Mostra i principali dati dell'item

Role of PPARy in the complex interplay between brest cancer cells and tumor microenvironment

dc.contributor.authorGionfriddo, Giulia
dc.contributor.authorAndò, Sebastiano
dc.contributor.authorBonofiglio, Daniela
dc.date.accessioned2019-10-31T15:47:23Z
dc.date.available2019-10-31T15:47:23Z
dc.date.issued2019-03-21
dc.identifier.urihttp://hdl.handle.net/10955/1761
dc.identifier.urihttps://doi.org/10.13126/unical.it/dottorati/1761
dc.descriptionDottorato di Ricerca in Medicina Traslazionale. Ciclo XXXIen_US
dc.description.abstractStromal 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 patientsen_US
dc.description.sponsorshipUniversità della Calabriaen_US
dc.language.isoenen_US
dc.relation.ispartofseriesMED/05;
dc.subjectClinical patologyen_US
dc.subjectBreast-Canceren_US
dc.subjectPeroxisomesen_US
dc.subjectClinical patologyBreastCancerPeroxisomesen_US
dc.titleRole of PPARy in the complex interplay between brest cancer cells and tumor microenvironmenten_US
dc.typeThesisen_US


Files in questo item

Questo item appare nelle seguenti collezioni

Mostra i principali dati dell'item