Background: Metastatic cancer cells exploit Epithelial-mesenchymal-transition (EMT) to enhance their migration, invasion, and resistance to treatments. Recent studies highlight that elevated levels of copper are implicated in cancer progression and metastasis. Clinical trials using copper chelators are associated with improved patient survival; however, the molecular mechanisms by which copper depletion inhibits tumor progression and metastasis are poorly understood. This remains a major hurdle to the clinical translation of copper chelators. Here, we propose that copper chelation inhibits metastasis by reducing TGF-β levels and EMT signaling. Given that many drugs targeting TGF-β have failed in clinical trials, partly because of severe side effects arising in patients, we hypothesized that copper chelation therapy might be a less toxic alternative to target the TGF-β/EMT axis. Results: Our cytokine array and RNA-seq data suggested a link between copper homeostasis, TGF-β and EMT process. To validate this hypothesis, we performed single-cell imaging, protein assays, and in vivo studies. Here, we used the copper chelating agent TEPA to block copper trafficking. Our in vivo study showed a reduction of TGF-β levels and metastasis to the lung in the TNBC mouse model. Mechanistically, TEPA significantly downregulated canonical (TGF-β/SMAD2&3) and non-canonical (TGF-β/PI3K/AKT, TGF-β/RAS/RAF/MEK/ERK, and TGF-β/WNT/β-catenin) TGF-β signaling pathways. Additionally, EMT markers of MMP-9, MMP-14, Vimentin, β-catenin, ZEB1, and p-SMAD2 were downregulated, and EMT transcription factors of SNAI1, ZEB1, and p-SMAD2 accumulated in the cytoplasm after treatment. Conclusions: Our study suggests that copper chelation therapy represents a potentially effective therapeutic approach for targeting TGF-β and inhibiting EMT in a diverse range of cancers.

Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer / Poursani E.M.; Mercatelli D.; Raninga P.; Bell J.L.; Saletta F.; Kohane F.V.; Neumann D.P.; Zheng Y.; Rouaen J.R.C.; Jue T.R.; Michniewicz F.T.; Schadel P.; Kasiou E.; Tsoli M.; Cirillo G.; Waters S.; Shai-Hee T.; Cazzoli R.; Brettle M.; Slapetova I.; Kasherman M.; Whan R.; Souza-Fonseca-Guimaraes F.; Vahdat L.; Ziegler D.; Lock J.G.; Giorgi F.M.; Khanna K.K.; Vittorio O.. - In: CELL & BIOSCIENCE. - ISSN 2045-3701. - ELETTRONICO. - 13:1(2023), pp. 132.132-132.150. [10.1186/s13578-023-01083-7]

Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer

Mercatelli D.;Giorgi F. M.;
2023

Abstract

Background: Metastatic cancer cells exploit Epithelial-mesenchymal-transition (EMT) to enhance their migration, invasion, and resistance to treatments. Recent studies highlight that elevated levels of copper are implicated in cancer progression and metastasis. Clinical trials using copper chelators are associated with improved patient survival; however, the molecular mechanisms by which copper depletion inhibits tumor progression and metastasis are poorly understood. This remains a major hurdle to the clinical translation of copper chelators. Here, we propose that copper chelation inhibits metastasis by reducing TGF-β levels and EMT signaling. Given that many drugs targeting TGF-β have failed in clinical trials, partly because of severe side effects arising in patients, we hypothesized that copper chelation therapy might be a less toxic alternative to target the TGF-β/EMT axis. Results: Our cytokine array and RNA-seq data suggested a link between copper homeostasis, TGF-β and EMT process. To validate this hypothesis, we performed single-cell imaging, protein assays, and in vivo studies. Here, we used the copper chelating agent TEPA to block copper trafficking. Our in vivo study showed a reduction of TGF-β levels and metastasis to the lung in the TNBC mouse model. Mechanistically, TEPA significantly downregulated canonical (TGF-β/SMAD2&3) and non-canonical (TGF-β/PI3K/AKT, TGF-β/RAS/RAF/MEK/ERK, and TGF-β/WNT/β-catenin) TGF-β signaling pathways. Additionally, EMT markers of MMP-9, MMP-14, Vimentin, β-catenin, ZEB1, and p-SMAD2 were downregulated, and EMT transcription factors of SNAI1, ZEB1, and p-SMAD2 accumulated in the cytoplasm after treatment. Conclusions: Our study suggests that copper chelation therapy represents a potentially effective therapeutic approach for targeting TGF-β and inhibiting EMT in a diverse range of cancers.
2023
Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer / Poursani E.M.; Mercatelli D.; Raninga P.; Bell J.L.; Saletta F.; Kohane F.V.; Neumann D.P.; Zheng Y.; Rouaen J.R.C.; Jue T.R.; Michniewicz F.T.; Schadel P.; Kasiou E.; Tsoli M.; Cirillo G.; Waters S.; Shai-Hee T.; Cazzoli R.; Brettle M.; Slapetova I.; Kasherman M.; Whan R.; Souza-Fonseca-Guimaraes F.; Vahdat L.; Ziegler D.; Lock J.G.; Giorgi F.M.; Khanna K.K.; Vittorio O.. - In: CELL & BIOSCIENCE. - ISSN 2045-3701. - ELETTRONICO. - 13:1(2023), pp. 132.132-132.150. [10.1186/s13578-023-01083-7]
Poursani E.M.; Mercatelli D.; Raninga P.; Bell J.L.; Saletta F.; Kohane F.V.; Neumann D.P.; Zheng Y.; Rouaen J.R.C.; Jue T.R.; Michniewicz F.T.; Schadel P.; Kasiou E.; Tsoli M.; Cirillo G.; Waters S.; Shai-Hee T.; Cazzoli R.; Brettle M.; Slapetova I.; Kasherman M.; Whan R.; Souza-Fonseca-Guimaraes F.; Vahdat L.; Ziegler D.; Lock J.G.; Giorgi F.M.; Khanna K.K.; Vittorio O.
File in questo prodotto:
File Dimensione Formato  
Copper chelation suppresses epithelial-mesenchymal transition by inhibition of canonical and non-canonical TGF-β signaling pathways in cancer.pdf

accesso aperto

Tipo: Versione (PDF) editoriale
Licenza: Licenza per Accesso Aperto. Creative Commons Attribuzione (CCBY)
Dimensione 10.2 MB
Formato Adobe PDF
10.2 MB Adobe PDF Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11585/940359
Citazioni
  • ???jsp.display-item.citation.pmc??? 2
  • Scopus 2
  • ???jsp.display-item.citation.isi??? 2
social impact