Please use this identifier to cite or link to this item: https://hdl.handle.net/10955/1761
Title: Role of PPARy in the complex interplay between brest cancer cells and tumor microenvironment
Authors: Gionfriddo, Giulia
Andò, Sebastiano
Bonofiglio, Daniela
Keywords: Clinical patology
Breast-Cancer
Peroxisomes
Clinical patologyBreastCancerPeroxisomes
Issue Date: 21-Mar-2019
Series/Report no.: MED/05;
Abstract: Stromal Derived Factor-1α (SDF-1α) and its cognate receptor CXCR4 play a key role in mediating breast cancer cell invasion and metastasis. Therefore, drugs able to inhibit CXCR4 activation may add critical tools to reduce tumor progression, especially in the most aggressive form of the breast cancer disease. Peroxisome Proliferator-Activated Receptor (PPAR) γ, a member of the nuclear receptor superfamily, has been found to downregulate CXCR4 gene expression in different cancer cells, however the molecular mechanism underlying this effect is not fully understood. Here, we identified a novel PPARγ-mediated mechanism that negatively regulates CXCR4 expression in both epithelial and stromal breast cancer cells. We found that ligand-activated PPARγ downregulated CXCR4 transcriptional activity through the recruitment of the silencing mediator of retinoid and thyroid hormone receptor (SMRT) corepressor onto a newly identified PPAR response element (PPRE) within the CXCR4 promoter in breast cancer cell lines. As a consequence, the PPARγ agonist rosiglitazone (BRL) significantly inhibited cell migration and invasion and this effect was PPARγ-mediated, since it was reversed in the presence of the PPARγ antagonist GW9662. According to the ability of Cancer-Associated Fibroblasts (CAFs), the most abundant component of breast cancer stroma, to secrete high levels of SDF-1 α, BRL reduced migratory promoting activities induced by conditioned media (CM) derived from CAFs and affected CXCR4 downstream signaling pathways activated by CAF-CM. In addition, CAFs exposed to BRL showed a decreased expression of CXCR4, a reduced motility and invasion along with a phenotype characterized by an altered morphology. A further component of the tumor microenvironment, that contributes to breast cancer progression and metastasis, is represented by Tumor Associated-Macrophages (TAMs), which phenotype is shaped by complex interactions with breast cancer cells. We found that the PPARγ ligand BRL, as well as DHA conjugates to ethanolamine and serotonin DHEA and DHA-5-HT respectively, were able to counteract the effects of CM derived from breast cancer cells on macrophage polarization. Collectively, our findings provide novel insights into the role of PPARγ in inhibiting breast cancer progression and further highlight the utility of PPARγ ligands for future therapies aimed at targeting both cancer and surrounding stromal cells in breast cancer patients
Description: Dottorato di Ricerca in Medicina Traslazionale. Ciclo XXXI
URI: http://hdl.handle.net/10955/1761
https://doi.org/10.13126/unical.it/dottorati/1761
Appears in Collections:Dipartimento di Farmacia e Scienze della Salute e della Nutrizione - Tesi di Dottorato

Files in This Item:
File Description SizeFormat 
PhD Thesis Gionfriddo Giulia.pdf2,72 MBAdobe PDFView/Open


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.