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:

Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient

Abstract

Enteroendocrine cells (EECs) control a wide range of physiological processes linked to metabolism1. We show that EEC hormones are differentially expressed between crypts (for example, Glp1) and villi (for example, secretin). As demonstrated by single-cell mRNA sequencing using murine Lgr5+ cell-derived organoids, BMP4 signals alter the hormone expression profiles of individual EECs to resemble those found in the villus. Accordingly, BMP4 induces hormone switching of EECs migrating up the crypt–villus axis in vivo. Our findings imply that EEC lineages in the small intestine exhibit a more flexible hormone repertoire than previously proposed. We also describe a protocol to generate human EECs in organoids and demonstrate a similar regulation of hormone expression by BMP signalling. These findings establish alternative strategies to target EECs with therapeutically relevant hormone production through BMP 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: Enteroendocrine cells switch hormone expression while migrating from crypt to villus.
Fig. 2: Activation of BMP signalling induces a villus-like hormone signature in mouse EECs.
Fig. 3: scRNA-seq reveals BMP-regulated plasticity among different EEC subtypes.
Fig. 4: Manipulation of BMP gradient alters hormone expression in mice.
Fig. 5: The human EEC differentiation protocol implies conserved BMP-controlled hormone expression.

Similar content being viewed by others

References

  1. Furness, J. B., Rivera, L. R., Cho, H.-J., Bravo, D. M. & Callaghan, B. The gut as a sensory organ. Nat. Rev. Gastroenterol. Hepatol. 1010, 729–740 (2013).

    Article  CAS  Google Scholar 

  2. Connor, T. M. O. et al. The role of substance P in inflammatory disease. J. Cell. Physiol. 201, 167–180 (2004).

    Article  CAS  Google Scholar 

  3. Egerod, K. L. et al. A major lineage of enteroendocrine cells coexpress CCK, secretin, GIP, GLP-1, PYY, and neurotensin but not somatostatin. Endocrinology 153, 5782–5795 (2012).

    Article  PubMed  CAS  Google Scholar 

  4. Haber, A. L. et al. A single-cell survey of the small intestinal epithelium. Nature 551, 333–339 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  PubMed  CAS  Google Scholar 

  6. Sato, T. et al. Single Lgr5 stem cells build crypt–villus structures in vitro without a mesenchymal niche. Nature 459, 262–265 (2009).

    Article  PubMed  CAS  Google Scholar 

  7. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–5 (2015).

    Article  PubMed  CAS  Google Scholar 

  8. Basak, O. et al. Induced quiescence of Lgr5+ stem cells in intestinal organoids enables differentiation of hormone-producing enteroendocrine cells. Cell Stem Cell 20, 177–190.e4 (2017).

    Article  PubMed  CAS  Google Scholar 

  9. Roth, K. A. & Gordon, J. I. Spatial differentiation of the intestinal epithelium: analysis of enteroendocrine cells containing immunoreactive serotonin, secretin, and substance P in normal and transgenic mice. Proc. Natl Acad. Sci. USA 87, 6408–6412 (1990).

    Article  PubMed  CAS  Google Scholar 

  10. Grunddal, K. V. et al. Neurotensin is coexpressed, coreleased, and acts together with GLP-1 and PYY in enteroendocrine control of metabolism. Endocrinology 157, 176–194 (2016).

    Article  PubMed  CAS  Google Scholar 

  11. Harris, J. A. et al. Anatomical characterization of Cre driver mice for neural circuit mapping and manipulation. Front. Neural Circuits 8, 78 (2014).

    Article  CAS  Google Scholar 

  12. Aiken, K. D. & Roth, K. A. Temporal differentiation and migration of substance P, serotonin, and secretin immunoreactive enteroendocrine cells in the mouse proximal small intestine. Dev. Dyn. 194, 303–310 (1992).

    Article  PubMed  CAS  Google Scholar 

  13. Haramis, A.-P. G. De novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine. Science 303, 1684–1686 (2004).

    Article  PubMed  CAS  Google Scholar 

  14. Beumer, J. & Clevers, H. Regulation and plasticity of intestinal stem cells during homeostasis and regeneration. Development 143, 3639–3649 (2016).

    Article  PubMed  CAS  Google Scholar 

  15. Camacho, S. et al. Anti-obesity and anti-hyperglycemic effects of cinnamaldehyde via altered ghrelin secretion and functional impact on food intake and gastric emptying. Sci. Rep. 5, 7919 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Habib, A. M. et al. Overlap of endocrine hormone expression in the mouse intestine revealed by transcriptional profiling and flow cytometry. Endocrinology 153, 3054–3065 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Svendsen, B. et al. GLP1- and GIP-producing cells rarely overlap and differ by bombesin receptor-2 expression and responsiveness. J. Endocrinol. 228, 39–48 (2016).

    Article  PubMed  CAS  Google Scholar 

  18. Muraro, M. J. et al. A Single-cell transcriptome atlas of the human pancreas. Cell Syst. 3, 385–394 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Hashimshony, T. et al. CEL-Seq2: sensitive highly-multiplexed single-cell RNA-Seq. Genome Biol. 17, 77 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Grün, D. et al. De novo prediction of stem cell identity using single-cell transcriptome data. Cell Stem Cell 19, 266–277 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Nozawa, K. et al. TRPA1 regulates gastrointestinal motility through serotonin release from enterochromaffin cells. Proc. Natl Acad. Sci. USA 106, 13 (2009).

    Article  CAS  Google Scholar 

  22. Zhang, X.-X. et al. Neuroendocrine hormone amylin in diabetes. World J. Diabetes 7, 189–197 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Gross, S. et al. The novel enterochromaffin marker Lmx1a regulates serotonin biosynthesis in enteroendocrine cell lineages downstream of Nkx2.2. Development 143, 2616–2628 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Zhang, J., McKenna, L. B., Bogue, C. W. & Kaestner, K. H. The diabetes gene Hhex maintains δ-cell differentiation and islet function. Genes Dev. 28, 829–834 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Kaaij, L. T. et al. DNA methylation dynamics during intestinal stem cell differentiation reveals enhancers driving gene expression in the villus. Genome Biol. 14, R50 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Hollnagel, A., Oehlmann, V., Heymer, J., Rüther, U. & Nordheim, A. Id genes are direct targets of bone morphogenetic protein induction in embryonic stem cells. J. Biol. Chem. 274, 19838–19845 (1999).

    Article  PubMed  CAS  Google Scholar 

  27. Jenny, M. et al. Neurogenin3 is differentially required for endocrine cell fate specification in the intestinal and gastric epithelium. EMBO J. 21, 6338–6347 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Whissell, G. et al. The transcription factor GATA6 enables self-renewal of colon adenoma stem cells by repressing BMP gene expression. Nat. Cell Biol. 16, 695–707 (2014).

    Article  PubMed  CAS  Google Scholar 

  29. Qi, Z. et al. BMP restricts stemness of intestinal Lgr5+ stem cells by directly suppressing their signature genes. Nat. Commun. 8, 13824 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Sato, T. et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology 141, 1762–1772 (2011).

    Article  PubMed  CAS  Google Scholar 

  31. Clevers, H. The intestinal crypt, a prototype stem cell compartment. Cell 154, 274–284 (2013).

    Article  PubMed  CAS  Google Scholar 

  32. Fothergill, L. J., Callaghan, B., Hunne, B., Bravo, D. M. & Furness, J. B. Costorage of enteroendocrine hormones evaluated at the cell and subcellular levels in male mice. Endocrinology 158, 2113–2123 (2017).

    Article  PubMed  Google Scholar 

  33. Aiken, K. D., Kisslinger, J. A. & Roth, K. A. Immunohistochemical studies indicate multiple enteroendocrine cell differentiation pathways in the mouse proximal small intestine. Dev. Dyn. 201, 63–70 (1994).

    Article  PubMed  CAS  Google Scholar 

  34. Bellono, N. W. et al. Enterochromaffin cells are gut chemosensors that couple to sensory neural pathways. Cell 170, 185–198.e16 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Bohorquez, D. V. et al. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J. Clin. Investig. 125, 782–786 (2015).

    Article  PubMed  Google Scholar 

  36. Beumer, J. & Clevers, H. Induction of different human enteroendocrine cells in intestinal organoids. Protoc. Exch. https://doi.org/10.1038/protex.2018.077 (2018).

  37. Farin, H. F. et al. Visualization of a short-range Wnt gradient in the intestinal stem-cell niche. Nature 530, 340–343 (2016).

    Article  PubMed  CAS  Google Scholar 

  38. Muñoz, J. et al. The Lgr5 intestinal stem cell signature: robust expression of proposed quiescent ‘+4’ cell markers. EMBO J. 31, 3079–3091 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Grün, D., Kester, L. & van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. van der Elst, R. van der Linden and Y. Bar-Ephraim for their help with FACS experiments. B.A. is supported by NWO/VENI 863.15.015.

Author information

Authors and Affiliations

Authors

Contributions

J.B. and H.C. conceived and designed the project. J.B. designed and performed all experiments. B.A. performed analysis of RNA-seq data. F.R. and F.G. generated the Gcg and Gip reporter mice. H.Z. generated the Tac1 reporter mouse, and T.N.N. assisted in providing the tissue. J.H.V.E supervised and performed the mouse experiments, with the help of M.V.d.B. Y.P. assisted in histology preparation. J.B., B.A. and H.C. wrote the manuscript with input from all other authors.

Corresponding author

Correspondence to Hans Clevers.

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.

Integrated supplementary information

Supplementary Figure 1 Crypt-to-villus heterogeneity and lineage relationship of enteroendocrine cells.

a, RNA was isolated from crypts and villi of the proximal and distal SI. qPCR analysis was performed on selected hormones. Tac1 and Gcg transcripts are enriched in the crypts of mice, while Pyy and Sct are increased in the villus. Alpi, a marker of enterocytes, confirmed enrichment of villus fractions. Sample size represents n = 2 mice, and the mean values are shown. b, Intestine of Tac1iresCre/RosaAi14 mice reveals that Tac1+ expressing cells give rise to Serotonin (Ser) expressing cells and are thus part of the Enterochromaffin lineage. Gip, Sst and Cck expressing EECs are rarely derived from Tac1, while the vast majority of Serotonin producing ECs are derived from Tac1. Quantification in Fig. 1g (n = 4 mice). Scale bar is 50 µm.

Supplementary Figure 2 Signaling through the BMP4-BMPR1a/BMPR2 axis induces hormone switching in EECs without causing apoptosis.

a, Organoids were differentiated into enteroendocrine cells (EECs) for 4 days by the inhibition of Wnt, Notch and MEK signaling. On the background of the EEC differentiation cocktail, we inhibited Hedgehog (Vismodegib), activated BMP (Noggin withdrawal), inhibited Wnt (Rspo withdrawal), inhibited TGFbeta (A83) or activated TGFbeta (TGFβ1) signaling. qPCR analysis of selected hormones shows that activation of BMP signaling selectively induces Sct but represses Gcg expression, while having a neutral effect on Chga. Results are shown relative to control organoids in ENR medium. Sample size represents n = 2 biologically independent experiments, and the mean values are shown. b, Secreted Glp1 and Sct peptides after stimulation with forskolin were measured by ELISA in EEC differentiation medium with or without BMP4. Secreted Glp1 was diminished in the background of BMP activation, while Sct increased. Experiment was performed in in proximal and distal organoids. Sample size represents n = 2 biologically independent experiments, and the mean values are shown. c, Addition of BMPR1a inhibitor LDN193189 to a 4-day EEC differentiation protocol containing BMP4 reverses the increase in Sct production, as shown by immunostaining. Image is presented as a maximum projection. Experiment was performed three times independently with similar results. Scale bar is 50 µm. d, qPCR analysis of selected hormones of the same experiment as in c. BMPR1a inhibition reverses all changes induced by BMP4 addition to the standard EEC differentiation cocktail (EEC BMPlow). Sample size represents n = 1 biologically independent experiment, and the average value of a technical duplicate is shown. e, Organoids derived from GcgVenus mice were imaged for 3 days in the presence and absence of BMP4. Individual L-cells shut down Gcg transcription when stimulated with BMP4, while control cells maintain Gcg positivity throughout the imaging timewindow. Imaging started 6 hours after addition of BMP4. f, Quantification of e. The number of Venus+ cells imaged and measured is depicted. The mean intensity of all cells at each timepoint is shown, and error bars represent SD. SD was derived from n = 16 cells in -BMP4 treatment and n = 28 cells in +BMP4 treatment. g, Organoids from GcgVenus and Tac1iresCre/RosaAi14 mice were differentiated to EECs in the absence or presence of BMP, and co-stained for cleaved caspase-3 to assess apoptosis. Experiment has been repeated two times with similar results. Scale bar is 50 µm.

Supplementary Figure 3 Single cell transcriptome analyses of EEC subtypes after BMP activation.

a, FACS plots display the gating strategy used to sort live, single cell (DAPI-negative, right plots) subtypes of EECs from reporter organoids (left plots) in different BMP activation contexts. b, Venus intensity levels of Goblet cells (Muc2+), Enterocytes (Alpi+) and Gcg-high cells. Mean intensity is shown for each group, together with the number of cells in each group (n = 82 Gcg-high cells, n = 13 Goblet cells and n = 23 enterocytes). c, Cells with a Venus intensity of more than 2.5 * 104, predictive for Gcg expression (Fig S4a,b), were selected from the dataset. Transcripts for Gcg, Sct and Pyy are plotted (upper plots). High Venus positivity is predictive for Gcg expression in BMPlow conditions, but not after BMP treatment. Cells with at least 100 Sst (middle plots) or Gip (lower plots) were selected from the dataset. Gip+ and Sst+ cells upregulate Pyy expression upon BMP treatment, while only cells producing Gip can co-express Sct. d, Expression levels of selected markers in the t-SNE space. Different transcription factors, receptors and secreted peptides were uniquely expressed by subtypes of EECs. BMP target genes associate with BMP4 treated samples.

Supplementary Figure 4 BMP signalling induces switches in peptide profile of individual EECs and blocks initial EEC specification.

a, Tac1iresCre/RosaAi14 and GcgVenus – reporter organoids were treated with a MEK inhibitor, while receiving Noggin (-BMP) or BMP4 (+BMP). The numbers of each marker positive cells are presented (in 10 organoids), and the co-localization of these hormones with tdTomato or Venus was quantified. Sample size represents n = 2 biologically independent experiments, and the mean values are shown. BMP activation dampens the generation of new EECs and induces switches in the peptide profile of individual EECs. b, Individual EECs in Tac1iresCre/RosaAi14 and GcgVenus – reporter organoids were followed using live cell imaging in the absence and presence of BMP signals. After 60 h, organoids were fixed inside the BME and stained for different hormones, as indicated. The experiment was performed two times independently with similar results. c, Mouse intestinal organoids were EEC differentiated for 4 days in BMP low or high conditions. qPCR analysis reveals Neurog3 is downregulated after BMP treatment (data is shown relative to BMP untreated sample). Sample size represents n = 2 biologically independent experiments, and the mean expression values are shown. d, Mouse intestinal organoids were EEC differentiated for 4 days in BMP low conditions, or for 3 days in BMP low and 24 h in BMP high conditions. qPCR analysis of selected hormones (data is shown relative to day 3 EEC differentiation). Initial EEC generation in BMP low conditions and subsequent induction of BMP signalling for 24 h is most efficient in generating Villus-enriched hormones. Sample size represents n = 2 biologically independent experiments, and the mean expression values are shown.

Supplementary Figure 5 A transgenic mouse model with a disrupted BMP gradient displays altered hormone expression.

a, Immunostaining of intestines from VillinNoggin mice. Tac1+ cells were scattered along the whole crypt-villus axis, while Sct+ cell numbers greatly reduced. Experiment has been repeated twice with a sample size of 2 mice per genotype. b, Quantification of a. Sample size represents n = 2 mice per genotype. Mean values are depicted. c, VillinNoggin mice show an increased number of Glp1+ L-cells in the villus. d, Quantification of c. Sample size represents n = 2 mice per genotype. Mean values are depicted. Scale bar is 50 µm.

Supplementary Information

Supplementary Information

Supplementary Figures 1–5 and legends for Supplementary Tables 1 and 2.

Reporting Summary

Supplementary Table 1

qPCR primers

Supplementary Table 2

Statistics source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Beumer, J., Artegiani, B., Post, Y. et al. Enteroendocrine cells switch hormone expression along the crypt-to-villus BMP signalling gradient. Nat Cell Biol 20, 909–916 (2018). https://doi.org/10.1038/s41556-018-0143-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0143-y

This article is cited by

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