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:

Neuropilin-2 regulates androgen-receptor transcriptional activity in advanced prostate cancer

Abstract

Aberrant transcriptional activity of androgen receptor (AR) is one of the dominant mechanisms for developing of castration-resistant prostate cancer (CRPC). Analyzing AR-transcriptional complex related to CRPC is therefore important towards understanding the mechanism of therapy resistance. While studying its mechanism, we observed that a transmembrane protein called neuropilin-2 (NRP2) plays a contributory role in forming a novel AR-transcriptional complex containing nuclear pore proteins. Using immunogold electron microscopy, high-resolution confocal microscopy, chromatin immunoprecipitation, proteomics, and other biochemical techniques, we delineated the molecular mechanism of how a specific splice variant of NRP2 becomes sumoylated upon ligand stimulation and translocates to the inner nuclear membrane. This splice variant of NRP2 then stabilizes the complex between AR and nuclear pore proteins to promote CRPC specific gene expression. Both full-length and splice variants of AR have been identified in this specific transcriptional complex. In vitro cell line-based assays indicated that depletion of NRP2 not only destabilizes the AR-nuclear pore protein interaction but also inhibits the transcriptional activities of AR. Using an in vivo bone metastasis model, we showed that the inhibition of NRP2 led to the sensitization of CRPC cells toward established anti-AR therapies such as enzalutamide. Overall, our finding emphasize the importance of combinatorial inhibition of NRP2 and AR as an effective therapeutic strategy against treatment refractory prostate cancer.

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: NRP2 localization detected within the nucleus.
Fig. 2: NRP2 present in the nuclear membrane of the cell.
Fig. 3: VEGF-C-mediated retrograde transport and post-translational SUMOylation of NRP2B.
Fig. 4: NRP2 Mass-Spectrometry predicted its interactions with various nuclear-associated proteins.
Fig. 5: Analysis of AR-NRP2 regulated gene expression.
Fig. 6: AR-DNA interaction decreases following NRP2 depletion.
Fig. 7: Inhibition of nuclear transport of NRP2 increased AR-targeted therapeutic efficacy.

Similar content being viewed by others

References

  1. Powers E, Karachaliou GS, Kao C, Harrison MR, Hoimes CJ, George DJ, et al. Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol. 2020;13:144.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Schmidt KT, Huitema ADR, Chau CH, Figg WD. Resistance to second-generation androgen receptor antagonists in prostate cancer. Nat Rev Urol. 2021;18:209–26.

    Article  CAS  PubMed  Google Scholar 

  3. Imamura Y, Sadar MD. Androgen receptor targeted therapies in castration-resistant prostate cancer: Bench to clinic. Int J Urol. 2016;23:654–65.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Tucci M, Zichi C, Buttigliero C, Vignani F, Scagliotti GV, Di Maio M. Enzalutamide-resistant castration-resistant prostate cancer: challenges and solutions. Onco Targets Ther. 2018;11:7353–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Giacinti S, Poti G, Roberto M, Macrini S, Bassanelli M, DIP F, et al. Molecular Basis of Drug Resistance and Insights for New Treatment Approaches in mCRPC. Anticancer Res. 2018;38:6029–39.

    Article  CAS  PubMed  Google Scholar 

  6. Roumiguie M, Paoletti X, Neuzillet Y, Mathieu R, Vincendeau S, Kleinclauss F, et al. Apalutamide, darolutamide and enzalutamide in nonmetastatic castration-resistant prostate cancer: a meta-analysis. Future Oncol. 2021;17:1811–23.

    Article  CAS  PubMed  Google Scholar 

  7. Tucci M, Leone G, Buttigliero C, Zichi C, DI Stefano RF, Pignataro D, et al. Hormonal treatment and quality of life of prostate cancer patients: new evidence. Minerva Urol Nefrol. 2018;70:144–51.

    PubMed  Google Scholar 

  8. Vander Ark A, Cao J, Li X. Mechanisms and approaches for overcoming enzalutamide resistance in prostate cancer. Front Oncol. 2018;8:180.

    Article  Google Scholar 

  9. He Y, Wei T, Ye Z, Orme JJ, Lin D, Sheng H, et al. A noncanonical AR addiction drives enzalutamide resistance in prostate cancer. Nat Commun. 2021;12:1521.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Schweizer MT, Haugk K, McKiernan JS, Gulati R, Cheng HH, Maes JL, et al. A phase I study of niclosamide in combination with enzalutamide in men with castration-resistant prostate cancer. PloS ONE. 2018;13:e0198389.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Shafran JS, Andrieu GP, Gyorffy B, Denis GV. BRD4 regulates metastatic potential of castration-resistant prostate cancer through AHNAK. Mol Cancer Res. 2019;17:1627–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Asangani IA, Dommeti VL, Wang X, Malik R, Cieslik M, Yang R, et al. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature. 2014;510:278–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Coleman DJ, Gao L, Schwartzman J, Korkola JE, Sampson D, Derrick DS, et al. Maintenance of MYC expression promotes de novo resistance to BET bromodomain inhibition in castration-resistant prostate cancer. Sci Rep. 2019;9:3823.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Wang L, Xu M, Kao CY, Tsai SY, Tsai MJ. Small molecule JQ1 promotes prostate cancer invasion via BET-independent inactivation of FOXA1. J Clin Investig. 2020;130:1782–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sulpice E, Plouet J, Berge M, Allanic D, Tobelem G, Merkulova-Rainon T. Neuropilin-1 and neuropilin-2 act as coreceptors, potentiating proangiogenic activity. Blood. 2008;111:2036–45.

    Article  CAS  PubMed  Google Scholar 

  16. Parker MW, Linkugel AD, Goel HL, Wu T, Mercurio AM, Vander, et al. Structural basis for VEGF-C binding to neuropilin-2 and sequestration by a soluble splice form. Structure. 2015;23:677–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Rossignol M, Gagnon ML, Klagsbrun M. Genomic organization of human neuropilin-1 and neuropilin-2 genes: identification and distribution of splice variants and soluble isoforms. Genomics. 2000;70:211–22.

    Article  CAS  PubMed  Google Scholar 

  18. Fricker M, Hollinshead M, White N, Vaux D. Interphase nuclei of many mammalian cell types contain deep, dynamic, tubular membrane-bound invaginations of the nuclear envelope. J Cell Biol. 1997;136:531–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Drozdz MM, Vaux DJ. Shared mechanisms in physiological and pathological nucleoplasmic reticulum formation. Nucleus. 2017;8:34–45.

    Article  CAS  PubMed  Google Scholar 

  20. Ibarra A, Hetzer MW. Nuclear pore proteins and the control of genome functions. Genes Dev. 2015;29:337–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rodriguez-Navarro S, Fischer T, Luo MJ, Antunez O, Brettschneider S, Lechner J, et al. Sus1, a functional component of the SAGA histone acetylase complex and the nuclear pore-associated mRNA export machinery. Cell. 2004;116:75–86.

    Article  CAS  PubMed  Google Scholar 

  22. Garcia-Oliver E, Garcia-Molinero V, Rodriguez-Navarro S. mRNA export and gene expression: the SAGA-TREX-2 connection. Biochim Biophys Acta. 2012;1819:555–65.

    Article  CAS  PubMed  Google Scholar 

  23. Labade AS, Karmodiya K, Sengupta K. HOXA repression is mediated by nucleoporin Nup93 assisted by its interactors Nup188 and Nup205. Epigenetics Chromatin. 2016;9:54.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Sump B, Brickner JH. Nup98 regulation of histone methylation promotes normal gene expression and may drive leukemogenesis. Genes Dev. 2017;31:2201–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Franks TM, Hetzer MW. The role of Nup98 in transcription regulation in healthy and diseased cells. Trends Cell Biol. 2013;23:112–7.

    Article  CAS  PubMed  Google Scholar 

  26. Liang Y, Franks TM, Marchetto MC, Gage FH, Hetzer MW. Dynamic association of NUP98 with the human genome. PLoS Genet. 2013;9:e1003308.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dutta S, Roy S, Polavaram NS, Baretton GB, Muders MH, Batra S, et al. NRP2 transcriptionally regulates its downstream effector WDFY1. Sci Rep. 2016;6:23588.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Coutinho I, Day TK, Tilley WD, Selth LA. Androgen receptor signaling in castration-resistant prostate cancer: a lesson in persistence. Endocr Relat Cancer. 2016;23:T179–97.

    Article  CAS  PubMed  Google Scholar 

  29. Sharma NL, Massie CE, Ramos-Montoya A, Zecchini V, Scott HE, Lamb AD, et al. The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. Cancer Cell. 2013;23:35–47.

    Article  CAS  PubMed  Google Scholar 

  30. Massie CE, Lynch A, Ramos-Montoya A, Boren J, Stark R, Fazli L, et al. The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO J. 2011;30:2719–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tan KN, Avery VM, Carrasco-Pozo C. Metabolic roles of androgen receptor and Tip60 in androgen-dependent prostate cancer. Int J Mol Sci. 2020;21:6622.

    Article  PubMed Central  CAS  Google Scholar 

  32. Borkowetz A, Froehner M, Rauner M, Conrad S, Erdmann K, Mayr T, et al. Neuropilin-2 is an independent prognostic factor for shorter cancer-specific survival in patients with Acinar adenocarcinoma of the prostate. Int J Cancer. 2020;146:2619–27.

    Article  CAS  PubMed  Google Scholar 

  33. Palancade B, Liu X, Garcia-Rubio M, Aguilera A, Zhao X, Doye V. Nucleoporins prevent DNA damage accumulation by modulating Ulp1-dependent sumoylation processes. Mol Biol Cell. 2007;18:2912–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ruben GJ, Kirkland JG, MacDonough T, Chen M, Dubey RN, Gartenberg MR, et al. Nucleoporin mediated nuclear positioning and silencing of HMR. PloS ONE. 2011;6:e21923.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Radman-Livaja M, Ruben G, Weiner A, Friedman N, Kamakaka R, Rando OJ. Dynamics of Sir3 spreading in budding yeast: secondary recruitment sites and euchromatic localization. EMBO J. 2011;30:1012–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kuhn TM, Capelson M. Nuclear pore proteins in regulation of chromatin state. Cells. 2019;8:1414.

    Article  PubMed Central  CAS  Google Scholar 

  37. Kuhn TM, Pascual-Garcia P, Gozalo A, Little SC, Capelson M. Chromatin targeting of nuclear pore proteins induces chromatin decondensation. J Cell Biol. 2019;218:2945–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Raices M, D’Angelo MA. Nuclear pore complexes and regulation of gene expression. Curr Opin cell Biol. 2017;46:26–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ptak C, Aitchison JD, Wozniak RW. The multifunctional nuclear pore complex: a platform for controlling gene expression. Curr Opin Cell Biol. 2014;28:46–53.

    Article  CAS  PubMed  Google Scholar 

  40. Dieppois G, Stutz F. Connecting the transcription site to the nuclear pore: a multi-tether process that regulates gene expression. J Cell Sci. 2010;123:1989–99.

    Article  CAS  PubMed  Google Scholar 

  41. Ibarra A, Benner C, Tyagi S, Cool J, Hetzer MW. Nucleoporin-mediated regulation of cell identity genes. Genes Dev. 2016;30:2253–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kitazawa T, Rijli FM. Nuclear pore protein meets transcription factor in neural fate. Neuron. 2017;96:259–61.

    Article  CAS  PubMed  Google Scholar 

  43. Gomez-Cavazos JS, Hetzer MW. The nucleoporin gp210/Nup210 controls muscle differentiation by regulating nuclear envelope/ER homeostasis. J Cell Biol. 2015;208:671–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. D’Angelo MA. Nuclear pore complexes as hubs for gene regulation. Nucleus. 2018;9:142–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Raices M, Bukata L, Sakuma S, Borlido J, Hernandez LS, Hart DO, et al. Nuclear pores regulate muscle development and maintenance by assembling a localized Mef2C complex. Dev Cell. 2017;41:540–54.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Holzer K, Ori A, Cooke A, Dauch D, Drucker E, Riemenschneider P, et al. Nucleoporin Nup155 is part of the p53 network in liver cancer. Nat Commun. 2019;10:2147.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Rodriguez-Bravo V, Pippa R, Song WM, Carceles-Cordon M, Dominguez-Andres A, Fujiwara N, et al. Nuclear pores promote lethal prostate cancer by increasing POM121-driven E2F1, MYC, and AR nuclear import. Cell. 2018;174:1200–15.e20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Su Y, Pelz C, Huang T, Torkenczy K, Wang X, Cherry A, et al. Post-translational modification localizes MYC to the nuclear pore basket to regulate a subset of target genes involved in cellular responses to environmental signals. Genes Dev. 2018;32:1398–419.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gemmill RM, Nasarre P, Nair-Menon J, Cappuzzo F, Landi L, D'Incecco A, et al. The neuropilin 2 isoform NRP2b uniquely supports TGFbeta-mediated progression in lung cancer. Sci Signal. 2017;10:eaag0528.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Savoy RM, Chen L, Siddiqui S, Melgoza FU, Durbin-Johnson B, Drake C, et al. Transcription of Nrdp1 by the androgen receptor is regulated by nuclear filamin A in prostate cancer. Endocr Relat Cancer. 2015;22:369–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Stanton MJ, Dutta S, Zhang H, Polavaram NS, Leontovich AA, Honscheid P, et al. Autophagy control by the VEGF-C/NRP-2 axis in cancer and its implication for treatment resistance. Cancer research. 2013;73:160–71.

    Article  CAS  PubMed  Google Scholar 

  52. Polavaram NS, Dutta S, Islam R, Bag AK, Roy S, Poitz D, et al. Tumor- and osteoclast-derived NRP2 in prostate cancer bone metastases. Bone Res. 2021;9:24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kersemans V, Cornelissen B, Allen PD, Beech JS, Smart SC. Subcutaneous tumor volume measurement in the awake, manually restrained mouse using MRI. J Magn Reson Imaging. 2013;37:1499–504.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank all stuff members of the Advanced Microscopy Core and Genomics Core Facility of UNMC. We also extend our sincere thanks to the UNMC Bioinformatics and Bio-statistician Core for their support to analyze the data. The raw data has been deposited in Gene Expression Omnibus with the accession number GSE205150.

Funding

This work was supported by grants for SD (1R21CA241234-01, NE-LB506, Lageschulte Fund), KD (R01CA182435, R01CA239343, DoD W81XWH2110628), MHM and LHH (DFG project number 273676790), and MHM (DFG project number 416001651). MM is funded by the Rudolf-Becker-Foundation for his professorship. The construction of the prostate cancer tissue microarray was funded by the DFG Forschergruppe-1586 SKELMET to LCH and SF.

Author information

Authors and Affiliations

Authors

Contributions

SD and KD has designed the project and SD performed most of the work. NSP, RI, SB, SB, SR have assisted some of the work. TM has developed the NRP2-HA-tagged plasmid. SAA did the mass spec. AD took the electron microscopic images. MI, AB, SC, SF, MW, GBB, LCH, MHM were involved in TMA development and staining of NRP2. PG help us in promoter assay. PG, KJP, SKB and MHM critically evaluated the work and time to time provide there suggestion. DLK performed and analyzed the ChIP-seq.

Corresponding authors

Correspondence to Samikshan Dutta or Kaustubh Datta.

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dutta, S., Polavaram, N.S., Islam, R. et al. Neuropilin-2 regulates androgen-receptor transcriptional activity in advanced prostate cancer. Oncogene 41, 3747–3760 (2022). https://doi.org/10.1038/s41388-022-02382-y

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41388-022-02382-y

This article is cited by

Search

Quick links