Cargando…

Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression

BACKGROUND: Protein disulphide isomerases (PDIs) play an important role in cancer progression. However, the relative contribution of the various isoforms of PDI in tumorigenesis is not clear. METHODS: The content of PDI isoforms in 22 cancer cells lines was investigated using LC–MS/MS-based proteomi...

Descripción completa

Detalles Bibliográficos
Autores principales: Kurpińska, Anna, Suraj-Prażmowska, Joanna, Stojak, Marta, Jarosz, Joanna, Mateuszuk, Łukasz, Niedzielska-Andres, Ewa, Smolik, Magdalena, Wietrzyk, Joanna, Kalvins, Ivars, Walczak, Maria, Chłopicki, Stefan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9208212/
https://www.ncbi.nlm.nih.gov/pubmed/35725466
http://dx.doi.org/10.1186/s12935-022-02631-w
_version_ 1784729694878302208
author Kurpińska, Anna
Suraj-Prażmowska, Joanna
Stojak, Marta
Jarosz, Joanna
Mateuszuk, Łukasz
Niedzielska-Andres, Ewa
Smolik, Magdalena
Wietrzyk, Joanna
Kalvins, Ivars
Walczak, Maria
Chłopicki, Stefan
author_facet Kurpińska, Anna
Suraj-Prażmowska, Joanna
Stojak, Marta
Jarosz, Joanna
Mateuszuk, Łukasz
Niedzielska-Andres, Ewa
Smolik, Magdalena
Wietrzyk, Joanna
Kalvins, Ivars
Walczak, Maria
Chłopicki, Stefan
author_sort Kurpińska, Anna
collection PubMed
description BACKGROUND: Protein disulphide isomerases (PDIs) play an important role in cancer progression. However, the relative contribution of the various isoforms of PDI in tumorigenesis is not clear. METHODS: The content of PDI isoforms in 22 cancer cells lines was investigated using LC–MS/MS-based proteomic analysis. The effects of PDIA1, PDIA3 and PDIA17 inhibition on the proliferation, migration and adhesion of MCF-7 and MDA-MB-231 cells, identified as high and low PDIA17 expressing cells, respectively, were assessed using novel aromatic N-sulphonamides of aziridine-2-carboxylic acid derivatives as PDI inhibitors. RESULTS: PDIA1 and PDIA3 were the most abundant in cancer cell lysates and were also detected extracellularly in breast cancer cells (MDA-MB-231 and MCF-7). Some cancer cell lines (e.g., MCF-7, HT-29) showed upregulated expression of PDIA17, whereas in others (e.g., MDA-MB-231, 67NR), PDIA17 was not detected. The simultaneous inhibition of PDIA1 and PDIA3 showed similar anti-proliferative effects in MCF-7 and MDA-MB-231 breast cancer cells. However, the inhibition of PDIA1 and PDIA17 in the MCF-7 cell line resulted in more effective anti-adhesive and anti-proliferative effects. CONCLUSIONS: PDIA1 and PDIA3 represent major isoforms of multiple cancer cells, and their non-selective inhibition displays significant anti-proliferative effects irrespective of whether or not PDIA17 is present. The more pronounced anti-adhesive effects of PDI inhibition in hormone-sensitive MCF-7 cells featured by higher levels of PDIs when compared to triple-negative MDA-MB-231 cells suggests that targeting extracellular PDIA1 and PDIA3 with or without additional PDIA17 inhibition may represent a strategy for personalized anti-adhesive, anti-metastatic therapy in cancers with high PDI expression. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-022-02631-w.
format Online
Article
Text
id pubmed-9208212
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-92082122022-06-21 Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression Kurpińska, Anna Suraj-Prażmowska, Joanna Stojak, Marta Jarosz, Joanna Mateuszuk, Łukasz Niedzielska-Andres, Ewa Smolik, Magdalena Wietrzyk, Joanna Kalvins, Ivars Walczak, Maria Chłopicki, Stefan Cancer Cell Int Research BACKGROUND: Protein disulphide isomerases (PDIs) play an important role in cancer progression. However, the relative contribution of the various isoforms of PDI in tumorigenesis is not clear. METHODS: The content of PDI isoforms in 22 cancer cells lines was investigated using LC–MS/MS-based proteomic analysis. The effects of PDIA1, PDIA3 and PDIA17 inhibition on the proliferation, migration and adhesion of MCF-7 and MDA-MB-231 cells, identified as high and low PDIA17 expressing cells, respectively, were assessed using novel aromatic N-sulphonamides of aziridine-2-carboxylic acid derivatives as PDI inhibitors. RESULTS: PDIA1 and PDIA3 were the most abundant in cancer cell lysates and were also detected extracellularly in breast cancer cells (MDA-MB-231 and MCF-7). Some cancer cell lines (e.g., MCF-7, HT-29) showed upregulated expression of PDIA17, whereas in others (e.g., MDA-MB-231, 67NR), PDIA17 was not detected. The simultaneous inhibition of PDIA1 and PDIA3 showed similar anti-proliferative effects in MCF-7 and MDA-MB-231 breast cancer cells. However, the inhibition of PDIA1 and PDIA17 in the MCF-7 cell line resulted in more effective anti-adhesive and anti-proliferative effects. CONCLUSIONS: PDIA1 and PDIA3 represent major isoforms of multiple cancer cells, and their non-selective inhibition displays significant anti-proliferative effects irrespective of whether or not PDIA17 is present. The more pronounced anti-adhesive effects of PDI inhibition in hormone-sensitive MCF-7 cells featured by higher levels of PDIs when compared to triple-negative MDA-MB-231 cells suggests that targeting extracellular PDIA1 and PDIA3 with or without additional PDIA17 inhibition may represent a strategy for personalized anti-adhesive, anti-metastatic therapy in cancers with high PDI expression. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-022-02631-w. BioMed Central 2022-06-20 /pmc/articles/PMC9208212/ /pubmed/35725466 http://dx.doi.org/10.1186/s12935-022-02631-w Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Kurpińska, Anna
Suraj-Prażmowska, Joanna
Stojak, Marta
Jarosz, Joanna
Mateuszuk, Łukasz
Niedzielska-Andres, Ewa
Smolik, Magdalena
Wietrzyk, Joanna
Kalvins, Ivars
Walczak, Maria
Chłopicki, Stefan
Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title_full Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title_fullStr Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title_full_unstemmed Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title_short Comparison of anti-cancer effects of novel protein disulphide isomerase (PDI) inhibitors in breast cancer cells characterized by high and low PDIA17 expression
title_sort comparison of anti-cancer effects of novel protein disulphide isomerase (pdi) inhibitors in breast cancer cells characterized by high and low pdia17 expression
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9208212/
https://www.ncbi.nlm.nih.gov/pubmed/35725466
http://dx.doi.org/10.1186/s12935-022-02631-w
work_keys_str_mv AT kurpinskaanna comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT surajprazmowskajoanna comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT stojakmarta comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT jaroszjoanna comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT mateuszukłukasz comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT niedzielskaandresewa comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT smolikmagdalena comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT wietrzykjoanna comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT kalvinsivars comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT walczakmaria comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression
AT chłopickistefan comparisonofanticancereffectsofnovelproteindisulphideisomerasepdiinhibitorsinbreastcancercellscharacterizedbyhighandlowpdia17expression