In the tumor microenvironment, cancer cells experience hypoxia resulting in the accumulation of misfolded/unfolded proteins largely in the endoplasmic reticulum (ER). Consequently, ER proteotoxicity elicits unfolded protein response (UPR) as an adaptive mechanism to resolve ER stress. In addition to canonical UPR, proteotoxicity also stimulates the selective, autophagy-dependent, removal of discrete ER domains loaded with misfolded proteins to further alleviate ER stress. These mechanisms can favor cancer cell growth, metastasis, and long-term survival. Our investigations reveal that during hypoxia-induced ER stress, the ER-phagy receptor FAM134B targets damaged portions of ER into autophagosomes to restore ER homeostasis in cancer cells. Loss of FAM134B in breast cancer cells results in increased ER stress and reduced cell proliferation. Mechanistically, upon sensing hypoxia-induced proteotoxic stress, the ER chaperone BiP forms a complex with FAM134B and promotes ER-phagy. To prove the translational implication of our mechanistic findings, we identified vitexin as a pharmacological agent that disrupts FAM134B-BiP complex, inhibits ER-phagy, and potently suppresses breast cancer progression in vivo.

Cancer cells adapt FAM134B/BiP mediated ER-phagy to survive hypoxic stress / Chipurupalli, Sandhya; Ganesan, Raja; Martini, Giulia; Mele, Luigi; Reggio, Alessio; Esposito, Marianna; Kannan, Elango; Namasivayam, Vigneshwaran; Grumati, Paolo; Desiderio, Vincenzo; Robinson, Nirmal. - In: CELL DEATH & DISEASE. - ISSN 2041-4889. - 13:4(2022). [10.1038/s41419-022-04813-w]

Cancer cells adapt FAM134B/BiP mediated ER-phagy to survive hypoxic stress

Esposito, Marianna;Grumati, Paolo;
2022

Abstract

In the tumor microenvironment, cancer cells experience hypoxia resulting in the accumulation of misfolded/unfolded proteins largely in the endoplasmic reticulum (ER). Consequently, ER proteotoxicity elicits unfolded protein response (UPR) as an adaptive mechanism to resolve ER stress. In addition to canonical UPR, proteotoxicity also stimulates the selective, autophagy-dependent, removal of discrete ER domains loaded with misfolded proteins to further alleviate ER stress. These mechanisms can favor cancer cell growth, metastasis, and long-term survival. Our investigations reveal that during hypoxia-induced ER stress, the ER-phagy receptor FAM134B targets damaged portions of ER into autophagosomes to restore ER homeostasis in cancer cells. Loss of FAM134B in breast cancer cells results in increased ER stress and reduced cell proliferation. Mechanistically, upon sensing hypoxia-induced proteotoxic stress, the ER chaperone BiP forms a complex with FAM134B and promotes ER-phagy. To prove the translational implication of our mechanistic findings, we identified vitexin as a pharmacological agent that disrupts FAM134B-BiP complex, inhibits ER-phagy, and potently suppresses breast cancer progression in vivo.
2022
Cancer cells adapt FAM134B/BiP mediated ER-phagy to survive hypoxic stress / Chipurupalli, Sandhya; Ganesan, Raja; Martini, Giulia; Mele, Luigi; Reggio, Alessio; Esposito, Marianna; Kannan, Elango; Namasivayam, Vigneshwaran; Grumati, Paolo; Desiderio, Vincenzo; Robinson, Nirmal. - In: CELL DEATH & DISEASE. - ISSN 2041-4889. - 13:4(2022). [10.1038/s41419-022-04813-w]
File in questo prodotto:
Non ci sono file associati a questo prodotto.

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/11588/899904
Citazioni
  • ???jsp.display-item.citation.pmc??? ND
  • Scopus 17
  • ???jsp.display-item.citation.isi??? 15
social impact