Activating Transcription Factor 4 Modulates TGFβ-Induced Aggressiveness in Triple-Negative Breast Cancer via SMAD2/3/4 and mTORC2 Signaling.

TitleActivating Transcription Factor 4 Modulates TGFβ-Induced Aggressiveness in Triple-Negative Breast Cancer via SMAD2/3/4 and mTORC2 Signaling.
Publication TypeJournal Article
Year of Publication2018
AuthorsGonzález-González A, Muñoz-Muela E, Marchal JA, Cara FE, Molina MP, Cruz-Lozano M, Jiménez G, Verma A, Ramírez A, Qian W, Chen W, Kozielski AJ, Elemento O, Martín-Salvago MD, Luque RJ, Rosa-Garrido C, Landeira D, Quintana-Romero M, Rosato RR, García MA, Ramirez-Tortosa CL, Kim H, Rodriguez-Aguayo C, Lopez-Berestein G, Sood AK, Lorente JA, Sánchez-Rovira P, Chang JC, Granados-Principal S
JournalClin Cancer Res
Volume24
Issue22
Pagination5697-5709
Date Published2018 11 15
ISSN1078-0432
KeywordsActivating Transcription Factor 4, Animals, Cell Line, Tumor, Cell Movement, Cell Proliferation, Computational Biology, Disease Models, Animal, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Heterografts, Humans, Immunohistochemistry, Mechanistic Target of Rapamycin Complex 2, Mice, Models, Biological, Prognosis, RNA, Small Interfering, Signal Transduction, Smad Proteins, Transcriptome, Transforming Growth Factor beta, Triple Negative Breast Neoplasms
Abstract

On the basis of the identified stress-independent cellular functions of activating transcription factor 4 (ATF4), we reported enhanced ATF4 levels in MCF10A cells treated with TGFβ1. is overexpressed in patients with triple-negative breast cancer (TNBC), but its impact on patient survival and the underlying mechanisms remain unknown. We aimed to determine effects on patients with breast cancer survival and TNBC aggressiveness, and the relationships between TGFβ and ATF4. Defining the signaling pathways may help us identify a cell signaling-tailored gene signature. Patient survival data were determined by Kaplan-Meier analysis. Relationship between TGFβ and ATF4, their effects on aggressiveness (tumor proliferation, metastasis, and stemness), and the underlying pathways were analyzed in three TNBC cell lines and using patient-derived xenografts (PDX). overexpression correlated with TNBC patient survival decrease and a SMAD-dependent crosstalk between ATF4 and TGFβ was identified. expression inhibition reduced migration, invasiveness, mammosphere-forming efficiency, proliferation, epithelial-mesenchymal transition, and antiapoptotic and stemness marker levels. In PDX models, silencing decreased metastases, tumor growth, and relapse after chemotherapy. ATF4 was shown to be active downstream of SMAD2/3/4 and mTORC2, regulating TGFβ/SMAD and mTOR/RAC1-RHOA pathways independently of stress. We defined an eight-gene signature with prognostic potential, altered in 45% of 2,509 patients with breast cancer. ATF4 may represent a valuable prognostic biomarker and therapeutic target in patients with TNBC, and we identified a cell signaling pathway-based gene signature that may contribute to the development of combinatorial targeted therapies for breast cancer. .

DOI10.1158/1078-0432.CCR-17-3125
Alternate JournalClin. Cancer Res.
PubMed ID30012564