Cargando…

Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms

Patients with metastatic prostate cancer frequently develop bone metastases that elicit significant skeletal morbidity and increased mortality. The high tropism of prostate cancer cells for bone and their tendency to induce the osteoblastic-like phenotype are a result of a complex interplay between...

Descripción completa

Detalles Bibliográficos
Autores principales: Ribelli, Giulia, Simonetti, Sonia, Iuliani, Michele, Rossi, Elisabetta, Vincenzi, Bruno, Tonini, Giuseppe, Pantano, Francesco, Santini, Daniele
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8711264/
https://www.ncbi.nlm.nih.gov/pubmed/34966687
http://dx.doi.org/10.3389/fonc.2021.789885
_version_ 1784623335168016384
author Ribelli, Giulia
Simonetti, Sonia
Iuliani, Michele
Rossi, Elisabetta
Vincenzi, Bruno
Tonini, Giuseppe
Pantano, Francesco
Santini, Daniele
author_facet Ribelli, Giulia
Simonetti, Sonia
Iuliani, Michele
Rossi, Elisabetta
Vincenzi, Bruno
Tonini, Giuseppe
Pantano, Francesco
Santini, Daniele
author_sort Ribelli, Giulia
collection PubMed
description Patients with metastatic prostate cancer frequently develop bone metastases that elicit significant skeletal morbidity and increased mortality. The high tropism of prostate cancer cells for bone and their tendency to induce the osteoblastic-like phenotype are a result of a complex interplay between tumor cells and osteoblasts. Although the role of osteoblasts in supporting prostate cancer cell proliferation has been reported by previous studies, their precise contribution in tumor growth remains to be fully elucidated. Here, we tried to dissect the molecular signaling underlining the interactions between castration-resistant prostate cancer (CRPC) cells and osteoblasts using in vitro co-culture models. Transcriptomic analysis showed that osteoblast-conditioned media (OCM) induced the overexpression of genes related to cell cycle in the CRPC cell line C4-2B but, surprisingly, reduced androgen receptor (AR) transcript levels. In-depth analysis of AR expression in C4-2B cells after OCM treatment showed an AR reduction at the mRNA (p = 0.0047), protein (p = 0.0247), and functional level (p = 0.0029) and, concomitantly, an increase of C4-2B cells in S-G2-M cell cycle phases (p = 0.0185). An extensive proteomic analysis revealed in OCM the presence of some molecules that reduced AR activation, and among these, Matrix metalloproteinase-1 (MMP-1) was the only one able to block AR function (0.1 ng/ml p = 0.006; 1 ng/ml p = 0.002; 10 ng/ml p = 0.0001) and, at the same time, enhance CRPC proliferation (1 ng/ml p = 0.009; 10 ng/ml p = 0.033). Although the increase of C4-2B cell growth induced by MMP-1 did not reach the proliferation levels observed after OCM treatment, the addition of Vorapaxar, an MMP-1 receptor inhibitor (Protease-activated receptor-1, PAR-1), significantly reduced C4-2B cell cycle (0.1 μM p = 0.014; 1 μM p = 0.0087). Overall, our results provide a novel AR-independent mechanism of CRPC proliferation and suggest that MMP-1/PAR-1 could be one of the potential pathways involved in this process.
format Online
Article
Text
id pubmed-8711264
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-87112642021-12-28 Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms Ribelli, Giulia Simonetti, Sonia Iuliani, Michele Rossi, Elisabetta Vincenzi, Bruno Tonini, Giuseppe Pantano, Francesco Santini, Daniele Front Oncol Oncology Patients with metastatic prostate cancer frequently develop bone metastases that elicit significant skeletal morbidity and increased mortality. The high tropism of prostate cancer cells for bone and their tendency to induce the osteoblastic-like phenotype are a result of a complex interplay between tumor cells and osteoblasts. Although the role of osteoblasts in supporting prostate cancer cell proliferation has been reported by previous studies, their precise contribution in tumor growth remains to be fully elucidated. Here, we tried to dissect the molecular signaling underlining the interactions between castration-resistant prostate cancer (CRPC) cells and osteoblasts using in vitro co-culture models. Transcriptomic analysis showed that osteoblast-conditioned media (OCM) induced the overexpression of genes related to cell cycle in the CRPC cell line C4-2B but, surprisingly, reduced androgen receptor (AR) transcript levels. In-depth analysis of AR expression in C4-2B cells after OCM treatment showed an AR reduction at the mRNA (p = 0.0047), protein (p = 0.0247), and functional level (p = 0.0029) and, concomitantly, an increase of C4-2B cells in S-G2-M cell cycle phases (p = 0.0185). An extensive proteomic analysis revealed in OCM the presence of some molecules that reduced AR activation, and among these, Matrix metalloproteinase-1 (MMP-1) was the only one able to block AR function (0.1 ng/ml p = 0.006; 1 ng/ml p = 0.002; 10 ng/ml p = 0.0001) and, at the same time, enhance CRPC proliferation (1 ng/ml p = 0.009; 10 ng/ml p = 0.033). Although the increase of C4-2B cell growth induced by MMP-1 did not reach the proliferation levels observed after OCM treatment, the addition of Vorapaxar, an MMP-1 receptor inhibitor (Protease-activated receptor-1, PAR-1), significantly reduced C4-2B cell cycle (0.1 μM p = 0.014; 1 μM p = 0.0087). Overall, our results provide a novel AR-independent mechanism of CRPC proliferation and suggest that MMP-1/PAR-1 could be one of the potential pathways involved in this process. Frontiers Media S.A. 2021-12-13 /pmc/articles/PMC8711264/ /pubmed/34966687 http://dx.doi.org/10.3389/fonc.2021.789885 Text en Copyright © 2021 Ribelli, Simonetti, Iuliani, Rossi, Vincenzi, Tonini, Pantano and Santini https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Ribelli, Giulia
Simonetti, Sonia
Iuliani, Michele
Rossi, Elisabetta
Vincenzi, Bruno
Tonini, Giuseppe
Pantano, Francesco
Santini, Daniele
Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title_full Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title_fullStr Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title_full_unstemmed Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title_short Osteoblasts Promote Prostate Cancer Cell Proliferation Through Androgen Receptor Independent Mechanisms
title_sort osteoblasts promote prostate cancer cell proliferation through androgen receptor independent mechanisms
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8711264/
https://www.ncbi.nlm.nih.gov/pubmed/34966687
http://dx.doi.org/10.3389/fonc.2021.789885
work_keys_str_mv AT ribelligiulia osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT simonettisonia osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT iulianimichele osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT rossielisabetta osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT vincenzibruno osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT toninigiuseppe osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT pantanofrancesco osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms
AT santinidaniele osteoblastspromoteprostatecancercellproliferationthroughandrogenreceptorindependentmechanisms