Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of subepithelial mesenchymal cells that induce IgA and diversify gut microbiota

Abstract

Immunoglobulin A (IgA) maintains a symbiotic equilibrium with intestinal microbes. IgA induction in the gut-associated lymphoid tissues (GALTs) is dependent on microbial sampling and cellular interaction in the subepithelial dome (SED). However it is unclear how IgA induction is predominantly initiated in the SED. Here we show that previously unrecognized mesenchymal cells in the SED of GALTs regulate bacteria-specific IgA production and diversify the gut microbiota. Mesenchymal cells expressing the cytokine RANKL directly interact with the gut epithelium to control CCL20 expression and microfold (M) cell differentiation. The deletion of mesenchymal RANKL impairs M cell–dependent antigen sampling and B cell–dendritic cell interaction in the SED, which results in a reduction in IgA production and a decrease in microbial diversity. Thus, the subepithelial mesenchymal cells that serve as M cell inducers have a fundamental role in the maintenance of intestinal immune homeostasis.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: RANKL regulates epithelial CCL20 expression in the FAE of GALTs.
Figure 2: Membrane-bound RANKL induces M cell differentiation and CCL20 expression in the FAE of GALTs.
Figure 3: RANKL+ mesenchymal cells in contact with the FAE show distinct gene expression profiles.
Figure 4: Subepithelial mesenchymal cells have a role as MCi cells.
Figure 5: RANKL produced by mesenchymal cells is essential for B cell–DC interaction and antigen sampling in the SED.
Figure 6: Mesenchymal RANKL regulates germinal center formation in PPs.
Figure 7: Mesenchymal RANKL induces bacteria-specific IgA responses in the gut.
Figure 8: RANKL supplied by mesenchymal cells facilitates diversification of the gut microbiota.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Lozupone, C.A., Stombaugh, J.I., Gordon, J.I., Jansson, J.K. & Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220–230 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Fagarasan, S., Kawamoto, S., Kanagawa, O. & Suzuki, K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu. Rev. Immunol. 28, 243–273 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Fagarasan, S. et al. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science 298, 1424–1427 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Kawamoto, S. et al. Foxp3+ T cells regulate immunoglobulin A selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 41, 152–165 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Palm, N.W. et al. Immunoglobulin A coating identifies colitogenic bacteria in inflammatory bowel disease. Cell 158, 1000–1010 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pabst, O. New concepts in the generation and functions of IgA. Nat. Rev. Immunol. 12, 821–832 (2012).

    Article  CAS  PubMed  Google Scholar 

  7. Reboldi, A. et al. IgA production requires B cell interaction with subepithelial dendritic cells in Peyer's patches. Science 352, aaf4822 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Mueller, S.N. & Germain, R.N. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat. Rev. Immunol. 9, 618–629 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Owen, R.L. & Jones, A.L. Epithelial cell specialization within human Peyer's patches: an ultrastructural study of intestinal lymphoid follicles. Gastroenterology 66, 189–203 (1974).

    Article  CAS  PubMed  Google Scholar 

  10. Hase, K. et al. Uptake through glycoprotein 2 of FimH+ bacteria by M cells initiates mucosal immune response. Nature 462, 226–230 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Kanaya, T. et al. The Ets transcription factor Spi-B is essential for the differentiation of intestinal microfold cells. Nat. Immunol. 13, 729–736 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Knoop, K.A. et al. RANKL is necessary and sufficient to initiate development of antigen-sampling M cells in the intestinal epithelium. J. Immunol. 183, 5738–5747 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Rios, D. et al. Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol. 9, 907–916 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Mabbott, N.A., Donaldson, D.S., Ohno, H., Williams, I.R. & Mahajan, A. Microfold (M) cells: important immunosurveillance posts in the intestinal epithelium. Mucosal Immunol. 6, 666–677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Tanaka, Y. et al. Selective expression of liver and activation-regulated chemokine (LARC) in intestinal epithelium in mice and humans. Eur. J. Immunol. 29, 633–642 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Kong, Y.Y. et al. OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397, 315–323 (1999).

    Article  CAS  PubMed  Google Scholar 

  17. Fata, J.E. et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell 103, 41–50 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Yasuda, H. et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc. Natl. Acad. Sci. USA 95, 3597–3602 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lum, L. et al. Evidence for a role of a tumor necrosis factor-α (TNF-α)-converting enzyme-like protease in shedding of TRANCE, a TNF family member involved in osteoclastogenesis and dendritic cell survival. J. Biol. Chem. 274, 13613–13618 (1999).

    Article  CAS  PubMed  Google Scholar 

  20. Nakashima, T. et al. Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-κB ligand: modulation of the expression by osteotropic factors and cytokines. Biochem. Biophys. Res. Commun. 275, 768–775 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Hikita, A. et al. Negative regulation of osteoclastogenesis by ectodomain shedding of receptor activator of NF-κB ligand. J. Biol. Chem. 281, 36846–36855 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Cella, M., Miller, H. & Song, C. Beyond NK cells: the expanding universe of innate lymphoid cells. Front. Immunol. 5, 282 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Cella, M., Otero, K. & Colonna, M. Expansion of human NK-22 cells with IL-7, IL-2, and IL-1β reveals intrinsic functional plasticity. Proc. Natl. Acad. Sci. USA 107, 10961–10966 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Totsuka, T. et al. RANK-RANKL signaling pathway is critically involved in the function of CD4+CD25+ regulatory T cells in chronic colitis. J. Immunol. 182, 6079–6087 (2009).

    Article  CAS  PubMed  Google Scholar 

  25. Taylor, R.T. et al. Lymphotoxin-independent expression of TNF-related activation-induced cytokine by stromal cells in cryptopatches, isolated lymphoid follicles, and Peyer's patches. J. Immunol. 178, 5659–5667 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Katakai, T. et al. Organizer-like reticular stromal cell layer common to adult secondary lymphoid organs. J. Immunol. 181, 6189–6200 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Katakai, T. Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer. Front. Immunol. 3, 200 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  28. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003).

    Article  CAS  PubMed  Google Scholar 

  29. Geske, M.J., Zhang, X., Patel, K.K., Ornitz, D.M. & Stappenbeck, T.S. Fgf9 signaling regulates small intestinal elongation and mesenchymal development. Development 135, 2959–2968 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Hamada, H. et al. Identification of multiple isolated lymphoid follicles on the antimesenteric wall of the mouse small intestine. J. Immunol. 168, 57–64 (2002).

    Article  CAS  PubMed  Google Scholar 

  32. Bouskra, D. et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 456, 507–510 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Tsuji, M. et al. Requirement for lymphoid tissue-inducer cells in isolated follicle formation and T cell-independent immunoglobulin A generation in the gut. Immunity 29, 261–271 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Golovkina, T.V., Shlomchik, M., Hannum, L. & Chervonsky, A. Organogenic role of B lymphocytes in mucosal immunity. Science 286, 1965–1968 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Ebisawa, M. et al. CCR6hiCD11cint B cells promote M-cell differentiation in Peyer's patch. Int. Immunol. 23, 261–269 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kimura, S. et al. Visualization of the entire differentiation process of murine M cells: suppression of their maturation in cecal patches. Mucosal Immunol. 8, 650–660 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Debard, N., Sierro, F., Browning, J. & Kraehenbuhl, J.P. Effect of mature lymphocytes and lymphotoxin on the development of the follicle-associated epithelium and M cells in mouse Peyer's patches. Gastroenterology 120, 1173–1182 (2001).

    Article  CAS  PubMed  Google Scholar 

  38. Liu, Y. et al. CCL20 mediates RANK/RANKL-induced epithelial-mesenchymal transition in endometrial cancer cells. Oncotarget 7, 25328–25339 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Ivanov, I.I. et al. Induction of intestinal TH17 cells by segmented filamentous bacteria. Cell 139, 485–498 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Suzuki, K. et al. Aberrant expansion of segmented filamentous bacteria in IgA-deficient gut. Proc. Natl. Acad. Sci. USA 101, 1981–1986 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Franke, A. et al. Genome-wide meta-analysis increases to 71 the number of confirmed Crohn's disease susceptibility loci. Nat. Genet. 42, 1118–1125 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Franchimont, N. et al. Increased expression of receptor activator of NF-κB ligand (RANKL), its receptor RANK and its decoy receptor osteoprotegerin in the colon of Crohn's disease patients. Clin. Exp. Immunol. 138, 491–498 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Madison, B.B. et al. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J. Biol. Chem. 277, 33275–33283 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Nakashima, T. et al. Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat. Med. 17, 1231–1234 (2011).

    Article  CAS  PubMed  Google Scholar 

  45. Hanada, R. et al. Central control of fever and female body temperature by RANKL/RANK. Nature 462, 505–509 (2009).

    Article  CAS  PubMed  Google Scholar 

  46. Oh-Hora, M. et al. Agonist-selected T cell development requires strong T cell receptor signaling and store-operated calcium entry. Immunity 38, 881–895 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Nitta, T. et al. The thymic cortical epithelium determines the TCR repertoire of IL-17-producing γδT cells. EMBO Rep. 16, 638–653 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sawa, S. et al. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat. Immunol. 12, 320–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Fletcher, A.L. et al. Reproducible isolation of lymph node stromal cells reveals site-dependent differences in fibroblastic reticular cells. Front. Immunol. 2, 35 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Donaldson, D.S., Bradford, B.M., Artis, D. & Mabbott, N.A. Reciprocal regulation of lymphoid tissue development in the large intestine by IL-25 and IL-23. Mucosal Immunol. 8, 582–595 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to T. Yamada (Tokyo Institute of Technology), H. Mori (Tokyo Institute of Technology), K. Hase (Keio University), Y. Obata (Keio University), H. Kiyono (the University of Tokyo), Y. Morishita and H. Tanaka (Okayama University) for insightful discussion and valuable technical assistance. We thank all our laboratory members for discussion. We thank S. Nitta, Y. Nakayama, K. Kaneki, Y. Ogiwara, K. Kusubata, R. Yanobu-Takanashi and K. Nakano for technical assistance. D. Kioussis (UK National Institute for Medical Research) provided Vav-iCre mice. Supported in part by a Grant-in-Aid for Specially Promoted Research from the Japan Society for Promotion of Science (JSPS) (15H05703); grants for Exploratory Research for Advanced Technology (ERATO) Program and PRESTO from JST; Yakult Bio-Science Foundation; and JSPS KAKENHI (25111503 and 16H05202). K.N. was supported by Japanese Society for Immunology (JSI) Kibou Scholarship for Doctoral Students in Immunology.

Author information

Authors and Affiliations

Authors

Contributions

K.N. performed most of the experiments, interpreted the results and prepared the manuscript. S.S. provided advice on project planning and data interpretation and contributed to the manuscript preparation. T. Nitta, T.O., M.T., J.M.P. and T. Nakashima provided genetically modified mice and contributed to data interpretation and discussion. H.T. directed the project and wrote the manuscript.

Corresponding author

Correspondence to Hiroshi Takayanagi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of Tnfsf11ΔS/ΔS mice by CRISPR–Cas9-mediated gene editing.

(a) Scheme of the Tnfsf11ΔS allele. The Tnfsf11 cleavage sites were genetically trimmed by CRISPR/Cas9-mediated homology-directed repair. (b) The target sequences of sgRNA1 and sgRNA2, and the sequences of single-stranded oligodeoxynucleotides (ssODNs). (c) Flow cytometry analysis of RANKL expression in lymphocytes in PPs. Histograms show RANKL expression patterns in the ILC3s or T cells of the PPs from Tnfsf11ΔS/ΔS and Tnfsf11+/+ mice after four hours of stimulation with PMA/ionomycin (n=8,5 and 3 mice respectively). (d) Representative histological sections of RANKL+ mesenchymal cell in the SED of PPs from Tnfsf11+/+ and Tnfsf11ΔS/ΔS mice. Sections were stained for podoplanin, RANKL and DAPI. Scale bars represent 10 μm. (e) Q-PCR analysis of the Tnfsf11 transcripts in MAdCAM-1podoplanin+CD21CD31CD45 mesenchymal cells in PPs (n=5 and 3 mice respectively). Statistical analyses were carried out using Student’s t test (e) or analysis of variance (ANOVA) with Turkey’s multiple-comparison test (c). Error bars denote the mean ± SEM. NS, not significant (p>0.05); *P<0.05 and **P<0.01.

Supplementary Figure 2 Gating strategy for mesenchymal stromal cells and lymphocytes in PPs.

(a) Flow cytometry analysis of CD45EpCAMTer119 stromal subsets in PPs. The CD45CD21/35 fraction was enriched with a MACS Column before staining in order to eliminate CD45+ hematopoietic cells and CD21/35+ follicular dendritic cells. LECs, lymphatic endothelial cells; BECs, blood endothelial cells; DN, double negative cells. (b) Flow cytometry analysis of RANKL expression in CD45+ lymphocytes in PPs after four hours of stimulation with or without PMA/ionomycin. The numbers indicate the mean ± SEM. The data are from three independent experiments (n=3 mice).

Supplementary Figure 3 Histological analysis of mesenchymal cell populations in PPs.

(a) MAdCAM-1 expressing mesenchymal stromal cells in PPs. Blood endothelial cells (BECs), follicular dendritic cells (FDCs) and marginal reticular cells (MRCs) expressed MAdCAM-1. Scale bars, 100 μm (left and middle), 25 μm (right). (b) Analysis of T cell-containing interfollicular zones in the PPs from Tnfsf11fl/Δ, Tnfsf11fl/Δ Twist2-Cre, Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice. Scale bars, 200 μm. (c) Identification of RANKL expressing mesenchymal cells in the SED of PPs. Scale bars, 25 μm. (d) HE staining of the small or large intestine of Tnfsf11fl/Δ, Tnfsf11fl/Δ Twist2-Cre, Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice. Scale bars, 50 μm.

Supplementary Figure 4 Histological identification of RANKL+ mesenchymal cells in the large intestine.

(a) Representative images of colonic patches. RANKL+podoplanin+ mesenchymal cells were found to be in contact with the FAE (upper panels). MAdCAM-1 was expressed by podoplanin blood endothelial cells (BECs), but MAdCAM-1+podoplanin+ MRCs were not observed (lower panels). Scale bars, 50, 20, 20 or 20 μm (left to right). (b,c) Representative sections of colonic ILFs (b) and cecal patches (c). RANKL+podoplanin+ mesenchymal cells interacting with the FAE were observed in colonic ILFs and cecal patches, while MAdCAM-1+podoplanin+ MRCs were identified only in cecal patches. The dashed lines indicate the FAE basal membrane. Scale bars, 50 μm (b, left), 100 μm (c, left), 15 μm (middle and right). (d) Representative histological images of the FAE of colonic patches, colonic ILFs and cecal patches of Tnfsf11+/+, Tnfsf11ΔS/ΔS and Tnfsf11fl/Δ Twist2-Cre mice. Scale bars, 10 μm. (e) Representative sections of the colonic patches, colonic ILFs and cecal patches from Tnfsf11fl/Δ and Tnfsf11fl/Δ Twist2-Cre mice. Scale bars, 100 μm (upper and lower panels), 50 μm (middle panels). (f) Representative images of the large intestinal lamina propria (LILP) obtained from Tnfsf11fl/Δ, Tnfsf11fl/Δ Twist2-Cre, Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice. Scale bars, 25 μm.

Supplementary Figure 5 Mesenchymal cell–specific deletion of RANKL in Tnfsf11fl/Δ; Twist2-Cre mice.

(a ,b,c) Histological sections of the SED of the PPs from Twist2-Cre CAG-tdTomato mice. The dashed lines indicate the FAE basal membrane. Scale bars, 5 μm (a), 25 μm (b,c). (d) Q-PCR analysis of the Pigr transcripts in the FAE of PPs from Tnfsf11fl/Δ and Tnfsf11fl/Δ Twist2-Cre mice (n=8 PPs). (e) Q-PCR analysis of the Tnfsf11 transcripts in MAdCAM-1podoplanin+CD21CD31CD45 mesenchymal cells (n=5 and 4 mice, respectively). (f) Flow cytometry analysis of RANKL expression in ILC3s in PPs from Tnfsf11fl/Δ and Tnfsf11fl/Δ Twist2-Cre mice after four hours of stimulation with PMA/ionomycin. (g) Mean fluorescence intensity (MFI) of RANKL expression in ILC3s and T cells (n=3 mice). (h) Q-PCR analysis of the Tnfsf11 transcripts in the MAdCAM-1podoplanin+CD21CD31CD45 mesenchymal cells of Tnfsf11fl/Δ and Tnfsf11fl/Δ Vav-iCre mice (n=4 mice). (i,j) Flow cytometry analysis of RANKL expression in ILC3s (i) and T cells (j) in PPs from Tnfsf11+/+, Tnfsf11fl/Δ and Tnfsf11fl/Δ Vav-iCre mice after four hours of stimulation with PMA/ionomycin (n=23,9,7 and 4 mice respectively). Statistical analyses were carried out using Student’s t test (d,g,h) or analysis of variance (ANOVA) with Dunnett's multiple-comparison test (i,j) Error bars denote the mean ± SEM. NS, not significant (p>0.05); *P<0.01 and **P<0.001.

Supplementary Figure 6 Epithelial RANK is crucial for IgA induction in the gut.

(a) Assessment of antigen sampling in PPs. Ligated loop assay was performed in the Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice (n=12 slides). (b) The number of IgD+ pre-GC B cells interacting with CD11c+ DCs in the SED quantified by histological sections (n=8 and 7 slides respectively). (c) Q-PCR analysis of the Aicda transcripts in the B220+ B cells sorted from PPs (n=7 mice). (d) B cell-containing ILFs in the terminal ileum quantified by whole-mount immunostaining (n=12 and 13 mice respectively). (e) The size of PPs measured by stereomicroscopy (n=33 and 31 PPs respectively). (f) Histological analysis of GL7+ germinal center formation in PPs. Scale bars, 200 μm. (g,h,i) Flow cytometry analysis of GL7+ GC B cells in B220+ cells (g), IgA+ B cells in B220+ cells (h) and PD-1+CXCR5+ Tfh cells in CD3+CD4+ cells (i) in the PPs (n=8 and 9 mice). (j) IgM+ and IgA+ cells in CD45+CD3 cells in the SILP (n=9 mice). (k) The fecal IgA and serum IgA, IgG and IgM levels measured by ELISA (n=7 mice). (l) Cholera toxin-specific IgA production determined by ELISA (n=6 mice). (m) The percentages of IgA-coated bacteria in the total bacteria determined by flow cytometry (n=7 mice). The data are from 3 (m), 4 (e), 6(i), 7(g,h,j) and 10 (d) independent experiments. Statistical analyses were carried out using Student’s t test, and the data are shown as the mean ± SEM. NS, not significant (p>0.05); *P<0.05, **P<0.01 and ***P<0.001. 4-6 week old mice were used along with littermate controls. A similar reduction in IgA production was observed in 8-10 week old Tnfrsf11afl/Δ Vil1-Cre mice.

Supplementary Figure 7 Analysis of B cell homeostasis in M cell–deficient mice.

(a,b) Flow cytometry analysis of B cells in the bone marrow (BM), superficial lymph nodes (sLNs), peritoneal cavity (PC), peripheral blood (PB) or spleen obtained from Tnfsf11fl/Δ, Tnfsf11fl/Δ Twist2-Cre (a), Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice (b) (n=4 mice). (c,d) Flow cytometry analysis of CD11c+1-A/1-E+ dendritic cells in Peyer’s patches (PPs) (n=3 mice) (c) (n=7 mice) (d). Statistical analyses were carried out using Student’s t test, and the data are shown as the mean ± SEM. NS, not significant (p>0.05).

Supplementary Figure 8 Epithelial RANK is required for the diversification of the gut microbiota.

(a) Relative abundance (major order; family) in the fecal pellets from Tnfrsf11afl/Δ and Tnfrsf11afl/Δ Vil1-Cre mice (n=12 fecal pellets from 6 mice). (b) Relative abundance of the 16S rRNA gene of SFB among the total bacteria (n=17 and 24 mice respectively). (c) Differences between bacterial communities. PCoA and PERMANOVA comparisons of Bray-Curtis distances are shown (n=12 fecal pellets from 6 mice). (d) Diversity of bacterial species shown by a chao1 refraction measure based on 1,000-10,000 sequences (n=12 fecal pellets from 6 mice). Statistical analyses were carried out using Student’s t test (b,d). Error bars denote the mean ± SEM. *P<0.05 and **P<0.01. 4-7 week old mice were separately caged after weaning and used along with littermate controls. (e) Schematic model of the IgA production against the gut microbiota organized by MCi cells in GALTs. Membrane-bound RANKL provided by MCi cells induces M cell differentiation and CCL20 production by enterocytes in the FAE. CCL20 and the microbial antigens sampled by M cells cooperatively stimulate the B cell-DC interaction required for the initiation of IgA class switching. IgA is secreted into the lumen and shapes the gut microbiota. MRCs, another mesenchymal stromal subset in GALTs, express high levels of CXCL13 and may guide B cells from the SED to follicles so as to promote germinal center formation.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 2616 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nagashima, K., Sawa, S., Nitta, T. et al. Identification of subepithelial mesenchymal cells that induce IgA and diversify gut microbiota. Nat Immunol 18, 675–682 (2017). https://doi.org/10.1038/ni.3732

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3732

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology