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 duct luminal epithelium controls adipocyte thermogenic programme

Abstract

Sympathetic activation during cold exposure increases adipocyte thermogenesis via the expression of mitochondrial protein uncoupling protein 1 (UCP1)1. The propensity of adipocytes to express UCP1 is under a critical influence of the adipose microenvironment and varies between sexes and among various fat depots2,3,4,5,6,7. Here we report that mammary gland ductal epithelial cells in the adipose niche regulate cold-induced adipocyte UCP1 expression in female mouse subcutaneous white adipose tissue (scWAT). Single-cell RNA sequencing shows that glandular luminal epithelium subtypes express transcripts that encode secretory factors controlling adipocyte UCP1 expression under cold conditions. We term these luminal epithelium secretory factors ‘mammokines’. Using 3D visualization of whole-tissue immunofluorescence, we reveal sympathetic nerve–ductal contact points. We show that mammary ducts activated by sympathetic nerves limit adipocyte UCP1 expression via the mammokine lipocalin 2. In vivo and ex vivo ablation of mammary duct epithelium enhance the cold-induced adipocyte thermogenic gene programme in scWAT. Since the mammary duct network extends throughout most of the scWAT in female mice, females show markedly less scWAT UCP1 expression, fat oxidation, energy expenditure and subcutaneous fat mass loss compared with male mice, implicating sex-specific roles of mammokines in adipose thermogenesis. These results reveal a role of sympathetic nerve-activated glandular epithelium in adipocyte UCP1 expression and suggest that mammary duct luminal epithelium has an important role in controlling glandular adiposity.

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: Deconstruction of mgWAT shows cold-induced remodelling of mammary duct epithelium.
Fig. 2: SNS fibres directly innervate mammary ductal epithelium.
Fig. 3: Mammary ductal cells directly inhibit adipocyte thermogenesis.
Fig. 4: LCN2 preserves mgWAT adiposity.

Similar content being viewed by others

Data availability

Source data for all figures are provided with the paper. The scRNA-seq dataset (Fig. 1) generated for this paper has been uploaded to the Gene Expression Omnibus under accession number GSE231394. The bulk RNA-sequencing dataset (Fig. 4) generated for this paper has been uploaded to the GEO under accession number GSE121098 (GSM7287577GSM7287584). Source data are provided with this paper.

References

  1. Chi, J. et al. Three-dimensional adipose tissue imaging reveals regional variation in beige fat biogenesis and PRDM16-dependent sympathetic neurite density. Cell Metab. 27, 226–236.e3 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Zhang, F. et al. An adipose tissue atlas: an image-guided identification of human-like BAT and beige depots in rodents. Cell Metab. 27, 252–262.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Rajbhandari, P. et al. IL-10 signaling remodels adipose chromatin architecture to limit thermogenesis and energy expenditure. Cell 172, 218–233.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Knights, A. J., Wu, J. & Tseng, Y.-H. The heating microenvironment: intercellular cross talk within thermogenic adipose tissue. Diabetes 69, 1599 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Li, J. et al. Neurotensin is an anti-thermogenic peptide produced by lymphatic endothelial cells. Cell Metab. 33, 1449–1465.e6 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hu, B. et al. γδ T cells and adipocyte IL-17RC control fat innervation and thermogenesis. Nature 578, 610–614 (2020).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wang, Q. et al. IL-27 signalling promotes adipocyte thermogenesis and energy expenditure. Nature 600, 314–318 (2021).

    Article  ADS  CAS  PubMed  Google Scholar 

  8. Gjorevski, N. & Nelson, C. M. Integrated morphodynamic signalling of the mammary gland. Nat. Rev. Mol. Cell Biol. 12, 581–593 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. McNally, S. & Martin, F. Molecular regulators of pubertal mammary gland development. Ann. Med. 43, 212–234 (2011).

    Article  PubMed  Google Scholar 

  10. 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 

  11. Wang, Q. A. & Scherer, P. E. Remodeling of murine mammary adipose tissue during pregnancy, lactation, and involution. J. Mammary Gland Biol. Neoplasia 24, 207–212 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Landskroner-Eiger, S., Park, J., Israel, D., Pollard, J. W. & Scherer, P. E. Morphogenesis of the developing mammary gland: stage-dependent impact of adipocytes. Dev. Biol. 344, 968–978 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang, Q. A. et al. Reversible de-differentiation of mature white adipocytes into preadipocyte-like precursors during lactation. Cell Metab. 28, 282–288.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  15. Li, C. M.-C. et al. Aging-associated alterations in mammary epithelia and stroma revealed by single-cell RNA sequencing. Cell Rep. 33, 108566 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Emont, M. P. et al. A single-cell atlas of human and mouse white adipose tissue. Nature 603, 926–933 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. The Tabula Muris Consortium. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Article  ADS  CAS  Google Scholar 

  18. The Tabula Muris Consortium. A single-cell transcriptomic atlas characterizes ageing tissues in the mouse. Nature 583, 590–595 (2020).

    Article  PubMed Central  Google Scholar 

  19. Aryal, B. et al. Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis. JCI Insight 3, e97918 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Arner, E. et al. Ceruloplasmin is a novel adipokine which is overexpressed in adipose tissue of obese subjects and in obesity-associated cancer cells. PLoS ONE 9, e80274 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  21. Chella Krishnan, K. et al. Sex-specific metabolic functions of adipose lipocalin-2. Mol. Metab. 30, 30–47 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sun, W. Y. et al. Lipocalin-2 derived from adipose tissue mediates aldosterone-induced renal injury. JCI Insight 3, e120196 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Deis, J. A. et al. Adipose lipocalin 2 overexpression protects against age-related decline in thermogenic function of adipose tissue and metabolic deterioration. Mol. Metab. 24, 18–29 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ishii, A. et al. Obesity-promoting and anti-thermogenic effects of neutrophil gelatinase-associated lipocalin in mice. Sci. Rep. 7, 15501 (2017).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  25. Wang, G. X. et al. The brown fat-enriched secreted factor Nrg4 preserves metabolic homeostasis through attenuation of hepatic lipogenesis. Nat. Med. 20, 1436–1443 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Maffei, M., Barone, I., Scabia, G. & Santini, F. The multifaceted haptoglobin in the context of adipose tissue and metabolism. Endocr. Rev. 37, 403–416 (2016).

    Article  CAS  PubMed  Google Scholar 

  27. Kumar, K. G. et al. Identification of adropin as a secreted factor linking dietary macronutrient intake with energy homeostasis and lipid metabolism. Cell Metab. 8, 468–481 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Choi, C. H. J. et al. LRG1 is an adipokine that promotes insulin sensitivity and suppresses inflammation. eLife 11, e81559 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  29. MacCannell, A., Futers, S., Moran, A., Whitehead, A. & Roberts, L. Lrg1 is a driver of brown adipose tissue dysfunction in obesity. FASEB J. https://doi.org/10.1096/fasebj.2021.35.S1.00285 (2021).

  30. Rajbhandari, P. et al. Single cell analysis reveals immune cell-adipocyte crosstalk regulating the transcription of thermogenic adipocytes. eLife 8, e49501 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Gargiulo, L. et al. A novel effect of β-adrenergic receptor on mammary branching morphogenesis and its possible implications in breast cancer. J. Mammary Gland Biol. Neoplasia 22, 43–57 (2017).

    Article  PubMed  Google Scholar 

  32. Marchetti, B. et al. β-adrenergic receptors in the rat mammary gland during pregnancy and lactation: characterization, distribution, and coupling to adenylate cyclase. Endocrinology 126, 565–574 (1990).

    Article  CAS  PubMed  Google Scholar 

  33. Nedvetsky, P. I., Kwon, S. H., Debnath, J. & Mostov, K. E. Cyclic AMP regulates formation of mammary epithelial acini in vitro. Mol. Biol. Cell 23, 2973–2981 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cao, Q., Jing, J., Cui, X., Shi, H. & Xue, B. Sympathetic nerve innervation is required for beigeing in white fat. Physiol. Rep. 7, e14031 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Jiang, H., Ding, X., Cao, Y., Wang, H. & Zeng, W. Dense intra-adipose sympathetic arborizations are essential for cold-induced beiging of mouse white adipose tissue. Cell Metab. 26, 686–692.e3 (2017).

    Article  CAS  PubMed  Google Scholar 

  36. Walker, M. R. et al. Alveolar progenitor cells in the mammary gland are dependent on the β4 integrin. Dev. Biol. 457, 13–19 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Feng, Y., Manka, D. R., Wagner, K.-U. & Khan, S. A. Estrogen receptor-α expression in the mammary epithelium is required for ductal and alveolar morphogenesis in mice. Proc. Natl Acad. Sci. USA 104, 14718–14723 (2007).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lawson, D. A., Werb, Z., Zong, Y. & Goldstein, A. S. The cleared mammary fat pad transplantation assay for mammary epithelial organogenesis. Cold Spring Harb. Protoc. 2015, pdb.prot078071 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Dompmartin, A. et al. Radio-opaque ethylcellulose-ethanol is a safe and efficient sclerosing agent for venous malformations. Eur. Radiol. 21, 2647–2656 (2011).

    Article  PubMed  Google Scholar 

  40. Zhang, W. Y., Li, Z. S. & Jin, Z. D. Endoscopic ultrasound-guided ethanol ablation therapy for tumors. World J. Gastroenterol. 19, 3397–3403 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kuang, M. et al. Ethanol ablation of hepatocellular carcinoma up to 5.0 cm by using a multipronged injection needle with high-dose strategy. Radiology 253, 552–561 (2009).

    Article  PubMed  Google Scholar 

  42. Kenyon, E. et al. Ductal tree ablation by local delivery of ethanol prevents tumor formation in an aggressive mouse model of breast cancer. Breast Cancer Res. 21, 129 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Parks, B. W. et al. Genetic control of obesity and gut microbiota composition in response to high-fat, high-sucrose diet in mice. Cell Metab. 17, 141–152 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Oakes, S. R. et al. The Ets transcription factor Elf5 specifies mammary alveolar cell fate. Genes Dev. 22, 581–586 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bauters, D., Van Hul, M. & Lijnen, H. R. Macrophage elastase (MMP-12) in expanding murine adipose tissue. Biochim. Biophys. Acta 1830, 2954–2959 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Lee, J. T. et al. Macrophage metalloelastase (MMP12) regulates adipose tissue expansion, insulin sensitivity, and expression of inducible nitric oxide synthase. Endocrinology 155, 3409–3420 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Shao, M. et al. Zfp423 maintains white adipocyte identity through suppression of the beige cell thermogenic gene program. Cell Metab. 23, 1167–1184 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gavalda-Navarro, A. et al. Lipopolysaccharide-binding protein is a negative regulator of adipose tissue browning in mice and humans. Diabetologia 59, 2208–2218 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Sun, W. et al. snRNA-seq reveals a subpopulation of adipocytes that regulates thermogenesis. Nature 587, 98–102 (2020).

    Article  ADS  CAS  PubMed  Google Scholar 

  53. Kiefer, F. W. et al. Retinaldehyde dehydrogenase 1 regulates a thermogenic program in white adipose tissue. Nat. Med. 18, 918–925 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Mina, A. I. et al. CalR: a web-based analysis tool for indirect calorimetry experiments. Cell Metab. 28, 656–666.e1 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  56. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Love, M. I., Anders, S., Kim, V. & Huber, W. RNA-seq workflow: gene-level exploratory analysis and differential expression. F1000Res 4, 1070 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://arxiv.org/abs/1802.03426 (2018).

  62. van der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    MATH  Google Scholar 

  63. Blondel, V. D., Guillaume, J.-L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. 2008, P10008 (2008).

    Article  MATH  Google Scholar 

  64. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  PubMed  Google Scholar 

  65. Moerman, T. et al. GRNBoost2 and Arboreto: efficient and scalable inference of gene regulatory networks. Bioinformatics 35, 2159–2161 (2019).

    Article  CAS  PubMed  Google Scholar 

  66. Alvarsson, A. et al. Optical clearing and 3D analysis optimized for mouse and human pancreata. Bio Protoc. 11, e4103 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Jardé, T. et al. Wnt and neuregulin1/ErbB signalling extends 3D culture of hormone responsive mammary organoids. Nat. Commun. 7, 13207 (2016).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank former P.R. laboratory members S. Sadeh and S. Hart for technical assistance with metabolic chambers and RNAscope; P. Cohen and C. H. J. Choi at The Rockefeller University for the Lrg1-KO mice. LSFM and confocal microscopy were performed at the Microscopy and Advances Bioimaging Core at ISMMS. A.A. is supported by senior postdoctoral fellowship from the Charles H. Revson Foundation (grant no. 18-25), a fellowship from Sweden–America Foundation (Ernst O. Eks fond), and a postdoctoral scholarship from the Swedish Society for Medical Research (SSMF). S.A.S. is supported by American Diabetes Association Pathway to Stop Diabetes Grant ADA no. 1-17-ACE-31, NIH (R01NS097184, OT2OD024912 and R01DK124461) and Department of Defense (W81XWH-20-1-0345, W81XWH-20-1-0156). L.G. is supported by R00HL150234. A.J.L. is supported by NIH U01 AG070959 and U54 DK120342. X.Y. is supported by NIH R01 DK117850. P.R. is supported by R00DK114571, NIDDK-supported Einstein-Sinai Diabetes Research Center (DRC) Pilot and Feasibility Award, and Diabetes Action Research and Education Foundation (DREF) grant no. 501 (PR). P.W. is a member of the Human Islet and Adenovirus Core of the Einstein-Sinai Diabetes Research Center (ES-DRC) supported by NIHP30DK020541. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.

Author information

Authors and Affiliations

Authors

Contributions

S.P., N.Z.R.S. and L.C.S. performed RNAscope, cell culture, indirect calorimetry, immunofluorescence microscopy, intraductal injections, mouse experiments, qPCR and data analysis and FACS under the supervision of P.R. L.C.S. and D.A. performed scRNA-seq data analysis under the supervision of X.Y. and P.R. A.A. performed iDISCO and data analysis under the supervision of S.A.S. S.J.D. performed intraductal injections and microscopy under the supervision of P.R. and A.K.R. K.C.K. and N.K.T. performed RNA-sequencing and LCN2-related animal experiments under the supervision of A.J.L. and K.C.K. A.C. performed organoid experiments under the supervision of C.B. I.S.A., G.D. and I.C. prepared single-cell suspensions of mgWAT SVF cells under the supervision of P.R. and X.Y. E.H. performed cellular respirometry measurements under the supervision of L.G. N.Z.R.S. and C.H.C. performed cold exposure experiments under the supervision of P.R. P.W. generated adenoviruses. P.R. conceived the project and wrote the manuscript with input from A.J.B., C.B., S.A.S, A.J.L. and X.Y.

Corresponding author

Correspondence to Prashant Rajbhandari.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks the anonymous reviewer(s) for their contribution to the peer review of this work.

Additional information

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 Cold-associated increase in cell percentages of luminal epithelium subtypes.

a) UMAP plots of integrated single cell data from this study and 8 external datasets (see Methods and Supplementary Table 1). Each point represents a single cell and are colored by dataset. b) UMAP plots of integrated single cell data from this study and 8 external datasets (see Methods). Each point represents a single cell and clusters are colored by cell type. c) Expression of known canonical markers for cell types in the SVF and mammary gland. Color corresponds to average expression level and size corresponds to percentage of cells which express the gene within the cluster. d) Differentially expressed genes between COLD (24 hr) treated mice and RT animals across all cell types. Significant DEGs (adjusted p-value < 0.05) are highlighted with the average log fold change between 4 degree and RT indicated on the y-axis. Cell types are ordered on the x-axis based on the number of significant DEGs. e) Relative fractions of cell types within each sample. Dots indicate average relative fractions across all samples. f) Aggregated UMAP plot of subclustering of luminal single cells from RT and cold-exposed mice. g) Differentially expressed genes between COLD treated mice and RT animals across Luminal-HS-AV. Select significant DEGs (adjusted p-value < 0.05) are highlighted with the average log fold change between 4 degree and RT indicated on the y-axis. Genes indicated by red arrows encode for secreted factors. h) UMAP and violin plots of normalized gene expression levels for genes of interest in luminal cells. Violin plots also show expression levels of Adrb1, Adrb2, and Adrb3 in luminal subtypes. i–k) tSNE plots of cell type clusters and normalized gene expression levels for genes of interest across multiple datasets including female mgWAT SVFs (i), male iWAT SVFs (j), and mature mgWAT adipocytes (k). Statistics for scRNA-seq data (d,g) was derived using Wilcoxon rank sum test between cold vs RT cells for cells and the adjusted p-value is based on Bonferroni correction.

Extended Data Fig. 2 SNS fibers directly innervate ductal epithelial cells.

a) Light sheet microscopy fluorescence (LSFM) images of mgWAT isolated from female mice exposed to RT or COLD for 24 h and stained with TH antibody (SNS fibers) and EPCAM antibody (ductal cells). N = 6,6. Scale bar at 200 μm. b) Confocal images of mgWAT isolated from female mice exposed to RT or COLD for 24 h and stained for EPCAM and TH antibodies. Merged staining of EPCAM and TH represent neuroductal points. Representative images of 6 mice per condition. Scale bar at 100 μm. c-j) Quantification of indicated parameters of images from (b). N = 6,5. Unpaired Student’s t Test (j). Data are represented as mean ± S.E.M.

Source data

Extended Data Fig. 3 Mammary gland ductal epithelium inhibits cold-induced adipocyte thermogenesis.

a) Representative FACS plot of CD49f and EPCAM expression in EPCAM bead selected EPCAM+ or EPCAM-ve epithelial cells from RT or cold mice. 2 mice per condition and 4 mammary fat pads per mouse. Representative data from 4 independent experiments. b) Real-time qPCR of indicated genes in beige differentiated SVFs from mgWAT treated with indicated concentration of Iso for 5 h. c) Real-time qPCR of indicated genes from beige differentiated primary mgWAT SVF (Parent) and EPCAM-ve (EPCAM-NEG) cells isolated from cold exposed mice treated with and without 10 µM isoproterenol (ISO) for 24 h ex vivo. d) Images showing cell morphology of D0-D9 beige differentiated 10T1/2 and NMuMG (0, 5, and 10%) mixture cells. Yellow enclosures show NMuMG epithelial cells surrounded by 10T1/2 cells. Scale bar at 100 μm. e) Real-time qPCR of indicated genes from beige differentiated and Iso treated 10T1/2 and 5-10% NMuMG mixture cells. f) Images showing cell morphology of D0-D8 beige differentiated 10T1/2 and NMuMG (0-10%) mixture cells. Yellow enclosures show NMuMG epithelial cells surrounded by 10T1/2 cells. Scale bar at 100 μm. g) Real-time qPCR of indicated genes from beige differentiated and Iso treated 10T1/2 and 2.5-10% NMuMG mixture cells. h–i) Real-time qPCR of indicated genes from a transwell system of beige differentiated norepinephrine (NE) treated (h) or Iso treated (APC) (i) APCs in the presence (APC:NMuMG) or absence (APC) of NMuMG. j) Average oxygen consumption rate (OCR) in APC and APC:NMuMG pretreated with (NE-APC and NE:APC:NMuMG) and without (Ctrl APC and Ctrl-APC:NMuMG) 10 μM norepinephrine (NE) followed by acute treatment with NE (10 μM), 5 μM oligomycin (Oligo), 1 μM FCCP, and 5 μM antimycin A (AA). Mean of N = 6,6. k) Fold increase in OCR from basal respiration (J) with and without acute NE injection. N = 6,6. l) Average OCR normalized to protein level (μg protein) in SVF and SVF:NMuMG with 10 μM NE, 5 μM Oligo, 1 μM FCCP, and 5 μM AA. N = 9,9. m) Real-time qPCR of indicated genes from a transwell system of beige differentiated (Ctrl 10T1/2) or Iso treated (Iso-10T1/2) 10T1/2 cells in the presence of NMuMG with (Iso-10T1/2:NMuMG) or without iso (Ctrl-10T1/2:NMuMG). Results are from three independent experiments. *, p < 0.05; **, p < 0.01, ***, p < 0.001 comparing Iso-10T1/2 and Iso-10T1/2:NMuMG. Unpaired Student’s t Test (E,G,H,I), 2way ANOVA Bonferroni posthoc test (k,l,m), *, p < 0.05, **, p < 0.01; ***, p < 0.001, ns, not significant. Data are represented as mean ± S.E.M. Cartoon models in h,i,m created using BioRender.

Source data

Extended Data Fig. 4 Maintenance of adiposity in female mice under cold exposure.

a) Images showing cell morphology of beige differentiated SVFs isolated from male and female iWATs. Scale bar at 100 μm. b) Real-time qPCR of indicated genes from the scWATs of 24 h cold exposed male and female adrenalectomized mice. N = 6,6. c–e) Individual mice data for oxygen consumption (VO2 ml/hr), and energy expenditure (EE kcal/hr) (c), respiratory exchange ratio (RER) and generalized linear model (GLM)-based regression plots of RER with total body weight (Total), lean mass (Lean) and fat mass (Fat) as co-variates (Two-way ANCOVA) showing p-value for Mass effect, Group effect, and Interaction effect (d), and total food consumed (kcal), locomotor activity (beam breaks), and total distance in cage (m) (e) of male and female mice exposed to 22 °C for 21 h and 4 °C for 24 h in Sable Promethion metabolic chambers (12 h light/dark cycle, 45 h total duration, white bar represents light cycle and grey bar represents night cycle). N = 6,6. f and g) Body weight (f) and lean mass (g) of male and female mice before (RT) and after 24 h cold exposure (COLD). N = 6,6. h) Weights of indicated fat depots in cold exposed male and female mice. N = 8,8. i and j) Gross appearance, H&E staining, and immunostaining of UCP1 of male dorsolumbar iWATs and female dorsolumbar mgWATs exposed to 24 h cold (i) and Western blot of UCP1 in iWAT from 24 h cold-exposed males and females (j). Actin was used as a loading control and UCP1 bands were quantified and normalized to Actin intensity. N = 4,4. Scale bar at 200 μm. Unpaired Student’s t Test (b,f,h) and ANCOVA (d) *, p < 0.05, **, p < 0.01; ***, p < 0.001, n.s., not significant. Data are represented as mean ± S.E.M.

Source data

Extended Data Fig. 5 Mammary duct ablation potentiates the beiging of mgWAT.

a) Whole tissue images and H&E sections of mgWATs of WT and Esr1KO cold exposed mice. Blue asterisks represent same samples at different magnifications. Scale bar at 100 μm. b and c) Cartoon showing intraductal injection into the 4th and 5th nipples of female mice (b) with adjacent cartoon showing injection of Evan’s blue or GFP expressing virus for confirmation ductal-specific injection (c). Scale bar at 100 μm d) Intraductal injection of Krt8-Dtr mice with contralateral injection of DTA or PBS and immunohistochemistry with DAPI or UCP1. White arrows indicated UCP1 signals in the stroma and yellow arrows indicated UCP1 signal in ducts. Scale bar at 50 μm e) H&E staining of mgWAT from mice intraductally injected with PBS, 70% EtOH, or 0.5% Trypsin. Yellow arrows indicate ablated ducts. Red asterixis indicate images used in Main Fig. 3j. Representative section from 7 mice. Scale bar at 50 μm. f) Immunofluorescence microscopy for UCP1 or DAPI in mice intraductally injected with PBS or EtOH. Dotted yellow outlines are ductal epithelium. Scale bar at 100 μm. g) Real-time qPCR analysis of indicated genes from mgWATs of mice intraductally injected with PBS or Trypsin. N = 9,9. Data are represented as mean ± S.E.M. Cartoon models in b,d,g created using BioRender.

Source data

Extended Data Fig. 6 LCN2 is a cold-induced mammokine involved in blocking cold-induced adipocyte UCP1 expression.

a and b) RNAScope FISH of indicated probes from mgWAT of RT or 24 h cold exposed (Cold) mice. Yellow asterisk indicate image used in Main Fig. 1f for Enho RNAScope. Scale bar at 50 μm. c) Real-time qPCR of indicated genes from mgWAT beige differentiated SVFs treated with 0-100 μm recombinant Adropin (Top) or 0-80 ng/ml recombinant LRG1 (Bottom) for 24 h. d) Real-time qPCR of indicated genes from mgWATs derived from 24 h cold-exposed WT or LRG1KO mice. N = 7,8. e) Gene correlation plots of Ucp1 and Lcn2 expression in WATs from male and female mice from the HMDP studies. f) Real-time qPCR of indicated genes from 24 h cold-exposed male and female iWATs. N = 6,6. g) tSNE plots of cell type clusters and normalized gene expression levels for Lcn2 across scRNA-seq datasets of female mgWAT SVFs and male iWAT SVFs. h) Real-time qPCR of Lcn2 in mgWAT EPCAM+ and EPCAM- cells from RT or 24 h cold-exposed mice. Inset shows violin plot of Lcn2 from scRNA-seq data. i) RNAscope FISH of indicated probes from mgWAT of 24 h cold exposed or Iso treated mice. Yellow asterisks indicate images used in Main Fig. 4b. Scale bar at 50 μm. j) Immunofluorescence microscopy for LCN2 or DAPI in mice exposed to 24 h cold or RT. Yellow outlines indicated ductal epithelium. Scale bar at 50 μm. k) The top five transcription factor regulons for luminal cell types at RT and COLD identified using SCENIC program. l) Subnetworks of transcription factor regulons of interest for Bhlhe41 in hormone sensing luminal cells (Up) and Elf5 in luminal alveolar cells (Down) derived using SCENIC. Transcription factors indicated in diamonds in the center and genes in the regulon in the surrounding circles. Genes in red were derived from the RT data, genes in blue were derived from the COLD data, and genes with both were present in the regulons in both conditions. Red arrow shows cold-induced mammokine Lcn2. m and n) Body weights (n) and scWAT weights (m) of 24 h cold exposed WT and Lcn2 KO male and female mice. N = 6,6 o) Real-time qPCR of indicated genes in organoids derived from K8rtTA-DTA mice treated with and without ISO and Doxycycline (Dox). Results from 3 organoid experiments. p-t) Confocal images of immunostaining of indicated antibodies in organoids derived from K8rtTA-DTA mice treated with and without Iso and Dox. Scale bar at 10 μm. Representative images from 3 organoid experiments. Each point represents three independent experiments (c, h, o). Unpaired two-tailed Student’s t test *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001. Data are represented as mean ± S.E.M.

Source data

Extended Data Fig. 7 LCN2 is a mammokine involved in blocking cold-induced adipocyte UCP1 expression.

a) Real-time qPCR of indicated genes from mgWAT of AAV-GFP or AAV-LCN2 treated Lcn2 KO mice. N = 4,4. b) Gross appearance and H&E section of mgWAT from RT and cold-exposed LCN2KO mice (Top) and WT or LCN2KO mice exposed to 24 h cold (Bottom). Scale bar at 100 μm. c) Real-time qPCR of indicated genes from 24 h cold exposed females (Top) and males (bottom) WT and LCN2KO scWATs. N = 6,6. d) Intraductal injection of Ad-GFP or Ad-LCN2 contralaterally in the 4th and 5th nipple and immunofluorescence microscopy of GFP or LCN2 from Ad-GFP or Ad-LCN2 injected mice with indicated viral concentrations and barplot showing intensity of GFP or LCN2 from microscopy. Scale bar at 100 μm. Yellow asterisks indicate images also used in Main Fig. 4g. Representative image of two independent injections. For bar plots, quantification of intensity was calculated as a mean from multiple ducts from 1 image except for 2X1010 PFU for Ad-GFP (N = 3) and 2X109 for Ad-LCN2. (N = 2). Unpaired two-tailed Student’s t test *, p < 0.05; **, p < 0.01; ***, p < 0.001. Data are represented as mean ± S.E.M. Cartoon model in D created using BioRender.

Source data

Supplementary information

Supplementary Fig. 1

Gel source data for Extended Data Fig. 4j.

Reporting Summary

Supplementary Fig. 2

Cell selection and FACS sequential gating strategy. Middle gated panel in page 7 corresponds to Extended Data Fig. 2a.

Supplementary Table 1

Publicly available single-cell datasets used in this study

Supplementary Table 2

Curated set of canonical marker genes used in this study.

Supplementary Table 3

Real-time qPCR primer sequences.

Supplementary Video 1

LSFM video of mgWAT isolated from female mice exposed to RT for 24 h and stained with TH antibody (SNS fibres) and EPCAM antibody (ductal cells). Representative mgWAT video for 6 mice per condition.

Supplementary Video 2

LSFM video of mgWAT isolated from female mice exposed to cold for 24 h and stained with TH antibody (SNS fibres) and EPCAM antibody (ductal cells). Representative mgWAT video for 6 mice per condition.

Supplementary Video 3

Confocal video of mgWAT isolated from female mice exposed to RT for 24 h and stained for EPCAM and TH antibodies. Merged staining of EPCAM and TH represent neuroductal points. Representative video for 6 mice per condition

Supplementary Video 4

Confocal video of mgWAT isolated from female mice exposed to cold (24 h) and stained for EPCAM and TH antibodies. Merged staining of EPCAM and TH represent neuroductal points. Representative video for 6 mice per condition

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

Patel, S., Sparman, N.Z.R., Arneson, D. et al. Mammary duct luminal epithelium controls adipocyte thermogenic programme. Nature 620, 192–199 (2023). https://doi.org/10.1038/s41586-023-06361-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-06361-5

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