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:

Single-cell analysis reveals transcriptomic remodellings in distinct cell types that contribute to human prostate cancer progression

Abstract

Prostate cancer shows remarkable clinical heterogeneity, which manifests in spatial and clonal genomic diversity. By contrast, the transcriptomic heterogeneity of prostate tumours is poorly understood. Here we have profiled the transcriptomes of 36,424 single cells from 13 prostate tumours and identified the epithelial cells underlying disease aggressiveness. The tumour microenvironment (TME) showed activation of multiple progression-associated transcriptomic programs. Notably, we observed promiscuous KLK3 expression and validated the ability of cancer cells in altering T-cell transcriptomes. Profiling of a primary tumour and two matched lymph nodes provided evidence that KLK3 ectopic expression is associated with micrometastases. Close cell–cell communication exists among cells. We identified an endothelial subset harbouring active communication (activated endothelial cells, aECs) with tumour cells. Together with sequencing of an additional 11 samples, we showed that aECs are enriched in castration-resistant prostate cancer and promote cancer cell invasion. Finally, we created a user-friendly web interface for users to explore the sequenced data.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Overview of the single-cell landscape for prostate cancer.
Fig. 2: Purified signature derived from single-cell data and the association with survival.
Fig. 3: scRNA-seq reveals heterogeneity in immune components.
Fig. 4: Tumour-derived EVs convey ectopic KLK3 expression in T cells.
Fig. 5: Identifying an activated EC cell subset in stromal cells.
Fig. 6: aEC cells undergo ECM remodelling and are enriched in CRPC.

Similar content being viewed by others

Data availability

Data have been deposited in the Gene Expression Omnibus (GEO) under accession no. GSE141445 and the Genome Sequence Archive for Human (GSA-Human) under accession HRA000312 and can be accessed at www.pradcellatlas.com. For gene expression analysis in T cells, scRNA-seq data from the following NCBI GEO accessions were used: GSE99254 (NSCLC)66, GSE108989 (CRC)67 GSE98638 (HCC)51 and GSE103322 (HNSCC)10. For survival analysis, bulk RNA-seq data from the following studies were used: TCGA (333 samples, http://firebrowse.org/?cohort=PRAD)4, ref. 17 (131 samples, MSKCC, https://doi.org/10.1016/j.ccr.2010.05.026), ref. 68 (294 samples, GSE70770), ref. 69 (79 samples, https://doi.org/10.1172/JCI20032/) and Changhai 2020 (136 samples, www.cpgea.com)70. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

All R packages used are available online, as described in the Methods. Customized code for data analysis and plotting are available on GitHub (https://github.com/chensujun/scRNA).

References

  1. Ferlay, J. et al. Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int. J. Cancer 144, 1941–1953 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Weiner, A. B., Matulewicz, R. S., Eggener, S. E. & Schaeffer, E. M. Increasing incidence of metastatic prostate cancer in the United States (2004–2013). Prostate Cancer Prostatic Dis. 19, 395–397 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Chen, S. et al. Widespread and functional RNA circularization in localized prostate cancer. Cell 176, 831–843 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Cancer Genome Atlas Research Network The molecular taxonomy of primary prostate cancer. Cell 163, 1011–1025 (2015).

  5. You, S. et al. Integrated classification of prostate cancer reveals a novel luminal subtype with poor outcome. Cancer Res. 76, 4948–4958 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhao, S. G. et al. Associations of luminal and basal subtyping of prostate cancer with prognosis and response to androgen deprivation therapy. JAMA Oncol. 3, 1663–1672 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck. Cancer Cell 171, 1611–1624 (2017).

    CAS  Google Scholar 

  11. Kumar, M. P. et al. Analysis of single-cell RNA-seq identifies cell–cell communication associated with tumor characteristics. Cell Rep. 25, 1458–1468 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  15. Yaari, G., Bolen, C. R., Thakar, J. & Kleinstein, S. H. Quantitative set analysis for gene expression: a method to quantify gene set differential expression including gene–gene correlations. Nucleic Acids Res. 41, e170 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Henry, G. H. et al. A cellular anatomy of the normal adult human prostate and prostatic urethra. Cell Rep. 25, 3530–3542 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Taylor, B. S. et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fraser, M. et al. Genomic hallmarks of localized, non-indolent prostate cancer. Nature 541, 359–364 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Espiritu, S. M. G. et al. The evolutionary landscape of localized prostate cancers drives clinical aggression. Cell 173, 1003–1013 (2018).

    Article  CAS  PubMed  Google Scholar 

  20. Pressinotti, N. C. et al. Differential expression of apoptotic genes PDIA3 and MAP3K5 distinguishes between low- and high-risk prostate cancer. Mol. Cancer 8, 130 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Isella, C. et al. Stromal contribution to the colorectal cancer transcriptome. Nat. Genet. 47, 312–319 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Ragnum, H. B. et al. The tumour hypoxia marker pimonidazole reflects a transcriptional programme associated with aggressive prostate cancer. Br. J. Cancer 112, 382–390 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Scialdone, A. et al. Computational assignment of cell-cycle stage from single-cell transcriptome data. Methods 85, 54–61 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Cuzick, J. et al. Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol. 12, 245–255 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Aran, D., Sirota, M. & Butte, A. J. Systematic pan-cancer analysis of tumour purity. Nat. Commun. 6, 8971 (2015).

    Article  CAS  PubMed  Google Scholar 

  26. Novershtern, N. et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell 144, 296–309 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Muller, S. et al. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome Biol. 18, 234 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Hayes, A. R., Brungs, D. & Pavlakis, N. Osteoclast inhibitors to prevent bone metastases in men with high-risk, non-metastatic prostate cancer: a systematic review and meta-analysis. PLoS ONE 13, e0191455 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Fiorino, C. & Harrison, R. E. E-cadherin is important for cell differentiation during osteoclastogenesis. Bone 86, 106–118 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Tsuchida, K. et al. Activin signaling as an emerging target for therapeutic interventions. Cell Commun. Signal. 7, 15 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sugatani, T. et al. Ligand trap of the activin receptor type IIA inhibits osteoclast stimulation of bone remodeling in diabetic mice with chronic kidney disease. Kidney Int. 91, 86–95 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Alon, R. A sweet solution: glycolysis-dependent Treg cell migration. Immunity 47, 805–807 (2017).

    Article  CAS  PubMed  Google Scholar 

  33. Xu, T. et al. Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism. Nature 548, 228–233 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. van Loosdregt, J. et al. Canonical Wnt signaling negatively modulates regulatory T cell function. Immunity 39, 298–310 (2013).

    Article  PubMed  Google Scholar 

  35. Wild, C. A. et al. HMGB1 conveys immunosuppressive characteristics on regulatory and conventional T cells. Int. Immunol. 24, 485–494 (2012).

    Article  CAS  PubMed  Google Scholar 

  36. Harjunpaa, H., Llort Asens, M., Guenther, C. & Fagerholm, S. C. Cell adhesion molecules and their roles and regulation in the immune and tumor microenvironment. Front. Immunol. 10, 1078 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tung, K. H., Ernstoff, M. S., Allen, C. & Shu, S. A review of exosomes and their role in the tumor microenvironment and host–tumor ‘macroenvironment’. J. Immunol. Sci. 3, 4–8 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Bartoschek, M. et al. Spatially and functionally distinct subclasses of breast cancer-associated fibroblasts revealed by single cell RNA sequencing. Nat. Commun. 9, 5150 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Ayala, G. et al. Reactive stroma as a predictor of biochemical-free recurrence in prostate cancer. Clin. Cancer Res. 9, 4792–4801 (2003).

    CAS  PubMed  Google Scholar 

  40. Vellanki, R. N., Zhang, L. & Volchuk, A. OASIS/CREB3L1 is induced by endoplasmic reticulum stress in human glioma cell lines and contributes to the unfolded protein response, extracellular matrix production and cell migration. PLoS ONE 8, e54060 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Varrault, A. et al. Identification of Plagl1/Zac1 binding sites and target genes establishes its role in the regulation of extracellular matrix genes and the imprinted gene network. Nucleic Acids Res. 45, 10466–10480 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sieweke, M. H., Tekotte, H., Frampton, J. & Graf, T. MafB is an interaction partner and repressor of Ets-1 that inhibits erythroid differentiation. Cell 85, 49–60 (1996).

    Article  CAS  PubMed  Google Scholar 

  43. Sinha, M. et al. Direct conversion of injury-site myeloid cells to fibroblast-like cells of granulation tissue. Nat. Commun. 9, 936 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ochiya, T., Takenaga, K. & Endo, H. Silencing of S100A4, a metastasis-associated protein, in endothelial cells inhibits tumor angiogenesis and growth. Angiogenesis 17, 17–26 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Schubert, K., Gutknecht, D., Koberle, M., Anderegg, U. & Saalbach, A. Melanoma cells use Thy-1 (CD90) on endothelial cells for metastasis formation. Am. J. Pathol. 182, 266–276 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Finkenzeller, G., Hager, S. & Stark, G. B. Effects of bone morphogenetic protein 2 on human umbilical vein endothelial cells. Microvasc. Res. 84, 81–85 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. de Sousa Mesquita, A. P., de Araujo Lopes, S., Pernambuco Filho, P. C. A., Nader, H. B. & Lopes, C. C. Acquisition of anoikis resistance promotes alterations in the Ras/ERK and PI3K/Akt signaling pathways and matrix remodeling in endothelial cells. Apoptosis 22, 1116–1137 (2017).

    Article  PubMed  Google Scholar 

  48. Wojno, K. J. & Epstein, J. I. The utility of basal cell-specific anti-cytokeratin antibody (34βE12) in the diagnosis of prostate cancer. A review of 228 cases. Am. J. Surg. Pathol. 19, 251–260 (1995).

    Article  CAS  PubMed  Google Scholar 

  49. Parsons, J. K., Gage, W. R., Nelson, W. G. & De Marzo, A. M. p63 protein expression is rare in prostate adenocarcinoma: implications for cancer diagnosis and carcinogenesis. Urology 58, 619–624 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Heck, M. M. et al. Topography of lymph node metastases in prostate cancer patients undergoing radical prostatectomy and extended lymphadenectomy: results of a combined molecular and histopathologic mapping study. Eur. Urol. 66, 222–229 (2014).

    Article  PubMed  Google Scholar 

  51. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 (2017).

    Article  CAS  PubMed  Google Scholar 

  52. Guo, X. et al. A linc1405/Eomes complex promotes cardiac mesoderm specification and cardiogenesis. Cell Stem Cell 22, 893–908 (2018).

    Article  CAS  PubMed  Google Scholar 

  53. Jeppesen, D. K. et al. Reassessment of exosome composition. Cell 177, 428–445 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Minciacchi, V. R. et al. Large oncosomes contain distinct protein cargo and represent a separate functional class of tumor-derived extracellular vesicles. Oncotarget 6, 11327–11341 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Vagner, T. et al. Large extracellular vesicles carry most of the tumour DNA circulating in prostate cancer patient plasma. J. Extracell. Vesicles 7, 1505403 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhou, B. et al. Low-background acyl-biotinyl exchange largely eliminates the coisolation of non-S-acylated proteins and enables deep S-acylproteomic analysis. Anal. Chem. 91, 9858–9866 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884–i890 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with BioConductor. F1000Res. 5, 2122 (2016).

    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. Taylor, R. A., Toivanen, R. & Risbridger, G. P. Stem cells in prostate cancer: treating the root of the problem. Endocr. Relat. Cancer 17, R273–R285 (2010).

    Article  CAS  PubMed  Google Scholar 

  61. 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 

  62. P’ng, C. et al. BPG: seamless, automated and interactive visualization of scientific data. BMC Bioinf. 20, 42 (2019).

    Article  Google Scholar 

  63. Davis, S. & Meltzer, P. S. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics 23, 1846–1847 (2007).

    Article  PubMed  Google Scholar 

  64. Wan, Y. W., Allen, G. I. & Liu, Z. TCGA2STAT: simple TCGA data access for integrated statistical analysis in R. Bioinformatics 32, 952–954 (2016).

    Article  CAS  PubMed  Google Scholar 

  65. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  CAS  PubMed  Google Scholar 

  67. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Ross-Adams, H. et al. Integration of copy number and transcriptomics provides risk stratification in prostate cancer: a discovery and validation cohort study. EBioMedicine 2, 1133–1144 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Glinsky, G. V., Glinskii, A. B., Stephenson, A. J., Hoffman, R. M. & Gerald, W. L. Gene expression profiling predicts clinical outcome of prostate cancer. J. Clin. Invest. 113, 913–923 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, J. et al. A genomic and epigenomic atlas of prostate cancer in Asian populations. Nature 580, 93–99 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the National Key R&D Plan of the China Precision Medicine Project (2017YFC0908002 to S.R.), the National Natural Science Foundation of China (81872105 to S.R.), the Princess Margaret Cancer Foundation (886012001223 to H.H.H.), the Canada Foundation for Innovation and Ontario Research Fund (CFI32372 to H.H.H.), a NSERC discovery grant (498706 to H.H.H.), a Canadian Cancer Society Innovation Grant (703800 to H.H.H.), Prostate Cancer Canada (TAG2018-2061, RS2016-1022 and D2016-1115 to H.H.H.), CIHR operating grants (142246, 152863, 152864 and 159567 to H.H.H.) and a Terry Fox New Frontiers Program Project Grant (1090 P3 to H.H.H.). H.H.H. was supported by an OMIR Early Researcher Award and CIHR New Investigator Award. H.H.H. holds the Joey and Toby Tanenbaum Brazilian Ball Chair in Prostate Cancer. J.W. was partially funded by the National Natural Funding of China (81272404, 81772806 and 81972744). G.Z. and S.W. were supported by the Sanming Project of Medicine in Shenzhen (SZSM201601043). Y.L. was funded by the National Natural Science Foundation of China (31801111) and the Dream Mentor–Outstanding Young Talent Program (fkyq1910). P.C.B. was supported by the NIH/NCI under award nos. P30CA016042, 1U01CA214194-01 and 1U24CA248265-01. We thank L.W. Chung from Cedars-Sinai for providing the C4-2B cell line.

Author information

Authors and Affiliations

Authors

Contributions

The studies were designed by S.C., G.Z., J.W., M.F., D.D.V., H.H.H. and S.R. Experiments were performed by Y.-T.X., N.Z., X.B., Y. Yang, F.W., C.W., Y.Z., Y. Yu, K.D., J.M., Y.L., F.S., H.L.Y., M.L. and W.C. Data analysis was carried out by S.C., G.Z., B.Z., F.L., W.C., D.C., Q.G., Z.Y., S.W., M.F., P.C.B., D.D.D.C., T.v.d.K., Z.J. and A.B. The first draft of the manuscript was written by S.C., G.Z., S.R. and H.H.H. All authors revised and approved the manuscript.

Corresponding authors

Correspondence to Jianhua Wang, Housheng Hansen He or Shancheng Ren.

Ethics declarations

Competing interests

B.Z., W.C., C.W., D.C. and Q.G. are co-founders for Novel Bioinformatics Co., Ltd. All other authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks Mark A. Rubin for his 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 Single cell characterization and CNV inference analysis of prostate tumours.

tSNE view of 36,424 single cells, color coded by sample (a), cluster (b) and broad lineage (c). d, Inferred CNA for cells in tumour 156. e, the CNA score and correlation for each cell in the indicated sample. Red, CNA score > 0.04 and CNA correlation >0.4; blue, CNA score <0.04 and CNA correlation <0.4; black, all remaining cells. f, Percentage of genes showing strong CNV (averaged CNA score > 0.1 in putative malignant cells) in our data (PCa, n = 13 tumor samples) and that from Tirosh et al. (Mel, n = 14 tumor samples). P value calculated with two-sided Mann-Whitney U test. g, tSNE view of 36,424 single cells, color coded by inferred cell malignancy identity. For box plots, center line represents the median and box limits represent upper and lower quartiles, and whiskers depicts 1.5× the interquartile range (IQR), extreme values outside of this range is shown as individual points. Statistical data for Extended Data Fig. 1f are provided in the source data.

Source data

Extended Data Fig. 2 Gene expression and signature analysis for different cell types.

a, Heatmap shows the relative abundance of 27 genes in the ‘T cell costimulation’ process in each cell. The top color bar indicates cell types and the left color bar shows the mean UMI of genes. b, tSNE view of 36,424 single cells, color coded by epithelial subtypes. c, Smoothened distribution of PAM50 signature score, cells grouped by annotated cell type. d, Smoothened distribution of high Gleason Score (GS) related signature, cells grouped by annotated cell type. Signature score calculated as the mean of z-score for the 19 high GS related genes identified by Pressinotti et al.20. e, Smoothened distribution of 3 high GS related individual genes; cells grouped by annotated cell type. Y-axis shows normalized UMI (nUMI) in logarithm scale.

Extended Data Fig. 3 Characterizing different epithelial cell derived programs.

a, tSNE view of 23,674 epithelial cells, colour-coded by sample. b, Smoothened distribution for four prostate cancer related signatures in epithelial cell clusters as defined in Fig. 2a. P-values for cluster 10 compared to all other cells for Luminal A, Luminal B are <2.2 × 10−16; CLESs are 0.26 and 0.14, respectively; P-values for cluster 12 compared to all other cells for Luminal A, Luminal B, Hypoxia and PCS1 are <2.2 × 10−16: CLESs are 0.04, 0.79, 0.88 and 0.995, respectively. P values are two-sided and not adjusted for multiple comparisons. c, Smoothened distribution of the indicated signature score, TME and epithelial cells grouped by their assigned cell types and clusters, respectively. P-values (Mann-Whitney U test, two-sided) for cluster 12 CellCycle and cluster 10 basal/intermediate signatures compared to all the rest cells are < 2.2 × 10−16; CLESs are 0.998 and 0.98, respectively. d, GO terms enriched in CellCycle subtype (left) and contingency table showing number of cells in G2/M for CellCycle subtype compared to all the other cells (right), One-sided P value calculated with Fisher’s exact test. OR = 5.7. e, Comparison of BCR-free rate between the high and low groups stratified using CellCycle signature across multiple datasets. P-values are calculated using Cox proportional hazard model (CoxPH) and not adjusted for multiple comparisons. Numbers in brackets show 95% CI for PH.

Extended Data Fig. 4 Characterizing the basal/intermediate signature.

a, Comparison of BCR-free rate between the high and low groups stratified using Basal/Intermediate signature in the indicated datasets. P-values are calculated using Cox proportional hazard model (CoxPH) and not adjusted for multiple comparisons. Numbers in brackets show 95% confidence interval (CI) for hazard ratio (HR). b, Correlation between tumour purity corrected CCL2 expression and basal/intermediate signature in TCGA. c, Correlation between macrophage, T cell and basal/intermediate signature across multiple datasets. Two-sided P values calculated for Spearman’s rank correlation and not adjusted for multiple comparisons.

Extended Data Fig. 5 Single cell transcriptome reveals immune cell heterogeneity.

a, Schematics for all C6 specific incoming signals. b, Percentage of C6 TAM cells in each sample. c, Differentially activated metabolism-related pathways. d, tSNE view of 3,116 T cells, color coded by the average expression of lipid mediator, glycogen metabolism and glycolysis genes. e, Smoothened distribution of AR signature gene abundance, cells grouped by cluster. f, Genes in module 61. Line length and circle size corresponds to expression correlation between KLK3 and the indicated gene. Statistical data relevant to Extended Data Fig. 5b are provided in the source data.

Source data

Extended Data Fig. 6 Analysis of KLK3 expression in T cells.

Flow cytometry sorting strategy (a) and statistics (b) of PSMA+ CD8 T cells. Relative expression of KLK3 in the indicated prostate cancer cell lines c) and EVs (d) derived from them. e, Examination of KLK3 protein (PSA, red) in T cells (CD8+, green) after co-culture with EVs derived from the indicated prostate cancer cell lines by fluorescence microscopy. Scale bars, 10μm. Data show representative results of two repeats. f, Immunoblot for small EV-enriched (CD81) and depleted proteins (GM130) in C4-2B EVs. EVs collected by differential centrifugation followed by density gradient purification. 1µg of protein was loaded for whole cell lysate (WCL). Same volume was loaded for S-EV fractions. g, rtPCR for the indicated probes in C4-2B small EVs collected by differential centrifugation and floated in different density fractions. tSNE view showing cells from the high-risk prostate cancer patient (SC001H), color coded by the tissue source (h) or cell type (i). Distribution of KLK3 expression in different cell types for samples from Batch 1 data (j) (n = 22,667, 7,495 and 629 cells for epi., immune and stroma groups, respectively), tumour tissue (k) (n = 700, 4,038 and 594 cells for epi., immune and stroma groups, respectively) and left LN (l) (n = 153, 1,954 and 2 cells for epi., immune and stroma groups, respectively) from Batch 2. Two-sided P values are calculated using Mann-Whitney U test. Y-axis of j-l represents natural logarithm scale. For box plots, center line represents the median and box limits represent upper and lower quartiles, and whiskers depicts 1.5× the IQR, extreme values outside of this range is shown as individual points. Statistical data relevant to Extended Data Fig. 6c-d, g are provided in the source data. Unprocessed Western Blots relevant to Extended Data Fig. 6f are provided in the source data.

Source data

Extended Data Fig. 7 Single cell characterization of stromal components.

a, Hierarchical clustering using all subtype marker genes, color coded by subtypes. b, Correlation between EMT score in epithelial cells and percentage of ACTA2+ CAF. Two-sided P values calculated for Spearman’s rank correlation and not adjusted for multiple comparisons. c, tSNE view CAF cells, color coded by activation level of the indicated TFs (AUC). d, Overlap of the unique incoming cell communication pairs from epithelial cells to fibroblast, regular EC and aEC. e, Top 5 most enriched pathways for each cluster.

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Table 1. Patient sample clinical information. Supplementary Table 2. GO enrichment for each co-expression module shown in Fig. 3h. Supplementary Table 3. GO enrichment for the top 150 marker genes for each CAF subtype. Related to Extended Data Fig. 7b.

Source data

Source Data Fig. 4

Statistical source data relevant to Fig. 4b–d.

Source Data Fig. 6

Statistical source data relevant to Fig. 6g,h.

Source Data Extended Data Fig. 1

Statistical source data relevant to Extended Data Fig. 1f.

Source Data Extended Data Fig. 5

Statistical source data relevant to Extended Data Fig. 5b.

Source Data Extended Data Fig. 6

Statistical source data relevant to Extended Data Fig. 6c,d and f,g.

Source Data Extended Data Fig. 6

Unprocessed western blots relevant to Extended Data Fig. 6f.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, S., Zhu, G., Yang, Y. et al. Single-cell analysis reveals transcriptomic remodellings in distinct cell types that contribute to human prostate cancer progression. Nat Cell Biol 23, 87–98 (2021). https://doi.org/10.1038/s41556-020-00613-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-00613-6

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