J Cancer 2022; 13(10):3031-3043. doi:10.7150/jca.72793 This issue Cite

Research Paper

Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells

Su Hyun Lee1, Ji Sun Lee1, Jae Hyeon Park1, Sungpil Yoon1, Kwang Youl Lee2✉, Hyung Sik Kim1✉

1. School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon, 16419, Republic of Korea.
2. College of Pharmacy, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea.

Citation:
Lee SH, Lee JS, Park JH, Yoon S, Lee KY, Kim HS. Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells. J Cancer 2022; 13(10):3031-3043. doi:10.7150/jca.72793. https://www.jcancer.org/v13p3031.htm
Other styles

File import instruction

Abstract

Graphic abstract

Hedgehog (Hh) signaling pathway plays an essential role in embryonic development, tissue regeneration, and stem cell renewal. In particular, terminal effectors of the Hh signaling pathway are associated with the regulation of glioma-associated oncogene homolog 1 (GLI1) transcription factors. Overexpression of GLI1 is closely associated with poor prognosis in breast cancer. The Hh-GLI1 signaling pathway is activated and participates in the tumorigenesis and progression of breast cancer, especially in the aggressive subtype of triple-negative breast cancer (TNBC). However, the role of GLI1 in regulating TNBC metabolism remains unclear. This study aimed to explore the functional role of GLI1 in glycolytic metabolism in TNBC. Immunohistochemical analysis of GLI1 expression in a tissue microarray revealed significant correlations between GLI1 expression and advanced tumor stage and grade. GLI1 expression levels were drastically increased in MDA-MB-231 cells compared to those in other cell lines. Inhibition of GLI1 expression using GLI1 small interfering RNA (siRNA) in MDA-MB-231 cells resulted in a significant reduction in cell proliferation and induced cell cycle arrest at the G1 phase. Furthermore, GLI1 downregulation significantly reduced the expression of glycolysis-regulated proteins. GLI1 knockdown resulted in reduced glycolytic rates and extracellular lactate levels. Moreover, metabolic stress after GLI1 knockdown activated the energy sensor, adenosine monophosphate-activated protein kinase, which subsequently resulted in autophagy induction. In conclusion, this study indicates that targeting GLI1 reprograms the tumor glucose metabolism to suppress breast cancer cell growth and proliferation.

Keywords: autophagy, glioma-associated oncogene, glycolytic rates, hedgehog (Hh) signaling, TNBC


Citation styles

APA
Lee, S.H., Lee, J.S., Park, J.H., Yoon, S., Lee, K.Y., Kim, H.S. (2022). Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells. Journal of Cancer, 13(10), 3031-3043. https://doi.org/10.7150/jca.72793.

ACS
Lee, S.H.; Lee, J.S.; Park, J.H.; Yoon, S.; Lee, K.Y.; Kim, H.S. Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells. J. Cancer 2022, 13 (10), 3031-3043. DOI: 10.7150/jca.72793.

NLM
Lee SH, Lee JS, Park JH, Yoon S, Lee KY, Kim HS. Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells. J Cancer 2022; 13(10):3031-3043. doi:10.7150/jca.72793. https://www.jcancer.org/v13p3031.htm

CSE
Lee SH, Lee JS, Park JH, Yoon S, Lee KY, Kim HS. 2022. Glycolytic Metabolic Remodeling by the Truncate of Glioma-Associated Oncogene Homolog 1 in Triple-Negative Breast Cancer Cells. J Cancer. 13(10):3031-3043.

This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
Popup Image