American Association for Cancer Research
Browse
10780432ccr180517-sup-197057_3_supp_4939651_pcw6hf.pdf (3.08 MB)

Supplementary Figures 1-20 from Extracellular HMGA1 Promotes Tumor Invasion and Metastasis in Triple-Negative Breast Cancer

Download (3.08 MB)
journal contribution
posted on 2023-03-31, 20:21 authored by Olga Méndez, Vicente Peg, Cándida Salvans, Mireia Pujals, Yolanda Fernández, Ibane Abasolo, José Pérez, Ana Matres, Marta Valeri, Josep Gregori, Laura Villarreal, Simó Schwartz, Santiago Ramon y Cajal, Josep Tabernero, Javier Cortés, Joaquín Arribas, Josep Villanueva

Supplementary Figures 1-20

Funding

Instituto Salud Carlos III

FEDER

BCRF

Instituto de Salud Carlos III

Fundació la Marató de TV3

SGR

History

ARTICLE ABSTRACT

The study of the cancer secretome suggests that a fraction of the intracellular proteome could play unanticipated roles in the extracellular space during tumorigenesis. A project aimed at investigating the invasive secretome led us to study the alternative extracellular function of the nuclear protein high mobility group A1 (HMGA1) in breast cancer invasion and metastasis. Antibodies against HMGA1 were tested in signaling, adhesion, migration, invasion, and metastasis assays using breast cancer cell lines and xenograft models. Fluorescence microscopy was used to determine the subcellular localization of HMGA1 in cell lines, xenograft, and patient-derived xenograft models. A cohort of triple-negative breast cancer (TNBC) patients was used to study the correlation between subcellular localization of HMGA1 and the incidence of metastasis. Our data show that treatment of invasive cells with HMGA1-blocking antibodies in the extracellular space impairs their migration and invasion abilities. We also prove that extracellular HMGA1 (eHMGA1) becomes a ligand for the Advanced glycosylation end product-specific receptor (RAGE), inducing pERK signaling and increasing migration and invasion. Using the cytoplasmic localization of HMGA1 as a surrogate marker of secretion, we showed that eHMGA1 correlates with the incidence of metastasis in a cohort of TNBC patients. Furthermore, we show that HMGA1 is enriched in the cytoplasm of tumor cells at the invasive front of primary tumors and in metastatic lesions in xenograft models. Our results strongly suggest that eHMGA1 could become a novel drug target in metastatic TNBC and a biomarker predicting the onset of distant metastasis.

Usage metrics

    Clinical Cancer Research

    Categories

    Keywords

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC