Cargando…

Obesity modulates the immune macroenvironment associated with breast cancer development

Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironmen...

Descripción completa

Detalles Bibliográficos
Autores principales: Núñez-Ruiz, Aleida, Sánchez-Brena, Flor, López-Pacheco, Cynthia, Acevedo-Domínguez, Naray A., Soldevila, Gloria
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9041840/
https://www.ncbi.nlm.nih.gov/pubmed/35472214
http://dx.doi.org/10.1371/journal.pone.0266827
_version_ 1784694581437136896
author Núñez-Ruiz, Aleida
Sánchez-Brena, Flor
López-Pacheco, Cynthia
Acevedo-Domínguez, Naray A.
Soldevila, Gloria
author_facet Núñez-Ruiz, Aleida
Sánchez-Brena, Flor
López-Pacheco, Cynthia
Acevedo-Domínguez, Naray A.
Soldevila, Gloria
author_sort Núñez-Ruiz, Aleida
collection PubMed
description Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8(+) T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39(+), PD-1(+) and CCR8(+) cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity.
format Online
Article
Text
id pubmed-9041840
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-90418402022-04-27 Obesity modulates the immune macroenvironment associated with breast cancer development Núñez-Ruiz, Aleida Sánchez-Brena, Flor López-Pacheco, Cynthia Acevedo-Domínguez, Naray A. Soldevila, Gloria PLoS One Research Article Growing evidence demonstrates a strong correlation between obesity and an increased risk of breast cancer, although the mechanisms involved have not been completely elucidated. Some reports have described a crosstalk between adipocytes, cancer cells, and immune cells within the tumor microenvironment, however, it is currently unknown whether obesity can promote tumor growth by inducing systemic alterations of the immune cell homeostasis in peripheral lymphoid organs and adipose tissue. Here, we used the E0771 breast cancer cell line in a mouse model of diet-induced obesity to analyze the immune subpopulations present in the tumors, visceral adipose tissue (VAT), and spleen of lean and obese mice. Our results showed a significant reduction in the frequency of infiltrating CD8(+) T cells and a decreased M1/M2 macrophage ratio, indicative of the compromised anti-tumoral immune response reported in obesity. Despite not finding differences in the percentage or numbers of intratumoral Tregs, phenotypic analysis showed that they were enriched in CD39(+), PD-1(+) and CCR8(+) cells, compared to the draining lymph nodes, confirming the highly immunosuppressive profile of infiltrating Tregs reported in established tumors. Analysis of peripheral T lymphocytes showed that tumor development in obese mice was associated to a significant increase in the percentage of peripheral Tregs, which supports the systemic immunosuppressive effect caused by the tumor. Interestingly, evaluation of immune subpopulations in the VAT showed that the characteristic increase in the M1/M2 macrophage ratio reported in obesity, was completely reversed in tumor-bearing mice, resembling the M2-polarized profile found in the microenvironment of the growing tumor. Importantly, VAT Tregs, which are commonly decreased in obese mice, were significantly increased in the presence of breast tumors and displayed significantly higher levels of Foxp3, indicating a regulatory feedback mechanism triggered by tumor growth. Altogether, our results identify a complex reciprocal relationship between adipocytes, immune cells, and the tumor, which may modulate the immune macroenvironment that promotes breast cancer development in obesity. Public Library of Science 2022-04-26 /pmc/articles/PMC9041840/ /pubmed/35472214 http://dx.doi.org/10.1371/journal.pone.0266827 Text en © 2022 Núñez-Ruiz et al https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Núñez-Ruiz, Aleida
Sánchez-Brena, Flor
López-Pacheco, Cynthia
Acevedo-Domínguez, Naray A.
Soldevila, Gloria
Obesity modulates the immune macroenvironment associated with breast cancer development
title Obesity modulates the immune macroenvironment associated with breast cancer development
title_full Obesity modulates the immune macroenvironment associated with breast cancer development
title_fullStr Obesity modulates the immune macroenvironment associated with breast cancer development
title_full_unstemmed Obesity modulates the immune macroenvironment associated with breast cancer development
title_short Obesity modulates the immune macroenvironment associated with breast cancer development
title_sort obesity modulates the immune macroenvironment associated with breast cancer development
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9041840/
https://www.ncbi.nlm.nih.gov/pubmed/35472214
http://dx.doi.org/10.1371/journal.pone.0266827
work_keys_str_mv AT nunezruizaleida obesitymodulatestheimmunemacroenvironmentassociatedwithbreastcancerdevelopment
AT sanchezbrenaflor obesitymodulatestheimmunemacroenvironmentassociatedwithbreastcancerdevelopment
AT lopezpachecocynthia obesitymodulatestheimmunemacroenvironmentassociatedwithbreastcancerdevelopment
AT acevedodomingueznaraya obesitymodulatestheimmunemacroenvironmentassociatedwithbreastcancerdevelopment
AT soldevilagloria obesitymodulatestheimmunemacroenvironmentassociatedwithbreastcancerdevelopment