Further, another study demonstrates that dietary-derived sugar, D-mannose, induces Treg cell generation in both human and mouse cells by promoting TGF-β activation. Supplement of D-mannose represses proinflammatory responses in animal models of autoimmunity (81). Moreover, dietary fibers can be fermented and converted into SCFAs through gut microbiota. Various studies suggest that SCFAs stimulate Treg cell differentiation, expansion and accumulation through activation of different G protein-coupled receptors such as GPR43 (58), GPR109A (82) and GPR15 (83). Tryptophan is another critical food component as an essential amino acid. It can be metabolized to kynurenin through IECs which modulates Treg cell development (84). Tryptophan is also the precursor of vitamin B3. Vitamin B3 binds to its receptor GPR109A on macrophages and DCs in the gut, leading to differentiation of Treg cells. Loss of GPR109A results in elevated levels of intestinal inflammation (82). Collectively, these pieces of data indicate that both dietary components modulate Treg cell generation and function in the gut, providing insight of dietary-based therapies in controlling intestinal inflammation.

Treg Cells Modulate Intestinal Epithelium Barrier Functions

Foxp3+ Treg cells play a critical role in regulating IEC homeostasis and intestinal barrier integrity. Although various of cellular sources contribute to intestinal IL-10, as one major effector molecule from Treg cells, Treg cell-derived IL-10 has been demonstrated to play a key role for the maintenance of mucosal immune homeostasis (85, 86). Recent study indicates that Treg cells are required for intestinal stem cells (ISC) renewal via IL-10. Loss of Treg cells results in decreased ISC frequency with elevated levels of IEC differentiation (35). T cell-derived IL-10 has been reported to regulate the IECs function via inhibiting their fucosylation (87). Further, IL-10 is demonstrated to suppress Fas-mediated IEC apoptosis (88), and protects IEC from endoplasmic reticulum stress for epithelium barrier integrity (89, 90). Given the immune regulatory function of Treg cells in the gut, they are thus able to control epithelium barrier function indirectly by impacting other immune cells. For instance, it is known that Treg cells control the abundance of Th17 cells in the gut. And intestinal Th17 cells-derived cytokines such as IL-17 and IL-22, are beneficial for mucosal barrier function (9193). Moreover, a previous study indicated that Treg cells improve intestinal barrier function by regulating neutrophil infiltration during heatstroke (94). Treg cells have also been shown to enhance intestinal barrier function by repressing type 2 responses during food allergy (95). The process of generating pTreg cells from naïve T cells carrying environmental antigen specific TCRs is important since it can prevent these T cells from eliciting harmful immune responses. pTreg cell deficient mice exhibit spontaneous inflammation in the GI tract associated with altered microbiota (96). Hence, the reciprocal interactions between IEC and Treg cells are delicately balanced by the gut microenvironment while controlling intestinal barrier physiology.

Conclusions

Intestinal Treg cells are critical for establishing gut tolerance and host defense. The heterogenicity of these Treg cells are beneficial for protecting the intestinal tissue from various sources of insults. Importantly, the IECs play a key role in connecting environmental cues to tissue immune system for the induction, expansion and function of Treg cells. While the role of IEC as non-canonical APC has been studied, further investigation is still required to illustrate the molecular mechanism of IEC-Treg cell crosstalk. These include correlation of spatial expression pattern of MHC-II on IECs with Treg cell distribution, intracellular signaling pathways of antigen process and presentation by IECs and how specific mediators produced by IECs mediate Treg cells generation and function. Moreover, because of the heterogeneity of IEC population, it will be essential to interrogate in detail that whether and how Treg cells regulate different enterocyte subsets for mucosal neuroendocrinal responses beyond intestinal barrier function. The understanding of the cellular and molecular mechanisms responsible for reciprocal regulation between Treg cells and IECs could provide new insights into how Treg cells control tissue homeostasis on different barrier surfaces for development of therapeutic interventions.

Author Contributions

CW and ZJ wrote the manuscript. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by the Intramural Research Program of the NCI, NIH, United States.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T Cells: Mechanisms of Differentiation and Function. Annu Rev Immunol (2012) 30:531–64. doi: 10.1146/annurev.immunol.25.022106.141623

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ Regulatory T Cells in the Human Immune System. Nat Rev Immunol (2010) 10:490–500. doi: 10.1038/nri2785

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Bluestone JA, Abbas AK. Natural Versus Adaptive Regulatory T Cells. Nature Reviews. Immunology (2003) 3:253–7. doi: 10.1038/nri1032

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Curotto de Lafaille MA, Lafaille JJ. Natural and Adaptive Foxp3+ Regulatory T Cells: More of the Same or a Division of Labor? Immunity (2009) 30:626–35. doi: 10.1016/j.immuni.2009.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 Programs the Development and Function of CD4+CD25+ Regulatory T Cells. Nat Immunol (2003) 4:330–6. doi: 10.1038/ni904

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Zheng Y, Rudensky AY. Foxp3 in Control of the Regulatory T Cell Lineage. Nat Immunol (2007) 8:457–62. doi: 10.1038/ni1455

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Wildin RS, Ramsdell F, Peake J, Faravelli F, Casanova JL, Buist N, et al. X-Linked Neonatal Diabetes Mellitus, Enteropathy and Endocrinopathy Syndrome is the Human Equivalent of Mouse Scurfy. Nat Genet (2001) 27:18–20. doi: 10.1038/83707

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, et al. Severe Food Allergy as a Variant of IPEX Syndrome Caused by a Deletion in a Noncoding Region of the FOXP3 Gene. Gastroenterology (2007) 132:1705–17. doi: 10.1053/j.gastro.2007.02.044

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Bennett CL, Christie J, Ramsdell F, Brunkow ME, Ferguson PJ, Whitesell L, et al. The Immune Dysregulation, Polyendocrinopathy, Enteropathy, X-Linked Syndrome (IPEX) is Caused by Mutations of FOXP3. Nat Genet (2001) 27:20–1. doi: 10.1038/83713

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Okumura R, Takeda K. Roles of Intestinal Epithelial Cells in the Maintenance of Gut Homeostasis. Exp Mol Med (2017) 49:e338. doi: 10.1038/emm.2017.20

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Tanoue T, Atarashi K, Honda K. Development and Maintenance of Intestinal Regulatory T Cells. Nature Reviews. Immunology (2016) 16:295–309. doi: 10.1038/nri.2016.36

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Pabst O. Trafficking of Regulatory T Cells in the Intestinal Immune System. Int Immunol (2013) 25:139–43. doi: 10.1093/intimm/dxs113

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Mosconi I, Dubey LK, Volpe B, Esser-von Bieren J, Zaiss MM, Lebon L, et al. Parasite Proximity Drives the Expansion of Regulatory T Cells in Peyer's Patches Following Intestinal Helminth Infection. Infect Immun (2015) 83:3657–65. doi: 10.1128/IAI.00266-15

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Cassani B, Villablanca EJ, Quintana FJ, Love PE, Lacy-Hulbert A, Blaner WS, et al. Gut-Tropic T Cells That Express Integrin Alpha4beta7 and CCR9 are Required for Induction of Oral Immune Tolerance in Mice. Gastroenterology (2011) 141:2109–18. doi: 10.1053/j.gastro.2011.09.015

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Sun H, Kuk W, Rivera-Nieves J, Lopez-Ramirez MA, Eckmann L, Ginsberg MH. Beta7 Integrin Inhibition Can Increase Intestinal Inflammation by Impairing Homing of CD25(hi)FoxP3(+) Regulatory T Cells. Cell Mol Gastroenterol Hepatol (2020) 9:369–85. doi: 10.1016/j.jcmgh.2019.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Chen HH, Sun AH, Ojcius DM, Hu WL, Ge YM, Lin X, et al. Eosinophils From Murine Lamina Propria Induce Differentiation of Naive T Cells Into Regulatory T Cells via TGF-Beta1 and Retinoic Acid. PloS One (2015) 10:e0142881. doi: 10.1371/journal.pone.0142881

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Lee J, Kim D, Min B. Tissue Resident Foxp3(+) Regulatory T Cells: Sentinels and Saboteurs in Health and Disease. Front Immunol (2022) 13:865593. doi: 10.3389/fimmu.2022.865593

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Russler-Germain EV, Rengarajan S, Hsieh CS. Antigen-Specific Regulatory T-Cell Responses to Intestinal Microbiota. Mucosal Immunol (2017) 10:1375–86. doi: 10.1038/mi.2017.65

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kim KS, Hong SW, Han D, Yi J, Jung J, Yang BG, et al. Dietary Antigens Limit Mucosal Immunity by Inducing Regulatory T Cells in the Small Intestine. Science (2016) 351:858–63. doi: 10.1126/science.aac5560

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Nutsch K, Chai JN, Ai TL, Russler-Germain E, Feehley T, Nagler CR, et al. Rapid and Efficient Generation of Regulatory T Cells to Commensal Antigens in the Periphery. Cell Rep (2016) 17:206–20. doi: 10.1016/j.celrep.2016.08.092

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Yi J, Jung J, Hong SW, Lee JY, Han D, Kim KS, et al. Unregulated Antigen-Presenting Cell Activation by T Cells Breaks Self Tolerance. Proc Natl Acad Sci United States America (2019) 116:1007–16. doi: 10.1073/pnas.1818624116

CrossRef Full Text | Google Scholar

22. Lee HM, Bautista JL, Scott-Browne J, Mohan JF, Hsieh CS. A Broad Range of Self-Reactivity Drives Thymic Regulatory T Cell Selection to Limit Responses to Self. Immunity (2012) 37:475–86. doi: 10.1016/j.immuni.2012.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Haribhai D, Chatila TA, Williams CB. Immunotherapy With Itreg and Ntreg Cells in a Murine Model of Inflammatory Bowel Disease. Methods Mol Biol (2016) 1422:197–211. doi: 10.1007/978-1-4939-3603-8_19

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Haribhai D, Lin W, Edwards B, Ziegelbauer J, Salzman NH, Carlson MR, et al. A Central Role for Induced Regulatory T Cells in Tolerance Induction in Experimental Colitis. J Immunol (2009) 182:3461–8. doi: 10.4049/jimmunol.0802535

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Iliev ID, Mileti E, Matteoli G, Chieppa M, Rescigno M. Intestinal Epithelial Cells Promote Colitis-Protective Regulatory T-Cell Differentiation Through Dendritic Cell Conditioning. Mucosal Immunol (2009) 2:340–50. doi: 10.1038/mi.2009.13

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Chen X, Song CH, Feng BS, Li TL, Li P, Zheng PY, et al. Intestinal Epithelial Cell-Derived Integrin Alphabeta6 Plays an Important Role in the Induction of Regulatory T Cells and Inhibits an Antigen-Specific Th2 Response. J leukoc. Biol (2011) 90:751–9. doi: 10.1189/jlb.1210696

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Harrison OJ, Srinivasan N, Pott J, Schiering C, Krausgruber T, Ilott NE, et al. Epithelial-Derived IL-18 Regulates Th17 Cell Differentiation and Foxp3(+) Treg Cell Function in the Intestine. Mucosal Immunol (2015) 8:1226–36. doi: 10.1038/mi.2015.13

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Meinicke H, Bremser A, Brack M, Akeus P, Pearson C, Bullers S, et al. Tumour-Associated Changes in Intestinal Epithelial Cells Cause Local Accumulation of KLRG1(+) GATA3(+) Regulatory T Cells in Mice. Immunology (2017) 152:74–88. doi: 10.1111/imm.12750

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Kelly J, Weir DG, Feighery C. Differential Expression of HLA-D Gene Products in the Normal and Coeliac Small Bowel. Tissue Antigens (1988) 31:151–60. doi: 10.1111/j.1399-0039.1988.tb02076.x

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Madrigal L, Lynch S, Feighery C, Weir D, Kelleher D, O'Farrelly C. Flow Cytometric Analysis of Surface Major Histocompatibility Complex Class II Expression on Human Epithelial Cells Prepared From Small Intestinal Biopsies. J Immunol Methods (1993) 158:207–14. doi: 10.1016/0022-1759(93)90216-t

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Scott H, Solheim BG, Brandtzaeg P, Thorsby E. HLA-DR-Like Antigens in the Epithelium of the Human Small Intestine. Scand J Immunol (1980) 12:77–82. doi: 10.1111/j.1365-3083.1980.tb00043.x

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Wiman K, Curman B, Forsum U, Klareskog L, Malmnas-Tjernlund U, Rask L, et al. Occurrence of Ia Antigens on Tissues on non-Lymphoid Origin. Nature (1978) 276:711–3. doi: 10.1038/276711a0

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, et al. A Single-Cell Survey of the Small Intestinal Epithelium. Nature (2017) 551:333–9. doi: 10.1038/nature24489

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Jamwal DR, Laubitz D, Harrison CA, Figliuolo da Paz V, Cox CM, Wong R, et al. Intestinal Epithelial Expression of MHCII Determines Severity of Chemical, T-Cell-Induced, and Infectious Colitis in Mice. Gastroenterology (2020) 159:1342–1356 e1346. doi: 10.1053/j.gastro.2020.06.049

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Biton M, Haber AL, Rogel N, Burgin G, Beyaz S, Schnell A, et al. ). T Helper Cell Cytokines Modulate Intestinal Stem Cell Renewal and Differentiation. Cell (2018) 175:1307–1320 e1322. doi: 10.1016/j.cell.2018.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Farin HF, Karthaus WR, Kujala P, Rakhshandehroo M, Schwank G, Vries RG, et al. Paneth Cell Extrusion and Release of Antimicrobial Products is Directly Controlled by Immune Cell-Derived IFN-Gamma. J Exp Med (2014) 211:1393–405. doi: 10.1084/jem.20130753

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Koyama M, Mukhopadhyay P, Schuster IS, Henden AS, Hulsdunker J, Varelias A, et al. MHC Class II Antigen Presentation by the Intestinal Epithelium Initiates Graft-Versus-Host Disease and Is Influenced by the Microbiota. Immunity (2019) 51:885–98.e887. doi: 10.1016/j.immuni.2019.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Thelemann C, Eren RO, Coutaz M, Brasseit J, Bouzourene H, Rosa M, et al. Interferon-Gamma Induces Expression of MHC Class II on Intestinal Epithelial Cells and Protects Mice From Colitis. PloS One (2014) 9:e86844. doi: 10.1371/journal.pone.0086844

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Wosen JE, Ilstad-Minnihan A, Co JY, Jiang W, Mukhopadhyay D, Fernandez-Becker NQ, et al. Human Intestinal Enteroids Model MHC-II in the Gut Epithelium. Front Immunol (2019) 10:1970. doi: 10.3389/fimmu.2019.01970

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Westendorf AM, Bruder D, Hansen W, Buer J. Intestinal Epithelial Antigen Induces CD4+ T Cells With Regulatory Phenotype in a Transgenic Autoimmune Mouse Model. Ann New York Acad Sci (2006) 1072:401–6. doi: 10.1196/annals.1326.035

CrossRef Full Text | Google Scholar

41. Stephens WZ, Kubinak JL, Ghazaryan A, Bauer KM, Bell R, Buhrke K, et al. Epithelial-Myeloid Exchange of MHC Class II Constrains Immunity and Microbiota Composition. Cell Rep (2021) 37:109916. doi: 10.1016/j.celrep.2021.109916

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Trobonjaca Z, Leithauser F, Moller P, Bluethmann H, Koezuka Y, MacDonald HR, et al. MHC-II-Independent CD4+ T Cells Induce Colitis in Immunodeficient RAG-/- Hosts. J Immunol (2001) 166:3804–12. doi: 10.4049/jimmunol.166.6.3804

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Sujino T, London M, Hoytema van Konijnenburg DP, Rendon T, Buch T, Silva HM, et al. Tissue Adaptation of Regulatory and Intraepithelial CD4(+) T Cells Controls Gut Inflammation. Science (2016) 352:1581–6. doi: 10.1126/science.aaf3892

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Prakhar P, Alvarez-DelValle J, Keller H, Crossman A, Tai X, Park YK, et al. The Small Intestine Epithelium Exempts Foxp3+ Tregs From Their IL-2 Requirement for Homeostasis and Effector Function. JCI Insight (2021) 6:e149656. doi: 10.1172/jci.insight.149656

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Gaboriau-Routhiau V, Rakotobe S, Lecuyer E, Mulder I, Lan A, Bridonneau C, et al. The Key Role of Segmented Filamentous Bacteria in the Coordinated Maturation of Gut Helper T Cell Responses. Immunity (2009) 31:677–89. doi: 10.1016/j.immuni.2009.08.020

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria. Cell (2009) 139:485–98. doi: 10.1016/j.cell.2009.09.033

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Lecuyer E, Rakotobe S, Lengline-Garnier H, Lebreton C, Picard M, Juste C, et al. Segmented Filamentous Bacterium Uses Secondary and Tertiary Lymphoid Tissues to Induce Gut IgA and Specific T Helper 17 Cell Responses. Immunity (2014) 40:608–20. doi: 10.1016/j.immuni.2014.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Atarashi K, Tanoue T, Ando M, Kamada N, Nagano Y, Narushima S, et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell (2015) 163:367–80. doi: 10.1016/j.cell.2015.08.058

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. Peripheral Education of the Immune System by Colonic Commensal Microbiota. Nature (2011) 478:250–4. doi: 10.1038/nature10434

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Round JL, Mazmanian SK. Inducible Foxp3+ Regulatory T-Cell Development by a Commensal Bacterium of the Intestinal Microbiota. Proc Natl Acad Sci United States America (2010) 107:12204–9. doi: 10.1073/pnas.0909122107

CrossRef Full Text | Google Scholar

51. Shen Y, Giardino Torchia ML, Lawson GW, Karp CL, Ashwell JD, Mazmanian SK. Outer Membrane Vesicles of a Human Commensal Mediate Immune Regulation and Disease Protection. Cell Host Microbe (2012) 12:509–20. doi: 10.1016/j.chom.2012.08.004

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Atarashi K, Tanoue T, Shima T, Imaoka A, Kuwahara T, Momose Y, et al. Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species. Science (2011), 331:337–41. doi: 10.1126/science.1198469

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Chiba T, Seno H. Indigenous Clostridium Species Regulate Systemic Immune Responses by Induction of Colonic Regulatory T Cells. Gastroenterology (2011) 141:1114–6. doi: 10.1053/j.gastro.2011.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, et al. Treg Induction by a Rationally Selected Mixture of Clostridia Strains From the Human Microbiota. Nature (2013), 500:232–6. doi: 10.1038/nature12331

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites Produced by Commensal Bacteria Promote Peripheral Regulatory T-Cell Generation. Nature (2013) 504:451–5. doi: 10.1038/nature12726

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, et al. Commensal Microbe-Derived Butyrate Induces the Differentiation of Colonic Regulatory T Cells. Nature (2013) 504:446–50. doi: 10.1038/nature12721

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Narushima S, Sugiura Y, Oshima K, Atarashi K, Hattori M, Suematsu M, et al. Characterization of the 17 Strains of Regulatory T Cell-Inducing Human-Derived Clostridia. Gut. Microbes (2014) 5:333–9. doi: 10.4161/gmic.28572

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, et al. The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis. Science (2013) 341:569–73. doi: 10.1126/science.1241165

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hang S, Paik D, Yao L, Kim E, Trinath J, Lu J, et al. Bile Acid Metabolites Control TH17 and Treg Cell Differentiation. Nature (2019) 576:143–8. doi: 10.1038/s41586-019-1785-z

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Di Giacinto C, Marinaro M, Sanchez M, Strober W, Boirivant M. Probiotics Ameliorate Recurrent Th1-Mediated Murine Colitis by Inducing IL-10 and IL-10-Dependent TGF-Beta-Bearing Regulatory Cells. . J Immunol (2005) 174:3237–46. doi: 10.4049/jimmunol.174.6.3237

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Karimi K, Inman MD, Bienenstock J, Forsythe P. Lactobacillus Reuteri-Induced Regulatory T Cells Protect Against an Allergic Airway Response in Mice. Am J Respir Crit Care Med (2009) 179:186–93. doi: 10.1164/rccm.200806-951OC

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Tang C, Kamiya T, Liu Y, Kadoki M, Kakuta S, Oshima K, et al. Inhibition of Dectin-1 Signaling Ameliorates Colitis by Inducing Lactobacillus-Mediated Regulatory T Cell Expansion in the Intestine. Cell Host Microbe (2015) 18:183–97. doi: 10.1016/j.chom.2015.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Gehlhaar A, Inala A, Llivichuzhca-Loja D, Silva TN, Adegboye CY, O'Connell AE, et al. Insights Into the Role of Commensal-Specific T Cells in Intestinal Inflammation. J Inflammation Res (2022) 15:1873–87. doi: 10.2147/JIR.S288288

CrossRef Full Text | Google Scholar

64. Leonardi I, Gao IH, Lin WY, Allen M, Li XV, Fiers WD, et al. Mucosal Fungi Promote Gut Barrier Function and Social Behavior via Type 17 Immunity. Cell (2022) 185:831–846 e814. doi: 10.1016/j.cell.2022.01.017

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Bacher P, Hohnstein T, Beerbaum E, Rocker M, Blango MG, Kaufmann S, et al. Human Anti-Fungal Th17 Immunity and Pathology Rely on Cross-Reactivity Against Candida Albicans. Cell (2019) 176:1340–1355 e1315. doi: 10.1016/j.cell.2019.01.041

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, et al. Pathogen-Induced Human TH17 Cells Produce IFN-Gamma or IL-10 and are Regulated by IL-1beta. Nature (2012) 484:514–8. doi: 10.1038/nature10957

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Hall AB, Tolonen AC, Xavier RJ. Human Genetic Variation and the Gut Microbiome in Disease. Nat Rev Genet (2017) 18:690–9. doi: 10.1038/nrg.2017.63

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Abdel-Gadir A, Stephen-Victor E, Gerber GK, Noval Rivas M, Wang S, Harb H, et al. Microbiota Therapy Acts via a Regulatory T Cell MyD88/RORgammat Pathway to Suppress Food Allergy. . Nat Med (2019) 25:1164–74. doi: 10.1038/s41591-019-0461-z

PubMed Abstract | CrossRef Full Text | Google Scholar

69. DePaolo RW, Abadie V, Tang F, Fehlner-Peach H, Hall JA, Wang W, et al. Co-Adjuvant Effects of Retinoic Acid and IL-15 Induce Inflammatory Immunity to Dietary Antigens. Nature (2011) 471:220–4. doi: 10.1038/nature09849

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Kang SG, Lim HW, Andrisani OM, Broxmeyer HE, Kim CH. Vitamin A Metabolites Induce Gut-Homing FoxP3+ Regulatory T Cells. J Immunol (2007) 179:3724–33. doi: 10.4049/jimmunol.179.6.3724

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Kang SW, Kim SH, Lee N, Lee WW, Hwang KA, Shin MS, et al. 1,25-Dihyroxyvitamin D3 Promotes FOXP3 Expression via Binding to Vitamin D Response Elements in its Conserved Noncoding Sequence Region. J Immunol (2012) 188:5276–82. doi: 10.4049/jimmunol.1101211

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Yamaguchi T, Hirota K, Nagahama K, Ohkawa K, Takahashi T, Nomura T, et al. Control of Immune Responses by Antigen-Specific Regulatory T Cells Expressing the Folate Receptor. Immunity (2007) 27:145–59. doi: 10.1016/j.immuni.2007.04.017

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Kinoshita M, Kayama H, Kusu T, Yamaguchi T, Kunisawa J, Kiyono H, et al. Dietary Folic Acid Promotes Survival of Foxp3+ Regulatory T Cells in the Colon. . J Immunol (2012) 189:2869–78. doi: 10.4049/jimmunol.1200420

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Oyarce K, Campos-Mora M, Gajardo-Carrasco T, Pino-Lagos K. Vitamin C Fosters the In Vivo Differentiation of Peripheral CD4(+) Foxp3(-) T Cells Into CD4(+) Foxp3(+) Regulatory T Cells But Impairs Their Ability to Prolong Skin Allograft Survival. Front Immunol (2018) 9:112. doi: 10.3389/fimmu.2018.00112

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Hernandez AL, Kitz A, Wu C, Lowther DE, Rodriguez DM, Vudattu N, et al. Sodium Chloride Inhibits the Suppressive Function of FOXP3+ Regulatory T Cells. J Clin Invest (2015) 125:4212–22. doi: 10.1172/JCI81151

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Safa K, Ohori S, Borges TJ, Uehara M, Batal I, Shimizu T, et al. Salt Accelerates Allograft Rejection Through Serum- and Glucocorticoid-Regulated Kinase-1-Dependent Inhibition of Regulatory T Cells. J Am Soc Nephrol JASN (2015) 26:2341–7. doi: 10.1681/ASN.2014090914

CrossRef Full Text | Google Scholar

77. Wu C, Chen Z, Xiao S, Thalhamer T, Madi A, Han T, et al. SGK1 Governs the Reciprocal Development of Th17 and Regulatory T Cells. Cell Rep (2018) 22:653–65. doi: 10.1016/j.celrep.2017.12.068

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Wu C, Yosef N, Thalhamer T, Zhu C, Xiao S, Kishi Y, et al. Induction of Pathogenic TH17 Cells by Inducible Salt-Sensing Kinase SGK1. Nature (2013) 496:513–7. doi: 10.1038/nature11984

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, et al. Salt-Responsive Gut Commensal Modulates TH17 Axis and Disease. Nature (2017) 551:585–9. doi: 10.1038/nature24628

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Farez MF, Fiol MP, Gaitan MI, Quintana FJ, Correale J. Sodium Intake is Associated With Increased Disease Activity in Multiple Sclerosis. J Neurol Neurosurg Psychiatry (2015) 86:26–31. doi: 10.1136/jnnp-2014-307928

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Zhang D, Chia C, Jiao X, Jin W, Kasagi S, Wu R, et al. D-Mannose Induces Regulatory T Cells and Suppresses Immunopathology. Nat Med (2017) 23:1036–45. doi: 10.1038/nm.4375

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Singh N, Gurav A, Sivaprakasam S, Brady E, Padia R, Shi H, et al. Activation of Gpr109a, Receptor for Niacin and the Commensal Metabolite Butyrate, Suppresses Colonic Inflammation and Carcinogenesis. Immunity (2014) 40:128–39. doi: 10.1016/j.immuni.2013.12.007

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Kim SV, Xiang WV, Kwak C, Yang Y, Lin XW, Ota M, et al. GPR15-Mediated Homing Controls Immune Homeostasis in the Large Intestine Mucosa. Science (2013) 340:1456–9. doi: 10.1126/science.1237013

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Mezrich JD, Fechner JH, Zhang X, Johnson BP, Burlingham WJ, Bradfield CA. An Interaction Between Kynurenine and the Aryl Hydrocarbon Receptor can Generate Regulatory T Cells. J Immunol (2010) 185:3190–8. doi: 10.4049/jimmunol.0903670

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Chaudhry A, Samstein RM, Treuting P, Liang Y, Pils MC, Heinrich JM, et al. Interleukin-10 Signaling in Regulatory T Cells is Required for Suppression of Th17 Cell-Mediated Inflammation. Immunity (2011) 34:566–78. doi: 10.1016/j.immuni.2011.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Rubtsov YP, Rasmussen JP, Chi EY, Fontenot J, Castelli L, Ye X, et al. Regulatory T Cell-Derived Interleukin-10 Limits Inflammation at Environmental Interfaces. Immun (2008) 28:546–58. doi: 10.1016/j.immuni.2008.02.017

CrossRef Full Text | Google Scholar

87. Goto Y, Lamichhane A, Kamioka M, Sato S, Honda K, Kunisawa J, et al. IL-10-Producing CD4(+) T Cells Negatively Regulate Fucosylation of Epithelial Cells in the Gut. Sci Rep (2015) 5:15918. doi: 10.1038/srep15918

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Bharhani MS, Borojevic R, Basak S, Ho E, Zhou P, Croitoru K. IL-10 Protects Mouse Intestinal Epithelial Cells From Fas-Induced Apoptosis via Modulating Fas Expression and Altering Caspase-8 and FLIP Expression. Am J Physiol Gastrointest. Liver Physiol (2006) 291:G820–9. doi: 10.1152/ajpgi.00438.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Hasnain SZ, Tauro S, Das I, Tong H, Chen AC, Jeffery PL, et al. IL-10 Promotes Production of Intestinal Mucus by Suppressing Protein Misfolding and Endoplasmic Reticulum Stress in Goblet Cells. Gastroenterology (2013) 144:357–368 e359. doi: 10.1053/j.gastro.2012.10.043

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Shkoda A, Ruiz PA, Daniel H, Kim SC, Rogler G, Sartor RB, et al. Interleukin-10 Blocked Endoplasmic Reticulum Stress in Intestinal Epithelial Cells: Impact on Chronic Inflammation. Gastroenterology (2007) 132:190–207. doi: 10.1053/j.gastro.2006.10.030

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Blaschitz C, Raffatellu M. Th17 Cytokines and the Gut Mucosal Barrier. J Clin Immunol (2010) 30:196–203. doi: 10.1007/s10875-010-9368-7

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Eyerich K, Dimartino V, Cavani A. IL-17 and IL-22 in Immunity: Driving Protection and Pathology. Eur J Immunol (2017) 47:607–14. doi: 10.1002/eji.201646723

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Keir M, Yi Y, Lu T, Ghilardi N. The Role of IL-22 in Intestinal Health and Disease. J Exp Med (2020) 217:e20192195. doi: 10.1084/jem.20192195

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Hu J, Kang H, Liu C, Hu P, Yang M, Zhou F. Regulatory T Cells Could Improve Intestinal Barrier Dysfunction in Heatstroke. Inflammation (2019) 42:1228–38. doi: 10.1007/s10753-019-00983-6

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bae MJ, Shin HS, See HJ, Jung SY, Kwon DA, Shon DH. Baicalein Induces CD4(+)Foxp3(+) T Cells and Enhances Intestinal Barrier Function in a Mouse Model of Food Allergy. Sci Rep (2016) 6:32225. doi: 10.1038/srep32225

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, et al. Extrathymically Generated Regulatory T Cells Control Mucosal TH2 Inflammation. Nature (2012) 482:395–9. doi: 10.1038/nature10772

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: regulatory (treg) cell, intestinal epithelia cell, intestinal barrier, microbiota, dietary antigen

Citation: Jiang Z and Wu C (2022) Reciprocal Interactions Between Regulatory T Cells and Intestinal Epithelial Cells. Front. Immunol. 13:951339. doi: 10.3389/fimmu.2022.951339

Received: 23 May 2022; Accepted: 09 June 2022;
Published: 04 July 2022.

Edited by:

Xuyu Zhou, Chinese Academy of Sciences (CAS), China

Reviewed by:

Ye Zheng, Salk Institute for Biological Studies, United States

Copyright © 2022 Jiang and Wu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zhiqiang Jiang, Jiangzhq5@mail.sysu.edu.cn; Chuan Wu, chuan.wu@nih.gov

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.