1. Academic Validation
  2. Exosomal circSIPA1L3-mediated intercellular communication contributes to glucose metabolic reprogramming and progression of triple negative breast cancer

Exosomal circSIPA1L3-mediated intercellular communication contributes to glucose metabolic reprogramming and progression of triple negative breast cancer

  • Mol Cancer. 2024 Jun 8;23(1):125. doi: 10.1186/s12943-024-02037-4.
Yiran Liang # 1 Fangzhou Ye # 1 Dan Luo 1 Li Long 1 2 Yajie Wang 1 Yuhan Jin 1 Lei Wang 1 Yaming Li 1 Dianwen Han 1 Bing Chen 3 Wenjing Zhao 3 Lijuan Wang 3 Qifeng Yang 4 5 6
Affiliations

Affiliations

  • 1 Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, 250012, P.R. China.
  • 2 Department of Breast Surgery, Mianyang Central Hospital, Mianyang, Sichuan, 621000, P.R. China.
  • 3 Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China.
  • 4 Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Wenhua Xi Road No. 107, Jinan, Shandong, 250012, P.R. China. qifengy_sdu@163.com.
  • 5 Biological Resource Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, P.R. China. qifengy_sdu@163.com.
  • 6 Research Institute of Breast Cancer, Shandong University, Jinan, Shandong, 250012, P.R. China. qifengy_sdu@163.com.
  • # Contributed equally.
Abstract

Background: Breast Cancer is the most common malignant tumor, and metastasis remains the major cause of poor prognosis. Glucose metabolic reprogramming is one of the prominent hallmarks in Cancer, providing nutrients and energy to support dramatically elevated tumor growth and metastasis. Nevertheless, the potential mechanistic links between glycolysis and breast Cancer progression have not been thoroughly elucidated.

Methods: RNA-seq analysis was used to identify glucose metabolism-related circRNAs. The expression of circSIPA1L3 in breast Cancer tissues and serum was examined by qRT-PCR, and further assessed its diagnostic value. We also evaluated the prognostic potential of circSIPA1L3 by analyzing a cohort of 238 breast Cancer patients. Gain- and loss-of-function experiments, transcriptomic analysis, and Molecular Biology experiments were conducted to explore the biological function and regulatory mechanism of circSIPA1L3.

Results: Using RNA-seq analysis, circSIPA1L3 was identified as the critical mediator responsible for metabolic adaption upon energy stress. Gain- and loss-of-function experiments revealed that circSIPA1L3 exerted a stimulative effect on breast Cancer progression and glycolysis, which could also be transported by exosomes and facilitated malignant behaviors among breast Cancer cells. Significantly, the elevated lactate secretion caused by circSIPA1L3-mediated glycolysis enhancement promoted the recruitment of tumor associated macrophage and their tumor-promoting roles. Mechanistically, EIF4A3 induced the cyclization and cytoplasmic export of circSIPA1L3, which inhibited ubiquitin-mediated IGF2BP3 degradation through enhancing the UPS7-IGF2BP3 interaction. Furthermore, circSIPA1L3 increased mRNA stability of the lactate export carrier SLC16A1 and the glucose intake enhancer RAB11A through either strengthening their interaction with IGF2BP3 or sponging miR-665, leading to enhanced glycolytic metabolism. Clinically, elevated circSIPA1L3 expression indicated unfavorable prognosis base on the cohort of 238 breast Cancer patients. Moreover, circSIPA1L3 was highly expressed in the serum of breast Cancer patients and exhibited high diagnostic value for breast Cancer patients.

Conclusions: Our study highlights the oncogenic role of circSIPA1L3 through mediating glucose metabolism, which might serve as a promising diagnostic and prognostic biomarker and potential therapeutic target for breast Cancer.

Keywords

Breast cancer; Exosome; Glycolysis; IGF2BP3; Stemness; circSIPA1L3.

Figures
Products