Your browser doesn't support javascript.
loading
Therapeutic inhibition of USP9x-mediated Notch signaling in triple-negative breast cancer.
Jaiswal, Arushi; Murakami, Kiichi; Elia, Andrew; Shibahara, Yukiko; Done, Susan J; Wood, Stephen A; Donato, Nicholas J; Ohashi, Pamela S; Reedijk, Michael.
Afiliação
  • Jaiswal A; Department of Medical Biophysics, University of Toronto, Toronto, ON M1C 1A4, Canada.
  • Murakami K; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada.
  • Elia A; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada.
  • Shibahara Y; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada.
  • Done SJ; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M1C 1A4, Canada.
  • Wood SA; Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada.
  • Donato NJ; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M1C 1A4, Canada.
  • Ohashi PS; Laboratory Medicine Program, University Health Network, Toronto, ON M5G 2C4, Canada.
  • Reedijk M; Griffith Institute for Drug Discovery, Griffith University, Brisbane QLD 4111, Australia.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article em En | MEDLINE | ID: mdl-34518219
ABSTRACT
Triple-negative breast cancer (TNBC) is a breast cancer subtype that lacks targeted treatment options. The activation of the Notch developmental signaling pathway, which is a feature of TNBC, results in the secretion of proinflammatory cytokines and the recruitment of protumoral macrophages to the tumor microenvironment. While the Notch pathway is an obvious therapeutic target, its activity is ubiquitous, and predictably, anti-Notch therapies are burdened with significant on-target side effects. Previously, we discovered that, under conditions of cellular stress commonly found in the tumor microenvironment, the deubiquitinase USP9x forms a multiprotein complex with the pseudokinase tribbles homolog 3 (TRB3) that together activate the Notch pathway. Herein, we provide preclinical studies that support the potential of therapeutic USP9x inhibition to deactivate Notch. Using a murine TNBC model, we show that USP9x knockdown abrogates Notch activation, reducing the production of the proinflammatory cytokines, C-C motif chemokine ligand 2 (CCL2) and interleukin-1 beta (IL-1ß). Concomitant with these molecular changes, a reduction in tumor inflammation, the augmentation of antitumor immune response, and the suppression of tumor growth were observed. The pharmacological inhibition of USP9x using G9, a partially selective, small-molecule USP9x inhibitor, reduced Notch activity, remodeled the tumor immune landscape, and reduced tumor growth without associated toxicity. Proving the role of Notch, the ectopic expression of the activated Notch1 intracellular domain rescued G9-induced effects. This work supports the potential of USP9x inhibition to target Notch in metabolically vulnerable tissues like TNBC, while sparing normal Notch-dependent tissues.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Ubiquitina Tiolesterase / Receptores Notch / Neoplasias de Mama Triplo Negativas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transdução de Sinais / Ubiquitina Tiolesterase / Receptores Notch / Neoplasias de Mama Triplo Negativas Tipo de estudo: Prognostic_studies Limite: Animals / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article