Abstract

Identification of a novel squalene epoxidase function in breast cancer brain metastasis.

Author
Shu-Ping Wang Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Taipei City, Taiwan info_outline Shu-Ping Wang, Jia-Yun Yeh, Pony Yu-Ling Lee, Kun-Yuan Lin, Chao Di Chang, Chih-Chieh Yang, Shan-Yun Cheng, Ya Wen Hung, Yung-Lung Yu
Full text
Authors Shu-Ping Wang Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Taipei City, Taiwan info_outline Shu-Ping Wang, Jia-Yun Yeh, Pony Yu-Ling Lee, Kun-Yuan Lin, Chao Di Chang, Chih-Chieh Yang, Shan-Yun Cheng, Ya Wen Hung, Yung-Lung Yu Organizations Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Taipei City, Taiwan, Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan, Pharmacology Discovery Services Taiwan, Ltd., partner lab of Eurofins, New Taipei City, Taiwan, Pharmacology Discovery Services Ltd., Taiwan, New Taipei City, Taiwan, Institute of Translational Medicine and New Drug Development, China Medical University, Taichung, Taiwan Abstract Disclosures Research Funding Institutional Funding Institute of Biomedical Sciences, Academia Sinica, Taiwan and Institute of Translational Medicine and New Drug Development, China Medical University, Taiwan, Pharmacology Discovery Services Taiwan, Ltd., New Taipei City, Taiwan Background: Brain metastases are serious complications of breast cancer and there is no effective treatment. The major therapeutic issues can be attributed to the unique brain microenvironment as well as the difficulty of drug delivery to the brain due to the blood-brain barrier (BBB). BBB limits access of nutrients from the circulation and thereby makes the brain in a special condition that is hypoxic and composed of depleted metabolites, growth factors, and proteins. Nevertheless, tumor cells that are able to metastasize to the brain can necessitate metabolic adaptations for their nutrient availability and thereby succeed to colonize in the brain. However, it is still poorly understood how breast cancer cells alter their metabolic systems to manipulate brain metastasis by overcoming nutritional limitations. Methods: To probe the molecular mechanism of breast cancer to brain metastasis, we compared gene signatures in primary breast tumors and breast-to-brain metastatic tumors, and identified that SQLE (encodes squalene epoxidase, the second rate-limiting enzyme in the cholesterol biosynthesis) could be a leading-edge gene in breast-to-brain metastasis. Using the brain metastatic MDA-MB-231-BR (231-BR) and its parental MDA-MB-231 (231) cell line expressing SQLE shRNAs, we evaluated the effects of SQLE loss on cancer cell migration, invasion, and stemness by wound-healing, transwell invasion/migration, and tumorsphere formation assays. The in vitro cell-based BBB model and in vivo mouse models that develop 231-BR brain metastases were established to verify our hypothesis. Results: While loss of SQLE greatly attenuated cell invasiveness and stemness in both 231 and 231-BR cells, loss of SQLE could only affect the cell migration activity on 231 cells but not 231-BR cells. Since the ability to invade, migrate, and penetrate is critical for invasion of cancer cells, our results strongly imply the novel function of SQLE in breast cancer cell invasion, penetration, and colonization in the brain. Our RNA-seq data further identified a subset of SQLE-affected genes that is uniquely enriched in 231-BR cells and favors brain extravasation and colonization. The in vitro cell-based BBB model and in vivo mouse models that develop 231-BR brain extravasation and colonization finally verified our hypothesis that SQLE may play a unique function to promote breast cancer metastasis into the brain. Conclusions: Our findings provide new insights into contributions by SQLE in breast-to-brain extravasation and colonization and indicate that targeting SQLE may represent a therapeutic opportunity for breast cancer brain metastases.
Clinical status
Pre-clinical

6 organizations

1 drug

1 target

Drug
SQLE
Target
SQLE