Cargando…

NADPH oxidase 1 supports proliferation of colon cancer cells by modulating reactive oxygen species-dependent signal transduction

Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O(2)(˙̄), is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decreas...

Descripción completa

Detalles Bibliográficos
Autores principales: Juhasz, Agnes, Markel, Susan, Gaur, Shikha, Liu, Han, Lu, Jiamo, Jiang, Guojian, Wu, Xiwei, Antony, Smitha, Wu, Yongzhong, Melillo, Giovanni, Meitzler, Jennifer L., Haines, Diana C., Butcher, Donna, Roy, Krishnendu, Doroshow, James H.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Biochemistry and Molecular Biology 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5427267/
https://www.ncbi.nlm.nih.gov/pubmed/28330872
http://dx.doi.org/10.1074/jbc.M116.768283
Descripción
Sumario:Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O(2)(˙̄), is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decrease NOX1 expression stably in HT-29 human colon cancer cells. The 80–90% decrease in NOX1 expression achieved by RNAi produced a significant decline in ROS production and a G(1)/S block that translated into a 2–3-fold increase in tumor cell doubling time without increased apoptosis. The block at the G(1)/S checkpoint was associated with a significant decrease in cyclin D(1) expression and profound inhibition of mitogen-activated protein kinase (MAPK) signaling. Decreased steady-state MAPK phosphorylation occurred concomitant with a significant increase in protein phosphatase activity for two colon cancer cell lines in which NOX1 expression was knocked down by RNAi. Diminished NOX1 expression also contributed to decreased growth, blood vessel density, and VEGF and hypoxia-inducible factor 1α (HIF-1α) expression in HT-29 xenografts initiated from NOX1 knockdown cells. Microarray analysis, supplemented by real-time PCR and Western blotting, revealed that the expression of critical regulators of cell proliferation and angiogenesis, including c-MYC, c-MYB, and VEGF, were down-regulated in association with a decline in hypoxic HIF-1α protein expression downstream of silenced NOX1 in both colon cancer cell lines and xenografts. These studies suggest a role for NOX1 in maintaining the proliferative phenotype of some colon cancers and the potential of NOX1 as a therapeutic target in this disease.