Cargando…

The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells

The tumor suppressor p53 is a key mediator of cellular stress and DNA damage response cascades and is activated after exposure to ionizing radiation. Amplifying wild-type p53 expression by targeting negative regulators such as MDM2 in combination with external beam radiotherapy (EBRT) may result in...

Descripción completa

Detalles Bibliográficos
Autores principales: Mortensen, Anja Charlotte Lundgren, Spiegelberg, Diana, Brown, Christopher John, Lane, David Philip, Nestor, Marika
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6764291/
https://www.ncbi.nlm.nih.gov/pubmed/31616635
http://dx.doi.org/10.3389/fonc.2019.00923
_version_ 1783454347898650624
author Mortensen, Anja Charlotte Lundgren
Spiegelberg, Diana
Brown, Christopher John
Lane, David Philip
Nestor, Marika
author_facet Mortensen, Anja Charlotte Lundgren
Spiegelberg, Diana
Brown, Christopher John
Lane, David Philip
Nestor, Marika
author_sort Mortensen, Anja Charlotte Lundgren
collection PubMed
description The tumor suppressor p53 is a key mediator of cellular stress and DNA damage response cascades and is activated after exposure to ionizing radiation. Amplifying wild-type p53 expression by targeting negative regulators such as MDM2 in combination with external beam radiotherapy (EBRT) may result in increased therapeutic effects. The novel stapled peptide PM2 prevents MDM2 from suppressing wild-type p53, and is thus a promising agent for therapeutic combination with EBRT. Effects of PM2 and potential PM2-induced radiosensitivity were assessed in a panel of cancer cell lines using 2D cell viability assays. Western Blot and flow cytometric analyses were used to investigate the mechanisms behind the observed effects in samples treated with PM2 and EBRT. Finally, PM2-treatment combined with EBRT was evaluated in an in vitro 3D spheroid model. PM2-therapy decreased cell viability in wild-type p53, HPV-negative cell lines. Western Blotting and flow cytometry confirmed upregulation of p53, as well as initiation of p53-mediated apoptosis measured by increased cleaved caspase-3 and Noxa activity. Furthermore, 3D in vitro tumor spheroid experiments confirmed the superior effects of the combination, as the only treatment regime resulting in growth inhibition and complete spheroid disintegration. We conclude that PM2 induces antitumorigenic effects in wt p53 HPV-negative cancer cells and potentiates the effects of EBRT, ultimately resulting in tumor eradication in a 3D spheroid model. This strategy shows great potential as a new wt p53 specific tumor-targeting compound, and the combination of PM2 and EBRT could be a promising strategy to increase therapeutic effects and decrease adverse effects from radiotherapy.
format Online
Article
Text
id pubmed-6764291
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-67642912019-10-15 The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells Mortensen, Anja Charlotte Lundgren Spiegelberg, Diana Brown, Christopher John Lane, David Philip Nestor, Marika Front Oncol Oncology The tumor suppressor p53 is a key mediator of cellular stress and DNA damage response cascades and is activated after exposure to ionizing radiation. Amplifying wild-type p53 expression by targeting negative regulators such as MDM2 in combination with external beam radiotherapy (EBRT) may result in increased therapeutic effects. The novel stapled peptide PM2 prevents MDM2 from suppressing wild-type p53, and is thus a promising agent for therapeutic combination with EBRT. Effects of PM2 and potential PM2-induced radiosensitivity were assessed in a panel of cancer cell lines using 2D cell viability assays. Western Blot and flow cytometric analyses were used to investigate the mechanisms behind the observed effects in samples treated with PM2 and EBRT. Finally, PM2-treatment combined with EBRT was evaluated in an in vitro 3D spheroid model. PM2-therapy decreased cell viability in wild-type p53, HPV-negative cell lines. Western Blotting and flow cytometry confirmed upregulation of p53, as well as initiation of p53-mediated apoptosis measured by increased cleaved caspase-3 and Noxa activity. Furthermore, 3D in vitro tumor spheroid experiments confirmed the superior effects of the combination, as the only treatment regime resulting in growth inhibition and complete spheroid disintegration. We conclude that PM2 induces antitumorigenic effects in wt p53 HPV-negative cancer cells and potentiates the effects of EBRT, ultimately resulting in tumor eradication in a 3D spheroid model. This strategy shows great potential as a new wt p53 specific tumor-targeting compound, and the combination of PM2 and EBRT could be a promising strategy to increase therapeutic effects and decrease adverse effects from radiotherapy. Frontiers Media S.A. 2019-09-19 /pmc/articles/PMC6764291/ /pubmed/31616635 http://dx.doi.org/10.3389/fonc.2019.00923 Text en Copyright © 2019 Mortensen, Spiegelberg, Brown, Lane and Nestor. http://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
Mortensen, Anja Charlotte Lundgren
Spiegelberg, Diana
Brown, Christopher John
Lane, David Philip
Nestor, Marika
The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title_full The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title_fullStr The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title_full_unstemmed The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title_short The Stapled Peptide PM2 Stabilizes p53 Levels and Radiosensitizes Wild-Type p53 Cancer Cells
title_sort stapled peptide pm2 stabilizes p53 levels and radiosensitizes wild-type p53 cancer cells
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6764291/
https://www.ncbi.nlm.nih.gov/pubmed/31616635
http://dx.doi.org/10.3389/fonc.2019.00923
work_keys_str_mv AT mortensenanjacharlottelundgren thestapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT spiegelbergdiana thestapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT brownchristopherjohn thestapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT lanedavidphilip thestapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT nestormarika thestapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT mortensenanjacharlottelundgren stapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT spiegelbergdiana stapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT brownchristopherjohn stapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT lanedavidphilip stapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells
AT nestormarika stapledpeptidepm2stabilizesp53levelsandradiosensitizeswildtypep53cancercells