INVESTIGADORES
PROIETTI ANASTASI Cecilia Jazmin
artículos
Título:
Halting ErbB-2 isoforms retrograde transport to the nucleus as a new theragnostic approach for triple-negative breast cancer
Autor/es:
SANTIAGO MADERA; FRANCO IZZO; MARIA FLORENCIA CHERVO; AGUSTINA DUPONT; VIOLETA CHIAUZZI; SOFIA BRUNI; EZEQUIEL PETRILLO; SHARON MERIN; MARA DE MARTINO; DIEGO MONTERO; CLAUDIO LEVIT; GABRIEL LEBERSZTEIN; FABIANA ANFUSO; AGUSTINA ROLDÁN DEAMICIS; FLORENCIA MERCOGLIANO; CECILIA J PROIETTI; ROXANA SCHILLACI; PATRICIA V ELIZALDE; ROSALIA INES CORDO RUSSO
Revista:
Cell Death & Disease
Editorial:
Nature
Referencias:
Año: 2022
ISSN:
2041-4889
Resumen:
Triple-negative breast cancer (TNBC) is clinically defined by the absence of estrogen and progesterone receptors and the lack of membrane overexpression or gene amplification of receptor tyrosine kinase ErbB-2/HER2. Due to TNBC heterogeneity, clinical biomarkers and targeted therapies for this disease remain elusive. We demonstrated that ErbB-2 is localized in the nucleus (NErbB-2) of TNBC cells and primary tumors, from where it drives growth. We also discovered that TNBC expresses both wild-type ErbB-2 (WTErbB-2) and alternative ErbB-2 isoform c (ErbB-2c). Here, we revealed that the inhibitors of the retrograde transport Retro-2 and its cyclic derivative Retro-2.1 evict both WTErbB-2 and ErbB-2c from the nucleus of BC cells and tumors. Using BC cells from several molecular subtypes, as well as normal breast cells, we demonstrated that Retro-2 specifically blocks proliferation of BC cells expressing NErbB-2. Importantly, Retro-2 eviction of both ErbB-2 isoforms from the nucleus resulted in a striking growth abrogation in multiple TNBC preclinical models, including tumor explants and xenografts. Our mechanistic studies in TNBC cells revealed that Retro-2 induces a differential accumulation of WTErbB-2 at the early endosomes and the plasma membrane, and of ErbB-2c at the Golgi, shedding new light both on Retro-2 action on endogenous protein cargoes undergoing retrograde transport, and on the biology of ErbB-2 splicing variants. In addition, we revealed that the presence of a functional signal peptide and a nuclear export signal (NES), both located at the N-terminus of WTErbB-2, and absent in ErbB-2c, accounts for the differential subcellular distribution of ErbB-2 isoforms upon Retro-2 treatment. Our present discoveries provide evidence for the rational repurposing of Retro-2 as a novel therapeutic agent for TNBC.