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:

Sex differences orchestrated by androgens at single-cell resolution

Abstract

Sex differences in mammalian complex traits are prevalent and are intimately associated with androgens1,2,3,4,5,6,7. However, a molecular and cellular profile of sex differences and their modulation by androgens is still lacking. Here we constructed a high-dimensional single-cell transcriptomic atlas comprising over 2.3 million cells from 17 tissues in Mus musculus and explored the effects of sex and androgens on the molecular programs and cellular populations. In particular, we found that sex-biased immune gene expression and immune cell populations, such as group 2 innate lymphoid cells, were modulated by androgens. Integration with the UK Biobank dataset revealed potential cellular targets and risk gene enrichment in antigen presentation for sex-biased diseases. This study lays the groundwork for understanding the sex differences orchestrated by androgens and provides important evidence for targeting the androgen pathway as a broad therapeutic strategy for sex-biased diseases.

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: A single-cell transcriptomic atlas of sex differences and androgen effects.
Fig. 2: Molecular characterization of sex differences and androgen effects.
Fig. 3: Identification of AASB gene expression.
Fig. 4: Androgens modulate sex-biased immune compartments.
Fig. 5: Cell-type and risk gene enrichment for sex-biased diseases.

Similar content being viewed by others

Data availability

Customizable visualizations and analyses can be executed in our web tool (https://casadbtools.com). The raw data for scRNA-seq have been deposited in the GSA (https://ngdc.cncb.ac.cn/gsa/) under CRA006610. The processed data for scRNA-seq have been deposited in the OMIX under OMIX001083. The mm10 genome reference was obtained from: https://ftp.ensembl.org/pub/release-93/fasta/mus_musculus/dna/Mus_musculus.GRCm38.dna.primary_assembly.fa.gzSource data are provided with this paper.

Code availability

The codes used for analysing and visualizing the data in this study are available in the GitHub repository (https://github.com/lifei176/Single-cell-atlas-of-sex-differences-and-androgen-effects)45 and in Zenodo (https://doi.org/10.5281/zenodo.10784644)46.

References

  1. Khramtsova, E. A., Davis, L. K. & Stranger, B. E. The role of sex in the genomics of human complex traits. Nat. Rev. Genet. 20, 173–190 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Oliva, M. et al. The impact of sex on gene expression across human tissues. Science 369, eaba3066 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lopes-Ramos, C. M. et al. Sex differences in gene expression and regulatory networks across 29 human tissues. Cell Rep. 31, 107795 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Naqvi, S. et al. Conservation, acquisition, and functional impact of sex-biased gene expression in mammals. Science 365, eaaw7317 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gershoni, M. & Pietrokovski, S. The landscape of sex-differential transcriptome and its consequent selection in human adults. BMC Biol. 15, 7 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Yang, X. et al. Tissue-specific expression and regulation of sexually dimorphic genes in mice. Genome Res. 16, 995–1004 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Mauvais-Jarvis, F. et al. Sex and gender: modifiers of health, disease, and medicine. Lancet 396, 565–582 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Watson, P. A., Arora, V. K. & Sawyers, C. L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat. Rev. Cancer 15, 701–711 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Beer, T. M. et al. Enzalutamide in metastatic prostate cancer before chemotherapy. N. Engl. J. Med. 371, 424–433 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Schmidt, K. T., Huitema, A. D. R., Chau, C. H. & Figg, W. D. Resistance to second-generation androgen receptor antagonists in prostate cancer. Nat. Rev. Urol. 18, 209–226 (2021).

    Article  CAS  PubMed  Google Scholar 

  11. Vellano, C. P. et al. Androgen receptor blockade promotes response to BRAF/MEK-targeted therapy. Nature 606, 797–803 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  12. Yang, C. et al. Androgen receptor-mediated CD8+ T cell stemness programs drive sex differences in antitumor immunity. Immunity 55, 1–16 (2022).

    Article  CAS  Google Scholar 

  13. De Gendt, K. et al. A Sertoli cell-selective knockout of the androgen receptor causes spermatogenic arrest in meiosis. Proc. Natl Acad. Sci. USA 101, 1327–1332 (2004).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  14. Pihlajamaa, P. et al. Tissue-specific pioneer factors associate with androgen receptor cistromes and transcription programs. EMBO J. 33, 312–326 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hartig, S. M. et al. Feed-forward inhibition of androgen receptor activity by glucocorticoid action in human adipocytes. Chem. Biol. 19, 1126–1141 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Freshour, J. R., Chase, S. E. & Vikstrom, K. L. Gender differences in cardiac ACE expression are normalized in androgen-deprived male mice. Am. J. Physiol. Heart Circ. Physiol. 283, H1997–H2003 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Chuang, K. H. et al. Neutropenia with impaired host defense against microbial infection in mice lacking androgen receptor. J. Exp. Med. 206, 1181–1199 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Spits, H. & Mjosberg, J. Heterogeneity of type 2 innate lymphoid cells. Nat. Rev. Immunol. 22, 701–712 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Jacquelot, N., Seillet, C., Vivier, E. & Belz, G. T. Innate lymphoid cells and cancer. Nat. Immunol. 23, 371–379 (2022).

    Article  CAS  PubMed  Google Scholar 

  20. Ebbo, M., Crinier, A., Vely, F. & Vivier, E. Innate lymphoid cells: major players in inflammatory diseases. Nat. Rev. Immunol. 17, 665–678 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Buonocore, S. et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 464, 1371–1375 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Neill, D. R. et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464, 1367–1370 (2010).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Cephus, J. Y. et al. Testosterone attenuates group 2 innate lymphoid cell-mediated airway inflammation. Cell Rep. 21, 2487–2499 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Laffont, S. et al. Androgen signaling negatively controls group 2 innate lymphoid cells. J. Exp. Med. 214, 1581–1592 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Blanquart, E. et al. Targeting androgen signaling in ILC2s protects from IL-33-driven lung inflammation, independently of KLRG1. J. Allergy Clin. Immunol. 149, 237–251.e12 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Backman, J. D. et al. Exome sequencing and analysis of 454,787 UK Biobank participants. Nature 599, 628–634 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  28. Costa, A. R. et al. The sex bias of cancer. Trends Endocrinol. Metab. 31, 785–799 (2020).

    Article  CAS  PubMed  Google Scholar 

  29. Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Natri, H., Garcia, A. R., Buetow, K. H., Trumble, B. C. & Wilson, M. A. The pregnancy pickle: evolved immune compensation due to pregnancy underlies sex differences in human diseases. Trends Genet. 35, 478–488 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Bal, S. M., Golebski, K. & Spits, H. Plasticity of innate lymphoid cell subsets. Nat. Rev. Immunol. 20, 552–565 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Alyamani, M. et al. Deep androgen receptor suppression in prostate cancer exploits sexually dimorphic renal expression for systemic glucocorticoid exposure. Ann. Oncol. 31, 369–376 (2020).

    Article  CAS  PubMed  Google Scholar 

  33. Li, J. et al. Aberrant corticosteroid metabolism in tumor cells enables GR takeover in enzalutamide resistant prostate cancer. eLife 6, e20183 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Seale, J. V., Wood, S. A., Atkinson, H. C., Harbuz, M. S. & Lightman, S. L. Gonadal steroid replacement reverses gonadectomy-induced changes in the corticosterone pulse profile and stress-induced hypothalamic–pituitary–adrenal axis activity of male and female rats. J. Neuroendocrinol. 16, 989–998 (2004).

    Article  CAS  PubMed  Google Scholar 

  35. Li, F. et al. Distinct mechanisms for TMPRSS2 expression explain organ-specific inhibition of SARS-CoV-2 infection by enzalutamide. Nat. Commun. 12, 866 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lun, A. T. L. et al. EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol. 20, 63 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Zappia, L. & Oshlack, A. Clustering trees: a visualization for evaluating clusterings at multiple resolutions. Gigascience 7, giy083 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Miller, S. A. et al. LSD1 and aberrant DNA methylation mediate persistence of enteroendocrine progenitors that support BRAF-mutant colorectal cancer. Cancer Res. 81, 3791–3805 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. 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 

  42. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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 

  44. Wang, K., Li, M. & Hakonarson, H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Li, F. Single cell atlas of sex differences and androgen effects. GitHub https://github.com/lifei176/Single-cell-atlas-of-sex-differences-and-androgen-effects (2024).

  46. Li, F. Single cell atlas of sex differences and androgen effects. Zenodo https://doi.org/10.5281/zenodo.10784644 (2024).

Download references

Acknowledgements

We thank B. Wu, G. Chen and W. Bian for providing technical help at the CEMCS (SIBCB) Core Facility, the Genome Tagging Project Center at the CEMCS for technical support, and H. Dong for the schematic drawing. This study was supported by the National Key Research and Development Program of China (no. 2023YFC2506401, 2020YFA0509002, 2022YFC2504602 and 2022YFA1004800), the National Science Fund for Distinguished Young Scholars (32125013 and T2125002), the National Natural Science Foundation of China (92253304, 82241230, 82341007, T2341007, 12131020, 31930022 and T2350003), the Basic Frontier Science Research Program of Chinese Academy of Sciences (ZDBS-LY-SM015), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDA16020905, XDB38040400), the Shanghai Science and Technology Committee (21XD1424200, 21ZR1470100 and 23JS1401300), the Shanghai Municipal Science and Technology Major Project, the China National Postdoctoral Program for Innovative Talents (BX2021306), the China Postdoctoral Science Foundation (2023M743484 and 2023000162), the Innovative Research Team of High-level Local Universities in Shanghai (SHSMUZDCX20211800), the Science and Technology Commission of Shanghai Municipality (23JS1401300), JST Moonshot R&D (JPMJMS2021), the New Cornerstone Science Foundation through the XPLORER PRIZE, the Guangdong Basic and Applied Basic Research Foundation (2023A1515011075), and the Shenzhen Bay Laboratory.

Author information

Authors and Affiliations

Authors

Contributions

D.G., F.B., L.C. and C.Y. conceived and designed the project. F.L., X.X., Q.J., X.-M.W., C.Y., L.C., F.B. and D.G. wrote the manuscript. F.L., X.X., Q.J., X.-M.W., Q.W., Y.C., C.Y., L.C., F.B. and D.G. provided edits to the manuscript. F.L., P.D. and M.H. performed the experiments. H.S., X.S., Y.P., J.X., D.L., W.W. and Y.Z. provided experimental support. F.L., X.X., Q.J. and X.-M.W. performed the computational and statistical analyses. Z.Z. constructed the web tool.

Corresponding authors

Correspondence to Chen Yu, Luonan Chen, Fan Bai or Dong Gao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Darragh Duffy and the other, 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 AR-positive cells are prevalent across tissues.

a, b, Immunochemical staining for AR across tissues in male mice (a) and female mice (b). c, Schematic for the generation of the systemic AR knockout in male mice. d, Immunochemistry staining on AR across multiple tissues of Rosa26CreERT2/+; Arflox/Y male mice 2 weeks post tamoxifen or corn oil treatment. Scale bar represents 50 μm. In a, b and d, experiments were repeated independently with similar results for three times.

Extended Data Fig. 2 Effects of androgens on body weight and tissue weight.

a, Quantification of the changes in body weight of MS (n = 15), MC (n = 16), FS (n = 27) and FD (n = 29) mice. n denotes the number of mice. Data are mean ± s.e.m. b, Volcano plot showing the weight changes in each tissue induced by androgen deprivation in male mice. The tissues with decreased weight in response to androgen deprivation are marked in blue, while the tissues with increased weight in response to androgen deprivation are marked in red. c, Heatmap showing the weight changes in each tissue induced by androgen deprivation in male mice. d, Volcano plot showing the changes in the weight of each tissue induced by androgen supplementation in female mice. The tissues with decreased weight in response to androgen supplementation are marked in blue, while the tissues with increased weight in response to androgen supplementation are marked in red. e, Heatmap showing the weight changes in each tissue induced by androgen supplementation in female mice. f, Summary of the effects of androgens on the weight of each tissue. g, Representative tissues, including the salivary gland (androgen-promoting, left), thymus (androgen-suppressing, middle) and liver (nonsignificant changes, right) tissue of MS, MC, FS and FD mice. In b, d, statistical comparisons were performed using two-tailed Student’s t test.

Source Data

Extended Data Fig. 3 Expression levels of representative markers.

a, Feature plot showing the RNA expression levels of representative markers for tissue-common cell lineages, including Ptprc for immune cells, Pecam1 for endothelial cells, Col1a1 for fibroblasts and Tagln for smooth muscle cells. b, Feature plot showing the RNA expression levels of representative markers for tissue-specific cell lineages, including Alb for hepatocytes in the liver, Lrp2 for proximal tubule cells in the kidney, Sftpc for AT2 cells in the lung and Krt20 for enterocytes in the colon and intestine.

Extended Data Fig. 4 Androgens modulate sex-biased cellular populations.

a, Scatter plot showing the correlation between sex differences (log2(MS vs. FS)) and the differences in the proportions of all the cell types across tissues between FD and FS (log2(FD vs. FS)). b, Similar to (a), but for the correlation between log2(MS vs. FS) and log2(MC vs. MS). c, Heatmap denoting the impacts of sex and androgens on the proportions of all cell types across tissues. The color key represents the log2-transformed fold change in the cell proportions. d, Dot plot showing the Pearson correlation efficiency calculated based on sex differences (log2(MS vs. FS)) and the differences in cell composition between MC and MS (log2(MC vs. MS)) (x-axis), as well as sex differences (log2(MS vs. FS)) and the differences in cell composition between FD and FS (log2(FD vs. FS)) (y-axis). The dot color represents the tissue type. e, Scatter plot showing the correlation between sex differences (log2(MS vs. FS)) and the differences in salivary cell composition between FD and FS (log2(FD vs. FS), left) or the differences in salivary cell composition between MC and MS (log2(MC vs. MS), right). f, Bar plot denoting the cellular composition of salivary gland across four conditions. g, UMAP plot showing the cell composition of salivary gland based on four conditions involving sex and androgens. Two thousand cells for each condition were randomly extracted for visualization. h, Bubble plot (left) and heatmap (right) denoting the impacts of sex and androgens on the proportions of all salivary cell types. The size indicates the proportion. The color key represents the log2-transformed fold change in cell proportions. In a, b, and e, P values are shown; the test statistic is based on Pearson’s product moment correlation coefficient and follows a t distribution; level of confidence interval is 0.95.

Extended Data Fig. 5 Cross-cell type effects of sex and androgens on biological pathways.

a-f, Dot plot showing the top 50 cross-cell type biological pathways based on the upregulated DEGs (a) and downregulated DEGs (b) between MS and FS, the upregulated DEGs (c) and downregulated DEGs (d) between MC and MS, and the upregulated DEGs (e) and downregulated DEGs (f) between FD and FS. The x-axis represents the number of cell types sharing the target biological pathway. Gradient color represents the number of tissues sharing the target biological pathway. g, h, Lollipop plot showing the remarkable androgen-associated genes in the “neutrophil chemotaxis” pathway (g) and the “T-cell activation” pathway (h). The x-axis represents the number of cell types sharing the target gene. i, Scatter plot showing the correlation of log2FC in the DEGs which locates on autosomes between sex differences and MC vs. MS based on all of the cell types from 17 tissues. j, Scatter plot showing the correlation of log2FC in the DEGs which locates on autosomes between sex differences and FD vs. FS based on all of the cell types from 17 tissues. In i, j, P values are shown; the test statistic is based on Pearson’s product moment correlation coefficient and follows a t distribution.

Extended Data Fig. 6 The effects of androgen supplementation on ovariectomized female mice and castrated male mice.

a, Schematic diagram of the experimental design to investigate the effects of androgens on the tissue weight, gene expression and cell composition in ovariectomized female mice (FOD vs. FO). b, Heatmap showing the tissue weight changes induced by androgen supplementation in ovariectomized female mice. c, Representative tissues, including the kidney and salivary gland (androgen-promoting), thymus and spleen (androgen-suppressing), and brain and lung (nonsignificant changes) of FO and FOD mice. d, Scatter plot showing the correlation of androgen supplementation-induced cell proportional differences between intact (FD vs. FS) and ovariectomized (FOD vs. FO) female mice across tissues. Level of confidence interval is 0.95. e, Scatter plot showing the correlation of DEG expression differences between FD vs. FS and FOD vs. FO mice (left). Heatmap denoting the log2FC in the expression of the DEGs from a single cell type across the four comparisons, where DEGs are colored according to their tissue of origin (right). f, Similar to (a) but for castrated male mice (MCD vs. MC). g, Similar to (b) but for androgen supplementation in castrated male mice. h, Similar to (c) but for MC and MCD mice. i, Scatter plot showing the correlation of cell proportional differences between the comparisons of MC vs MCD and MC vs. MS across tissues. Level of confidence interval is 0.95. j, Similar to (e) but for comparison between MC vs. MCD and MC vs. MS mice. In d, e, i, and j, P values are shown; the test statistic is based on Pearson’s product moment correlation coefficient and follows a t distribution. In each condition (FOD, FO and MCD), each tissue contained at least three biological replicates (three mice).

Source Data

Extended Data Fig. 7 AASB-DEGs and their functionally enriched pathways across tissues.

a, Network of all AASB-DEGs in each cell type across tissues. Red edges and blue edges represent the positive AASB-DEGs and the negative AASB-DEGs, respectively. The gray node represents the AASB-DEG, and the colored node represents the cell type. b, Scatter plot showing the correlation between the numbers of AASB-DEGs and Ar RNA expression. c, Scatter plot showing the correlation between the numbers of AASB-DEGs and Gapdh RNA expression. d, e, Representative cross-tissue AASB-DEGs and their relevant cell types, including Ace (positive AASB-DEG, d) and Hsd11b1 (positive AASB-DEG, e). f, Lollipop plot denoting the shared biological pathways across tissues based on positive (left top) or negative (left bottom) AASB-DEGs. Network of significantly enriched biological pathways based on AASB-DEGs in each cell type across tissues (right). In b, c, P values are shown; the test statistic is based on Pearson’s product moment correlation coefficient and follows a t distribution; level of confidence interval is 0.95.

Extended Data Fig. 8 Dissection of the effects of androgens on the immune compartment.

a, Dot plot showing the expression patterns of representative markers for nine major immune cell types. The dot size represents the percentage of cells with detectable gene expression. The color key represents the expression levels. b-d, Heatmap denoting sex differences (b) and the effects of androgen deprivation (c) and androgen supplementation (d) on immune cell proportions across tissues. The color key represents the log2-transformed fold change in the cell proportion. Light gray indicates that the comparison was performed on two cell types with cell numbers less than 10.

Extended Data Fig. 9 Enriched biological pathways based on disease-shared risk genes.

a, Pie chart showing the categorization of sex-biased diseases in the 17 tissues in our atlas. b, Heatmap denoting the differences in the incidence rates of sex-biased diseases between males and females. Representative male-biased and female-biased diseases are marked in red and blue, respectively. c, Network denoting the top 30 significantly enriched biological pathways based on the 662 risk genes that were shared by multiple sex-biased diseases. Node size represents gene number. Distinct biological pathway modules are marked in different colors. d, Dot plot denoting the significantly enriched biological pathways based on the 662 disease-shared risk genes of sex-biased diseases. Significantly enriched biological pathways were defined by the hypergeometric distribution and P values were adjusted by the Benjamini-Hochberg method (BH) by using EnrichGO function implanted in the clusterProfiler.

Extended Data Fig. 10 Sex and androgens influence MHC gene expressions.

a, b, Volcano plot denoting the MHC genes that showed significant differential expression (a) and their tissue of origin (b) based on the comparison of MS vs. FS. DEGs were defined by Wilcoxon Rank Sum test and P values were adjusted by the Bonferroni correction using the FindMarkers function implanted in Seurat. c, d, Volcano plot denoting the MHC genes that showed significant differential expression (c) and their tissue of origin (d) based on the comparison of MC vs. MS. DEGs were defined by Wilcoxon Rank Sum test and P values were adjusted by the Bonferroni correction using the FindMarkers function implanted in Seurat. e, f, Volcano plot denoting the MHC genes that showed significant differential expression (e) and their tissue of origin (f) based on the comparison of FD vs. FS. DEGs were defined by Wilcoxon Rank Sum test and P values were adjusted by the Bonferroni correction using the FindMarkers function implanted in Seurat. g, Gating strategy for bone marrow immature B cells (CD19+/B220+/AA4.1+/IgM+) and relative H2-Eb1 mRNA expression level quantified by qRT–PCR in the FACS-sorted bone marrow immature B cells of FD and FS mice. n = 6 mice examined per group. h, Gating strategy for spleen macrophages (CD45+/Ly6G+/F4/80+) and relative H2-Ab1 mRNA expression level quantified by qRT–PCR in the FACS-sorted spleen macrophages of FD and FS mice. n = 5 mice examined per group. i, Schematic to explain how we categorized human sex-biased diseases into five major groups using our scRNA-seq data. In g, h, Data are mean ± s.e.m. P values are shown; statistical comparisons were performed using two-tailed Student’s t test.

Source Data

Extended Data Fig. 11 Serum levels of corticosterone.

Serum levels of corticosterone in FD, FS, MS and MC mice (FD: n = 5 mice; FS: n = 13 mice; MS: n = 5 mice; MC: n = 6 mice). n denotes the number of mice. Data are mean ± s.e.m.

Source Data

Supplementary information

Supplementary Information

Reporting Summary

Supplementary Table 1

General information for atlas.

Supplementary Table 2

Cell proportion.

Supplementary Table 3

DEGs.

Supplementary Table 4

Significantly enriched biological pathways.

Supplementary Table 5

Information for scRNA-seq data of FOD, FO and MCD conditions.

Supplementary Table 6

AASB-DEGs and pathways.

Supplementary Table 7

Sex-biased diseases and their risk genes.

Supplementary Table 8

Cell-type enrichment for sex-biased diseases.

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

Li, F., Xing, X., Jin, Q. et al. Sex differences orchestrated by androgens at single-cell resolution. Nature (2024). https://doi.org/10.1038/s41586-024-07291-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41586-024-07291-6

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