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:

Mammary epithelial cells have lineage-rooted metabolic identities

Abstract

Cancer metabolism adapts the metabolic network of its tissue of origin. However, breast cancer is not a disease of a single origin. Multiple epithelial populations serve as the culprit cell of origin for specific breast cancer subtypes, yet our knowledge of the metabolic network of normal mammary epithelial cells is limited. Using a multi-omic approach, here we identify the diverse metabolic programmes operating in normal mammary populations. The proteomes of basal, luminal progenitor and mature luminal cell populations revealed enrichment of glycolysis in basal cells and of oxidative phosphorylation in luminal progenitors. Single-cell transcriptomes corroborated lineage-specific metabolic identities and additional intra-lineage heterogeneity. Mitochondrial form and function differed across lineages, with clonogenicity correlating with mitochondrial activity. Targeting oxidative phosphorylation and glycolysis with inhibitors exposed lineage-rooted metabolic vulnerabilities of mammary progenitors. Bioinformatics indicated breast cancer subtypes retain metabolic features of their putative cell of origin. Thus, lineage-rooted metabolic identities of normal mammary cells may underlie breast cancer metabolic heterogeneity and targeting these vulnerabilities could advance breast cancer therapy.

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: Proteomics illustrates distinct metabolic networks of human MECs.
Fig. 2: Human breast single-cell transcriptomics reveals epithelial cell-state-specific metabolic networks.
Fig. 3: Mitochondrial structure and function vary with mammary lineage.
Fig. 4: Enhanced mitochondrial respiration is characteristic of MEC progenitors.
Fig. 5: Metabolic inhibitors expose lineage-restricted vulnerabilities of MECs.
Fig. 6: Breast cancer subtypes retain metabolic features of specific primary MECs.

Similar content being viewed by others

Data availability

MULTI-seq data is available from the NCBI Gene Expression Omnibus under accession number GSE168660. The human mammary proteome is available for download from ftp://massive.ucsd.edu/MSV000087042/ (MassiVE identifier: MSV000087042). The mouse mammary proteome data (Extended Data Fig. 4a,b) are published25 and can be downloaded from ftp://massive.ucsd.edu/MSV000079330/ (MassiVE identifier: MSV000079330). Source data are provided with this paper.

Code availability

All proteomic codes and MULTI-seq codes are available at https://github.com/mcclo/Mahendralingam-et-al.-Nat-Metab/.

References

  1. Perou, C. M. et al. Molecular portraits of human breast tumours. Nature 406, 747–752 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Prat, A. et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 12, R68 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Sørlie, T. et al. Gene expression patterns of breast carcinomas distinguish tumour subclasses with clinical implications. Proc. Natl Acad. Sci. USA 98, 10869–10874 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Brauer, H. A. et al. Impact of tumor microenvironment and epithelial phenotypes on metabolism in breast cancer. Clin. Cancer Res. 19, 571–585 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Budczies, J. et al. Comparative metabolomics of estrogen receptor positive and estrogen receptor negative breast cancer: alterations in glutamine and beta-alanine metabolism. J. Proteomics 94, 279–288 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Cappelletti, V. et al. Metabolic footprints and molecular subtypes in breast cancer. Dis. Markers 2017, 1–19 (2017).

    Article  Google Scholar 

  7. Tang, X. et al. A joint analysis of metabolomics and genetics of breast cancer. Breast Cancer Res. 16, 415 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Terunuma, A. et al. MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J. Clin. Invest. 124, 398–412 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Kulkoyluoglu-Cotul, E., Arca, A. & Madak-Erdogan, Z. Crosstalk between estrogen signaling and breast cancer metabolism. Trends Endocrinol. Metab. 30, 25–38 (2019).

    Article  CAS  PubMed  Google Scholar 

  10. Zhang, D. et al. Proteomic study reveals that proteins involved in metabolic and detoxification pathways are highly expressed in HER-2/neu-positive breast cancer. Mol. Cell. Proteomics 4, 1686–1696 (2005).

    Article  CAS  PubMed  Google Scholar 

  11. DeBerardinis, R. J. & Chandel, N. S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Mayers, J. R. & Vander Heiden, M. G. Nature and nurture: what determines tumor metabolic phenotypes? Cancer Res. 77, 3131–3134 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Yuneva, M. O. et al. The metabolic profile of tumors depends on both the responsible genetic lesion and tissue type. Cell Metab. 15, 157–170 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hu, J. et al. Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol. 31, 522–529 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mayers, J. R. et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 353, 1161–1165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Oakes, S. R., Gallego-Ortega, D. & Ormandy, C. J. The mammary cellular hierarchy and breast cancer. Cell. Mol. Life Sci. 71, 4301–4324 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Tharmapalan, P., Mahendralingam, M., Berman, H. K. & Khokha, R. Mammary stem cells and progenitors: targeting the roots of breast cancer for prevention. EMBO J. 38, e100852 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Visvader, J. E. & Stingl, J. Mammary stem cells and the differentiation hierarchy: current status and perspectives. Genes Dev. 28, 1143–1158 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Inman, J. L., Robertson, C., Mott, J. D. & Bissell, M. J. Mammary gland development: cell fate specification, stem cells and the microenvironment. Development 142, 1028–1042 (2015).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lim, E. et al. Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers. Nat. Med. 15, 907–913 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Koren, S. et al. PIK3CAH1047R induces multipotency and multi-lineage mammary tumours. Nature 525, 114–118 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Molyneux, G. et al. BRCA1 basal-like breast cancers originate from luminal epithelial progenitors and not from basal stem cells. Cell Stem Cell 7, 403–417 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  25. Casey, A. E. et al. Mammary molecular portraits reveal lineage-specific features and progenitor cell vulnerabilities. J. Cell Biol. 217, 2951–2974 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Pellacani, D. et al. Analysis of normal human mammary epigenomes reveals cell-specific active enhancer states and associated transcription factor networks. Cell Rep. 17, 2060–2074 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Shiah, Y.-J. et al. A progesterone–CXCR4 axis controls mammary progenitor cell fate in the adult gland. Stem Cell Rep. 4, 313–322 (2015).

    Article  CAS  Google Scholar 

  28. Shehata, M. et al. Identifying the murine mammary cell target of metformin exposure. Commun. Biol. 2, 192 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  29. 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 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kannan, N. et al. Glutathione-dependent and -independent oxidative stress-control mechanisms distinguish normal human mammary epithelial cell subsets. Proc. Natl Acad. Sci. USA 111, 7789–7794 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Possemato, R. et al. Functional genomics reveal that the serine synthesis pathway is essential in breast cancer. Nature 476, 346–350 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Losman, J.-A. & Kaelin, W. G. What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer. Genes Dev. 27, 836–852 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Lin, K. H. et al. Systematic dissection of the metabolic–apoptotic interface in AML reveals heme biosynthesis to be a regulator of drug sensitivity. Cell Metab. 29, 1217–1231 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. McGinnis, C. S. et al. MULTI-seq: sample multiplexing for single-cell RNA sequencing using lipid-tagged indices. Nat. Methods 16, 619–626 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nguyen, Q. H. et al. Profiling human breast epithelial cells using single-cell RNA sequencing identifies cell diversity. Nat. Commun. 9, 2028 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Ehmsen, S. et al. Increased cholesterol biosynthesis is a key characteristic of breast cancer stem cells influencing patient outcome. Cell Rep. 27, 3927–3938 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Calvo, S. E., Clauser, K. R. & Mootha, V. K. MitoCarta2.0: an updated inventory of mammalian mitochondrial proteins. Nucleic Acids Res. 44, D1251–D1257 (2016).

    Article  CAS  PubMed  Google Scholar 

  38. Pham, A. H., McCaffery, J. M. & Chan, D. C. Mouse lines with photo-activatable mitochondria to study mitochondrial dynamics. Genesis 50, 833–843 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Folmes, C. D. L., Dzeja, P. P., Nelson, T. J. & Terzic, A. Metabolic plasticity in stem cell homeostasis and differentiation. Cell Stem Cell 11, 596–606 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cogliati, S. et al. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155, 160–171 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Katajisto, P. et al. Asymmetric apportioning of aged mitochondria between daughter cells is required for stemness. Science 348, 340–343 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wu, M.-J. et al. Epithelial–mesenchymal transition directs stem cell polarity via regulation of mitofusin. Cell Metab. https://doi.org/10.1016/j.cmet.2018.11.004 (2018).

  43. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Lindley, L. E. et al. The WNT-controlled transcriptional regulator LBH is required for mammary stem cell expansion and maintenance of the basal lineage. Development 142, 893–904 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Joshi, P. A. et al. RANK signaling amplifies WNT-responsive mammary progenitors through R-SPONDIN1. Stem Cell Rep. 5, 31–44 (2015).

    Article  CAS  Google Scholar 

  46. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sullivan, L. B. et al. Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells. Cell 162, 552–563 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Liu, X., Romero, I. L., Litchfield, L. M., Lengyel, E. & Locasale, J. W. Metformin targets central carbon metabolism and reveals mitochondrial requirements in human cancers. Cell Metab. 24, 728–739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Garcia-Bermudez, J. et al. Aspartate is a limiting metabolite for cancer cell proliferation under hypoxia and in tumours. Nat. Cell Biol. 20, 775–781 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Curtis, C. et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chowdhry, S. et al. NAD metabolic dependency in cancer is shaped by gene amplification and enhancer remodelling. Nature https://doi.org/10.1038/s41586-019-1150-2 (2019).

  52. Tang, Y.-C. et al. Aneuploid cell survival relies upon sphingolipid homeostasis. Cancer Res. 77, 5272–5286 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Locasale, J. W. et al. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat. Genet. 43, 869–874 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Russnes, H. G. et al. Genomic architecture characterizes tumor progression paths and fate in breast cancer patients. Sci. Transl. Med. 2, 38ra47 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Chakrabarti, R. et al. Notch ligand Dll1 mediates cross-talk between mammary stem cells and the macrophageal niche. Science https://doi.org/10.1126/science.aan4153 (2018).

  56. Joshi, P. A. et al. PDGFRα+ stromal adipocyte progenitors transition into epithelial cells during lobulo-alveologenesis in the murine mammary gland. Nat. Commun. 10, 1760 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Morsing, M. et al. Evidence of two distinct functionally specialized fibroblast lineages in breast stroma. Breast Cancer Res. 18, 108 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the Intersections between metabolism and cancer biology. Cell 168, 657–669 (2017).

    Article  PubMed Central  Google Scholar 

  59. Kim, J. & DeBerardinis, R. J. Mechanisms and implications of metabolic heterogeneity in cancer. Cell Metab. 30, 434–446 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Gong, Y. et al. Metabolic-pathway-based subtyping of triple-negative breast cancer reveals potential therapeutic targets. Cell Metab. 33, 51–64 (2021).

    Article  CAS  PubMed  Google Scholar 

  61. Ramakrishnan, R., Khan, S. A. & Badve, S. Morphological changes in breast tissue with menstrual cycle. Mod. Pathol. 15, 1348–1356 (2002).

    Article  PubMed  Google Scholar 

  62. Labarge, M. A., Garbe, J. C. & Stampfer, M. R. Processing of human reduction mammoplasty and mastectomy tissues for cell culture. J. Vis. Exp. https://doi.org/10.3791/50011 (2013).

  63. Eirew, P., Stingl, J. & Eaves, C. J. Quantitation of human mammary epithelial stem cells with in vivo regenerative properties using a subrenal capsule xenotransplantation assay. Nat. Protoc. 5, 1945–1956 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Kannan, N. et al. The luminal progenitor compartment of the normal human mammary gland constitutes a unique site of telomere dysfunction. Stem Cell Rep. 1, 28–37 (2013).

    Article  CAS  Google Scholar 

  65. Joshi, P. A. et al. Progesterone induces adult mammary stem cell expansion. Nature 465, 803–807 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  67. Wojtowicz, E. E. et al. Ectopic miR-125a expression induces long-term repopulating stem cell capacity in mouse and human hematopoietic progenitors. Cell Stem Cell 19, 383–396 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Bunn, A. & Korpela, M. An introduction to dplR. https://cran.r-project.org/web/packages/dplR/vignettes/intro-dplR.pdf (2021).

  69. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  PubMed  Google Scholar 

  70. Leek, J. T., Johnson, W. E., Parker, H. S., Jaffe, A. E. & Storey, J. D. Surrogate variable analysis R package version 3.10.0. http://bioconductor.org/packages/sva/ (2019).

  71. Chen, H. & Boutros, P. C. VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R. BMC Bioinformatics 12, 35 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Kuleshov, M. V. et al. Enrichr: a comprehensive gene-set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Vu, V. A biplot based on ggplot2. GitHub. https://github.com/vqv/ggbiplot/ (2019).

  75. Maechler, M., Rousseeux, P., Struyf, A., Hubert, M. & Hornik, K. cluster: Cluster analysis basics and extensions. R package version 2.0.7-1. https://CRAN.R-project.org/package=cluster (2021).

  76. Kolde, R. pheatmap: Pretty Heatmaps. https://rdrr.io/cran/pheatmap/ (2019).

  77. Neuwirth, E. RColorBrewer: ColorBrewer Palettes. https://cran.r-project.org/web/packages/RColorBrewer/index.html (2014).

  78. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Stuart, T. et al. Comprehensive Integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Lütge, A. CellMixS: evaluate cell-specific mMixing. Bioconductor version 3.12. https://doi.org/10.18129/B9.bioc.CellMixS (2021).

  82. Zhang, X. et al. CellMarker: a manually curated resource of cell markers in human and mouse. Nucleic Acids Res. 47, D721–D728 (2019).

    Article  CAS  PubMed  Google Scholar 

  83. Cao, Y., Wang, X. & Peng, G. SCSA: a cell type annotation tool for single-cell RNA-seq data. Front. Genet. 11, 490 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Zhang, A. W. et al. Probabilistic cell-type assignment of single-cell RNA-seq for tumor microenvironment profiling. Nat. Methods 16, 1007–1015 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Alquicira, J. & Powell, J. E. Nebulosa recovers single-cell gene expression signals by kernel density estimation. Bioinformatics https://doi.org/10.1093/bioinformatics/btab003 (2021).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ibrahim, M. M. & Kramann, R. genesorteR: feature ranking in clustered single-cell data. Preprint at bioRxiv https://doi.org/10.1101/676379 (2019).

  89. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Filby, A. et al. An imaging flow cytometric method for measuring cell division history and molecular symmetry during mitosis. Cytometry A 79, 496–506 (2011).

    Article  PubMed  Google Scholar 

  91. Ortyn, W. E. et al. Sensitivity measurement and compensation in spectral imaging. Cytometry A 69, 852–862 (2006).

    Article  PubMed  Google Scholar 

  92. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

  93. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Hänzelmann, S., Castelo, R. & Guinney, J. GSVA: gene-set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Crowell, H. L. et al. muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data. Nat. Commun. 11, 6077 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to the Nanoscale Biomedical Imaging Facility at SickKids and the Princess Margaret and SickKids flow cytometry core facilities. We thank Z. Gartner for generously providing MULTI-seq reagents. This work is supported by funding from Canadian Institutes of Health Research (CIHR), Canadian Breast Cancer Foundation (CBCF) and Canadian Cancer Society Research Institute (CCSRI) grants to the laboratories of R.K. and T.K. M.A.P. is supported by Carlos III National Health Institute funded by FEDER funds—a way to build Europe (PI18/01029) and the Government of Catalonia (CERCA programme and 2017SGR449). M.J.M. received a CIHR Masters award, P.T. received a CBCF/CCS Doctoral Fellowship, and C.W.M. received a CIHR Banting Postdoctoral Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

M.J.M., A.E.C., H.K., C.W.M., T.K. and R.K. conceived the study. D.P., J.C., C.J.E. and H.K.B. provided participant samples, and A.D.S., R.N.V. and M.K. provided technical resources. M.J.M., H.K., C.W.M., A.E.C., A.S., V.S., P.T. and R.S. performed experiments, biological analyses and data interpretation. C.W.M. performed MULTI-seq analyses. A.E.C., A.S. and T.K. performed mass spectrometry. K.A., L.P., V.I., M.G.-V., C.W.M. and M.A.P. performed bioinformatics, and H.K. performed Amnis imaging flow cytometry. M.J.M., H.K., P.T., C.W.M., T.K. and R.K. wrote the paper. T.K. and R.K. supervised the study.

Corresponding authors

Correspondence to Thomas Kislinger or Rama Khokha.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editors: Elena Bellafante; George Caputa. Nature Metabolism thanks Ralph DeBerardinis, Fernando Martin Belmonte and Senthil Muthuswamy 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

Extended Data Fig. 1 Characterization of proteomic datasets of primary FACS-purified human mammary epithelial cells.

a, Gating strategy for FACS-purifying human mammary epithelial cells (MECs). Total cells from dissected human breast tissue are gated to exclude debris. Doublet, dead cell and Lineage (Lin+) exclusion ensures sorting of single, live and nonimmune cells. b, Venn diagram summarizing the distribution of the total 6040 detected proteins across human MEC proteomes. The numbers in brackets are the total number of proteins detected in that cell type. Uniquely detected genes are highlighted for each lineage. c, Heatmap shows unsupervised hierarchical clustering and z-scores for protein abundance of a set of known marker proteins well established for distinguishing mammary epithelial cell types. d, Distribution of protein abundance detected within each MEC lineage. Each point represents a protein and is colour-coded by the decile its median density is found. e, Pathway analysis using Enrichr was performed on each decile for each MEC type. The top 2 GO Biological Processes per decile are presented with associated adjusted p-values (Fisher’s Exact test) in brackets.

Source data

Extended Data Fig. 2 Map of human mammary epithelial cell metabolism.

Metabolic map is adapted from a previously published template33. Proteins are color-coded to denote which mammary cell-type specific metabolic network exhibited their significantly increased abundance (Black = not significant or not detected; Light blue = luminal progenitors; Dark blue = mature luminal; Red = basal).

Extended Data Fig. 3 Age correlation with human proteome lineage signatures and expression of lineage-specific metabolic genes in scRNA-seq.

a, Linear regression of the mean metabolic protein abundance across age within each sample for the basal cell (BC), luminal progenitor (LP), and mature luminal (ML) metabolic signatures within their respective cell types. Linear equation (y), correlation coefficient (r2), and p-value are presented. Hormone status of each sample is represented by shapes indicated in legend. Error band represents 95% confidence interval and p-value was calculated by two-sided t-test. b, Violin plots show differentially expressed genes from Fig. 1f in basal, mature luminal, and luminal progenitor cells from our MULTI-seq dataset. Log10 expression values are presented with mean indicated by black lines. All groups are significantly different by one-way ANOVA followed by multiple pairwise comparisons, **p < 0.05, ***p < 0.01.

Extended Data Fig. 4 Characterization of human and mouse mammary mitochondria.

a, Heatmap showing unsupervised hierarchical clustering and z-scores of only metabolic proteins, determined by a curated list31, in our previously published mouse mammary proteomic dataset25. b, Heatmap showing unsupervised hierarchical clustering and z-scores of mitochondrial proteins present in the Mouse MitoCarta2.0 database in our previously published mouse mammary proteomic dataset25. c, Heatmap showing unsupervised hierarchical clustering and z-scores of only mitochondrial proteins present in the Human MitoCarta2.0 database in human mammary proteomes. d, Volcano plot highlighting the top 5 differentially expressed mitochondrial proteins in premenopausal compared to post-menopausal human samples within each of the basal (red), luminal progenitor (light blue), and mature luminal (blue) lineages. Significance is presented as log10(p-value) derived by two-sided t-test. e, Volcano plot highlighting the top 5 differentially expressed mitochondrial proteins in estrogentreated compared to estrogen + progesterone-treated murine mammary proteomes within each of the basal (red), luminal progenitor (light blue), and mature luminal (blue) lineages. Significance is presented as log10(p-value) derived by two-sided t-test.

Extended Data Fig. 5 Gating strategies of conventional flow and imaging flow cytometry.

a, Conventional flow gating strategy for mouse mammary epithelial cells. Total cells from dissected mouse mammary gland are gated to exclude debris, doublets, dead cells and Lineage (Lin + ) to yield single, live, total mammary epithelial cells. b, General workflow of Amnis imaging flow cytometry. c, Gating strategy for image processing using the IDEAS™ software. Pre-defined features including Gradient RMS, Aspect Ratio, Area, and Raw Centroid X based on brightfield (CH01) images were used to gate focused, nonclipped, single cell images. Saturated mito-Denda2 events (based on CH02) were removed. d, The resulting live (Zombie UV–), Lin– (lineage (CD45 + CD31 + Ter119 + )-depleted) population was separated based on EpCAM (CH12), CD49f (CH06), CD49b (CH11), and Sca-1 (CH04) cell surface markers to acquire basal, luminal progenitor (LP), mature (ML), and stromal populations. Finally, distribution of mito-Dendra2 intensity from the 4 populations was plotted. e, Histogram illustrating proportional cell distribution across 4 foci bins for each population (basal: B; luminal progenitor: LP; mature luminal: ML). Each dot represents a biological replicate (n = 4 mice), and data are presented as mean ± SD.

Extended Data Fig. 6 CFC enumeration with metabolic inhibitors and exogenous metabolite rescue.

a, b, Quantification of absolute CFC counts at various concentrations of the specified a) OXPHOS inhibitor or b) glycolysis inhibitor Basal colonies are red and luminal colonies are blue. Each dot represents a mouse and number of biological replicates per drug is shown in brackets. P-values were calculated using two-way ANOVA and Sidak’s multiple comparisons test. Data are mean ± SEM. * P ≤ 0.05; ** P ≤ 0.01; *** P ≤ 0.001; ****P ≤ 0.0001. c, Boxplots of relative count of basal (red) and luminal (blue) colonies after addition of exogenous metabolites (FCCP, pyruvate, aspartate or α-ketobutyrate) following rotenone treatment (filled). Relative values were determined by normalization with matching Rotenone-negative, vehicle control basal or luminal colony (n = 4 mice). The centre line represents the median, box limits are the first and third quartiles, whiskers extend to 1.5×interquartile range and the points beyond the whiskers are outliers.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mahendralingam, M.J., Kim, H., McCloskey, C.W. et al. Mammary epithelial cells have lineage-rooted metabolic identities. Nat Metab 3, 665–681 (2021). https://doi.org/10.1038/s42255-021-00388-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00388-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer