TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner

The influence of an inflammatory microenvironment on tumorigenesis has been widely accepted. Systemic conditions that favor the onset of an inflammatory landscape predispose the progression of breast cancer. Under obesity conditions, the endocrine function of adipose tissue is one of the main determ...

Full description

Bibliographic Details
Main Authors: María Jesús Vera, Francisco Guajardo, Felix A. Urra, Nicolás Tobar, Jorge Martínez
Format: Article
Language:English
Published: MDPI AG 2023-03-01
Series:Antioxidants
Subjects:
Online Access:https://www.mdpi.com/2076-3921/12/4/813
_version_ 1797606638891302912
author María Jesús Vera
Francisco Guajardo
Felix A. Urra
Nicolás Tobar
Jorge Martínez
author_facet María Jesús Vera
Francisco Guajardo
Felix A. Urra
Nicolás Tobar
Jorge Martínez
author_sort María Jesús Vera
collection DOAJ
description The influence of an inflammatory microenvironment on tumorigenesis has been widely accepted. Systemic conditions that favor the onset of an inflammatory landscape predispose the progression of breast cancer. Under obesity conditions, the endocrine function of adipose tissue is one of the main determinants of the production of local and systemic inflammatory mediators. Although these mediators can stimulate tumorigenesis and recruit inflammatory cells, as macrophages, the mechanism involved remains poorly understood. In the present work, we describe that the TNFα treatment of mammary preadipocytes from human normal patients blocks adipose differentiation and promotes the generation of pro-inflammatory soluble factors. The latter stimulate the mobilization of THP-1 monocytes and MCF-7 epithelial cancer cells in an MCP1/CCL2- and mitochondrial-ROS-dependent manner. Together, these results reaffirm the contribution of an inflammatory microenvironment and mtROS in the progression of breast cancer.
first_indexed 2024-03-11T05:18:02Z
format Article
id doaj.art-7afd3d09eb9e4c5195944104f420fd46
institution Directory Open Access Journal
issn 2076-3921
language English
last_indexed 2024-03-11T05:18:02Z
publishDate 2023-03-01
publisher MDPI AG
record_format Article
series Antioxidants
spelling doaj.art-7afd3d09eb9e4c5195944104f420fd462023-11-17T18:04:40ZengMDPI AGAntioxidants2076-39212023-03-0112481310.3390/antiox12040813TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent MannerMaría Jesús Vera0Francisco Guajardo1Felix A. Urra2Nicolás Tobar3Jorge Martínez4Molecular and Cell Biology Laboratory, INTA, University of Chile, Santiago 7830490, ChileMIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Santiago 8380453, ChileMIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Santiago 8380453, ChileMolecular and Cell Biology Laboratory, INTA, University of Chile, Santiago 7830490, ChileMolecular and Cell Biology Laboratory, INTA, University of Chile, Santiago 7830490, ChileThe influence of an inflammatory microenvironment on tumorigenesis has been widely accepted. Systemic conditions that favor the onset of an inflammatory landscape predispose the progression of breast cancer. Under obesity conditions, the endocrine function of adipose tissue is one of the main determinants of the production of local and systemic inflammatory mediators. Although these mediators can stimulate tumorigenesis and recruit inflammatory cells, as macrophages, the mechanism involved remains poorly understood. In the present work, we describe that the TNFα treatment of mammary preadipocytes from human normal patients blocks adipose differentiation and promotes the generation of pro-inflammatory soluble factors. The latter stimulate the mobilization of THP-1 monocytes and MCF-7 epithelial cancer cells in an MCP1/CCL2- and mitochondrial-ROS-dependent manner. Together, these results reaffirm the contribution of an inflammatory microenvironment and mtROS in the progression of breast cancer.https://www.mdpi.com/2076-3921/12/4/813migrationmonocytesbreast cancerOXPHOSadipocytes
spellingShingle María Jesús Vera
Francisco Guajardo
Felix A. Urra
Nicolás Tobar
Jorge Martínez
TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
Antioxidants
migration
monocytes
breast cancer
OXPHOS
adipocytes
title TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
title_full TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
title_fullStr TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
title_full_unstemmed TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
title_short TNF-Alpha Promotes an Inflammatory Mammary Microenvironment That Favors Macrophage and Epithelial Migration in a CCL2- and Mitochondrial-ROS-Dependent Manner
title_sort tnf alpha promotes an inflammatory mammary microenvironment that favors macrophage and epithelial migration in a ccl2 and mitochondrial ros dependent manner
topic migration
monocytes
breast cancer
OXPHOS
adipocytes
url https://www.mdpi.com/2076-3921/12/4/813
work_keys_str_mv AT mariajesusvera tnfalphapromotesaninflammatorymammarymicroenvironmentthatfavorsmacrophageandepithelialmigrationinaccl2andmitochondrialrosdependentmanner
AT franciscoguajardo tnfalphapromotesaninflammatorymammarymicroenvironmentthatfavorsmacrophageandepithelialmigrationinaccl2andmitochondrialrosdependentmanner
AT felixaurra tnfalphapromotesaninflammatorymammarymicroenvironmentthatfavorsmacrophageandepithelialmigrationinaccl2andmitochondrialrosdependentmanner
AT nicolastobar tnfalphapromotesaninflammatorymammarymicroenvironmentthatfavorsmacrophageandepithelialmigrationinaccl2andmitochondrialrosdependentmanner
AT jorgemartinez tnfalphapromotesaninflammatorymammarymicroenvironmentthatfavorsmacrophageandepithelialmigrationinaccl2andmitochondrialrosdependentmanner