Skip to main content

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

  • Article
  • Published:

Reconstruction of dynamic mammary mini gland in vitro for normal physiology and oncogenesis

Abstract

Organoid culture has been extensively exploited for normal tissue reconstruction and disease modeling. However, it is still challenging to establish organoids that mimic in vivo-like architecture, size and function under homeostatic conditions. Here we describe the development of a long-term adult stem cell-derived mammary mini gland culture system that supports robust three-dimensional outgrowths recapitulating the morphology, scale, cellular context and transcriptional heterogeneity of the normal mammary gland. The self-organization ability of stem cells and the stability of the outgrowths were determined by a coordinated combination of extracellular matrix, environmental signals and dynamic physiological cycles. We show that these mini glands were hormone responsive and could recapitulate the entire postnatal mammary development including puberty, estrus cycle, lactation and involution. We also observed that these mini glands maintained the presence of mammary stem cells and could also recapitulate the fate transition from embryonic bipotency to postnatal unipotency in lineage tracing assays. In addition, upon induction of oncogene expression in the mini glands, we observed tumor initiation in vitro and in vivo in a mouse model. Together, this study provides an experimental system that can support a dynamic miniature mammary gland for the study of physiologically relevant, complex biological processes.

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: Development of mammary mini gland with augmented branching patterns and stabilized structures.
Fig. 2: Mammary mini gland recapitulates the molecular identity and cellular heterogeneity of mammary gland.
Fig. 3: The mammary mini gland recapitulates the reproductive functions of mammary gland.
Fig. 4: The mammary mini gland maintains mammary stem cells.
Fig. 5: The mini gland is able to recapitulate stem cell fate transition.
Fig. 6: The mammary mini gland can be utilized to model cancer initiation.

Similar content being viewed by others

Data availability

Sequencing raw data and counts matrices are available under accession number GEO: GSE211527. Computer code for single-cell transcriptome analysis is available at Supplementary Code. Source data are provided with this paper.

References

  1. Rossi, G., Manfrin, A. & Lutolf, M. P. Progress and potential in organoid research. Nat. Rev. Genet. 19, 671–687 (2018).

    CAS  PubMed  Google Scholar 

  2. Hofer, M. & Lutolf, M. P. Engineering organoids. Nat. Rev. Mater. 6, 402–420 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Fatehullah, A., Tan, S. H. & Barker, N. Organoids as an in vitro model of human development and disease. Nat. Cell Biol. 18, 246–254 (2016).

    PubMed  Google Scholar 

  4. 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 

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

    PubMed  Google Scholar 

  6. Davis, F. M. et al. Single-cell lineage tracing in the mammary gland reveals stochastic clonal dispersion of stem/progenitor cell progeny. Nat. Commun. 7, 13053 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Jamieson, P. R. et al. Derivation of a robust mouse mammary organoid system for studying tissue dynamics. Development 144, 1065–1071 (2017).

    CAS  PubMed  Google Scholar 

  8. Zhang, L. et al. Establishing estrogen-responsive mouse mammary organoids from single Lgr5+ cells. Cell Signal 29, 41–51 (2017).

    PubMed  Google Scholar 

  9. Wang, J. et al. Endothelial Wnts control mammary epithelial patterning via fibroblast signaling. Cell Rep. 34, 108897 (2021).

    CAS  PubMed  Google Scholar 

  10. 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 

  11. Boras-Granic, K., Dann, P. & Wysolmerski, J. J. Embryonic cells contribute directly to the quiescent stem cell population in the adult mouse mammary gland. Breast Cancer Res. 16, 487 (2014).

    PubMed  PubMed Central  Google Scholar 

  12. 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 

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

    CAS  PubMed  Google Scholar 

  14. Cai, S. et al. A quiescent Bcl11b high stem cell population is required for maintenance of the mammary gland. Cell Stem Cell 20, 247–260 e245 (2017).

    CAS  PubMed  Google Scholar 

  15. Maller, O., Martinson, H. & Schedin, P. Extracellular matrix composition reveals complex and dynamic stromal-epithelial interactions in the mammary gland. J. Mammary Gland Biol. Neoplasia 15, 301–318 (2010).

    PubMed  Google Scholar 

  16. Ghajar, C. M. & Bissell, M. J. Extracellular matrix control of mammary gland morphogenesis and tumorigenesis: insights from imaging. Histochem. Cell Biol. 130, 1105–1118 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Nguyen-Ngoc, K. V. & Ewald, A. J. Mammary ductal elongation and myoepithelial migration are regulated by the composition of the extracellular matrix. J. Microsc. 251, 212–223 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Nguyen-Ngoc, K. V. et al. ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc. Natl Acad. Sci. USA 109, E2595–E2604 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Nguyen-Ngoc, K. V. et al. 3D culture assays of murine mammary branching morphogenesis and epithelial invasion. Methods Mol. Biol. 1189, 135–162 (2015).

    PubMed  PubMed Central  Google Scholar 

  20. Veltmaat, J. M. et al. Gli3-mediated somitic Fgf10 expression gradients are required for the induction and patterning of mammary epithelium along the embryonic axes. Development 133, 2325–2335 (2006).

    CAS  PubMed  Google Scholar 

  21. Mailleux, A. A. et al. Role of FGF10/FGFR2b signaling during mammary gland development in the mouse embryo. Development 129, 53–60 (2002).

    CAS  PubMed  Google Scholar 

  22. Zhang, X. et al. FGF ligands of the postnatal mammary stroma regulate distinct aspects of epithelial morphogenesis. Development 141, 3352–3362 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Ewald, A. J., Brenot, A., Duong, M., Chan, B. S. & Werb, Z. Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev. Cell 14, 570–581 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Neumann, N. M. et al. Coordination of receptor tyrosine kinase signaling and interfacial tension dynamics drives radial intercalation and tube elongation. Dev. Cell 45, 67–82 e66 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kleinberg, D. L., Feldman, M. & Ruan, W. IGF-I: an essential factor in terminal end bud formation and ductal morphogenesis. J. Mammary Gland Biol. Neoplasia 5, 7–17 (2000).

    CAS  PubMed  Google Scholar 

  26. Ki, M. H. et al. All-trans retinoic acid induced differentiation of rat mammary epithelial cells cultured in serum-free medium. Arch. Pharm. Res 21, 298–304 (1998).

    CAS  PubMed  Google Scholar 

  27. Cabezuelo, M. T., Zaragoza, R., Barber, T. & Vina, J. R. Role of vitamin A in mammary gland development and lactation. Nutrients https://doi.org/10.3390/nu12010080 (2019).

  28. McLean, A. C., Valenzuela, N., Fai, S. & Bennett, S. A. Performing vaginal lavage, crystal violet staining, and vaginal cytological evaluation for mouse estrous cycle staging identification. J. Vis. Exp. https://doi.org/10.3791/4389 (2012).

  29. Fata, J. E., Chaudhary, V. & Khokha, R. Cellular turnover in the mammary gland is correlated with systemic levels of progesterone and not 17β-estradiol during the estrous cycle. Biol. Reprod. 65, 680–688 (2001).

    CAS  PubMed  Google Scholar 

  30. Shehata, M. et al. Phenotypic and functional characterisation of the luminal cell hierarchy of the mammary gland. Breast Cancer Res. 14, R134 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Wiseman, B. S. & Werb, Z. Stromal effects on mammary gland development and breast cancer. Science 296, 1046–1049 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Mikaelian, I. et al. Expression of terminal differentiation proteins defines stages of mouse mammary gland development. Vet. Pathol. 43, 36–49 (2006).

    CAS  PubMed  Google Scholar 

  33. Asselin-Labat, M. L. et al. Gata-3 is an essential regulator of mammary-gland morphogenesis and luminal-cell differentiation. Nat. Cell Biol. 9, 201–209 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  35. Brisken, C. & Rajaram, R. D. Alveolar and lactogenic differentiation. J. Mammary Gland Biol. Neoplasia 11, 239–248 (2006).

    PubMed  Google Scholar 

  36. Schedin, P., Mitrenga, T. & Kaeck, M. Estrous cycle regulation of mammary epithelial cell proliferation, differentiation, and death in the Sprague–Dawley rat: a model for investigating the role of estrous cycling in mammary carcinogenesis. J. Mammary Gland Biol. Neoplasia 5, 211–225 (2000).

    CAS  PubMed  Google Scholar 

  37. Cai, C. et al. Amphiregulin mediates the hormonal regulation on Rspondin-1 expression in the mammary gland. Dev. Biol. 458, 43–51 (2020).

    CAS  PubMed  Google Scholar 

  38. Macias, H. & Hinck, L. Mammary gland development. Wiley Interdiscip. Rev. Dev. Biol. 1, 533–557 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Jena, M. K., Jaswal, S., Kumar, S. & Mohanty, A. K. Molecular mechanism of mammary gland involution: an update. Dev. Biol. 445, 145–155 (2019).

    CAS  PubMed  Google Scholar 

  40. Ichinose, R. R. & Nandi, S. Lobuloalveolar differentiation in mouse mammary tissues in vitro. Science 145, 496–497 (1964).

    CAS  PubMed  Google Scholar 

  41. Sumbal, J., Chiche, A., Charifou, E., Koledova, Z. & Li, H. Primary mammary organoid model of lactation and involution. Front. Cell Dev. Biol. 8, 68 (2020).

    PubMed  PubMed Central  Google Scholar 

  42. Nishimori, K. et al. Oxytocin is required for nursing but is not essential for parturition or reproductive behavior. Proc. Natl Acad. Sci. USA 93, 11699–11704 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kamikawa, A. & Seko, J. Physiological and pharmacological evaluation of oxytocin-induced milk ejection in mice. Exp. Anim. 69, 345–353 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Marti, A. et al. Mouse mammary gland involution is associated with cytochrome c release and caspase activation. Mech. Dev. 104, 89–98 (2001).

    CAS  PubMed  Google Scholar 

  45. Song, J. et al. Roles of Fas and Fas ligand during mammary gland remodeling. J. Clin. Invest. 106, 1209–1220 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Ge, Y. & Fuchs, E. Stretching the limits: from homeostasis to stem cell plasticity in wound healing and cancer. Nat. Rev. Genet. 19, 311–325 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Rios, A. C., Fu, N. Y., Lindeman, G. J. & Visvader, J. E. In situ identification of bipotent stem cells in the mammary gland. Nature 506, 322–327 (2014).

    CAS  PubMed  Google Scholar 

  48. Centonze, A. et al. Heterotypic cell–cell communication regulates glandular stem cell multipotency. Nature 584, 608–613 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Lin, E. Y. et al. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am. J. Pathol. 163, 2113–2126 (2003).

    PubMed  PubMed Central  Google Scholar 

  50. Acheampong, T., Kehm, R. D., Terry, M. B., Argov, E. L. & Tehranifar, P. Incidence trends of breast cancer molecular subtypes by age and race/ethnicity in the US from 2010 to 2016. JAMA Netw. Open 3, e2013226 (2020).

    PubMed  PubMed Central  Google Scholar 

  51. Prater, M., Shehata, M., Watson, C. J. & Stingl, J. Enzymatic dissociation, flow cytometric analysis, and culture of normal mouse mammary tissue. Methods Mol. Biol. 946, 395–409 (2013).

    CAS  PubMed  Google Scholar 

  52. Marampon, F. et al. MEK/ERK inhibitor U0126 affects in vitro and in vivo growth of embryonal rhabdomyosarcoma. Mol. Cancer Ther. 8, 543–551 (2009).

    CAS  PubMed  Google Scholar 

  53. Favata, M. F. et al. Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J. Biol. Chem. 273, 18623–18632 (1998).

    CAS  PubMed  Google Scholar 

  54. Allen, L. F., Sebolt-Leopold, J. & Meyer, M. B. CI-1040 (PD184352), a targeted signal transduction inhibitor of MEK (MAPKK). Semin. Oncol. 30, 105–116 (2003).

    CAS  PubMed  Google Scholar 

  55. Hayot, C. et al. Characterization of the activities of actin-affecting drugs on tumor cell migration. Toxicol. Appl. Pharmacol. 211, 30–40 (2006).

    CAS  PubMed  Google Scholar 

  56. Shoji, K., Ohashi, K., Sampei, K., Oikawa, M. & Mizuno, K. Cytochalasin D acts as an inhibitor of the actin-cofilin interaction. Biochem. Biophys. Res. Commun. 424, 52–57 (2012).

    CAS  PubMed  Google Scholar 

  57. Nemoto, S., Xiang, J., Huang, S. & Lin, A. Induction of apoptosis by SB202190 through inhibition of p38β mitogen-activated protein kinase. J. Biol. Chem. 273, 16415–16420 (1998).

    CAS  PubMed  Google Scholar 

  58. Maher, P. p38 mitogen-activated protein kinase activation is required for fibroblast growth factor-2-stimulated cell proliferation but not differentiation. J. Biol. Chem. 274, 17491–17498 (1999).

    CAS  PubMed  Google Scholar 

  59. TrimGalore v.0.6.7 (GitHub, 2023) https://github.com/FelixKrueger/TrimGalore

  60. FastQC v.0.11.9 (Babraham Institute, 2019) https://www.bioinformatics.babraham.ac.uk/projects/fastqc/

  61. Smith, T., Heger, A. & Sudbery, I. UMI-tools: modeling sequencing errors in unique molecular identifiers to improve quantification accuracy. Genome Res. 27, 491–499 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  63. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 e3529 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Matyash, V., Liebisch, G., Kurzchalia, T. V., Shevchenko, A. & Schwudke, D. Lipid extraction by methyl-tert-butyl ether for high-throughput lipidomics. J. Lipid Res. 49, 1137–1146 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. ELDA: extreme limiting dilution analysis (Walter and Eliza Hall Institute of Medical Research, 2009). http://bioinf.wehi.edu.au/software/elda/

Download references

Acknowledgements

We thank the Imaging Facility of Westlake University for assistance in mini gland cleared tissue imaging. We thank the Flow Cytometry Facility of Westlake University for assistance in FACS analysis and sorting. We thank the Westlake Animal Facility for mouse husbandry. This work was supported by National Natural Science Foundation NSF grant 32170803. This work was supported by Westlake Education Foundation.

Author information

Authors and Affiliations

Authors

Contributions

L.Y. and S.C. conceived the study and experimental design, performed and analyzed experiments, and wrote the manuscript. S.X. contributed to qRT–PCR, plasmid design and lentivirus production. H.B. and X.L. contributed to single-cell RNA-seq and single-cell RNA-seq analysis. J.L. participated in microscopy analysis. P.C., C.W., S.L., Z.L. and Y.G. assisted with animal experiment.

Corresponding author

Correspondence to Shang Cai.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Cedric Blanpain, Silvia Fre and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Madhura Mukhopadhyay, in collaboration with the Nature Methods team.

Additional information

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

Extended data

Extended Data Fig. 1 Optimization of culture conditions for murine mammary gland Organoid.

a, Confocal images of immunostaining of mammary gland at E13.5, E15.5, E17.5 and P1 showing the cellular heterogeneity, polarity and architecture during mammary gland development (top). Model representation of embryonic mammary rudiment formation from E13.5 to birth (bottom). b, Immunofluorescence staining for the indicated markers of organoid. c, Mammary organoid culture for extended time. d, Representative images of organoids cultured for 20d in sphere formation media. e, Bright-field (top and middle rows) and immunofluorescence (bottom row) images of organoids cultured for 3d in the indicated medium, showing the essential factors for symmetry breaking. f, Representative images of organoids cultured for 3d in the indicated inhibitors. g, Representative images (top and middle rows) of organoids cultured for 3d in basic media with/without addition of FGF2 or the indicated inhibitors. Bottom row: Immunofluorescence staining of the frozen section of the organoids. h, Immunofluorescence staining for the indicated markers of the frozen section of the organoids. i, Quantification of Ki67+ cells in mammary organoids under various conditions. n = 3, biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. j, Motility trajectory plots of individual cells after treatment of indicated factors. X and Y represent spatial coordinates. The unit for X and Y axis is μm. k, Quantification of the speed of movement for the individual cells under indicated treatments. Each panel displays superimposed trajectories of 20 individual cells. n = 3, biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. l, Time-lapse movie of mammary organoid formation and initiation of symmetry breaking. The red dashed line indicates the formation and healing of the cavity. m, Representative images of organoids cultured in collagen I. n, Representative images of organoid outgrowths in ECM of different ratio of Matrigel and Collagen I. The scale bar represents 500 μm (black) in b, c, d, e, f, g, m, n, 50 μm in a, b, e, g, h (white), l.

Source data

Extended Data Fig. 2 Establishment of mini gland culture system.

a, Brightfield images of organoids without feeder cells for 24 days in branching media. b, Immunofluorescence staining of organoid in branching media for indicated markers at d24. c, Representative FACS plots showing mammary cell population from digested organoids without feeder cells. d, Immunofluorescence staining of organoids for indicated markers without feeder cells after pseudo estrus cycles at d20. e, Representative FACS plots showing basal and luminal cell populations from digested organoids without feeder cells after pseudo estrus cycles. f, qRT-PCR analysis of gene expression of mammary gland markers in mammary organoids, compare with primary mammary glands. n = 3, biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. g, Tracking of the size of the mini gland during one month culture. n = 10, biological replicates. Data are presented as mean ± SEM. h, Immunofluorescence staining for indicated markers from the adult primary mammary gland (left) and mammary mini gland (right). i, Comparison of the TEB widths of the mammary mini-glands and the primary mammary glands. n = 10, biological replicates. Data are presented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. j, Comparison of the duct widths of the mammary mini glands and the primary mammary glands. n = 4, biological replicates. Data are presented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. k, Bright-field images of organoids cultured for 6d in the indicated medium. l, Bright-field images of mini glands cultured in the indicated medium, showing the effect of Fgf10 and Fgf2 in mini gland culture. The scale bar represents 500 μm in a, k, l, 50 μm in b, d, h.

Source data

Extended Data Fig. 3 Growth factor requirement of mini gland culture.

a, Representative images of mini gland outgrowths in medium of indicated composition with feeder cells and pseudo estrus cycle. b, Sizes of mini glands tracked from d0 to d14 after seeding in different culture conditions with feeder cells and pseudo estrus cycle. n = 3 biological replicates. c, Representative images of mini gland outgrowths in medium without serum of indicated composition with feeder cells and pseudo estrus cycle. Data are presented as mean ± SEM. The scale bar represents 500 μm in a, c.

Source data

Extended Data Fig. 4 Effect of Wnt signaling in mini gland morphogenesis.

a, Schematic diagram showing the introduction of pseudo-estrus cycles into the mammary culture system. b, Representative brightfield images and FACS plots showing mammary cell population from digested organoids without feeder cells. c, Schematic diagram showing the introduction of exogenous Wnt3a (200 ng/ml) and pseudo-estrus cycles into the mammary culture system. d, Representative brightfield images and FACS plots showing mammary cell population from digested organoids without feeder cells but with exogenous Wnt3a. e, Schematic diagram showing the introduction of stromal cells and pseudo-estrus cycles into the mammary culture system. f, Representative brightfield images and FACS plots showing basal and luminal cell populations from digested mini gland. g, Schematic diagram showing the introduction of exogenous Dkk1 (200 ng/ml), stromal cells and pseudo-estrus cycles into the mammary culture system. h, Representative brightfield images and FACS plots showing basal and luminal cell populations from digested mini gland with exogenous Dkk1. i, Comparison of the size of the mammary mini glands under indicated treatments. n = 4, biological replicates. Data are presented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. j, Quantification of the percentage of Sca1/+ and Cd61-/+ cells in luminal cells. n = 4, biological replicates. Data are represented as mean ± SEM. The scale bar represents 500 μm in b, d, f, h.

Source data

Extended Data Fig. 5 Molecular identity of mammary cells in mini gland in vitro.

a, Expression of Cd61 and Sca1 in luminal cells of the mini gland. b, Immunofluorescence staining of mini gland for indicated markers at d26. c, Brightfield images and representative FACS plots showing basal and luminal cell populations from dissociated mini gland. d, qRT-PCR analysis of gene expression of mammary gland markers in the basal-like and luminal-like cells of the mini gland. n = 3 biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. e, qRT-PCR analysis of gene expression of Sca1- and Sca1+ cell populations from luminal cells in mini gland, compared with primary mammary gland. n = 3, biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. The scale bar represents 500 μm in c, 50 μm in b.

Source data

Extended Data Fig. 6 Characterization of transcriptome of mini gland.

a, UMAP plots comparing the expression of cell identity markers in primary mammary gland epithelial cells and the mini gland. b, UMAP plot comparing the expression of cell identity markers in two basal subpopulations.

Extended Data Fig. 7 Capacity of mini gland undergoing estrus cycle.

a, Multiple pseudo-estrus cycles for mini gland culture. Dashed lines indicate that side branches forming at the primary ducts after estrus cycle. b, Representative images of mini gland outgrowth with or without FGF2 during pseudo estrus cycle. c, Growth curve of mini gland tracked from d0 to d14 with or without FGF2 during pseudo estrus cycle. n = 4 biological replicates. Data are presented as mean ± SEM. d, Transcriptional characterization of the pseudo estrus cycle. n = 3 biological replicates. Data are represented as mean ± SEM. The scale bar represents 500 μm in a, b.

Source data

Extended Data Fig. 8 Capacity of mini gland undergoing lactation.

a, Comparison between different lactation strategies. b, Immunofluorescence staining of E-cadherin in mini gland of lactation. White dashed lines indicate the binucleated cells. c, Lipid metabolomics of mini gland and mini gland in lactation. d, Bright-field images of mini gland morphogenesis under lactation induction and involution induction. e, H&E staining of mini gland morphogenesis under lactation induction and involution induction. f, Nile red staining of mini gland morphogenesis under lactation induction and involution induction. g, Immunofluorescence for indicated markers in mini gland under lactation induction and involution induction. h, Bright-field images of mini gland morphogenesis under two round of lactation induction and involution induction. The scale bar represents 500 μm in a, d, h, 50 μm in e, f, g, 10 μm in b.

Source data

Extended Data Fig. 9 Exploration of the dynamics of the stem cell fate transition using mini gland.

a, Bright field image of d6 organoids. b, The workflow to analyze the fate of labeled cells after 1d tracing. c, FACS dot plots of luminal cells and basal cells gated within the tdTomato positive and GFP positive populations in Krt14rtTA/TetO-Cre/mTmG organoids with 1day doxycycline induction. Data were collected 1 days after the induction. d, Confocal images of immunostaining of Krt14, Krt8 and GFP in 7d organoids with 1day doxycycline induction. Images showed clonal induction in mammary ducts (arrow). e, qRT-PCR analysis indicating that TNFa level is increased from d2 to d14. mRNA expression was normalized to β-actin; n = 3, Data are presented as mean ± SEM. f, Schematic diagram showing the workflow to study the fate transition with the treatment of TNFa. g, Bright-field image of d14 mini gland with or without the treatment of TNFa. h, Representative FACS dot plots of luminal cells and basal cells gated within the tdTomato positive and GFP positive population in Krt14rtTA/TetO-Cre/mTmG mini gland with the treatment of TNF after doxycycline induction at d6. i, FACS quantification of tdTomato positive and GFP positive basal cell (gray) and luminal cell (black) populations in Krt14rtTA/TetO-Cre/mTmG mini gland after doxycycline induction with TNF treatment; n = 3, Data are presented as mean ± SEM. j, Confocal images of immunostaining of Krt14, Krt8 and GFP in mini gland at d14 with the induction at d6 by 100 ng doxycycline per ml with or without TNF treatment. The staining showed clonal induction in mammary ducts (arrow). k, Schematic diagram showing the workflow to analyze the fate of cells in mini glands labeled at d14 with the treatment of anti-TNF blocking antibody adalimumab. l, Representative FACS dot plots of luminal cells and basal cells gated within the tdTomato positive and GFP positive populations in Krt14rtTA/TetO-Cre/mTmG mini gland after doxycycline induction at d14 and adalimumab treatment for 12 days. m, FACS quantification of tdTomato positive and GFP positive basal cells (gray) and luminal cells (black) after doxycycline induction at d14 with or without the treatment of adalimumab; n = 3, Data are presented as mean ± SEM. n, Confocal images of immunostaining of Krt14, Krt8 and GFP in 26d mini gland induced by 100 ng/ml Doxycycline at d14 with anti-TNF blocking antibody adalimumab treatment. Images showed clonal induction in mammary ducts (arrow). The scale bar represents 500 μm in a, g, 50 μm in d, j, n.

Source data

Extended Data Fig. 10 Long-term cultures and in vitro modelling breast cancer using mini gland.

a, Brightfield images of long-term culture of organoid without feeder cells. Organoid collapsed and formed many vacuolar structures (arrow). b, Percentage of well-maintained organoids over time, cultured without feeder cells. c, Representative FACS plots showing basal and luminal cell populations from 117d mini gland. d, Sphere formation from flow cytometry-sorted basal and luminal cells of the 117d mini gland. e, Brightfield images of the development of a mini gland into a branching structure. f, 293T cell lines were infected with mCherry-LSL-PyMT-GFP virus, and then with or without transient transfected Cre plasmid. g, qRT-PCR analysis of PyMT expression with or without transient transfection of Cre plasmid. n = 3, biological replicates. Data are represented as mean ± SEM. P values were calculated using two-sided unpaired Student’s t-test. h, Confocal images of immunostaining of Krt8 and GFP in d38 mini gland with a tumor-like structure and control cells with mock vector (without PyMT). i, Representative FACS dot plots, H&E staining, and confocal images of immunostaining of Krt14, Krt5, Krt8 and Ki67 in spontaneous mouse model of MMTV-PyMT. j, k, Representative H&E staining, and confocal images of immunostaining of Krt14, Krt5, Krt8 and Ki67 in d38 mini gland with a tumor-like structure. The scale bar represents 500 μm in a, d, e, 200um in f, 50 μm in h, i, j, k.

Source data

Supplementary information

Supplementary Information

The supplementary information for an example of the FACS gating strategy.

Reporting Summary

Supplementary Table 1

Summary of the experimental procedures.

Supplementary Table 2

Comparison with previous research of generation of mammary gland organoids versus our work.

Supplementary Table 3

Primers for qPCR.

Supplementary Video 1

Time-lapse movie of mammary organoid formation and initiation of symmetry breaking.

Supplementary Code

Supplementary code.

Source data

Source Data Fig. 1

Image source data, statistical source data.

Source Data Fig. 2

Image source data, statistical source data.

Source Data Fig. 3

Image source data.

Source Data Fig. 4

Image source data.

Source Data Fig. 5

Image source data, statistical source data.

Source Data Fig. 6

Image source data.

Source Data Extended Data Fig. 1

Image source data, statistical source data.

Source Data Extended Data Fig. 2

Image source data, statistical source data.

Source Data Extended Data Fig. 3

Image source data, statistical source data.

Source Data Extended Data Fig. 4

Image source data, statistical source data.

Source Data Extended Data Fig. 5

Image source data, statistical source data.

Source Data Extended Data Fig. 7

Image source data, statistical source data.

Source Data Extended Data Fig. 8

Image source data, statistical source data.

Source Data Extended Data Fig. 9

Image source data, statistical source data.

Source Data Extended Data Fig. 10

Image source data, statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yuan, L., Xie, S., Bai, H. et al. Reconstruction of dynamic mammary mini gland in vitro for normal physiology and oncogenesis. Nat Methods 20, 2021–2033 (2023). https://doi.org/10.1038/s41592-023-02039-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-02039-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