Abstract
Prostate epithelium develops from multipotent stem cells, which are replaced in adult life by different lineage-restricted basal and luminal unipotent stem cells. Deletion of Pten re-induces multipotency in basal cells (BCs); however, the molecular mechanisms regulating BC plasticity and tumor initiation are poorly understood. Here we showed that Pten deletion in BCs led to distinct cell fate reprogramming and tumor initiation in a regionalized manner. Single-cell RNA sequencing, ATAC-seq and in situ characterization revealed that following Pten deletion in anterior and dorsolateral prostates, BCs were highly plastic and reprogrammed into a hillock-like state, progressing into a proximal-like luminal state before giving rise to invasive tumors. This BC reprogramming was associated with the activation of innate immunity. Pharmacological targeting of interleukin-1, JAK–STAT and NF-κB as well as genetic deletion of Nfkb inhibit Pten-induced cell plasticity and reprogramming in a cellular autonomous manner, opening new opportunities for prevention and treatment of prostate cancer.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout








Similar content being viewed by others
Data availability
All raw sequencing datasets that support the findings of this study have been deposited in the GEO under accession number GSE270187 (scRNA-seq), GSE270189 (bulk RNA-seq, rapamycin treated), GSE270190 (bulk RNA-seq, cell fate upon PTEN deletion in BC), GSE286018 (bulk RNA-seq of P65/Pten knock BCs), GSE286019 (bulk RNA-seq of basal-derived and luminal-derived luminal tumors), GSE270191 (ATAC-seq of pooled BC/LC of WT and PTEN-deleted cells) and GSE288787 (ATAC-seq of BCs from different lobes upon PTEN deletion). Previously published human prostate cancer datasets re-analyzed in this study were available from GitHub repository of the author (https://github.com/franklinhuanglab/scRNA-seq-Analysis-of-Prostate-Cancer-Samples)38, the Prostate Cell Atlas (https://www.prostatecellatlas.org)39 and GEO GSE181294 (ref. 40) and GSE185344 (ref. 41). The data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.
Code availability
The codes used for data processing, downstream analysis and plotting for this paper are available on GitHub at https://github.com/yurasong/PTEN_codes.
References
Toivanen, R. & Shen, M. M. Prostate organogenesis: tissue induction, hormonal regulation and cell type specification. Development 144, 1382–1398 (2017).
Ousset, M. et al. Multipotent and unipotent progenitors contribute to prostate postnatal development. Nat. Cell Biol. 14, 1131–1138 (2012).
Blanpain, C. & Fuchs, E. Stem cell plasticity. Plasticity of epithelial stem cells in tissue regeneration. Science 344, 1242281 (2014).
Lu, T. L. et al. Conditionally ablated Pten in prostate basal cells promotes basal-to-luminal differentiation and causes invasive prostate cancer in mice. Am. J. Pathol. 182, 975–991 (2013).
Choi, N., Zhang, B., Zhang, L., Ittmann, M. & Xin, L. Adult murine prostate basal and luminal cells are self-sustained lineages that can both serve as targets for prostate cancer initiation. Cancer Cell 21, 253–265 (2012).
Wang, Z. A. et al. Lineage analysis of basal epithelial cells reveals their unexpected plasticity and supports a cell-of-origin model for prostate cancer heterogeneity. Nat. Cell Biol. 15, 274–283 (2013).
Kwon, O. J., Zhang, L., Ittmann, M. M. & Xin, L. Prostatic inflammation enhances basal-to-luminal differentiation and accelerates initiation of prostate cancer with a basal cell origin. Proc. Natl Acad. Sci. USA 111, E592–E600 (2014).
Centonze, A. et al. Heterotypic cell–cell communication regulates glandular stem cell multipotency. Nature 584, 608–613 (2020).
Karthaus, W. R. et al. Regenerative potential of prostate luminal cells revealed by single-cell analysis. Science 368, 497–505 (2020).
Guo, W. et al. Single-cell transcriptomics identifies a distinct luminal progenitor cell type in distal prostate invagination tips. Nat. Genet. 52, 908–918 (2020).
Crowley, L. et al. A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors. eLife 9, e59465 (2020).
Mevel, R. et al. RUNX1 marks a luminal castration-resistant lineage established at the onset of prostate development. eLife 9, e60225 (2020).
Wang, X. et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature 461, 495–500 (2009).
Shen, M. M. & Abate-Shen, C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev. 24, 1967–2000 (2010).
Wang, G., Zhao, D., Spring, D. J. & DePinho, R. A. Genetics and biology of prostate cancer. Genes Dev. 32, 1105–1140 (2018).
Li, W. & Shen, M. M. Prostate cancer cell heterogeneity and plasticity: Insights from studies of genetically-engineered mouse models. Semin. Cancer Biol. 82, 60–67 (2022).
Wang, S. et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer Cell 4, 209–221 (2003).
Goldstein, A. S., Huang, J., Guo, C., Garraway, I. P. & Witte, O. N. Identification of a cell of origin for human prostate cancer. Science 329, 568–571 (2010).
Stoyanova, T. et al. Prostate cancer originating in basal cells progresses to adenocarcinoma propagated by luminal-like cells. Proc. Natl Acad. Sci. USA 110, 20111–20116 (2013).
Lawson, D. A. et al. Basal epithelial stem cells are efficient targets for prostate cancer initiation. Proc. Natl Acad. Sci. USA 107, 2610–2615 (2010).
Wang, Z. A., Toivanen, R., Bergren, S. K., Chambon, P. & Shen, M. M. Luminal cells are favored as the cell of origin for prostate cancer. Cell Rep. 8, 1339–1346 (2014).
Lukacs, R. U., Goldstein, A. S., Lawson, D. A., Cheng, D. & Witte, O. N. Isolation, cultivation and characterization of adult murine prostate stem cells. Nat. Protoc. 5, 702–713 (2010).
Pearson, H. B. et al. Identification of Pik3ca mutation as a genetic driver of prostate cancer that cooperates with Pten loss to accelerate progression and castration-resistant growth. Cancer Discov. 8, 764–779 (2018).
Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).
Agarwal, S. et al. Identification of different classes of luminal progenitor cells within prostate tumors. Cell Rep. 13, 2147–2158 (2015).
Martin, P. et al. Prostate epithelial Pten/TP53 loss leads to transformation of multipotential progenitors and epithelial to mesenchymal transition. Am. J. Pathol. 179, 422–435 (2011).
Thorpe, L. M., Yuzugullu, H. & Zhao, J. J. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat. Rev. Cancer 15, 7–24 (2015).
Carver, B. S. et al. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell 19, 575–586 (2011).
Floc’h, N. et al. Dual targeting of the Akt/mTOR signaling pathway inhibits castration-resistant prostate cancer in a genetically engineered mouse model. Cancer Res. 72, 4483–4493 (2012).
Lamming, D. W. Inhibition of the mechanistic target of rapamycin (mTOR)-rapamycin and beyond. Cold Spring Harb. Perspect. Med. 6, a025924 (2016).
Henry, G. H. et al. A cellular anatomy of the normal adult human prostate and prostatic urethra. Cell Rep. 25, 3530–3542.e5 (2018).
Joseph, D. B. et al. Urethral luminal epithelia are castration-insensitive cells of the proximal prostate. Prostate 80, 872–884 (2020).
Baures, M. et al. Prostate luminal progenitor cells: from mouse to human, from health to disease. Nat. Rev. Urol. 19, 201–218 (2022).
Baures, M. et al. Transcriptomic signature and growth factor regulation of castration-tolerant prostate luminal progenitor cells. Cancers 14, 3775 (2022).
Chan, J. M. et al. Lineage plasticity in prostate cancer depends on JAK/STAT inflammatory signaling. Science 377, 1180–1191 (2022).
Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).
Li, F. et al. ERG orchestrates chromatin interactions to drive prostate cell fate reprogramming. J. Clin. Invest. 130, 5924–5941 (2020).
Song, H. et al. Single-cell analysis of human primary prostate cancer reveals the heterogeneity of tumor-associated epithelial cell states. Nat. Commun. 13, 141 (2022).
Tuong, Z. K. et al. Resolving the immune landscape of human prostate at a single-cell level in health and cancer. Cell Rep. 37, 110132 (2021).
Hirz, T. et al. Dissecting the immune suppressive human prostate tumor microenvironment via integrated single-cell and spatial transcriptomic analyses. Nat. Commun. 14, 663 (2023).
Wong, H. Y. et al. Single cell analysis of cribriform prostate cancer reveals cell intrinsic and tumor microenvironmental pathways of aggressive disease. Nat. Commun. 13, 6036 (2022).
Mitchell, C. A. et al. Stromal niche inflammation mediated by IL-1 signalling is a targetable driver of haematopoietic ageing. Nat. Cell Biol. 25, 30–41 (2023).
Tsujimura, A. et al. Proximal location of mouse prostate epithelial stem cells: a model of prostatic homeostasis. J. Cell Biol. 157, 1257–1265 (2002).
Burger, P. E. et al. Sca-1 expression identifies stem cells in the proximal region of prostatic ducts with high capacity to reconstitute prostatic tissue. Proc. Natl Acad. Sci. USA 102, 7180–7185 (2005).
Goldstein, A. S. et al. Trop2 identifies a subpopulation of murine and human prostate basal cells with stem cell characteristics. Proc. Natl Acad. Sci. USA 105, 20882–20887 (2008).
Kwon, O.-J., Zhang, L. & Xin, L. Stem cell antigen-1 identifies a distinct androgen-independent murine prostatic luminal cell lineage with bipotent potential. Stem Cells 34, 191–202 (2015).
Ali, A. et al. Prostate zones and cancer: lost in transition? Nat. Rev. Urol. 19, 101–115 (2022).
McNeal, J. E., Redwine, E. A., Freiha, F. S. & Stamey, T. A. Zonal distribution of prostatic adenocarcinoma: correlation with histologic pattern and direction of spread. Am. J. Surg. Pathol. 12, 897–906 (1988).
Lee, J. J. et al. Biologic differences between peripheral and transition zone prostate cancer. Prostate 75, 183–190 (2015).
Berquin, I. M., Min, Y., Wu, R., Wu, H. & Chen, Y. Q. Expression signature of the mouse prostate. J. Biol. Chem. 280, 36442–36451 (2005).
Garcia, A. J. et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression. Mol. Cell. Biol. 34, 2017–2028 (2014).
Toso, A. et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 9, 75–89 (2014).
Di Mitri, D. et al. Tumour-infiltrating Gr-1+ myeloid cells antagonize senescence in cancer. Nature 515, 134–137 (2014).
Calcinotto, A. et al. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 559, 363–369 (2018).
Lopez-Bujanda, Z. A. et al. Castration-mediated IL-8 promotes myeloid infiltration and prostate cancer progression. Nat. Cancer 2, 803–818 (2021).
Brina, D. et al. The Akt/mTOR and MNK/eIF4E pathways rewire the prostate cancer translatome to secrete HGF, SPP1 and BGN and recruit suppressive myeloid cells. Nat. Cancer 4, 1102–1121 (2023).
Lu, X. et al. Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 543, 728–732 (2017).
Deng, S. et al. Ectopic JAK–STAT activation enables the transition to a stem-like and multilineage state conferring AR-targeted therapy resistance. Nat. Cancer 3, 1071–1087 (2022).
Guo, W. et al. JAK/STAT signaling maintains an intermediate cell population during prostate basal cell fate determination. Nat. Genet. 56, 2776–2789 (2024).
Pitzen, S. P. et al. Comparative transcriptomics reveals a mixed basal, club, and hillock epithelial cell identity in castration-resistant prostate cancer. Proc. Natl Acad. Sci. USA 122, e2415308122 (2025).
Huang, F. W. et al. Club-like cells in proliferative inflammatory atrophy of the prostate. J. Pathol. 261, 85–95 (2023).
Kiviaho, A. et al. Single cell and spatial transcriptomics highlight the interaction of club-like cells with immunosuppressive myeloid cells in prostate cancer. Nat. Commun. 15, 9949 (2024).
Van Keymeulen, A. et al. Distinct stem cells contribute to mammary gland development and maintenance. Nature 479, 189–193 (2011).
Tikoo, A. et al. Physiological levels of Pik3caH1047R mutation in the mouse mammary gland results in ductal hyperplasia and formation of ERα-positive tumors. PLoS ONE 7, e36924 (2012).
Jonkers, J. et al. Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat. Genet. 29, 418–425 (2001).
Luedde, T. et al. IKK1 and IKK2 cooperate to maintain bile duct integrity in the liver. Proc. Natl Acad. Sci. USA 105, 9733–9738 (2008).
Leow, C. C., Wang, X.-D. & Gao, W.-Q. Novel method of generating prostate-specific Cre–LoxP gene switching via intraductal delivery of adenovirus. Prostate 65, 1–9 (2005).
DuPage, M., Dooley, A. L. & Jacks, T. Conditional mouse lung cancer models using adenoviral or lentiviral delivery of Cre recombinase. Nat. Protoc. 4, 1064–1072 (2009).
Park, S. et al. Novel mouse models of bladder cancer identify a prognostic signature associated with risk of disease progression. Cancer Res. 81, 5161–5175 (2021).
WHO Classification of Tumours Editorial Board. in Urinary and Male Genital Tumours: WHO Classification of Tumours 5th edn, Vol. 8, Ch. 4, 193–233 (International Agency for Research on Cancer, 2022).
Drost, J. et al. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 11, 347–358 (2016).
Jiang, C. et al. Collagen signaling and matrix stiffness regulate multipotency in glandular epithelial stem cells in mice. Nat. Commun. 15, 10482 (2024).
Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Anders, S., Pyl, P. T. & Huber, W. HTSeq-a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).
Dennis, G. et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 4, R60 (2003).
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).
Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).
McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).
Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).
Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).
Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
Pearson, K. LIII. On lines and planes of closest fit to systems of points in space. Lond. Edinb. Dubl. Phil. Mag. J. Sci. 2, 559–572 (1901).
McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: Uniform Manifold Approximation and Projection. J. Open Source Softw. 3, 861 (2018).
Haynes, W. in Encyclopedia of Systems Biology (eds Dubitzky, W. et al.) 78 (Springer, 2013).
Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genom. 19, 477 (2018).
Acknowledgements
We thank the ULB animal facility and ULB genomic core facility (F. Libert and A. Lefort) for help with sequencing, DIAPath–CMMI, supported by the Fonds Yvonne Boël and by the European Regional Development Fund and the Walloon region (S. Rorive and J. Allard), for helping with histology, and J.M. Vanderwinden and LiMif for helping with microscopy. We thank M. Pasparakis, University of Cologne for providing the P65fl/fl mice. C.J. is supported by a long-term EMBO Postdoctoral Fellowships (ALTF 982-2021) and Foundation Against Cancer (2023-041). Y.S. is supported by Télévie. C.B. is supported by the WEL Research Institute, FNRS, Télévie, Fond Erasme, Fondation Contre le Cancer, ULB Foundation, European Research Council, Fonds Yvonne Boël and the Foundation Baillet Latour. A.S. is supported by KULeuven (SymBioSys – C14/18/092) and the Fondation Contre le Cancer (2015-143). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.
Author information
Authors and Affiliations
Contributions
C.J. and C.B. designed the experiments and performed data analysis. C.J. performed most of the experiments and data analysis. Y.S. and A.S. performed the bioinformatic analysis. S.R. performed prostate tumor scoring. I.S. provided human prostate cancer samples. J.A. performed the H&E staining on mouse prostate tumors and IHC staining on human prostate cancers. E.T. performed the experiments on bulk RNA-seq of Pten deletion at 5 weeks. Z.Z. provided technical help. C.J. and C.D. performed sorting experiments. C.B. wrote the manuscript. All authors read and approved of the final paper.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Cancer thanks the anonymous reviewers 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
Extended Data Fig. 1 Pten deletion induced BC plasticity and tumor initiation in a cell of origin and region-specific manners.
(a) FACS gating strategy to analyze and isolate BCs and LCs from K5CreER/Ptenfl/fl/RosaYFP mice 6 weeks after TAM injection. Same strategy for all lineage tracing mice in this study. (b) Quantification of the % of Lin- YFP+ LCs in total YFP+ cells in VP and DLP at different time points after TAM injection in K5CreER/Ptenfl/fl/RosaYFP mice. n = 3 mice for 1w, 4w, 7w, 8w and 20w, n = 4 mice for 6w and 12w. (c) Representative FACS plot of CD49f and EpCAM expression in Lin- YFP+ prostate epithelial cells from K5CreER/RosaYFP mice 6 week after TAM administration. (d) Quantification of the % of Lin− YFP+ BCs and LCs in total BCs and LCs in VP, DLP and AP in in K5CreER/RosaYFP mice after 6-8 weeks TAM administration. n = 6 mice. (e) Quantification of the % of YFP + K14+ and YFP + K8+ in total K14+ and K8+ in VP, DLP and AP in K5CreER/RosaYFP mice after 4 weeks TAM administration. n = 2 mice. (f) Immunostainings of EdU (Red), GFP (Green), K14 (Grey) and K8 (Grey) in the prostate of K5CreER/Ptenfl/fl/RosaYFP at 4w, 6w and 12-week after TAM administration 24 h after EdU injection. Scale bar, 10 μm. n = 3 mice. (g) Quantification of percentage of EdU doublet following basal cell division giving rise BC-BC, BC-LC and LC-LC doublets. n = 152 EdU doublets (4w), n = 65 EdU doublets (6w) and n = 142 EdU (12w) doublets pooled from 3 mice. (h) Genetic strategy to lineage trace LC following Pten deletion (i) Quantification of Lin- YFP+ BCs and LCs in total BCs and LCs in VP, DLP and AP at different time points after TAM administration in K8CreER/Ptenfl/fl/RosaYFP mice. n = 3 mice. Graphs are mean ± s.e.m.
Extended Data Fig. 2 mTOR inhibition blocks multipotency and tumor initiation.
Representative immunostaining of the prostate from (a) K5CreER/Ptenfl/fl/RosaYFP mice 6 weeks after TAM administration (K5-PTEN 6w) and (d) K5CreER/Ptenfl/fl/Pik3caH1047R/RosaYFP mice 3 weeks after TAM administration (K5-PTEN-PIK 3w) treated with EtOH or Rapamycin (6 mg/kg, three injections per week) using anti-GFP (green), anti-K8 (red) and anti-K14 (grey) antibodies. Arrows indicate hybrid cells (YFP + K14 + K8 + ). Scale bar, 20 μm (upper row), 10 μm (lower row). n = 3 mice. Representative FACS plot of CD49f and EpCAM expression in Lin- YFP+ prostate epithelial cells of (b) K5-PTEN 6w mice and (e) K5-PTEN-PIK 3w mice treated with EtOH or Rapamycin. Quantification of % of LIN- YFP+ LCs in total YFP+ cells in the whole prostate of (c) K5-PTEN 6w mice and (f) K5-PTEN-PIK 3w mice treated with EtOH or Rapamycin. n = 6 for K5-PTEN 6w mice, n = 4 for K5-PTEN-PIK 3w mice. (g) H&E-stained histological sections and (h) quantification of different types of tumorigenic lesions along prostate tumor progression in DLP of K5-PTEN 6w mice treated with EtOH (n = 7 mice) or Rapamycin (n = 8 mice). Scale bar, 250 μm (upper row), 50 μm (lower row). Graphs are mean ± s.e.m. p values are derived from two-sided unpaired t-test.
Extended Data Fig. 3 Marker genes for each cell type in control and in K5-PTEN 6w prostate epithelial cells.
Cell populations found by scRNA-seq in CTL prostate epithelial cells: (a) UMAP dimensionality reduction plots of CTL prostate epithelial cells with colors representing unsupervised clustering. (b-g) UMAP plots colored by normalized gene expression for (b) BC marker genes, (c) Proximal marker genes, (d) Ventral marker genes, (e) Lateral marker genes, (f) Antero-Dorsal marker genes and (g) Proliferative marker genes. (h) UMAP dimensionality reduction plots for K5-PTEN 6w using Seurat: UMAP dimensionality reduction plots with colors representing unsupervised clustering. (i–o) UMAP plots colored by normalized gene expression for (i) BC marker genes, (j) Proximal marker genes, (k) HY BC Prox marker genes, (l) HY Nkx3.1 marker genes, (m) Ventral marker genes, (n) Antero-Dorsal marker genes and (o) Proliferative marker genes.
Extended Data Fig. 4 Cellular trajectory and in situ characterization of Hillock, Hybrid and proximal-like LCs.
(a) Cell populations of K5-PTEN 6w, which are used for trajectory inference: UMAP dimensionality reduction plots with different colors representing unsupervised clustering. (b) UMAP plot for the K5-PTEN 6w dataset showing the trajectory from BC to proximal LCs. (c-d) UMAP plot colored by the adjusted proportion of (c) BC-specific marker genes and (d) LC-specific marker genes. (e-h) Representative immunostaining of AP/DLP of CTL, K5CreER/Ptenfl/fl/RosaYFP mice 6 weeks after TAM administration, K5CreER/Ptenfl/fl/Pik3caH1047R/RosaYFP mice 3 weeks after TAM administration and K5CreER/Ptenfl/fl/P53fl/fl/RosaYFP mice 4 weeks after TAM administration using (e) anti-K13, (f) anti-Aqp3, (g) anti-K4, (h) anti-Trop2 and anti-GFP (green), anti-K14 (grey) and anti-K8 (grey) antibodies. Arrows indicate Hillock cells, Hybrid cells and Proximal LCs as indicated markers. n = 3 mice. Dash lines marked the expanded LCs. L: prostate lumen. Scale bar, 20 μm.
Extended Data Fig. 5 scRNA‑seq characterize hybrid basal–luminal cell states and lineage trajectory in K5-PTEN 6w prostate epithelial cells.
(a) Cell populations of K5-PTEN 6w, which are used for trajectory inference: UMAP dimensionality reduction plots with different colors representing unsupervised clustering. (b) UMAP plot for the K5-PTEN 6w dataset showing the trajectory from BC to ventral LCs. (c-d) UMAP plot colored by the adjusted proportion of (c) BC-specific marker genes and (d) LC-specific marker genes. (e) Representative images of immunostaining of the VP of K5CreER/Ptenfl/fl/RosaYFP mice 6 months after TAM administration using anti-GFP (green), anti-Nkx3.1 (red). Scale bar, 50 μm. n = 3 mice. (f) Representative images of immunostaining of the VP, DLP and AP of K8CreER/Ptenfl/fl/RosaYFP mice 6 months after TAM administration using anti-GFP (green), anti-K13 (red), anti-K4 (red) and anti-K14 (grey) antibodies. Scale bar, 50 μm. n = 3 mice. (g) Genes significantly upregulated on HY BC Prox compared to BC p63 low and BC p63 high in K5-PTEN 6w scRNA dataset. (h) Genes significantly upregulated in HY BC Prox compared to proximal luminal cells. (i) Genes significantly upregulated on HY Nkx3.1 compared to the BC Nkx3.1. (j) Genes significantly upregulated in HY Nkx3.1 compared to the ventral prostate. (k-m) UMAP plots colored by (Left) SCENIC AUCs for regulon activation and (Right) normalized gene expression of the TF for (k) Elf3, (l) Grhl3 and (m) Creb5. For g to j, red dots are significantly differentially expressed genes (FDR-adjusted P value < 0.01) and blue-labeled genes are uniquely expressed on hybrid population. P values were calculated using the Wilcoxon rank-sum test and adjusted using the Benjamini–Hochberg FDR method.
Extended Data Fig. 6 Temporal analysis of the activation of innate immunity and ATAC-seq and bulk RNA-seq on different lobes after Pten deletion in BC.
(a) Relative mRNA expression levels of Il1a and Cxcl2 were determined by quantitative RT–PCR in FACS-isolated BCs of AP/DLP from CD1 mice and YFP+ BCs from of K5CreER/Ptenfl/fl/RosaYFP mice at indicated time after TAM injection. mean ± s.e.m. n = 3 mice. p values are derived from one-way ANOVA with Dunnett test. ATAC-seq peaks of (b) Krt13 and (c) Aqp3 genes from FACS-isolated BCs of VP, DLP and AP of K5CreER/Ptenfl/fl/RosaYFP mice 6 weeks after TAM administration. Scale for visualization: 0-52 (Krt13) and 0-30 (Aqp3). Peaks which are upregulated at least 2-fold on AP and DLP compared to VP are highlighted with orange box. (d) TF motif enrichment analysis of peaks upregulated in BCs of AP/DLP compared to BCs of VP in K5-PTEN 6w. P values were calculated using a binomial test. (e) Relative Erg expression from bulk RNA-seq in FACS-isolated CTL BC, CTL LC from CTL mice and YFP+ BCs (BC_PTEN) and YFP+ LCs arising from Pten deleted BCs (LC_PTEN) from K5CreER/Ptenfl/fl/RosaYFP mice 5 weeks after TAM injection. n = 2. (f) Relative Erg expression from bulk RNA-seq of FACS-isolated LCs from AP/DLP versus VP in K5CreER/Ptenfl/fl/RosaYFP mice 6 months following TAM administration. (n = 3 for AP/DLP, n = 2 for VP).
Extended Data Fig. 7 Activation of innate immunity is a mTOR dependent.
(a) Venn diagram illustrating the number of 2-fold upregulated genes in FACS-isolated LIN- YFP+ BCs of K5CreER/Ptenfl/fl/Pik3caH1047R/RosaYFP mice 3 weeks after TAM injection (vehicle treated, BC_PTEN-PIK) compared to FACS-isolated LIN- BCs of CD1 mice (vehicle treated, BC_CTL) and the number of 2-fold downregulated genes in FACS-isolated LIN- YFP+ BCs of K5CreER/Ptenfl/fl/Pik3caH1047R/RosaYFP mice 3 weeks after TAM injection treated with Rapamycin (BC_PTEN-PIK Rap) compared to BC_PTEN-PIK. (b) GO analysis of genes upregulated more than 2-fold in FACS-isolated BC_PTEN-PIK compared to FACS-isolated BC_CTL. (c) GO analysis of genes downregulated more than 2-fold in FACS-isolated BC_PTEN-PIK Rap compared to FACS-isolated BC_PTEN-PIK. (d) Expression of genes on bulk RNA-seq in FACS-isolated BC_CTL, BC_PTEN-PIK and BC_PTEN-PIK Rap. n = 2 samples. (e) Representative immunostaining of myeloid cells within AP from K5CreER/Ptenfl/fl/Pik3caH1047R/RosaYFP mice treated with EtOH or Rapamycin 3 weeks after TAM injection using anti-GFP (green), anti-Ly6G (red) antibodies. Scale bar, 20 μm. (f) Representative immunostaining of myeloid cells within the DLP from K5CreER/Ptenfl/fl/RosaYFP mice and K5CreER/Ptenfl/fl/P53fl/fl/RosaYFP mice 4 weeks after TAM injection using anti-GFP (green), anti-Ly6G (red) antibodies. Scale bar, 50 μm. For b and c, P values were derived from two-tailed modified Fisher’s Exact Test and adjusted using the Benjamini–Hochberg method.
Extended Data Fig. 8 Regional reprogramming and activation of innate immunity in BC-derived mouse prostate tumors.
(a) UMAP dimensionality reduction plots with different colors representing unsupervised clustering of scRNA-seq of FACS-isolated LIN- YFP+ epithelial cells from K5CreER/Ptenfl/fl/RosaYFP mice 10 months after TAM administration: (b-h) UMAP plots colored by normalized gene expression for (b) Proximal marker genes, (c) Proliferative marker genes, (d) HY BC Prox marker genes, (e) Distal LC marker genes, (f) Chemokine marker genes, (g) Interferons marker genes, (h) MHC Class II antigens marker genes. (i–n) Representative immunostaining and quantification of (i, j) K13, (k, l) Aqp3 and (m, n) K4 in VP, DLP and AP of K5CreER/Ptenfl/fl/RosaYFP mice 6 months following TAM administration. Scale bar, 50 μm. n = 4 mice for K13 and Aqp3, n = 3 mice for K4. (o) Relative mRNA expression from bulk RNA-seq of the indicated genes in FACS-isolated YFP+ LCs from AP/DLP and VP in K5CreER/Ptenfl/fl/RosaYFP mice 6 months following TAM administration. (n = 3 for AP/DLP, n = 2 for VP). Representative immunostainings of AdeK5Cre infected AP of (p) Ptenfl/fl/P53/RosaYFP mice and (q) Ptenfl/fl/Pik3caH1047R/RosaYFP mice 2 months after virus injection using the indicated antibodies. Scale bar, 50 μm. (r) Quantification of the % of K13 + , Aqp3+ and K4+ in total YFP+ and Ly6G+ cells in AdeK5Cre injected AP of Ptenfl/fl/Pik3caH1047R/RosaYFP mice. n = 3 infections. Graphs are mean ± s.e.m. p values are derived from one-way ANOVA with Tukey’s test.
Extended Data Fig. 9 Regional reprogramming in human prostate tumors.
(a) Cell populations on ERG-driven human prostate cancer cells, human prostate cancer atlas, treatment-naïve prostate adenocarcinoma and invasive cribriform carcinoma and intraductal carcinoma data. (b) UMAP dimensionality reduction plots for KRT13 and WFDC2 with color scaling representing the level of gene expression. (c-f) Heatmap of reprogramming marker genes defined on K5-PTEN 6 W scRNA-seq data on (c) ERG-driven human prostate cancer cells, (d) human prostate cancer atlas, (e) treatment-naïve prostate adenocarcinoma and (f) invasive cribriform carcinoma and intraductal carcinoma data. (g-i) Slingshot pseudotime trajectory analysis illustrating the lineage trajectory on (g) ERG-driven human prostate cancer cells, (h) human prostate cancer atlas and (i) invasive cribriform carcinoma and intraductal carcinoma data. (j) Summary of IHC staining results for PIGR and AQP3 in prostate tumor samples from 136 patients, categorized by GG groups of prostate cancer. Green boxes indicate co-staining of PIGR and AQP3.
Extended Data Fig. 10 Model of the early step of prostate tumor initiation.
Cell plasticity, lineage infidelity and tumor progression following Pten deletion occurs in a region-specific manner during the early stage of prostate cancer initiation and are mediated by the activation of innate immunity in prostate basal stem cells.
Supplementary information
Software policy checklist
Software policy checklist includes the information of public Github repository (https://github.com/yurasong/PTEN_codes) where the workflow and codes used for this publication are available. This checklist includes summary of system requirements, installation/demo steps and license details.
Supplementary Table
List of antibodies used in this study.
Source data
Source Data Fig. 1
Numerical source data for Fig. 1.
Source Data Fig. 2
Numerical source data for Fig. 2.
Source Data Fig. 3
Numerical source data for Fig. 3.
Source Data Fig. 4
Numerical source data for Fig. 4.
Source Data Fig. 5
Numerical source data for Fig. 5.
Source Data Fig. 6
Numerical source data for Fig. 6.
Source Data Fig. 7
Numerical source data for Fig. 7.
Source Data Fig. 8
Numerical source data for Fig. 8.
Source Data Extended Data Fig. 1
Numerical source data for Extended Data Fig. 1.
Source Data Extended Data Fig. 2
Numerical source data for Extended Data Fig. 2.
Source Data Extended Data Fig. 4
Numerical source data for Extended Data Fig. 4.
Source Data Extended Data Fig. 5
Numerical source data for Extended Data Fig. 5.
Source Data Extended Data Fig. 6
Numerical source data for Extended Data Fig. 6.
Source Data Extended Data Fig. 7
Numerical source data for Extended Data Fig. 7.
Source Data Extended Data Fig. 8
Numerical source data for Extended Data Fig. 8.
Source Data Extended Data Fig. 9
Numerical source data for Extended Data Fig. 9.
Source Data Fig. 5
Unprocessed western blots for Fig. 5.
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.
About this article
Cite this article
Jiang, C., Song, Y., Rorive, S. et al. Innate immunity and the NF-κB pathway control prostate stem cell plasticity, reprogramming and tumor initiation. Nat Cancer 6, 1537–1558 (2025). https://doi.org/10.1038/s43018-025-00994-3
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s43018-025-00994-3