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.

  • Resource
  • Published:

Tracing the temporal-spatial transcriptome landscapes of the human fetal digestive tract using single-cell RNA-sequencing

A Publisher Correction to this article was published on 23 July 2018

This article has been updated

Abstract

The development of the digestive tract is critical for proper food digestion and nutrient absorption. Here, we analyse the main organs of the digestive tract, including the oesophagus, stomach, small intestine and large intestine, from human embryos between 6 and 25 weeks of gestation as well as the large intestine from adults using single-cell RNA-seq analyses. In total, 5,227 individual cells are analysed and 40 cell types clearly identified. Their crucial biological features, including developmental processes, signalling pathways, cell cycle, nutrient digestion and absorption metabolism, and transcription factor networks, are systematically revealed. Moreover, the differentiation and maturation processes of the large intestine are thoroughly investigated by comparing the corresponding transcriptome profiles between embryonic and adult stages. Our work offers a rich resource for investigating the gene regulation networks of the human fetal digestive tract and adult large intestine at single-cell resolution.

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: Global analysis of single-cell expression profiles of human fetal digestive system.
Fig. 2: Cell type identification and development tracking of human fetal oesophagus and stomach.
Fig. 3: Cell type identification and development tracking of human fetal small intestine.
Fig. 4: Cell type identification of fetal and adult large intestine and comparison of the cell type features between fetal and adult stages.
Fig. 5: Transcriptome dynamics and selected signalling pathway in the development of human fetal intestinal tract.
Fig. 6: Identification of organ-specific genes.
Fig. 7: Inferred master regulators and signal pathway regulation of the small intestine.
Fig. 8: Gene expression analysis of immune and endothelial cell and summary of small intestine development.

Similar content being viewed by others

Change history

  • 23 July 2018

    In the PDF version of this Resource originally published, on the author list the superscript number 9 to indicate Rui Wang was an equally contributing author was missing owing to a technical error. This has now been amended. In addition, the authors wish to replace Supplementary Table 2, as in the original version Group 1 immune cells were mis-classified into Group 46 immune cells, resulting in three groups of immune cells where there should have been four. Supplementary Table 2 has now been replaced.

References

  1. Zorn, A. M. & Wells, J. M. Vertebrate endoderm development and organ formation. Annu. Rev. Cell Dev. Biol. 25, 221–251 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Kraus, M. R. & Grapin-Botton, A. Patterning and shaping the endoderm in vivo and in culture. Curr. Opin. Genet. Dev. 22, 347–353 (2012).

    Article  PubMed  CAS  Google Scholar 

  3. Sherwood, R. I., Chen, T. Y. & Melton, D. A. Transcriptional dynamics of endodermal organ formation. Dev. Dyn. 238, 29–42 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Schedl, H. P. & Clifton, J. A. Solute and water absorption by the human small intestine. Nature 199, 1264–1267 (1963).

    Article  PubMed  CAS  Google Scholar 

  5. Schulz, M. D. et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature 514, 508–512 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Ugolev, A. Influence of the surface of the small intestine on enzymatic hydrolysis of starch by enzymes. Nature 188, 588–589 (1960).

    Article  PubMed  CAS  Google Scholar 

  7. Maynard, C. L., Elson, C. O., Hatton, R. D. & Weaver, C. T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 489, 231–241 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Jervis, E. L. & Levin, R. J. Anatomic adaptation of the alimentary tract of the rat to the hyperphagia of chronic alloxan-diabetes. Nature 210, 391–393 (1966).

    Article  PubMed  CAS  Google Scholar 

  10. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Park, Y. H. & Kim, N. Review of atrophic gastritis and intestinal metaplasia as a premalignant lesion of gastric cancer. J. Cancer Prev. 20, 25–40 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-Seq: single-cell RNA-seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).

    Article  PubMed  CAS  Google Scholar 

  13. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  PubMed  CAS  Google Scholar 

  14. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Tang, F. et al. RNA-seq analysis to capture the transcriptome landscape of a single cell. Nat. Protoc. 5, 516–535 (2010).

    Article  PubMed  CAS  Google Scholar 

  16. Fan, X. et al. Single-cell RNA-seq transcriptome analysis of linear and circular RNAs in mouse preimplantation embryos. Genome Biol. 16, 148 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Islam, S. et al. Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Res 21, 1160–1167 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Svensson, V. et al. Power analysis of single-cell RNA-sequencing experiments. Nat. Methods 14, 381–387 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Ziegenhain, C. et al. Comparative analysis of single-cell RNA sequencing methods. Mol. Cell 65, 631–643 (2017).

    Article  PubMed  CAS  Google Scholar 

  22. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Grun, 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 

  24. Grun, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Article  PubMed  CAS  Google Scholar 

  25. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Qiu, X. et al. Single-cell mRNA quantification and differential analysis with Census. Nat. Methods 14, 309–315 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Joost, S. et al. Single-cell transcriptomics reveals that differentiation and spatial signatures shape epidermal and hair follicle heterogeneity. Cell Syst. 3, 221–237 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Schumacher, M. A. et al. The use of murine-derived fundic organoids in studies of gastric physiology. J. Physiol. 593, 1809–1827 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. 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 

  30. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012–1026 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Mohammed, H. et al. Single-cell landscape of transcriptional heterogeneity and cell fate decisions during mouse early gastrulation. Cell Rep. 20, 1215–1228 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Noah, T. K., Donahue, B. & Shroyer, N. F. Intestinal development and differentiation. Exp. Cell Res. 317, 2702–2710 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Kearns, N. A. et al. Generation of organized anterior foregut epithelia from pluripotent stem cells using small molecules. Stem Cell Res. 11, 1003–1012 (2013).

    Article  PubMed  CAS  Google Scholar 

  34. Peters, H. et al. Isolation of the Pax9 cDNA from adult human esophagus. Mamm. Genome 8, 62–64 (1997).

    Article  PubMed  CAS  Google Scholar 

  35. Quante, M., Marrache, F., Goldenring, J. R. & Wang, T. C. TFF2 mRNA transcript expression marks a gland progenitor cell of the gastric oxyntic mucosa. Gastroenterology 139, 2018–2027 (2010).

    Article  PubMed  CAS  Google Scholar 

  36. Noguchi, T. K. et al. Generation of stomach tissue from mouse embryonic stem cells. Nat. Cell Biol. 17, 984–993 (2015).

    Article  PubMed  CAS  Google Scholar 

  37. Goodell, M. A., Nguyen, H. & Shroyer, N. Somatic stem cell heterogeneity: diversity in the blood, skin and intestinal stem cell compartments. Nat. Rev. Mol. Cell Biol. 16, 299–309 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Barker, N. Adult intestinal stem cells: critical drivers of epithelial homeostasis and regeneration. Nat. Rev. Mol. Cell Biol. 15, 19–33 (2014).

    Article  PubMed  CAS  Google Scholar 

  39. Rishniw, M. et al. Molecular aspects of esophageal development. Ann. NY Acad. Sci. 1232, 309–315 (2011).

    Article  PubMed  CAS  Google Scholar 

  40. Yu, W. Y., Slack, J. M. & Tosh, D. Conversion of columnar to stratified squamous epithelium in the developing mouse oesophagus. Dev. Biol. 284, 157–170 (2005).

    Article  PubMed  CAS  Google Scholar 

  41. Gao, Y., Chen, Y., Xu, D., Wang, J. & Yu, G. Differential expression of ANXA1 in benign human gastrointestinal tissues and cancers. BMC Cancer 14, 520 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Onozawa, H. et al. Annexin A1 is involved in resistance to 5-FU in colon cancer cells. Oncol. Rep. 37, 235–240 (2017).

    Article  PubMed  Google Scholar 

  43. Kim, T. H. & Shivdasani, R. A. Stomach development, stem cells and disease. Development 143, 554–565 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. McCracken, K. W. et al. Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature 516, 400–404 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Bartfeld, S. et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology 148, 126–136 (2015).

    Article  PubMed  Google Scholar 

  46. Wang, D. et al. Identification of multipotent mammary stem cells by protein C receptor expression. Nature 517, 81–84 (2015).

    Article  PubMed  CAS  Google Scholar 

  47. de Lau, W. et al. Peyer’s patch M cells derived from Lgr5+ stem cells require SpiB and are induced by RankL in cultured ‘miniguts’. Mol. Cell Biol. 32, 3639–3647 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Almohazey, D. The ErbB3 receptor tyrosine kinase negatively regulates Paneth cells by PI3K-dependent suppression of Atoh1. Cell Death Differ. 24, 855–865 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Beck, F. Homeobox genes in gut development. Gut 51, 450–454 (2002).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Merchant, J. L. Hedgehog signalling in gut development, physiology and cancer. J. Physiol. 590, 421–432 (2012).

    Article  PubMed  CAS  Google Scholar 

  51. Kim, B. M. & Choi, M. Y. New insights into the role of Hedgehog signaling in gastrointestinal development and cancer. Gastroenterology 137, 422–424 (2009).

    Article  PubMed  Google Scholar 

  52. Litingtung, Y., Lei, L., Westphal, H. & Chiang, C. Sonic hedgehog is essential to foregut development. Nat. Genet. 20, 58–61 (1998).

    Article  PubMed  CAS  Google Scholar 

  53. Motoyama, J. et al. Essential function of Gli2 and Gli3 in the formation of lung, trachea and oesophagus. Nat. Genet. 20, 54–57 (1998).

    Article  PubMed  CAS  Google Scholar 

  54. van den Brink, G. R. et al. Indian Hedgehog is an antagonist of Wnt signaling in colonic epithelial cell differentiation. Nat. Genet. 36, 277–282 (2004).

    Article  PubMed  CAS  Google Scholar 

  55. van den Brink, G. R. Hedgehog signaling in development and homeostasis of the gastrointestinal tract. Physiol. Rev. 87, 1343–1375 (2007).

    Article  PubMed  CAS  Google Scholar 

  56. Katoh, Y. & Katoh, M. Hedgehog signaling, epithelial-to-mesenchymal transition and miRNA (review). Int J. Mol. Med 22, 271–275 (2008).

    PubMed  CAS  Google Scholar 

  57. Li, L. et al. Single-cell RNA-seq analysis maps development of human germline cells and gonadal niche interactions. Cell Stem Cell 20, 891–892 (2017).

    Article  PubMed  CAS  Google Scholar 

  58. Siegel, P. M., Shu, W. & Massague, J. Mad upregulation and Id2 repression accompany transforming growth factor (TGF)-beta-mediated epithelial cell growth suppression. J. Biol. Chem. 278, 35444–35450 (2003).

    Article  PubMed  CAS  Google Scholar 

  59. Wang, R. N. et al. Bone morphogenetic protein (BMP) signaling in development and human diseases. Genes Dis. 1, 87–105 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Qi, Z. 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 

  61. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-seq. Bioinformatics 25, 1105–1111 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  PubMed  CAS  Google Scholar 

  63. Chung, N. C. & Storey, J. D. Statistical significance of variables driving systematic variation in high-dimensional data. Bioinformatics 31, 545–554 (2015).

    Article  PubMed  CAS  Google Scholar 

  64. Bodenhofer, U., Kothmeier, A. & Hochreiter, S. APCluster: an R package for affinity propagation clustering. Bioinformatics 27, 2463–2464 (2011).

    Article  PubMed  CAS  Google Scholar 

  65. Frey, B. J. & Dueck, D. Clustering by passing messages between data points. Science 315, 972–976 (2007).

    Article  PubMed  CAS  Google Scholar 

  66. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Scialdone, A. et al. Computational assignment of cell-cycle stage from single-cell transcriptome data. Methods 85, 54–61 (2015).

    Article  PubMed  CAS  Google Scholar 

  68. Lachmann, A., Giorgi, F. M., Lopez, G. & Califano, A. ARACNe-AP: gene network reverse engineering through adaptive partitioning inference of mutual information. Bioinformatics 32, 2233–2235 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Zhang, H. M. et al. AnimalTFDB 2.0: a resource for expression, prediction and functional study of animal transcription factors. Nucleic Acids Res. 43, 76–81 (2015).

    Article  CAS  Google Scholar 

  70. Zhang, H. M. et al. AnimalTFDB: a comprehensive animal transcription factor database. Nucleic Acids Res. 40, 144–149 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (31625018, 31230047, 31601177, 81521002), the Ministry of Science and Technology of China (2017YFA0102702), a General Financial Grant from the China Postdoctoral Science Foundation (2017M610703) and Shanghai Science and Technology Development Funds (16YF140940).

Author information

Authors and Affiliations

Authors

Contributions

F.T. and J.Q. conceived and supervised the project. S.G., L.Y., and J.L. performed the experiments, including single cell collection, library construction, immunofluorescence staining and so on, with the help of Ju.Y., Xi.Z., X.W., Y.W., X.Y.W, X.F., Ji.Y., Xu.Z., Y.G., S.H.G., W.W., C.W., Y.M., W.F. and L.W. Under the supervision of H.G. and with the help of X.J., R.W. performed bioinformatics analysis. S.G., F.T., R.W., J.L. and L.Y. wrote the manuscript with help from all of the authors.

Corresponding authors

Correspondence to Hao Ge, Jie Qiao or Fuchou Tang.

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 Non-epithelial Group Removal.

(a) Dot plot showing expression level of well-defined cell type marker genes in each group. (b) t-SNE showing the organ source of each group. In a and b, n = 4,089 cells. (c) Boxplot showing the expression levels of erythrocyte-specific markers between group 31 (n = 103 cells) and the control group (n = 54 cells). The black central line is the median, the boxes indicate the upper and lower quartiles, the whiskers indicate the 1.5 interquartile range. (d) t-SNE showing the expression patterns of muscle-specific marker gene MYLPF. n = 4,089 cells. (e) The correlation coefficient of four tissues at each stage. The result showed that the early stage organs were more similar to each other than the late stage. The correlation values are derived from Pearson pairwise correlation. 6W: n = 607 cells, 7W: n = 774 cells, 8W: n = 319 cells, 9W: n = 328 cells, 11W: n = 232 cells, 14W: n = 224 cells, 16W: n = 195 cells, 19W: n = 257 cells, 21W: n = 367 cells, 24W: n = 624 cells, 25W: n = 162 cells. The black central line is the median, the boxes indicate the upper and lower quartiles, the whiskers indicate the 1.5 interquartile range. (f) The analysis of cell cycle for the cells inside the circle (Groups 33-44, 48-51) and the cells outside of the circle (Groups 1-33 and 48) base on expression of sets of G1/S and G2/M genes derived from the scRNA-seq. The result showed that the proliferation index for the cells inside the circle was higher than that of the cells outside the circle according to the percentage of S + G2/M.

Supplementary Figure 2 Pseudotime construction in each organ and global expression patterns of selected cell-type-specific markers.

(a) Pseudotime construction of the four gastrointestinal tract organs. The cells are colored according to their actual developmental stages. (b-e) Smoothed spline showing the expression patterns of stem cell-, cell cycle- and organ-specific genes along the pseudotime for each organ. Colored lines and shaded regions represent the expression tendency of each gene and s.e.m., respectively. For example, the epithelial markers EPCAM and CDH1 showed relatively stable expression throughout the development of the human fetal esophagus (b). In the stomach from 6W to 25W, the expression levels of both the mucous neck cell marker TFF2, and the recently identified novel multipotent mammary stem cell marker PROCR, were gradually increased. (f) The Western blotting results show that the protein level of MUC2 was gradually increased during the development of S-Intes and L-Intes, while the protein level of OLFM4 was initially increased and subsequently decreased during the development of the S-Intes. The S-Intes and L-Intes of 7W, 14W and 24W embryos were used for Western blotting experiments. The western blotting experiments were independently repeated four times with similar results. The unprocessed gel data are provided in Supplementary Fig. 9. (g) After normalization, the pseudotimes for the four organs at the same time scale; the smoothed-line shows the cell number density along the normalized pseudotime in each organ. A majority of the cells in the esophagus and stomach reached relative mature state at the mid-developmental stage. However, the small and large intestines gradually matured throughout the entire developmental period from 6W to 25W. The color represents the organ source. (h) Line plot showing the actual stage information of cells along the normalized pseudotimes. (i) Analysis pipeline of the cell-type definition for each organ. To further resolve the cellular heterogeneity among these four organs and better identify the cell types, we combined StemID and K-nearest neighbor (KNN) in the subsequent analysis in each organ. For a-e and g-h, Esophagus: n = 773 cells, Stomach: n = 854 cells, S-Intes: n = 868 cells, L-Intes: n = 849 cells.

Supplementary Figure 3 Cell-Type identifications of the fetal esophagus and stomach.

(a) Boxplot shows the expression levels of cluster-specific genes and known esophageal cell-type markers. Different colors represent different cell types. Ciliated epithelial markers: NME5 and DNAI1. Basal cell markers: KRT6A and KRT6B. Secretory cell markers: MUC1 and MUC16. Cluster 1: n = 26 cells, Cluster 2: n = 18 cells, Cluster 3: n = 415 cells, Cluster 4: n = 314 cells. (b) Averaged expression levels of ANXA1 among the esophagus (n = 773 cells), stomach (n = 854 cells), S-Intes (n = 868 cells) and L-Intes (n = 849 cells). error bars, s.e.m.; *p<2.2×10-16;. (c) Immunostaining of ANXA1 of the esophagus at three different developmental stages (7W, 14W and 23W). White triangle indicates ANXA1+ cells. Dashed lines indicate the structure of the squamous epithelial layer. (d) Bar plot showing the expression levels of ANXA1 and EPCAM in all esophagus cells at single-cell resolution. The cells are ordered according to their AXNA1 and SOX2 expression levels. Early: n = 500 cells, Mid-: n = 138 cells, Late: n = 135 cells. (e) Immunostaining of ANXA1 in the 23W S-Intes. White triangle indicates ANXA1+ cells. Dashed lines indicate the villus. (f) The epithelial cells in the esophagus (n = 773) showed high ANXA1 expression, whereas in the S-Intes (n = 868) showed only marginal levels of ANXA1 expression (NS, not significant; *P<0.01). The precise p value from left to right — < 2.2×10-16, < 2.2×10-16, < 2.2×10-16, 0.2107, 0.0309. (g) The ANXA1 expressions between S-Intes endothelial cells (n = 18) from Groups 23-25 identified in the global t-SNE analysis and S-Intes epithelial cells (EPCAMHigh, n = 18) were compared. The endothelial cells in the S-Intes showed high ANXA1 expression, whereas the epithelial cells in S-Intes showed essentially no ANXA1 expression (***P<0.01). The precise p value from left to right — 2.611×10-7, 0.0009123, 7.832×10-6, 2.008×10-5. In a, f and g, the black central line is the median, the boxes indicate the upper and lower quartiles, the whiskers indicate the 1.5 interquartile range. In b, f and g, two-sided Mann-Whitney U test were used. (h) Immunostaining of PECAM1 and ANXA1 in the 24W S-Intes. In c, e and h, Scale bars, 25μm. The experiment was independently repeated twice with similar results.

Supplementary Figure 4 Immunostaining validation of the fetal S-Intes cell types.

Immunostaining of well-known cell type specific markers in 7W and 24W S-Intes. Enterocyte markers (CDH1, VIL1), Goblet cell markers (MUC2, TFF3), Endocrine cell markers (CHGA, PYY), Tuft maker and stem cell marker (DCLK1), Paneth cell marker (LYZ), Stem/progenitor cell markers (OLFM4, SOX9). Interestingly, the VIL1 and CDH1, two well-known enterocyte markers, showed distinct expression patterns in the 7W fetal S-Intes. VIL1 was expressed in columnar epithelial cells, whereas CDH1 was expressed in a minor proportion of the cells located in the putative mesenchymal layer. Moreover, MUC2 and TFF3 were primarily expressed in the villus whereas the SOX9 and OLFM4 were specifically expressed in the crypts. Scale bars, 25μm. All experiments were independently repeated twice with similar results.

Supplementary Figure 5 Immunostaining validation and proliferation index of the L-Intes cell types.

(a) Immunostaining of well-known cell-type markers in the 7W and 24W fetal L-Intes and adult L-Intes. Enterocyte markers (CDH1 and VIL1), goblet cell markers (MUC2 and TFF3), endocrine cell markers (CHGA, PYY), stem/progenitor cell markers (OLFM4 and SOX9), tuft marker and stem cell marker (DCLK1), and Paneth cell marker (LYZ). Scale bars, 25μm. Unexpectedly, the fetal L-Intes also expressed LYZ, which was previously reported to exist only in the Paneth cells of S-Intes. All experiments were independently repeated a minimum of twice with similar results. (b) Immunostaining of TFF3 and CHGA in the 24W L-Intes, 24W S-Intes and adult L-Intes. White triangle indicates TFF3+ CHGA+ cells. Scale bars, 25μm. All experiments were independently repeated twice with similar results. (c) Heatmap showing the expression levels of cell cycle-related genes in each L-Intes clusters. The cell cycle index is shown at the bottom of the heatmap. n = 849 cells.

Supplementary Figure 6 Cell-Type identification of adult large intestine and comparison between fetal and adult stages.

(a) Heatmap showing the averaged expression level of well-known cell-type markers and selected signaling pathway-related genes in the twelve clusters of adult L-Intes. The colors from blue to red represent the expression level from low to high. n = 1,303 cells. (b) A summary of the features of all adult large intestine cell types, including cell number, cell cycle index and cell type markers. Newly identified markers are shown in red, and the well-known markers are shown in black. (c) PCA of all stages of fetal L-Intes cells and all adult L-Intes cells (n = 2,218 cells).

Supplementary Figure 7 Identification of developmental stage-specific genes.

(a-c) Clustering of gene expression tendencies along the developmental stages of the esophagus, stomach and large intestine. Solid black and colored lines represented the expression tendencies of all genes and each gene, respectively. Esophagus: 7W (n = 168 cells), 14W (n = 65 cells), 19W (n = 57 cells); Stomach: 7W (n = 233 cells), 14W (n = 37 cells), 24W (n = 98 cells); L-Intes: 7W (n = 118 cells), 14W (n = 57 cells), 19W (n = 53 cells), 24W (n = 188 cells). (d) Schematic diagram of the interactions of epithelial and mesenchymal cells. (e) Bar plot showing S-Intes cell expression profiles of Hedgehog and other signaling pathway related genes at single-cell levels. Cells were ordered by the VIM expression level. The results showed the Hedgehog signaling-related genes (PTCH1, SMO and GLI2/3) tend to be expressed by the mesenchymal cells (VIMHigh cells) but not the epithelial cells, and the SFRP1 is indeed expressed by the mesenchymal cells that highly expressed Hedgehog signaling-related genes. Moreover, TGF-β signaling pathway-related genes ZEB1 and ZEB2 are highly expressed by the mesenchymal cells, which barely express CDH1. In addition, we observed that FGFR3 tend to be expressed by the epithelial cells, whereas FGFR1 tend to be expressed by the mesenchymal cells. n=868 cells. (f) Boxplot showing the expression levels of selected genes in S-Intes epithelial cells (EPCAMHigh cells) and S-Intes mesenchymal cells (VIMHigh cells). The top 50 cells highly expressing EPCAM and the top 50 cells highly expressing VIM were used for analysis. The black central line is the median, the boxes indicate the upper and lower quartiles, the whiskers indicate the 1.5 interquartile range.

Supplementary Figure 8 Transcription factor network analysis during fetal esophagus, stomach and L-Intes development.

(a, d and g) Top candidate master transcription regulators during esophagus, stomach and L-Intes development from early stage to mid-stage. Esophagus (early stage: n = 500 cells, mid- stage: n = 138 cells, late stage: n = 135 cells). Stomach (early stage: n = 586 cells, mid- stage: n = 75 cells, late stage: n = 193 cells). L-Intes (early stage: n = 481 cells, mid- stage: n = 95 cells, late stage: n = 273 cells). The violin plots show the expression levels of each master transcription regulator. The density of violin plots were scale to maximum of 1 by setting ‘scale = area’ and all violins have the same maximum width. (b, e and h) Top candidate master transcription regulators during esophagus, stomach and L-Intes development from mid-stage to late stage. The violin plots show the expression levels of each master transcription regulator. The density of violin plots were scale to maximum of 1 by setting ‘scale = area’ and all violins have the same maximum width. (c, f and i) Transcription factor correlation network during fetal esophagus, stomach and L-Intes development. Nodes (TFs) with more than three edges are shown, with each edge representing a high correlation (>0.3) between the related TFs. The correlation values are derived from Pearson pairwise correlation. Yellow, orange and red circles represent the highest average expression levels of genes in early, mid- and late stages, respectively. Esophagus: n = = 773 cells, Stomach: n = 854 cells, L-Intes: n = 849 cells.

Supplementary Figure 9

Unprocessed blots for related figures in Supplementary fig. 2f

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, S., Yan, L., Wang, R. et al. Tracing the temporal-spatial transcriptome landscapes of the human fetal digestive tract using single-cell RNA-sequencing. Nat Cell Biol 20, 721–734 (2018). https://doi.org/10.1038/s41556-018-0105-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0105-4

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