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.

  • Letter
  • Published:

Compartmentalized gut lymph node drainage dictates adaptive immune responses

Abstract

The intestinal immune system has the challenging task of tolerating foreign nutrients and the commensal microbiome, while excluding or eliminating ingested pathogens. Failure of this balance leads to conditions such as inflammatory bowel diseases, food allergies and invasive gastrointestinal infections1. Multiple immune mechanisms are therefore in place to maintain tissue integrity, including balanced generation of effector T (TH) cells and FOXP3+ regulatory T (pTreg) cells, which mediate resistance to pathogens and regulate excessive immune activation, respectively1,2,3,4. The gut-draining lymph nodes (gLNs) are key sites for orchestrating adaptive immunity to luminal perturbations5,6,7. However, it is unclear how they simultaneously support tolerogenic and inflammatory reactions. Here we show that gLNs are immunologically specific to the functional gut segment that they drain. Stromal and dendritic cell gene signatures and polarization of T cells against the same luminal antigen differ between gLNs, with the proximal small intestine-draining gLNs preferentially giving rise to tolerogenic responses and the distal gLNs to pro-inflammatory T cell responses. This segregation permitted the targeting of distal gLNs for vaccination and the maintenance of duodenal pTreg cell induction during colonic infection. Conversely, the compartmentalized dichotomy was perturbed by surgical removal of select distal gLNs and duodenal infection, with effects on both lymphoid organ and tissue immune responses. Our findings reveal that the conflict between tolerogenic and inflammatory intestinal responses is in part resolved by discrete gLN drainage, and encourage antigen targeting to specific gut segments for therapeutic immune modulation.

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

Fig. 1: The gLNs are metabolically and immunologically unique according to the gut segment drained.
Fig. 2: Migratory dendritic cell profiles and CD4+ T cell polarization differ between gLNs and follow proximal-to-distal gradients.
Fig. 3: Compartmentalization of CD4+ T cell fates permits vaccination via ileocaecal gLNs.
Fig. 4: Duodenal infection leads to a compartmentalized immune conflict in the duodenal gLNs and to compromised oral tolerance.

Similar content being viewed by others

Data availability

Source data for all figures are provided with the paper. For RNA-seq experiments the raw and processed data generated here can be obtained at the Gene Expression Omnibus database under the accession code: GSE121811.

References

  1. Honda, K. & Littman, D. R. The microbiota in adaptive immune homeostasis and disease. Nature 535, 75–84 (2016).

    Article  ADS  CAS  Google Scholar 

  2. Yang, Y. et al. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature 510, 152–156 (2014).

    Article  ADS  CAS  Google Scholar 

  3. Xu, M. et al. c-MAF-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont. Nature 554, 373–377 (2018).

    Article  ADS  CAS  Google Scholar 

  4. Mucida, D. et al. Oral tolerance in the absence of naturally occurring Tregs. J. Clin. Invest. 115, 1923–1933 (2005).

    Article  CAS  Google Scholar 

  5. Randolph, G. J., Ivanov, S., Zinselmeyer, B. H. & Scallan, J. P. The lymphatic system: integral roles in immunity. Annu. Rev. Immunol. 35, 31–52 (2017).

    Article  CAS  Google Scholar 

  6. Fonseca, D. M. et al. Microbiota-dependent sequelae of acute infection compromise tissue-specific immunity. Cell 163, 354–366 (2015).

    Article  Google Scholar 

  7. Cording, S. et al. The intestinal micro-environment imprints stromal cells to promote efficient Treg induction in gut-draining lymph nodes. Mucosal Immunol. 7, 359–368 (2014).

    Article  CAS  Google Scholar 

  8. Worbs, T. et al. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J. Exp. Med. 203, 519–527 (2006).

    Article  CAS  Google Scholar 

  9. Van den Broeck, W., Derore, A. & Simoens, P. Anatomy and nomenclature of murine lymph nodes: descriptive study and nomenclatory standardization in BALB/cAnNCrl mice. J. Immunol. Methods 312, 12–19 (2006).

    Article  Google Scholar 

  10. Carter, P. B. & Collins, F. M. The route of enteric infection in normal mice. J. Exp. Med. 139, 1189–1203 (1974).

    Article  CAS  Google Scholar 

  11. Houston, S. A. et al. The lymph nodes draining the small intestine and colon are anatomically separate and immunologically distinct. Mucosal Immunol. 9, 468–478 (2016).

    Article  CAS  Google Scholar 

  12. Gautreaux, M. D., Deitch, E. A. & Berg, R. D. Bacterial translocation from the gastrointestinal tract to various segments of the mesenteric lymph node complex. Infect. Immun. 62, 2132–2134 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Veenbergen, S. et al. Colonic tolerance develops in the iliac lymph nodes and can be established independent of CD103+ dendritic cells. Mucosal Immunol. 9, 894–906 (2016).

    Article  CAS  Google Scholar 

  14. Pezoldt, J. et al. Neonatally imprinted stromal cell subsets induce tolerogenic dendritic cells in mesenteric lymph nodes. Nat. Commun. 9, 3903 (2018).

    Article  ADS  Google Scholar 

  15. Hammerschmidt, S. I. et al. Stromal mesenteric lymph node cells are essential for the generation of gut-homing T cells in vivo. J. Exp. Med. 205, 2483–2490 (2008).

    Article  CAS  Google Scholar 

  16. Durai, V. & Murphy, K. M. Functions of murine dendritic cells. Immunity 45, 719–736 (2016).

    Article  CAS  Google Scholar 

  17. Esterházy, D. et al. Classical dendritic cells are required for dietary antigen-mediated induction of peripheral Treg cells and tolerance. Nat. Immunol. 17, 545–555 (2016).

    Article  Google Scholar 

  18. Atarashi, K. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).

    Article  ADS  CAS  Google Scholar 

  19. Sano, T. et al. An IL-23R/IL-22 circuit regulates epithelial serum amyloid A to promote local effector Th17 responses. Cell 163, 381–393 (2015).

    Article  CAS  Google Scholar 

  20. Atarashi, K. et al. Th17 Cell induction by adhesion of microbes to intestinal epithelial cells. Cell 163, 367–380 (2015).

    Article  CAS  Google Scholar 

  21. Hadis, U. et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity 34, 237–246 (2011).

    Article  CAS  Google Scholar 

  22. Balmer, M. L. et al. The liver may act as a firewall mediating mutualism between the host and its gut commensal microbiota. Sci. Transl. Med. 6, 237ra66 (2014).

    Article  Google Scholar 

  23. Bouziat, R. et al. Reovirus infection triggers inflammatory responses to dietary antigens and development of celiac disease. Science 356, 44–50 (2017).

    Article  ADS  CAS  Google Scholar 

  24. Mukai, K., Karasuyama, H., Kabashima, K., Kubo, M. & Galli, S. J. Differences in the importance of mast cells, basophils, IgE, and IgG versus that of CD4+ T cells and ILC2 cells in primary and secondary immunity to Strongyloides venezuelensis. Infect. Immun. 85, e00053-17 (2017).

    Article  Google Scholar 

  25. Tussiwand, R. et al. Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 42, 916–928 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all members of the Mucida laboratory, past and present, for assistance, discussions and reading of the manuscript; B. Reis for figure preparation; F. Matheis for propagating S. venezuelensis; A. Rogoz and S. Gonzalez for the maintenance of mice; T. Rendon and B. Lopez for genotyping; K. Gordon and K. Chosphel for assistance with cell sorting; the Rockefeller University Bio-imaging Research Center for assistance with microscopy and image analysis; the Rockefeller University Genomics Center for RNA sequencing; Rockefeller University employees for assistance; M. Nussenzweig, G. Victora and J. Lafaille and their respective laboratory members for discussions and suggestions; D. Littman and M. Xu (NYU) for 7B8tg mice and faeces from SFB monocolonized mice; and S. Galli and K. Matsushita (Stanford University) for providing S. venezuelensis and guidance on how to maintain it. This work was supported by a Swiss National Science Foundation postdoctoral fellowship and University of Chicago start-up funds (D.E.); a CAPES fellowship (M.C.C.C.); an NIH F31 Kirchstein Fellowship, Philip M. Levine Fellowship, and Kavli Neural Systems Institute Graduate Fellowship (P.A.M.); the Leona M. and Harry B. Helmsley Charitable Trust, the Crohn’s & Colitis Foundation of America Senior Research Award, the Burroughs Wellcome Fund PATH Award, and National Institute of Health grants R21AI31188, R01DK113375 and R01DK093674 (D.M.).

Reviewer information

Nature thanks Rodney Newberry and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

D.E. initiated, designed, performed and analysed the research, and wrote the paper. M.C.C.C. designed and performed the research. L.M. performed the dendritic cell sorting and guided the RNA-seq library preparation. P.A.M. performed RNA-seq data alignment and analyses and fast green injection. T.B.R.d.C. performed RNA-seq data alignment and analyses and designed the lymph node schematic. A.L. performed radioactive studies. M.E. prepared, imaged and analysed lymph nodes upon S. venezuelensis infection. A.M.C.F. initiated the S. venezuelensis model and contributed to the experimental design using the model. All authors edited the paper. D.M. initiated, designed and supervised the research, and wrote the paper.

Corresponding authors

Correspondence to Daria Esterházy or Daniel Mucida.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Extended analysis of gLN connectivity and drainage in the peritoneal cavity.

ae, 3D reconstruction of mouse lymphatics (anti-LYVE-1) after solvent clearing (iDISCO+) and light sheet microscopy of: the central chain of gLNs connected to the duodenum via afferent vessels (a), the D-gLNs with respect to the liver and pancreas (co-stained with anti-insulin; b), the distal intestine gLNs connected to the ileum (c), and individually dissected peritoneal gLNs (d) of SPF C57BL/6 mice. e, All peritoneal gLNs from germ-free and SPF C57BL/6 mice. fi, Fast green spreading upon injection into gLNs draining the duodenum (f), jejunum (g), ileum (h) and caecum-proximal colon (i) from SPF C57BL/6 mice. jm, Fast green spreading upon injection into the lymphatics in the muscularis of the duodenum (j), jejunum (k), ileum (l) and caecum (m) of 10-week-old mice. Pictures taken up to 15 min after dye injection. nq, 3D reconstruction of mouse lymphatic vessels (anti-LYVE-1) after solvent clearing (iDISCO+) and light sheet microscopy in the duodenum (n, o), ileum (p), and colon (q), highlighting the submucosal lymphatic network. In o the arrow denotes the direction of lymph flow from intestine to gLNs; CD11c was revealed by using ItgaxVenus mice and staining against GFP.

Extended Data Fig. 2 Imaging of intestinal lymphatic vessels and characterization of [3H]retinol absorption along the gut of SPF and germ-free mice.

a, 3D reconstruction of mouse lymphatic vessels (anti-LYVE-1) after iDISCO+ processing of villi and submucosa along the intestines of germ-free (GF) and SPF C57BL/6 mice; lymphatics protruding into the villi (lacteals) are indicated. b, Percentage of [3H]retinol absorption into lymph (L), portal vein serum (PV) or systemic serum (S) of mice 3 h after gavage with 1 µCi [3H]retinol in 100 µl olive oil, with or without pre-treatment with 5 µl Pluronic L-81 3 h before gavage (n = 4 per group). c, d, Percentage of [3H]retinol absorption into the duodenum (c) or duodenal gLN (d) of C57BL/6 mice 8 h after gavage with 1 µCi [3H]retinol in 100 µl olive oil with or without pre-treatment with 5 µl Pluronic L-81 3 h before gavage (n = 8 or 9 per group, as indicated). Data pooled from two independent experiments with 4 or 5 animals per group each. eg, Percentage of [3H]retinol absorption into systemic plasma (e), indicated intestinal tissue (f) or gLNs (g) from germ-free or SPF C57BL/6 mice 1 or 5 h after gavage with 1 µCi [3H]retinol in 100 µl olive oil (n = 3 per group). Data representative of two independent experiments. *P < 0.05, **P < 0.01, ***P < 0.005 (one-tailed t-test or ANOVA). g, comparison of results from germ-free mice by ANOVA; s, comparison of results from SPF mice by ANOVA; $, comparison at 1 h time point; #, comparison at 5 h time point.

Extended Data Fig. 3 Extended analysis of gLN stromal and dendritic cell differences according to gut segment drained.

a, Flow cytometry plot showing gating of FRCs and LECs for RNA-seq. Pre-gates indicated above. bg, Some differentially regulated pathways among LECs sorted from duodenal versus colonic gLNs (NES, normalized enrichment scores) (b); differentially expressed genes among LECs sorted from ileal versus colonic gLNs or ileal versus duodenal gLNs (c, d); some differentially regulated pathways among FRCs sorted from duodenal versus colonic gLNs (e); differentially expressed genes among FRCs sorted from ileal versus colonic gLNs or ileal versus duodenal gLNs (f, g), all from SPF C57BL/6 mice and identified by RNA-seq. Blue, metabolism-related pathways or genes; red, immunity-related pathways or genes. h, Flow cytometry plot showing gating of CD103+CD11b+ and CD103+CD11b dendritic cells for RNA-seq. Pre-gates indicated above. il, Differentially expressed genes identified by RNA-seq among CD103+CD11b+ (i, k) and CD103+CD11b (j, l) dendritic cells sorted from ileal versus colonic gLNs or ileal versus duodenal gLNs from SPF C57BL/6 mice. m, n, Some differentially regulated pathways among CD103+CD11b+ (m) and CD103+CD11b (n) dendritic cells sorted from duodenal versus colonic gLNs. oq, Mean fluorescence intensity of fluorescein isothiocyanate-positive boron-dipyrromethene-tagged aminoacetate (Aldefluor) in CD103+CD11b (o), CD103+CD11b+ (p), and CD8α+ or CD11b+ (q) dendritic cells from gLNs from germ-free and SPF C57BL/6 mice (n = 3) assessed by flow cytometry 30 min after the addition of substrate. r, Ratio of CD103+CD11b+ to CD103+CD11b dendritic cells in gLNs from SPF and germ-free mice (n = 4, representative of two independent experiments). *P < 0.05, **P < 0.01, ***P < 0.005; ns, not significant (one-tailed t-test or ANOVA).

Extended Data Fig. 4 Extended analysis of CD4+ T cells in the different gLNs of SPF and germ-free mice and activation of OT-II CD45.1 cells in all gLNs upon OVA gavage.

ae, Frequency of TCRβ+CD4+ cells among CD45+ cells (a), FOXP3+NRP1+ (b) and FOXP3+NRP1 cells (c) among TCRβ+CD4+ cells, RORγT+ cells among FOXP3+NRP1 cells (d) and FOXP3RORγT+ cells among TCRβ+CD4+ cells (e) in indicated gLNs from germ-free or SPF C57BL/6 mice (n = 4). Data representative of two independent experiments. fi, Frequency of CD25+ cells among CD45.1+ cells (f) and CD45.1+ cells among TCRβ+CD4+ cells (g), total CD45.1+ cells (h), and representative CFSE (carboxyfluorescein diacetate succinimidyl ester) dilution histogram (i) in indicated gLNs 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into SPF CD45.2 C57BL/6 host mice (n = 3), after gavage of OVA 48 h and 24 h before analysis.

Extended Data Fig. 5 Extended analysis of OT-II CD45.1 and dendritic cells upon OVA administration in different gLNs of SPF and germ-free mice.

a, b, Frequency of CD45.1+ cells among TCRβ+CD4+ cells (a) and flow cytometry plots for gLN FOXP3 and CD25 expression (b) in indicated gLNs 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into germ-free and SPF CD45.2 C57BL/6 host mice (n = 4) and after gavage of OVA 48 h and 24 h before analysis. ce, 125I recovery in indicated gLNs (c), intestinal tissue (d) and plasma (e) of germ-free and SPF C57BL/6 mice (n = 3 per time point and group), 1 h or 5 h after gavage with 4 × 106 CPM 125I-OVA in 50 mg cold OVA. f, g, Division index of CD45.1+ cells (f) and frequency of CD25+ cells among CD45.1+ cells (g) in indicated gLNs as in a, b. hm, Frequency of CD45.1+ cells among TCRβ+CD4+ cells (h), division index of CD45.1+ cells (i), total CD45.1+ cell count (j), frequency of FOXP3hi cells among CD45.1+ cells (k), representative flow cytometry plot for FOXP3hi and total FOXP3 gating amongst CD3+CD4+ cells in D-gLN (l) and frequency of FOXP3hi cells among CD45.2+TCRβ+CD4+ cells (m) in indicated gLNs or spleen, 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into SPF CD45.2 C57BL/6 host mice (n = 3) and after intravenous injection of OVA 48 h before analysis. n, Frequency of IL-12/23 p40+ cells among CD103+CD11b dendritic cells in indicated gLNs of germ-free and SPF C57BL/6 mice (n = 4 per group). #, fewer than 200 cells were recovered. *P < 0.05, **P < 0.01, ***P < 0.005; ns, not significant (one-tailed t-test or ANOVA).

Extended Data Fig. 6 Extended analysis of OT-II CD45.1 cell seeding and fate in gLNs upon intestinal OVA injection or infection.

ah, Frequency of FOXP3+RORγT (a), FOXP3+RORγT+ (b), FOXP3RORγT+ (c), FOXP3+CD25+ (d), or FOXP3CD25+ (e) cells among CD45.1+ OT-II cells, frequency of CD45.1+ OT-II cells among TCRβ+CD4+ cells (f), division index (g) and total TCRβ+CD4+ cell counts (h) in indicated gLNs of mice 48 h after duodenal or ileal injection with OVA or OVA–CT, performed 16 h after adoptive transfer of 1 × 106 naive OT-II cells into SPF CD45.2 C57BL/6 host mice (n = 3). il, Survival curve (sham n = 5, duo n = 6, ile n = 8; i), and frequency in ileal lamina propria of FOXP3RORγT+CD45.1+ (j), FOXP3RORγT+CD45.1+ (k) and FOXP3+RORγtCD45.1+ (l) cells among TCRβ+CD4+ cells (n = 4 per group) of mice infected with Stm-OVA 9 days after duodenal or ileal injection with OVA–CT versus sham operation, and 10 days after adoptive transfer of 1 × 105 naive OT-II cells into SPF CD45.2 C57BL/6 host mice. jl, Intestines removed 48 h after infection. *P < 0.05 (Mantel–Cox and Gehan–Breslow–Wilcoxon, i), ##P < 0.005 (logrank, i), *P < 0.05 (ANOVA, j–l). mp, Flow cytometry plot of CD25+ and CFSE (m), frequency of CD451+ OT-II cells among TCRβ+CD4+ cells (n), CD45.1+ division index (o) and total CD45.1+ cell counts (p) in indicated gLNs 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into SPF CD45.2 C57BL/6 mice infected with Citro-OVA 9 days earlier, and after gavage with OVA or PBS 48 h and 24 h before analysis (n = 5). Data representative of two independent experiments. qu, Frequency of total FOXP3+ (q), FOXP3CD25+ (r), and FOXP3RORγT+CD451+ (s) cells among CD45.1+ cells, frequency of CD45.1+ cells among TCRβ+CD4+ cells (t), and frequency of FOXP3RORγT+CD45.2+ cells among TCRβ+CD4+ cells (u) in indicated gLNs 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into SPF CD45.2 C57BL/6 host mice infected or not with Citrobacter 9 days earlier, and after gavage with OVA 48 h and 24 h before analysis (n = 5). ns, not significant in ANOVA or one-tailed t-test comparing gLNs of infected versus non-infected mice. *P < 0.05, **P < 0.01, ***P < 0.005 (ANOVA); #P < 0.05 (t-test).

Extended Data Fig. 7 Extended analysis of CD45.1+ 7B8tg cell fate in gLNs and gut upon removal of distal gLNs.

ac, Representative flow cytometry plots of RORγT+ and CD45.1+ cell frequency among CD3+CD4+ cells in indicated gLNs of sham or ΔicLN mice as quantified in Fig. 3g (a), and frequency of Vβ14+CD45.1+ cells among CD3+CD4+ cells (b) and RORγT+ cells among Vβ14+CD45.1+ (c) cells in indicated gLNs of mice with sham operation (n = 12) or surgical removal of the I- and C1-gLNs (ΔicLN, n = 14) 64 h (day 3, Fig. 3f) after adoptive transfer of 4 × 105 naive SFB-specific CD45.1+ 7B8tg cells into recently SFB-colonized SPF CD45.2 C57BL/6 Jax mice. Graph represents pooled data from three independent experiments with n = 4–5 per group each. d, e, Frequency of Vβ14+CD45.1+ cells among CD3+CD4+ cells (d) and RORγT+ cells among Vβ14+CD45.1+ cells (e) in indicated gLNs of Taconic sham or ΔicLN mice (n = 4) 64 h after adoptive transfer of 4 × 105 naive SFB-specific CD45.1+ cells. Note that SFB is a stable member of the microbial community in Taconic C57BL/6 mice through parental transmission. f, g, Total numbers of CD45+ cells in C2-gLN of recently SFB-colonized SPF mice (f) or Taconic mice (g) at point of harvest. hn, Frequency of Vβ14+CD45.1+ IL17a+ (h), Vβ14+CD45.1+RORγt+ (i), or Vβ14+CD45.1+ cells (j) among CD3+CD4+ cells, and of RORγT+ cells among Vβ14+CD45.1+ cells (k) and IL17a+ cells among RORγT+Vβ14+CD45.1+ cells (l) in lamina propria of indicated gut segments; frequency of Vβ14+CD45.1+ cells among CD3+CD4+ cells (m) and RORγT+ cells among Vβ14+CD45.1+ cells (n) in gLNs of sham or ΔicLN Jax mice (n = 4) 7 days after adoptive transfer of 5,000 naive CD45.1+ 7B8tg cells into recently SFB-colonized SPF CD45.2 C57BL/6 Jax mice. os, Frequency of Vβ14+CD45.1+ cells among CD3+CD4+ cells (o), RORγT+ cells among Vβ14+CD45.1+ cells (p) and IL17a+ cells among RORγT+Vβ14+CD45.1+ cells (q) in indicated lamina propria, or of Vβ14+CD45.1+ cells among CD3+CD4+ cells (r) and RORγT+ cells among Vβ14+CD45.1+ cells (s) in gLNs of sham or ΔicLN Taconic mice 7 days after adoptive transfer of 5,000 naive CD45.1+ 7B8tg cells (n = 4). Hash, fewer than 200 cells were recovered. *P < 0.05, **P < 0.01, ***P < 0.005 (one-tailed t-test or ANOVA); ns, not significant.

Extended Data Fig. 8 Effect of selective gLN removal on lymph flow, SFB colonization and oral tolerance.

a, Fast Green tracing of ileal lymphatic drainage to the gLNs of SPF C57BL/6 mice, 5 min after injection of 3 µl Fast Green in sham-operated (left, biological duplicates; Extended Data Fig. 2g) or ΔicLN (middle and right, biological triplicates) mice 3 weeks after surgery. Red arrows, sites of injection. b, c, Relative quantification of SFB-specific 16S in luminal contents of indicated gut segments from recently SFB-colonized SPF C57BL/6 Jax mice (b, n = 13 per group from four independent experiments) or parentally colonized Taconic mice (c, n = 7 per group from four independent experiments) after sham or ΔicLN surgery. di, Total eosinophils (d) and dendritic cells (e) in BALF and frequency of eosinophils (f) and dendritic cells (g) among CD45+ cells in lungs, and total IgE (h) and anti-OVA IgG1 (i) in serum from SPF C57BL/6 mice subjected to oral tolerance as in Fig. 4k but without S. venezuelensis infection, 14 days after sham surgery (n = 7), I- and C-gLN removal (ΔicLN, n = 5) or D-gLN removal (ΔdLN, n = 6). Data are representative of two independent experiments. ns, not significant (ANOVA).

Extended Data Fig. 9 Extended analysis of gLN swelling and restructuring, and dendritic cell and CD4+ T cell subset frequencies, upon S. venezuelensis infection.

a, Indicated gLN positions in non-infected (N.I.) SPF C57BL/6 mice or mice infected with 700 S. venezuelensis (S.v.) larvae 8 days earlier. b, 3D reconstruction of vasculature (anti-CD31) after solvent clearing (iDISCO+) and light sheet microscopy of indicated gLNs from non-infected SPF C57BL/6 mice or SPF C57BL/6 mice infected with 700 S. venezuelensis larvae 14 days earlier; scale bars, 500 μm. c, Quantification of mature S. venezuelensis worms in gut epithelium 8 days after infection with 700 larvae. n = 8, pooled from two independent experiments. dg, Frequency of CD11c+ cells among CD45+ cells (d), and CD11c+ (e) and CD8α+(f) cells among MHCIIintCD11c+ cells, or GATA3+FOXP3+ cells among TCRβ+CD4+ cells (g) in non-infected SPF C57BL/6 mice or mice infected with 700 S. venezuelensis larvae 8 days earlier; n = 5. Data are representative of three independent experiments. hl, Frequency of CD103+CD11b+ (h), CD103+CD11b (i), or CD103CD11b+ cells (j) among MHCIIintCD11c+ dendritic cells, and of GATA3+FOXP3 (k) or GATA3+FOXP3+ (l) cells among TCRβ+CD4+ cells in denoted gLNs of SPF C57BL/6 mice infected with 700 S. venezuelensis larvae 21–50 days earlier, as indicated (n = 4 per time point) or non-infected (day 21) SPF C57BL/6 mice. *P < 0.05, **P < 0.01, ***P < 0.005; ns, not significant (one-tailed t-test or ANOVA).

Extended Data Fig. 10 Extended analysis of OT-II CD45.1 cell seeding, activation and fate and oral tolerance upon S. venezuelensis infection.

a, Representative flow cytometry plot of FOXP3+ and CD25+ CD45.1 OT-II cells in gLNs 8 days after infection (or not) of mice with S. venezuelensis larvae. be, Frequency of total FOXP3CD25+ cells among CD45.1+ cells (b) and CD45.1+ cells among TCRβ+CD4+ cells (c), total CD45.1+ cells (d) and division index (e) in indicated gLNs 64 h after adoptive transfer of 1 × 106 naive CD45.1+ OT-II cells into CD45.2 SPF C57BL/6 mice (n = 4) infected (or not) with S. venezuelensis for 8 days and gavaged with OVA 48 h and 24 h before analysis. Data are representative of two independent experiments. f, g, Total dendritic cells in BALF (f) and frequency of dendritic cells among CD45+ cells in lung tissue (g) from non-infected SPF C57BL/6 mice or mice infected with S. venezuelensis during antigen feeding (+OVA groups) or no feeding (–OVA groups), 21 days after first immunization with OVA–alum (n = 13 for +OVA groups, n = 10 for –OVA groups). Data are pooled from two independent experiments. *P < 0.05, **P < 0.01, ***P < 0.005; ns, not significant (one-tailed t-test or ANOVA).

Supplementary information

Reporting Summary

Supplementary Tables

This zip folder contains Supplementary Tables 1-16 and the Supplementary Tables Legends.

Supplementary Data

This zip folder contains Source Data for Extended Data Figures 2-10 and Source Data for Figures 1-4.

Video 1

mLN lymphatics (LYVE-1, white) with APCs (CD11c-GFP, red) of 8 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 2

mLN chain lymphatics (LYVE-1, white) with APCs (CD11c-GFP, red) and insulin (green) of 9 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 3

mLN chain to duodenum lymphatics (LYVE-1, white) of 8 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 4

ileal and mLN lymphatics (LYVE-1, white) of 8 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 5

Duodenal and mLN lymphatics (LYVE-1, white) 9 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 6

Duodenal lymphatics (LYVE-1, white) of 9 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 7

Duodenal lymphatics (LYVE-1, white) with APCs (CD11c-GFP, red) of 7 week old SPF mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 8

Celiac (D1) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks old non-infected SPF C57BL/6 mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 9

Celiac (D1) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks SPF C57BL/6 mouse infected with 700 S. venezuelensis (S.v.) larvae 14 days prior to harvest. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 10

Duodenal (D2) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks old non-infected SPF C57BL/6 mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 11

Duodenal (D2) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks SPF C57BL/6 mouse infected with 700 S. venezuelensis (S.v.) larvae 14 days prior to harvest. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 12

Jejunal gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks old non-infected SPF C57BL/6 mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 13

Jejunal gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks SPF C57BL/6 mouse infected with 700 S. venezuelensis (S.v.) larvae 14 days prior to harvest. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 14

Ilial gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks old non-infected SPF C57BL/6 mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 15

Ilial gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks SPF C57BL/6 mouse infected with 700 S. venezuelensis (S.v.) larvae 14 days prior to harvest. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 16

Cecal-colonic (C1) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks old non-infected SPF C57BL/6 mouse. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Video 17

Cecal-colonic (C1) gLN vasculature (CD31, white) and lymphatics (LYVE-1, red) of 9 weeks SPF C57BL/6 mouse infected with 700 S. venezuelensis (S.v.) larvae 14 days prior to harvest. Antibody stained solvent cleared tissue followed by light sheet microscopy and 3D software reconstruction.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Esterházy, D., Canesso, M.C.C., Mesin, L. et al. Compartmentalized gut lymph node drainage dictates adaptive immune responses. Nature 569, 126–130 (2019). https://doi.org/10.1038/s41586-019-1125-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-1125-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

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