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.

Heterotypic cell–cell communication regulates glandular stem cell multipotency

Abstract

Glandular epithelia, including the mammary and prostate glands, are composed of basal cells (BCs) and luminal cells (LCs)1,2. Many glandular epithelia develop from multipotent basal stem cells (BSCs) that are replaced in adult life by distinct pools of unipotent stem cells1,3,4,5,6,7,8. However, adult unipotent BSCs can reactivate multipotency under regenerative conditions and upon oncogene expression3,9,10,11,12,13. This suggests that an active mechanism restricts BSC multipotency under normal physiological conditions, although the nature of this mechanism is unknown. Here we show that the ablation of LCs reactivates the multipotency of BSCs from multiple epithelia both in vivo in mice and in vitro in organoids. Bulk and single-cell RNA sequencing revealed that, after LC ablation, BSCs activate a hybrid basal and luminal cell differentiation program before giving rise to LCs—reminiscent of the genetic program that regulates multipotency during embryonic development7. By predicting ligand–receptor pairs from single-cell data14, we find that TNF—which is secreted by LCs—restricts BC multipotency under normal physiological conditions. By contrast, the Notch, Wnt and EGFR pathways were activated in BSCs and their progeny after LC ablation; blocking these pathways, or stimulating the TNF pathway, inhibited regeneration-induced BC multipotency. Our study demonstrates that heterotypic communication between LCs and BCs is essential to maintain lineage fidelity in glandular epithelial stem cells.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: LC ablation promotes BC multipotency in glandular epithelia.
Fig. 2: BSC multipotency is associated with a hybrid basal and luminal signature.
Fig. 3: TNF expression by LCs restricts BC multipotency under homeostatic conditions.
Fig. 4: Notch, Wnt and ErbB signalling pathways regulate BSC multipotency after LC ablation.

Data availability

Bulk RNA-seq data have been deposited in the NCBI Gene Expression Omnibus under accession number GSE127975. Single-cell RNA-seq data have been deposited in the Gene Expression Omnibus under accession number GSE148791. Data supporting the findings of this study are available within the article. Source data are provided with this paper.

References

  1. Lloyd-Lewis, B., Harris, O. B., Watson, C. J. & Davis, F. M. Mammary stem cells: premise, properties, and perspectives. Trends Cell Biol. 27, 556–567 (2017).

    PubMed  Google Scholar 

  2. Toivanen, R. & Shen, M. M. Prostate organogenesis: tissue induction, hormonal regulation and cell type specification. Development 144, 1382–1398 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Van Keymeulen, A. et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature 479, 189–193 (2011).

    ADS  PubMed  Google Scholar 

  4. Choi, N., Zhang, B., Zhang, L., Ittmann, M. & Xin, L. Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell 21, 253–265 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ousset, M. et al. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat. Cell Biol. 14, 1131–1138 (2012).

    CAS  PubMed  Google Scholar 

  6. Lu, C. P. et al. Identification of stem cell populations in sweat glands and ducts reveals roles in homeostasis and wound repair. Cell 150, 136–150 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Wuidart, A. et al. Early lineage segregation of multipotent embryonic mammary gland progenitors. Nat. Cell Biol. 20, 666–676 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Lilja, A. M. et al. Clonal analysis of Notch1-expressing cells reveals the existence of unipotent stem cells that retain long-term plasticity in the embryonic mammary gland. Nat. Cell Biol. 20, 677–687 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Shackleton, M. et al. Generation of a functional mammary gland from a single stem cell. Nature 439, 84–88 (2006).

    ADS  CAS  PubMed  Google Scholar 

  10. Stingl, J. et al. Purification and unique properties of mammary epithelial stem cells. Nature 439, 993–997 (2006).

    ADS  CAS  PubMed  Google Scholar 

  11. Prater, M. D. et al. Mammary stem cells have myoepithelial cell properties. Nat. Cell Biol. 16, 942–950 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Van Keymeulen, A. et al. Reactivation of multipotency by oncogenic PIK3CA induces breast tumour heterogeneity. Nature 525, 119–123 (2015).

    ADS  PubMed  Google Scholar 

  13. Koren, S. et al. PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 525, 114–118 (2015).

    ADS  CAS  PubMed  Google Scholar 

  14. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal–fetal interface in humans. Nature 563, 347–353 (2018).

    ADS  CAS  PubMed  Google Scholar 

  15. Kwon, O. J., Zhang, L., Ittmann, M. M. & Xin, L. Prostatic inflammation enhances basal-to-luminal differentiation and accelerates initiation of prostate cancer with a basal cell origin. Proc. Natl Acad. Sci. USA 111, E592–E600 (2014).

    CAS  PubMed  Google Scholar 

  16. Li, J. J. & Shen, M. M. Prostate stem cells and cancer stem cells. Cold Spring Harb. Perspect. Med. 9, a030395 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Toivanen, R., Mohan, A. & Shen, M. M. Basal progenitors contribute to repair of the prostate epithelium following induced luminal anoikis. Stem Cell Reports 6, 660–667 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Horton, C., Liu, Y., Yu, C., Xie, Q. & Wang, Z. A. Luminal-contact-inhibition of epithelial basal stem cell multipotency in prostate organogenesis and homeostasis. Biol. Open 8, bio045724 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Emmerson, E. & Knox, S. M. Salivary gland stem cells: a review of development, regeneration and cancer. Genesis 56, e23211 (2018).

    PubMed  PubMed Central  Google Scholar 

  20. Weng, P. L., Aure, M. H., Maruyama, T. & Ovitt, C. E. Limited regeneration of adult salivary glands after severe injury involves cellular plasticity. Cell Rep. 24, 1464–1470.e3 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. May, A. J. et al. Diverse progenitor cells preserve salivary gland ductal architecture after radiation-induced damage. Development 145, dev166363 (2018).

    PubMed  PubMed Central  Google Scholar 

  22. Pal, B. et al. Construction of developmental lineage relationships in the mouse mammary gland by single-cell RNA profiling. Nat. Commun. 8, 1627 (2017).

    ADS  PubMed  PubMed Central  Google Scholar 

  23. Bach, K. et al. Differentiation dynamics of mammary epithelial cells revealed by single-cell RNA sequencing. Nat. Commun. 8, 2128 (2017).

    ADS  PubMed  PubMed Central  Google Scholar 

  24. Giraddi, R. R. et al. Single-cell transcriptomes distinguish stem cell state changes and lineage specification programs in early mammary gland development. Cell Rep. 24, 1653–1666.e7 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kurita, T., Medina, R. T., Mills, A. A. & Cunha, G. R. Role of p63 and basal cells in the prostate. Development 131, 4955–4964 (2004).

    CAS  PubMed  Google Scholar 

  26. Signoretti, S. et al. p63 regulates commitment to the prostate cell lineage. Proc. Natl Acad. Sci. USA 102, 11355–11360 (2005).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yalcin-Ozuysal, O. et al. Antagonistic roles of Notch and p63 in controlling mammary epithelial cell fates. Cell Death Differ. 17, 1600–1612 (2010).

    CAS  PubMed  Google Scholar 

  28. Bernardo, G. M. et al. FOXA1 is an essential determinant of ERα expression and mammary ductal morphogenesis. Development 137, 2045–2054 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Choi, Y. S., Chakrabarti, R., Escamilla-Hernandez, R. & Sinha, S. Elf5 conditional knockout mice reveal its role as a master regulator in mammary alveolar development: failure of Stat5 activation and functional differentiation in the absence of Elf5. Dev. Biol. 329, 227–241 (2009).

    CAS  PubMed  Google Scholar 

  30. Fu, N. Y. et al. Identification of quiescent and spatially restricted mammary stem cells that are hormone responsive. Nat. Cell Biol. 19, 164–176 (2017).

    CAS  PubMed  Google Scholar 

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

    ADS  CAS  PubMed  Google Scholar 

  32. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    PubMed  PubMed Central  Google Scholar 

  34. Buono, K. D. et al. The canonical Notch/RBP-J signaling pathway controls the balance of cell lineages in mammary epithelium during pregnancy. Dev. Biol. 293, 565–580 (2006).

    CAS  PubMed  Google Scholar 

  35. Bouras, T. et al. Notch signaling regulates mammary stem cell function and luminal cell-fate commitment. Cell Stem Cell 3, 429–441 (2008).

    CAS  PubMed  Google Scholar 

  36. van Amerongen, R., Bowman, A. N. & Nusse, R. Developmental stage and time dictate the fate of Wnt/β-catenin-responsive stem cells in the mammary gland. Cell Stem Cell 11, 387–400 (2012).

    PubMed  Google Scholar 

  37. van Genderen, C. et al. Development of several organs that require inductive epithelial-mesenchymal interactions is impaired in LEF-1-deficient mice. Genes Dev. 8, 2691–2703 (1994).

    PubMed  Google Scholar 

  38. Chu, E. Y. et al. Canonical WNT signaling promotes mammary placode development and is essential for initiation of mammary gland morphogenesis. Development 131, 4819–4829 (2004).

    CAS  PubMed  Google Scholar 

  39. Forster, N. et al. Basal cell signaling by p63 controls luminal progenitor function and lactation via NRG1. Dev. Cell 28, 147–160 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Balko, J. M. et al. The receptor tyrosine kinase ErbB3 maintains the balance between luminal and basal breast epithelium. Proc. Natl Acad. Sci. USA 109, 221–226 (2012).

    ADS  CAS  PubMed  Google Scholar 

  41. Lafkas, D. et al. Therapeutic antibodies reveal Notch control of transdifferentiation in the adult lung. Nature 528, 127–131 (2015).

    ADS  CAS  PubMed  Google Scholar 

  42. Liu, J. et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl Acad. Sci. USA 110, 20224–20229 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tata, P. R. et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 503, 218–223 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lynch, T. J. et al. Submucosal gland myoepithelial cells are reserve stem cells that can regenerate mouse tracheal epithelium. Cell Stem Cell 22, 653–667 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Tata, A. et al. Myoepithelial cells of submucosal glands can function as reserve stem cells to regenerate airways after injury. Cell Stem Cell 22, 668–683 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Varela, L. M. & Ip, M. M. Tumor necrosis factor-alpha: a multifunctional regulator of mammary gland development. Endocrinology 137, 4915–4924 (1996).

    CAS  PubMed  Google Scholar 

  47. Lee, P. P., Hwang, J. J., Murphy, G. & Ip, M. M. Functional significance of MMP-9 in tumor necrosis factor-induced proliferation and branching morphogenesis of mammary epithelial cells. Endocrinology 141, 3764–3773 (2000).

    CAS  PubMed  Google Scholar 

  48. Youssef, K. K. et al. Adult interfollicular tumour-initiating cells are reprogrammed into an embryonic hair follicle progenitor-like fate during basal cell carcinoma initiation. Nat. Cell Biol. 14, 1282–1294 (2012).

    CAS  PubMed  Google Scholar 

  49. Ge, Y. et al. Stem cell lineage infidelity drives wound repair and cancer. Cell 169, 636–650 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Watson, J. K. et al. Clonal dynamics reveal two distinct populations of basal cells in slow-turnover airway epithelium. Cell Rep. 12, 90–101 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Weber, T. et al. Inducible gene expression in GFAP+ progenitor cells of the SGZ and the dorsal wall of the SVZ—a novel tool to manipulate and trace adult neurogenesis. Glia 59, 615–626 (2011).

    PubMed  Google Scholar 

  52. 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).

    CAS  PubMed  Google Scholar 

  53. Jardé, T. et al. Wnt and Neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nat. Commun. 7, 13207 (2016).

    ADS  PubMed  PubMed Central  Google Scholar 

  54. Krause, S., Brock, A. & Ingber, D. E. Intraductal injection for localized drug delivery to the mouse mammary gland. J. Vis. Exp. 80, e50692 (2013).

    Google Scholar 

  55. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  56. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

  57. Huang, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Google Scholar 

  58. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    CAS  PubMed  Google Scholar 

  59. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  61. McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Huynh-Thu, V. A., Irrthum, A., Wehenkel, L. & Geurts, P. Inferring regulatory networks from expression data using tree-based methods. PLoS One 5, e12776 (2010).

    ADS  PubMed  PubMed Central  Google Scholar 

  64. Angerer, P. et al. destiny: diffusion maps for large-scale single-cell data in R. Bioinformatics 32, 1241–1243 (2016).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the ULB animal facility and ULB genomic core facility (F. Libert and A. Lefort); J. M. Vanderwinden and LiMif for help with confocal microscopy; J. Rajagopal and P. R. Tata for their suggestions concerning the TetO-DTA mice; and B. Lloyd-Lewis for help with the organoid culture. This work was supported by the ERC and the FNRS. A.C. is supported by the FNRS/FRIA. S.L. is a long-term EMBO fellow. C.B. is supported by WELBIO, FNRS, TELEVIE, Fond Erasme, Fondation Contre le Cancer, ULB Foundation, European Research Council and the Foundation Baillet Latour. A. Sifrim, J.V.H. and T.V. are supported by KULeuven (SymBioSys – C14/18/092), the Fondation Contre le Cancer (2015-143) and FWO postdoctoral fellowships 12W7318N and I001818N.

Author information

Authors and Affiliations

Authors

Contributions

A.C., S.L. and C.B. designed the experiments and performed data analysis. A.C. and S.L. performed most of the biological experiments. E.T. performed the experiments and data analysis on prostate glands. A. Sifrim, M.M., Y.S., J.V.H. and T.V. performed the bioinformatic analysis. A.D. and G.B. provided technical help. C.D. performed FACS experiments. N.D. provided technical help with single-cell RNA sequencing. A.C., S.L., M.F., A.W. and A.V.K. performed immunostainings, blocking antibodies and small-molecule treatments and experiments with follow-up mice. A. Sahay contributed genetic tools. V.d.M. performed statistical analysis. C.W.S. provided the Notch antibodies. A.C., A.V.K. and C.B. wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Cédric Blanpain.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature thanks Jacco van Rheenen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 FACS analysis of MG epithelial cells.

ad, Unicellular suspension of MG cells from adult K5CreER/tdTomato/K8rtTA/TetO-DTA mice induced at 8 weeks stained for endothelial, immune and fibroblast markers (Lin+) (CD31, CD45, CD140a) in APC, EpCAM in Apc-Cy7 and CD29 in FITC, were gated to eliminate debris (a), doublets were discarded with gates (b), the living cells were gated by DAPI dye exclusion (c) and the non-epithelial Lin+ cells were discarded (d). CD24/CD29 and EpCAM/CD29 expression was studied in Lin. eg, CD24/CD29 gates, EpCAM/CD29 gates and the absence of leakiness of fluorescent reporter recombination before tamoxifen injection (e), after tamoxifen injection (f) and after DOX injection (g). The CD29lowCD24high and the CD29lowEpCAMhigh gate corresponds to LCs, the CD29highCD24low and the CD29highEpCAMlow gate corresponds to BCs, the CD29highEpCAMhigh correspond to the new population described in this manuscript. A total of 400,000 events is shown in the graphs. Percentages are calculated on the total LCs and BCs or LCs, CD29highEpCAMhigh and BCs shown. n = 3 independent experiments per condition. h, Graph showing the percentage of BCs that were tdTomato-labelled on the total number of BCs gated on CD24/CD29 before DOX injection (79.5 ± 2.2%; n = 20 mice, data are mean ± s.e.m.) after DOX injection (CD24 83.9 ± 2.6%; n = 23 mice, data are mean ± s.e.m.) and EpCAM/CD29 before DOX injection (84.8 ± 2.2%; n = 20 mice, data are mean ± s.e.m.) and after DOX injection (88.9 ± 1.9%; n = 23 mice, data are mean ± s.e.m.). P values are derived from unpaired two-sided t-tests.

Extended Data Fig. 2 DOX administration to K5CreER/tdTomato/K8rtTA/TetO-DTA mice promotes LC death, MG remodelling and proliferation.

a, Scheme summarizing the histology of the MG and its different lineages. b, c, FACS plot of CD29/CD24 expression in LintdTomato+ epithelial cells (b) and quantification of tdTomato+ LCs (c) in CTR mice and 1 week after DOX administration. The percentage of the gated population from all epithelial cells is shown. Number of mice analysed is shown in parentheses. P values are derived from unpaired two-sided t-tests. d, e, Confocal imaging (d) and FACS quantification (e) of immunostaining for tdTomato, cleaved caspase 3 (CC3) and K8 in K5CreER/tdTomato/K8rtTA/TetO-DTA MG after IDI with NaCl (0.29 ± 0.02 CC3+ cells) or 0.2 mg DOX (1.3 ± 0.4 CC3+ cells) and chased for 12 h (n = 6 mice per condition). Scale bar (d), 5 μm. P values are derived from unpaired two-sided t-tests. fj, Scheme of the different parameters measured to quantify MG remodelling (f), quantification and representation after ImageJ analysis of branch length, number of branches, branch points and terminal end buds of K5CreER/Rosa-tdTomato/K8rtTA/tet-O-DTA (g, h) or K5CreER/Rosa-tdTomato/K8rtTA contralateral MG (i, j) injected either with NaCl or DOX. Number of mice analysed is shown in parentheses. Scale bars, 100 μm. P values are derived from paired two-sided t-tests. k, Confocal imaging of immunostaining for tdTomato, E-cadherin and K14 of terminal end buds, branching points and ducts where LC replacement is occurring in a patchy and focal manner throughout the gland (n = 3 mice). Scale bars, 20 μm. l, m, Graphs showing the percentage of K8+CC3+ cells at 0 h, 12 h and 24 h (n = 3 mice per condition) after DOX injection (l) and FACS quantification of K8+CC3+ cells 12 h after different DOX doses (m). Number of mice analysed is shown in parentheses. n, o, Confocal imaging of immunostaining for tdTomato, K8 and CC3 (n) and FACS quantification of K8+tdTomato+ cells (o) in mice after intraductal injection with NaCl or DOX at different doses after 12 h. Number of mice analysed is shown in parentheses. p, q, Confocal imaging (p) and quantification (q) of immunostaining for tdTomato, Ki67 and K8 in K5CreER/tdTomato/K8rtTA/TetO-DTA MG after IDI with NaCl or 0.2 mg DOX and chased for 1 week. Scale bars, 5 μm. n = 3 mice per condition. P values are derived from unpaired two-sided t-tests. Data are mean ± s.e.m. For the immunofluorescence data, Hoechst nuclear staining is shown in blue.

Source data

Extended Data Fig. 3 DOX-induced DTA in LCs promotes inflammation, LC death, proliferation and activation of BC multipotency in MG in vivo and organoids in vitro.

a, b, Confocal imaging of immunostaining for tdTomato, CD68 and CD45 (a) and FACS quantification of CD45+CD68+ cells in MG cells from K5CreER/Rosa-tdTomato/K8rtTA/TetO-DTA CTR mice or DOX or DOX+DEX treated (DEX, dexamethasone) (b). n = 3 mice. P values are derived from unpaired two-sided t-tests. c, qRT–PCR analysis of the whole MG of K5CreER/Rosa-tdTomato/K8rtTA/TetO-DTA mice CTR, DOX, No DOX+DEX or DOX+DEX (n = 3 mice for each group), showing the upregulation of inflammation markers upon LC ablation and its normalization after DEX treatment. d, FACS quantification of tdTomato+ LCs in mice after DOX or DOX+DEX treatment (n = 5 mice). P values are derived from unpaired two-sided t-tests. e, f, Representative FACS plot of CD29/EpCAM expression in LinYFP+ epithelial cells of MG from K14rtTA/TetO-DTA/K8CreER/Rosa-YFP mice in CTR or one week after IDI of NaCl or DOX (e) and quantification of YFP+ cells in LCs and BCs (f) (n = 5 mice). g, h, Confocal imaging (g) and quantification of immunostaining (h) for tdTomato, CC3 and K8 in K5CreER/tdTomato/K8rtTA/TetO-DTA MG organoids without DOX (CTR) or 6 h after DOX. n = 4 independent experiments. P values are derived from unpaired two-sided t-tests. i, j, Confocal imaging (i) and quantification (j) of immunostaining for tdTomato, Ki67 and K8 in K5CreER/tdTomato/K8rtTA/TetO-DTA in organoids without DOX (CTR) or 24 h after DOX. n = 4 independent experiments. P values are derived from unpaired two-sided t-tests. k, Confocal imaging of immunostaining for tdTomato, K8 and K14 in K5CreER/tdTomato/K8rtTA/TetO-DTA CTR organoids or after DOX. Lower magnification of the images shown in Fig. 1d. n = 3 independent experiments. l, m, Confocal imaging (l) and quantification (m) of immunostaining for tdTomato, Ki67 and K8 in K5CreER/tdTomato/K8rtTA/TetO-DTA organoids treated with DOX, DOX+adeno-P21 or DOX+CDK1 inhibitor (CDK1-i) for 48 h and chased for 24 h. n, Quantification of tdTomato+K8+ cells in K5CreER/tdTomato/K8rtTA/TetO-DTA organoids treated with DOX, DOX+adeno-P21 or DOX+CDK1 inhibitor. For ln, n = 3 independent experiments. P values are derived from ANOVA followed by two-sided Dunnett’s tests. Data are mean ± s.e.m. For the immunofluorescence data, Hoechst nuclear staining is shown in blue. Scale bars, 10 μm.

Source data

Extended Data Fig. 4 DOX administration in adult K5CreER/tdTomato/K8rtTA/TetO-DTA mice promotes LC death and proliferation in the prostate, salivary gland and sweat gland.

a, Schematic representation of the histology of the salivary gland and its lineages. b, c, Confocal imaging (b) and quantification (c) of immunostaining for tdTomato, AQP5 and smooth muscle heavy chain myosin (SMH) of salivary gland acinar cells. n = 3 mice per condition. P values are derived from unpaired two-sided t-tests. d, e, Confocal imaging (d) and quantification (e) of immunostaining for tdTomato, K8 and SMH in the granulated duct showing the contribution of BCs to replenishing granulated ductal cells during homeostasis. n = 3 mice per condition. P values are derived from unpaired two-sided t-tests. f, Schematic representation of the histology of prostate and its lineages. gj, Confocal imaging (g, i) and quantification (h, j) of prostate immunostaining for tdTomato, K8 and CC3 or Ki67 in CTR or K5CreER/tdTomato/K8rtTA/TetO-DTA mice treated with DOX for 5 days and chased for 1 day. P values are derived from unpaired two-sided t-tests. n = 3 mice per condition. k, Schematic representation of the histology of the sweat gland and its different lineages. lo, Confocal imaging (l, n) and quantification (m, o) of sweat gland immunostaining for tdTomato, K8 and CC3 (l, m) or tdTomato, K8 and Ki67 (n, o) in CTR or K5CreER/tdTomato/K8rtTA/TetO-DTA mice treated with DOX for 5 days and chased for 1 day. P values are derived from unpaired two-sided t-tests. n = 3 mice per condition. ps, Confocal imaging (p, r) and quantification (q, s) of the intercalated duct of salivary gland immunostaining for tdTomato, K8 and CC3 (p, q) or tdTomato, K8 and Ki67 (r, s) in CTR or K5CreER/tdTomato/K8rtTA/TetO-DTA mice treated with DOX for 5 days and chased for 1 day. n = 4 for CC3 and n = 3 for Ki67 mice per condition. P values are derived from unpaired two-sided t-tests. Data are mean ± s.e.m. For the immunofluorescence data, Hoechst nuclear staining is shown in blue. Scale bars, 5 μm.

Source data

Extended Data Fig. 5 Transcriptional profiling associated with BC multipotency induced by LC ablation.

a, b, Gene ontology (GO) analysis of genes upregulated more than 2.5-fold in CD29highEpCAMhigh cells compared to LCs (a) or BCs (b). Histograms represent –log10 of the Benjamini P value. n = 2 independent experiments. c, d, Graphs representing mRNA expression measured by bulk population RNA sequencing of basal (c) and luminal (d) upregulated genes in FACS-isolated wild-type BCs, LC EXP, BC EXP, CD29highEpCAMhigh EXP. Fold change over wild-type LCs (c) or over wild-type BCs (d) of genes involved in different biological processes. n = 2 independent experiments. e, Confocal images of MGs from CTR mice or mice with IDI DOX. Immunostaining for K14, Snail2 and K8. f, Immunohistochemistry of Il33 in CTR MGs or MG after DOX IDI. g, Immunostaining for tdTomato, TnC and K14. h, Masson’s trichrome staining for collagen in CTR MGs or MG after DOX IDI. i, Immunostaining for tdTomato, Alcam and K14. n = 3 mice per condition. Hoechst nuclear staining in blue. Scale bars, 5 μm.

Source data

Extended Data Fig. 6 Single-cell transcriptional profiling reveals the hybrid features of CD29highEpCAMhigh cells associated with LC-ablation-induced BC multipotency.

a, Heat map of normalized scRNa-seq expression. Rows represent marker genes (ROC AUC >0.7 and log fold change >0.25) for clusters discovered through unsupervised clustering (resolution = 0.2) and columns represent individual cells, grouped by lineage and treatment condition. Colour values in the heat map represent normalized and scaled expression values. These data show the hybrid basal and luminal signature of the CD29highEpCAMhigh cells (Hybrid EXP) after LC ablation. b, t-SNE plots illustrating the expression of markers for BC and LC lineages (n = 337 cells). c, Graph showing the mRNA expression (read per million) of Tspan8 and Procr. n = 2 independent experiments. d, t-SNE and violin plots illustrating the expression for Tspan8. These data show that Tspan8 is expressed at similar levels in the different MG populations under physiological conditions and after LC ablation (n = 337 cells). e, t-SNE and violin plots illustrating the expression for Procr. These data show that Procr is expressed at very low levels in the different MG populations under physiological conditions and is not increased after LC ablation (n = 337 cells). For t-SNE plots, data points represent individual cells, the colour scaling represents the expression level of the respective marker gene (high, yellow; low, black). For violin plots the minima, maxima, centre and percentiles are provided in the Source Data. f, g, t-SNE plots for differentially activated regulons (FDR-corrected P value <0.01 from one-sided Kolmogorov–Smirnov test) as computed by SCENIC, data points represent individual cells (n = 337), the colour scaling represents the regulon AUC value as a measure of the number of transcription factor target genes being expressed (high yellow, low black).

Source data

Extended Data Fig. 7 Lineage trajectory inference using Slingshot.

a, b, Diffusion maps showing normalized expression of Krt14 (a, BC) and Krt8 (b, LC). c, Heat map representing the top 100 most significant (lowest adjusted P value) genes differentially expressed along trajectory 1 (n = 337 cells). FDR-corrected P values from F-test on Loess-term for pseudotime in the GAM fit. d, Diffusion maps showing normalized expression of markers for ER+ LC (Foxa1, Ly6a/Sca1 and Pgr). e, Heat map representing the top 100 most significant (lowest adjusted P value) genes differentially expressed along trajectory 2 (n = 337 cells). FDR-corrected P values from F-test on Loess-term for pseudotime in the GAM fit. f, Diffusion maps showing normalized expression of markers for ER- LC (Elf5, Hey1 and Kit). For the diffusion maps the colour bars represent normalized expression levels, going from black (low expression) to yellow (high expression). For the heat maps rows represent the genes, ordered by hierarchical clustering. Colours correspond to the Z-score scaled expression of a gene. Columns represent cells, which are ordered by their pseudotime value in the respective trajectory and their types (BC, hybrid or LC) in control or experimental condition (wild-type or EXP) are indicated at the top of the heat map.

Extended Data Fig. 8 Immunostaining of basal and luminal markers after LC ablation and antibody permeability.

af, Confocal imaging of immunostaining for E-cadherin, K5 and tdTomato (ac) or E-cadherin, K14 and tdTomato (df) in CTR (a, d) or K5CreER/tdTomato/K8rtTA/TetO-DTA mice after IDI of DOX and chased for 1 week (b, e) or 2 weeks (c, f) (n = 3 mice per condition). g, Quantification of the proportion of hybrid K8+K14+tdTomato+ cells and K8+tdTomato+ cells on the total K8+tdTomato+ cells at 1 week (n = 7 mice), 2 weeks (n = 4 mice), 4 weeks (n = 3 mice) and 7 weeks (n = 3 mice) after DOX treatment. The P value between 1 week and 7 weeks, derived from ANOVA followed by two-sided Dunnett’s test, is shown. h, i, Confocal imaging of immunostaining for K14, p63 and K8 (h) and quantification of p63+ cells in K8+K14+ cells (i) in CTR or K5CreER/tdTomato/K8rtTA/TetO-DTA mice after DOX IDI and chased for 1 week. n = 3 mice per condition. j, k, Confocal imaging of immunostaining for K14, Foxa1 and K8 (j) and quantification of Foxa1+ cells in K8+K14+ cells (k) in CTR or K5CreER/tdTomato/K8rtTA/TetO-DTA mice after DOX IDI and chased for 1 week. n = 3 mice per condition. l, Confocal imaging of immunostaining for K14 and anti-rat Alexa Fluor 488 (green) in CTR MG not injected or MG after IDI of rat anti-β4 integrin (CD104) and chased for 2 days. n = 3 mice per condition. Data are mean ± s.e.m. For the immunofluorescence data, Hoechst nuclear staining is shown in blue. Scale bars, 5 μm.

Source data

Extended Data Fig. 9 CellPhone-DB and scRNA-seq on the heterotypic interaction significant upon LC ablation.

a, Graph representing the mean expression and P values of the heterotypic ligand–receptor interaction significant only after LC ablation compared to the wild type condition (n = 242 cells). P values are derived from a one-sided permutation test. bd, t-SNE and violin plots illustrating the expression for the ligand–receptor couples WNT7B–FZD4 (b), JAG1–NOTCH1 (c) and NRG1–ERBB3 (d) in the different cell populations and experimental conditions (n = 337 cells). For violin plots the minima, maxima, centre and percentiles are provided in Source Data. e, t-SNE plots illustrating the expression of Jag2, Wnt6 and Nrg1 (n = 337 cells). Data points represent individual cells. The colour scaling represents the expression level of the respective marker gene (high, yellow; low, black).

Source data

Extended Data Fig. 10 Effect of administration of anti-DLL1, anti-JAG1 and anti-JAG2 blocking antibodies; LGK-974; sapitinib and afatinib on MG and MG organoids.

a, qRT–PCR analysis of Notch target gene Hey1 and Nrarp of FACS-isolated LCs and BCs of K5CreER/tdTomato/K8rtTA/TetO-DTA mice treated with IgG2a (CTR Ab) or anti-Dll1, Jag1, Jag2 blocking antibodies (n = 5 mice per condition), showing the efficient inhibition of Notch target genes after blocking antibody administration. P values are derived from unpaired two-sided t-tests. b, Immunostaining for CC10 in lung sections of K5CreER/tdTomato/K8rtTA/TetO-DTA mice treated with CTR IgG2a or anti-Dll1, Jag1 and Jag2 blocking antibodies, showing the disappearance of the goblet cells demonstrating the efficiency of Notch inhibition (n = 3 mice per condition). c, Representative FACS plot of CD21/CD23 in CD45+CD5 spleen cells in mice treated with CTR IgG2a or anti-Dll1, Jag1 and Jag2 blocking antibodies showing the disappearance of the marginal zone B cells (CD21highCD23) demonstrating the efficiency of Notch inhibition (n = 3 mice per condition). d, qRT–PCR analysis of Wnt target gene Axin2 of FACS-isolated LCs and BCs of K5CreER/tdTomato/K8rtTA/TetO-DTA CTR mice or treated with LGK-974, showing the efficient inhibition of Wnt target genes by LGK-974 (n = 3 mice per condition). P values are derived from unpaired two-sided t-tests. e, f, Confocal imaging (e) and quantification (f) of immunostaining for tdTomato, phospho-EGFR (p-EGFR) and K8 in CTR and K5CreER/tdTomato/K8rtTA/TetO-DTA MG after DOX IDI and chased for 1 week and mice after DOX IDI and treated with sapitinib (Sap) and afatinib (Afa) and chased for 1 week (n = 3 mice per condition). P values are derived from ANOVA followed by two-sided Dunnett’s tests. g, Quantification of the expression of Ki67 in MG after IDI of DOX in the absence or in the presence of different inhibitors. The number of independent mice analysed is shown in parentheses. P values are derived from ANOVA followed by two-sided Dunnett’s tests. h, FACS analysis of the LC/BC ratio of MG in control mice (n = 6 mice); mice treated with anti-Notch ligand antibodies (n = 3 mice); with LGK-974 (n = 4 mice) and anti-ErbB inhibitors (n = 3 mice) for 9 days showing that the inhibition of Notch or Wnt signalling does not affect the ratio between LCs and BCs whereas Erbb inhibitors decreased the proportion of LCs. P values are derived from ANOVA followed by two-sided Dunnett’s tests. i, FACS analysis of MG showing tdTomato+ cells in LCs after NaCl IDI (n = 4 mice); DOX IDI (n = 5 mice); NaCl or DOX IDI and anti-Notch ligand antibodies (n = 3 mice); NaCl or DOX IDI and Wnt inhibitor (n = 4 mice); NaCl or DOX IDI and anti-ErbB inhibitors (n = 3 mice); for 9 days. P values are derived from unpaired two-sided t-tests. jm, Confocal imaging of immunostaining for tdTomato, K8 and K14 of MG (jl) and organoids (m) after DOX or DOX+anti-Notch ligand antibodies (j); DOX+Wnt inhibitor (k); DOX+sapitinib/afatinib (l). n, Quantification of the expression of tdTomato in K8+ cells in MG organoids treated with DOX, DOX+anti-Notch ligand antibodies, DOX+LGK-974 or DOX+sapitinib/afatinib for 48 h and chased for 10 days with media and the corresponding inhibitor. Data are normalized over DOX. For m, n, n = 3 independent experiments. Data are mean ± s.e.m. P values are derived from ANOVA followed by two-sided Dunnett’s tests. Data are mean ± s.e.m. For the immunofluorescence data, Hoechst nuclear staining is shown in blue. Scale bars, 5 μm.

Source data

Supplementary information

Supplementary Table

Supplementary Table 1. Table presenting the primers used for qRT-PCR.

Reporting Summary

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Centonze, A., Lin, S., Tika, E. et al. Heterotypic cell–cell communication regulates glandular stem cell multipotency. Nature 584, 608–613 (2020). https://doi.org/10.1038/s41586-020-2632-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2632-y

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