Cargando…

eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model

Cells employ several adaptive mechanisms under conditions of accelerated cell division, such as the unfolded protein response (UPR). The UPR is composed of a tripartite signaling system that involves ATF6, PERK, and IRE1, which maintain protein homeostasis (proteostasis). However, deregulation of pr...

Descripción completa

Detalles Bibliográficos
Autores principales: González-Ortiz, Alina, Pulido-Capiz, Angel, Castañeda-Sánchez, César Y., Ibarra-López, Esmeralda, Galindo-Hernández, Octavio, Calderón-Fernández, Maritza Anahí, López-Cossio, Leslie Y., Díaz-Molina, Raul, Chimal-Vega, Brenda, Serafín-Higuera, Nicolás, Córdova-Guerrero, Iván, García-González, Victor
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9776898/
https://www.ncbi.nlm.nih.gov/pubmed/36552834
http://dx.doi.org/10.3390/cells11244069
_version_ 1784855971880763392
author González-Ortiz, Alina
Pulido-Capiz, Angel
Castañeda-Sánchez, César Y.
Ibarra-López, Esmeralda
Galindo-Hernández, Octavio
Calderón-Fernández, Maritza Anahí
López-Cossio, Leslie Y.
Díaz-Molina, Raul
Chimal-Vega, Brenda
Serafín-Higuera, Nicolás
Córdova-Guerrero, Iván
García-González, Victor
author_facet González-Ortiz, Alina
Pulido-Capiz, Angel
Castañeda-Sánchez, César Y.
Ibarra-López, Esmeralda
Galindo-Hernández, Octavio
Calderón-Fernández, Maritza Anahí
López-Cossio, Leslie Y.
Díaz-Molina, Raul
Chimal-Vega, Brenda
Serafín-Higuera, Nicolás
Córdova-Guerrero, Iván
García-González, Victor
author_sort González-Ortiz, Alina
collection PubMed
description Cells employ several adaptive mechanisms under conditions of accelerated cell division, such as the unfolded protein response (UPR). The UPR is composed of a tripartite signaling system that involves ATF6, PERK, and IRE1, which maintain protein homeostasis (proteostasis). However, deregulation of protein translation initiation could be associated with breast cancer (BC) chemoresistance. Specifically, eukaryotic initiation factor-4A (eIF4A) is involved in the unfolding of the secondary structures of several mRNAs at the 5′ untranslated region (5′-UTR), as well as in the regulation of targets involved in chemoresistance. Importantly, the tumor suppressor gene PDCD4 could modulate this process. This regulation might be disrupted in chemoresistant triple negative-BC (TNBC) cells. Therefore, we characterized the effect of doxorubicin (Dox), a commonly used anthracycline medication, on human breast carcinoma MDA-MB-231 cells. Here, we generated and characterized models of Dox chemoresistance, and chemoresistant cells exhibited lower Dox internalization levels followed by alteration of the IRE1 and PERK arms of the UPR and triggering of the antioxidant Nrf2 axis. Critically, chemoresistant cells exhibited PDCD4 downregulation, which coincided with a reduction in eIF4A interaction, suggesting a sophisticated regulation of protein translation. Likewise, Dox-induced chemoresistance was associated with alterations in cellular migration and invasion, which are key cancer hallmarks, coupled with changes in focal adhesion kinase (FAK) activation and secretion of matrix metalloproteinase-9 (MMP-9). Moreover, eIF4A knockdown via siRNA and its overexpression in chemoresistant cells suggested that eIF4A regulates FAK. Pro-atherogenic low-density lipoproteins (LDL) promoted cellular invasion in parental and chemoresistant cells in an MMP-9-dependent manner. Moreover, Dox only inhibited parental cell invasion. Significantly, chemoresistance was modulated by cryptotanshinone (Cry), a natural terpene purified from the roots of Salvia brandegeei. Cry and Dox co-exposure induced chemosensitization, connected with the Cry effect on eIF4A interaction. We further demonstrated the Cry binding capability on eIF4A and in silico assays suggest Cry inhibition on the RNA-processing domain. Therefore, strategic disruption of protein translation initiation is a druggable pathway by natural compounds during chemoresistance in TNBC. However, plasmatic LDL levels should be closely monitored throughout treatment.
format Online
Article
Text
id pubmed-9776898
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-97768982022-12-23 eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model González-Ortiz, Alina Pulido-Capiz, Angel Castañeda-Sánchez, César Y. Ibarra-López, Esmeralda Galindo-Hernández, Octavio Calderón-Fernández, Maritza Anahí López-Cossio, Leslie Y. Díaz-Molina, Raul Chimal-Vega, Brenda Serafín-Higuera, Nicolás Córdova-Guerrero, Iván García-González, Victor Cells Article Cells employ several adaptive mechanisms under conditions of accelerated cell division, such as the unfolded protein response (UPR). The UPR is composed of a tripartite signaling system that involves ATF6, PERK, and IRE1, which maintain protein homeostasis (proteostasis). However, deregulation of protein translation initiation could be associated with breast cancer (BC) chemoresistance. Specifically, eukaryotic initiation factor-4A (eIF4A) is involved in the unfolding of the secondary structures of several mRNAs at the 5′ untranslated region (5′-UTR), as well as in the regulation of targets involved in chemoresistance. Importantly, the tumor suppressor gene PDCD4 could modulate this process. This regulation might be disrupted in chemoresistant triple negative-BC (TNBC) cells. Therefore, we characterized the effect of doxorubicin (Dox), a commonly used anthracycline medication, on human breast carcinoma MDA-MB-231 cells. Here, we generated and characterized models of Dox chemoresistance, and chemoresistant cells exhibited lower Dox internalization levels followed by alteration of the IRE1 and PERK arms of the UPR and triggering of the antioxidant Nrf2 axis. Critically, chemoresistant cells exhibited PDCD4 downregulation, which coincided with a reduction in eIF4A interaction, suggesting a sophisticated regulation of protein translation. Likewise, Dox-induced chemoresistance was associated with alterations in cellular migration and invasion, which are key cancer hallmarks, coupled with changes in focal adhesion kinase (FAK) activation and secretion of matrix metalloproteinase-9 (MMP-9). Moreover, eIF4A knockdown via siRNA and its overexpression in chemoresistant cells suggested that eIF4A regulates FAK. Pro-atherogenic low-density lipoproteins (LDL) promoted cellular invasion in parental and chemoresistant cells in an MMP-9-dependent manner. Moreover, Dox only inhibited parental cell invasion. Significantly, chemoresistance was modulated by cryptotanshinone (Cry), a natural terpene purified from the roots of Salvia brandegeei. Cry and Dox co-exposure induced chemosensitization, connected with the Cry effect on eIF4A interaction. We further demonstrated the Cry binding capability on eIF4A and in silico assays suggest Cry inhibition on the RNA-processing domain. Therefore, strategic disruption of protein translation initiation is a druggable pathway by natural compounds during chemoresistance in TNBC. However, plasmatic LDL levels should be closely monitored throughout treatment. MDPI 2022-12-15 /pmc/articles/PMC9776898/ /pubmed/36552834 http://dx.doi.org/10.3390/cells11244069 Text en © 2022 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
González-Ortiz, Alina
Pulido-Capiz, Angel
Castañeda-Sánchez, César Y.
Ibarra-López, Esmeralda
Galindo-Hernández, Octavio
Calderón-Fernández, Maritza Anahí
López-Cossio, Leslie Y.
Díaz-Molina, Raul
Chimal-Vega, Brenda
Serafín-Higuera, Nicolás
Córdova-Guerrero, Iván
García-González, Victor
eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title_full eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title_fullStr eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title_full_unstemmed eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title_short eIF4A/PDCD4 Pathway, a Factor for Doxorubicin Chemoresistance in a Triple-Negative Breast Cancer Cell Model
title_sort eif4a/pdcd4 pathway, a factor for doxorubicin chemoresistance in a triple-negative breast cancer cell model
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9776898/
https://www.ncbi.nlm.nih.gov/pubmed/36552834
http://dx.doi.org/10.3390/cells11244069
work_keys_str_mv AT gonzalezortizalina eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT pulidocapizangel eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT castanedasanchezcesary eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT ibarralopezesmeralda eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT galindohernandezoctavio eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT calderonfernandezmaritzaanahi eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT lopezcossiolesliey eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT diazmolinaraul eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT chimalvegabrenda eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT serafinhigueranicolas eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT cordovaguerreroivan eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel
AT garciagonzalezvictor eif4apdcd4pathwayafactorfordoxorubicinchemoresistanceinatriplenegativebreastcancercellmodel