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:

Zeb1-controlled metabolic plasticity enables remodeling of chromatin accessibility in the development of neuroendocrine prostate cancer

Abstract

Cell plasticity has been found to play a critical role in tumor progression and therapy resistance. However, our understanding of the characteristics and markers of plastic cellular states during cancer cell lineage transition remains limited. In this study, multi-omics analyses show that prostate cancer cells undergo an intermediate state marked by Zeb1 expression with epithelial-mesenchymal transition (EMT), stemness, and neuroendocrine features during the development of neuroendocrine prostate cancer (NEPC). Organoid-formation assays and in vivo lineage tracing experiments demonstrate that Zeb1+ epithelioid cells are putative cells of origin for NEPC. Mechanistically, Zeb1 transcriptionally regulates the expression of several key glycolytic enzymes, thereby predisposing tumor cells to utilize glycolysis for energy metabolism. During this process, lactate accumulation-mediated histone lactylation enhances chromatin accessibility and cellular plasticity including induction of neuro-gene expression, which promotes NEPC development. Collectively, Zeb1-driven metabolic rewiring enables the epigenetic reprogramming of prostate cancer cells to license the adeno-to-neuroendocrine lineage transition.

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: Zeb1 is dynamically expressed during the course of NEPC development.
Fig. 2: Single-cell sequencing data reveal dynamic changes of Zeb1 expression during human and mouse NEPC development.
Fig. 3: Zeb1+ epithelioid cells exhibit a more accessible chromatin with an expression characteristic of neuroactive and EMT program.
Fig. 4: Zeb1+ epithelioid cells can generate organoids with neuroendocrine characteristics.
Fig. 5: Lineage tracing indicates that Zeb1+ epithelioid cells are the cellular origin of NEPC.
Fig. 6: Zeb1 promotes glycolysis and accumulation of lactate in PCa.
Fig. 7: Lactate accumulation in Zeb1+ epithelioid cells enhances histone lactylation conferring lineage plasticity in PCa.

Similar content being viewed by others

Data availability

RNA-seq, ATAC-seq and CUT & Tag data in this study have been deposited to the National Genomics Data Center, China National Center for Bioinformation with Bioproject number PRJCA020095.

References

  1. Quintanal-Villalonga A, Chan JM, Yu HA, Pe’er D, Sawyers CL, Sen T, et al. Lineage plasticity in cancer: a shared pathway of therapeutic resistance. Nat Rev Clin Oncol. 2020;17:360–71.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Davies AH, Beltran H, Zoubeidi A. Cellular plasticity and the neuroendocrine phenotype in prostate cancer. Nat Rev Urol. 2018;15:271–86.

    Article  CAS  PubMed  Google Scholar 

  3. Dong B, Miao J, Wang Y, Luo W, Ji Z, Lai H, et al. Single-cell analysis supports a luminal-neuroendocrine transdifferentiation in human prostate cancer. Commun Biol. 2020;3:778.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Soundararajan R, Paranjape AN, Maity S, Aparicio A, Mani SA. EMT, stemness and tumor plasticity in aggressive variant neuroendocrine prostate cancers. Biochim Biophys Acta Rev Cancer. 2018;1870:229–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Zou M, Toivanen R, Mitrofanova A, Floch N, Hayati S, Sun Y, et al. Transdifferentiation as a mechanism of treatment resistance in a mouse model of castration-resistant prostate cancer. Cancer Discov. 2017;7:736–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Santoni M, Conti A, Burattini L, Berardi R, Scarpelli M, Cheng L, et al. Neuroendocrine differentiation in prostate cancer: novel morphological insights and future therapeutic perspectives. Biochim Biophys Acta. 2014;1846:630–7.

    CAS  PubMed  Google Scholar 

  7. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22:298–305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Bakir B, Chiarella AM, Pitarresi JR, Rustgi AK. EMT, MET, plasticity, and tumor metastasis. Trends cell Biol. 2020;30:764–76.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Lu W, Kang Y. Epithelial-mesenchymal plasticity in cancer progression and metastasis. Dev Cell. 2019;49:361–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139:871–90.

    Article  CAS  PubMed  Google Scholar 

  11. Li R, Liang J, Ni S, Zhou T, Qing X, Li H, et al. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell. 2010;7:51–63.

    Article  CAS  PubMed  Google Scholar 

  12. Liu X, Sun H, Qi J, Wang L, He S, Liu J, et al. Sequential introduction of reprogramming factors reveals a time-sensitive requirement for individual factors and a sequential EMT-MET mechanism for optimal reprogramming. Nat Cell Biol. 2013;15:829–38.

    Article  CAS  PubMed  Google Scholar 

  13. Boumahdi S, de Sauvage FJ. The great escape: tumour cell plasticity in resistance to targeted therapy. Nat Rev Drug Discov. 2020;19:39–56.

    Article  CAS  PubMed  Google Scholar 

  14. Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, et al. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527:525–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Noman MZ, Janji B, Abdou A, Hasmim M, Terry S, Tan TZ, et al. The immune checkpoint ligand PD-L1 is upregulated in EMT-activated human breast cancer cells by a mechanism involving ZEB-1 and miR-200. Oncoimmunology. 2017;6:e1263412.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Kurrey NK, Jalgaonkar SP, Joglekar AV, Ghanate AD, Chaskar PD, Doiphode RY, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells. 2009;27:2059–68.

    Article  CAS  PubMed  Google Scholar 

  17. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527:472–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dongre A, Rashidian M, Reinhardt F, Bagnato A, Keckesova Z, Ploegh HL, et al. Epithelial-to-mesenchymal transition contributes to immunosuppression in breast carcinomas. Cancer Res. 2017;77:3982–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Arumugam T, Ramachandran V, Fournier KF, Wang H, Marquis L, Abbruzzese JL, et al. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009;69:5820–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Thakur C, Chen F. Connections between metabolism and epigenetics in cancers. Semin Cancer Biol. 2019;57:52–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Dai Z, Ramesh V, Locasale JW. The evolving metabolic landscape of chromatin biology and epigenetics. Nat Rev Genet. 2020;21:737–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. He Y, Ji Z, Gong Y, Fan L, Xu P, Chen X, et al. Numb/Parkin-directed mitochondrial fitness governs cancer cell fate via metabolic regulation of histone lactylation. Cell Rep. 2023;42:112033.

    Article  CAS  PubMed  Google Scholar 

  23. Wang X, Xu H, Cheng C, Ji Z, Zhao H, Sheng Y, et al. Identification of a Zeb1 expressing basal stem cell subpopulation in the prostate. Nat Commun. 2020;11:706.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Greenberg NM, DeMayo F, Finegold MJ, Medina D, Tilley WD, Aspinall JO, et al. Prostate cancer in a transgenic mouse. Proc Natl Acad Sci USA. 1995;92:3439–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Han M, Li F, Zhang Y, Dai P, He J, Li Y, et al. FOXA2 drives lineage plasticity and KIT pathway activation in neuroendocrine prostate cancer. Cancer Cell. 2022;40:1306–23 e8.

    Article  CAS  PubMed  Google Scholar 

  26. Qiu X, Mao Q, Tang Y, Wang L, Chawla R, Pliner HA, et al. Reversed graph embedding resolves complex single-cell trajectories. Nat Methods. 2017;14:979–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Faure L, Soldatov R, Kharchenko PV, Adameyko I. scFates: a scalable python package for advanced pseudotime and bifurcation analysis from single-cell data. Bioinformatics. 2023;39:btac746.

    Article  CAS  PubMed  Google Scholar 

  28. Guo W, Keckesova Z, Donaher JL, Shibue T, Tischler V, Reinhardt F, et al. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148:1015–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mazzu YZ, Liao Y, Nandakumar S, Sjostrom M, Jehane LE, Ghale R, et al. Dynamic expression of SNAI2 in prostate cancer predicts tumor progression and drug sensitivity. Mol Oncol. 2022;16:2451–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kahounova Z, Remsik J, Fedr R, Bouchal J, Mickova A, Slabakova E, et al. Slug-expressing mouse prostate epithelial cells have increased stem cell potential. Stem Cell Res. 2020;46:101844.

    Article  CAS  PubMed  Google Scholar 

  31. Wang Z, Wang T, Hong D, Dong B, Wang Y, Huang H, et al. Single-cell transcriptional regulation and genetic evolution of neuroendocrine prostate cancer. iScience. 2022;25:104576.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Aigner K, Dampier B, Descovich L, Mikula M, Sultan A, Schreiber M, et al. The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene. 2007;26:6979–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Vannier C, Mock K, Brabletz T, Driever W. Zeb1 regulates E-cadherin and Epcam (epithelial cell adhesion molecule) expression to control cell behavior in early zebrafish development. J Biol Chem. 2013;288:18643–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Feldker N, Ferrazzi F, Schuhwerk H, Widholz SA, Guenther K, Frisch I, et al. Genome-wide cooperation of EMT transcription factor ZEB1 with YAP and AP-1 in breast cancer. EMBO J. 2020;39:e103209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Izzo LT, Wellen KE. Histone lactylation links metabolism and gene regulation. Nature. 2019;574:492–3.

    Article  CAS  PubMed  Google Scholar 

  36. Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Li L, Chen K, Wang T, Wu Y, Xing G, Chen M, et al. Glis1 facilitates induction of pluripotency via an epigenome-metabolome-epigenome signalling cascade. Nat Metab. 2020;2:882–92.

    Article  CAS  PubMed  Google Scholar 

  38. Sun Y, Wang BE, Leong KG, Yue P, Li L, Jhunjhunwala S, et al. Androgen deprivation causes epithelial-mesenchymal transition in the prostate: implications for androgen-deprivation therapy. Cancer Res. 2012;72:527–36.

    Article  CAS  PubMed  Google Scholar 

  39. Miao L, Yang L, Li R, Rodrigues DN, Crespo M, Hsieh JT, et al. Disrupting androgen receptor signaling induces snail-mediated epithelial-mesenchymal plasticity in prostate cancer. Cancer Res. 2017;77:3101–12.

    Article  CAS  PubMed  Google Scholar 

  40. Wilson MM, Weinberg RA, Lees JA, Guen VJ. Emerging mechanisms by which EMT programs control stemness. Trends Cancer. 2020;6:775–80.

    Article  CAS  PubMed  Google Scholar 

  41. Yang J, Antin P, Berx G, Blanpain C, Brabletz T, Bronner M, et al. Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2020;21:341–52.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Postigo AA, Depp JL, Taylor JJ, Kroll KL. Regulation of Smad signaling through a differential recruitment of coactivators and corepressors by ZEB proteins. EMBO J. 2003;22:2453–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Malouf GG, Taube JH, Lu Y, Roysarkar T, Panjarian S, Estecio MR, et al. Architecture of epigenetic reprogramming following Twist1-mediated epithelial-mesenchymal transition. Genome Biol. 2013;14:R144.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Taube JH, Sphyris N, Johnson KS, Reisenauer KN, Nesbit TA, Joseph R, et al. The H3K27me3-demethylase KDM6A is suppressed in breast cancer stem-like cells, and enables the resolution of bivalency during the mesenchymal-epithelial transition. Oncotarget. 2017;8:65548–65.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Chaffer CL, Marjanovic ND, Lee T, Bell G, Kleer CG, Reinhardt F, et al. Poised chromatin at the ZEB1 promoter enables breast cancer cell plasticity and enhances tumorigenicity. Cell. 2013;154:61–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The study was supported by funds from the National Key R&D Program of China (2022YFA1302704 and 2023YFC1404101), the National Natural Science Foundation of China U23A20454, NSFC82372873, NSFC32022021, and NSFC81872406, Shanghai Pilot Program for Basic Research-Shanghai Jiao Tong University (21TQ1400225), the Program of Shanghai Academic/Technology Research Leader (21XD1422300), the Shanghai Municipal Education Commission–Gaofeng Clinical Medicine Grant Support (20181706), the Innovative research team of high-level local universities in Shanghai, 111 project (B21024), Peak Disciplines (Type IV) of Institutions of Higher Learning in Shanghai, the KC Wong foundation.

Author information

Authors and Affiliations

Authors

Contributions

HHZ and WQG conceived the study; DW performed the experiments; GD, XC, JW, and NW supported data analysis; KL, NJ, YS, YL, and XX assisted in animal experiments; HZ and PX helped in construction of plasmids; YH and YZ participated in immunostaining and imaging; CC, WB, KZ, and PZ helped in experiments design. HHZ, WQG, and DW interpreted the data and wrote the manuscript.

Corresponding authors

Correspondence to Wei-Qiang Gao or Helen He Zhu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

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

Wang, D., Du, G., Chen, X. et al. Zeb1-controlled metabolic plasticity enables remodeling of chromatin accessibility in the development of neuroendocrine prostate cancer. Cell Death Differ (2024). https://doi.org/10.1038/s41418-024-01295-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41418-024-01295-5

Search

Quick links