The University of Southampton
University of Southampton Institutional Repository

V-ATPase inhibition decreases mutant androgen receptor activity in castrate-resistant prostate cancer

V-ATPase inhibition decreases mutant androgen receptor activity in castrate-resistant prostate cancer
V-ATPase inhibition decreases mutant androgen receptor activity in castrate-resistant prostate cancer

Prostate cancer is critically dependent on androgen receptor (AR) signaling. Despite initial responsiveness to androgen deprivation, most patients with advanced prostate cancer subsequently progress to a clinically aggressive castrate-resistant prostate cancer (CRPC) phenotype, typically associated with expression of splice-variant or mutant AR forms. Although current evidence suggests that the vacuolar-ATPase (V-ATPase), a multiprotein complex that catalyzes proton transport across intracellular and plasma membranes, influences wild-type AR function, the effect of V-ATPase inhibition on variant AR function is unknown. Inhibition of V-ATPase reduced AR function in wild-type and mutant AR luciferase reporter models. In hormone-sensitive prostate cancer cell lines (LNCaP, DuCaP) and mutant AR CRPC cell lines (22Rv1, LNCaP-F877L/T878A), V-ATPase inhibition using bafilomycin-A1 and concanamycin-A reduced AR expression, and expression of AR target genes, at mRNA and protein levels. Furthermore, combining chemical V-ATPase inhibition with the AR antagonist enzalutamide resulted in a greater reduction in AR downstream target expression than enzalutamide alone in LNCaP cells. To investigate the role of individual subunit isoforms, siRNA and CRISPR-Cas9 were used to target the V 1C1 subunit in 22Rv1 cells. Whereas transfection with ATP6V1C1-targeted siRNA significantly reduced AR protein levels and function, CRISPR-Cas9–mediated V 1C1 knockout showed no substantial change in AR expression, but a compensatory increase in protein levels of the alternate V 1C2 isoform. Overall, these results indicate that V-ATPase dysregulation is directly linked to both hormone-responsive prostate cancer and CRPC via impact on AR function. In particular, V-ATPase inhibition can reduce AR signaling regardless of mutant AR expression.

1535-7163
739-748
Whitton, Bradleigh
352554cc-cbf8-4d58-8c8f-51721abfe743
Okamoto, Haruko
cea35380-7618-44c8-a268-47b0198cc7f9
Rose-Zerilli, Matthew
29603401-e310-4054-b818-8a542c361b9a
Packham, Graham
fdabe56f-2c58-469c-aadf-38878f233394
Crabb, Simon J
bcd1b566-7677-4f81-8429-3ab0e85f8373
Whitton, Bradleigh
352554cc-cbf8-4d58-8c8f-51721abfe743
Okamoto, Haruko
cea35380-7618-44c8-a268-47b0198cc7f9
Rose-Zerilli, Matthew
29603401-e310-4054-b818-8a542c361b9a
Packham, Graham
fdabe56f-2c58-469c-aadf-38878f233394
Crabb, Simon J
bcd1b566-7677-4f81-8429-3ab0e85f8373

Whitton, Bradleigh, Okamoto, Haruko, Rose-Zerilli, Matthew, Packham, Graham and Crabb, Simon J (2021) V-ATPase inhibition decreases mutant androgen receptor activity in castrate-resistant prostate cancer. Molecular Cancer Therapeutics, 20 (4), 739-748. (doi:10.1158/1535-7163.MCT-20-0662).

Record type: Article

Abstract

Prostate cancer is critically dependent on androgen receptor (AR) signaling. Despite initial responsiveness to androgen deprivation, most patients with advanced prostate cancer subsequently progress to a clinically aggressive castrate-resistant prostate cancer (CRPC) phenotype, typically associated with expression of splice-variant or mutant AR forms. Although current evidence suggests that the vacuolar-ATPase (V-ATPase), a multiprotein complex that catalyzes proton transport across intracellular and plasma membranes, influences wild-type AR function, the effect of V-ATPase inhibition on variant AR function is unknown. Inhibition of V-ATPase reduced AR function in wild-type and mutant AR luciferase reporter models. In hormone-sensitive prostate cancer cell lines (LNCaP, DuCaP) and mutant AR CRPC cell lines (22Rv1, LNCaP-F877L/T878A), V-ATPase inhibition using bafilomycin-A1 and concanamycin-A reduced AR expression, and expression of AR target genes, at mRNA and protein levels. Furthermore, combining chemical V-ATPase inhibition with the AR antagonist enzalutamide resulted in a greater reduction in AR downstream target expression than enzalutamide alone in LNCaP cells. To investigate the role of individual subunit isoforms, siRNA and CRISPR-Cas9 were used to target the V 1C1 subunit in 22Rv1 cells. Whereas transfection with ATP6V1C1-targeted siRNA significantly reduced AR protein levels and function, CRISPR-Cas9–mediated V 1C1 knockout showed no substantial change in AR expression, but a compensatory increase in protein levels of the alternate V 1C2 isoform. Overall, these results indicate that V-ATPase dysregulation is directly linked to both hormone-responsive prostate cancer and CRPC via impact on AR function. In particular, V-ATPase inhibition can reduce AR signaling regardless of mutant AR expression.

Text
MCT-20-0662R1_Merged_PDF - Accepted Manuscript
Download (4MB)

More information

Accepted/In Press date: 5 February 2021
e-pub ahead of print date: 9 February 2021
Published date: 1 April 2021
Additional Information: Funding Information: B. Whitton was funded by a doctoral studentship from The Urology Foundation, Wessex Medical Research, and The Gerald Kerkut Charitable Trust. This work was also supported by grants from Cancer Research UK (C2750/A23669). Funding Information: G. Packham reports grants from The Urology Foundation, Wessex Medical Research, The Gerald Kerkut Charitable Trust, and Cancer Research UK during the conduct of the study. S.J. Crabb reports personal fees from Roche, Janssen Cilag, MSD, Pfizer, and Bayer; grants and personal fees from AstraZeneca; and grants from Astex Pharmaceuticals and Clovis Oncology outside the submitted work. No potential conflicts of interest were disclosed by the other authors. Publisher Copyright: © 2021 American Association for Cancer Research.

Identifiers

Local EPrints ID: 447023
URI: http://eprints.soton.ac.uk/id/eprint/447023
ISSN: 1535-7163
PURE UUID: 73bd4512-1b42-4286-8a08-14828f7facd4
ORCID for Graham Packham: ORCID iD orcid.org/0000-0002-9232-5691
ORCID for Simon J Crabb: ORCID iD orcid.org/0000-0003-3521-9064

Catalogue record

Date deposited: 02 Mar 2021 17:30
Last modified: 17 Mar 2024 06:20

Export record

Altmetrics

Contributors

Author: Bradleigh Whitton
Author: Haruko Okamoto
Author: Matthew Rose-Zerilli
Author: Graham Packham ORCID iD
Author: Simon J Crabb ORCID iD

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×