4.8 Article

SOX2 mediates metabolic reprogramming of prostate cancer cells

期刊

ONCOGENE
卷 41, 期 8, 页码 1190-1202

出版社

SPRINGERNATURE
DOI: 10.1038/s41388-021-02157-x

关键词

-

资金

  1. University of Chicago Comprehensive Cancer Center Support Grant [P30CA014599]
  2. The Brinson Foundation
  3. Alvin Baum Family Fund
  4. The Pierce Foundation
  5. National Center for Advancing Translational Sciences [UL1TR002003]
  6. Department of Defense Prostate Cancer Research Program [W81XWH-14-2-0182, W81XWH-14-2-0183, W81XWH-14-2-0185, W81XWH14-2-0186, W81XWH-15-2-0062, W81XWH-18-1-0411]
  7. Goldblatt Foundation Fellowship
  8. Cancer Biology Training Grant [T32 CA 009594]
  9. CPRIT [RR170050]
  10. Welch Foundation [I-2005-20190330]
  11. Prostate Cancer Foundation [17YOUN12]
  12. [R01CA178431]
  13. [R00CA218885-04]

向作者/读者索取更多资源

This study investigated the impact of SOX2 expression on patient outcomes and its function within prostate cancer cells. The results revealed that SOX2 expression promotes metastatic progression and therapy resistance in prostate cancer, and affects the metabolic pathways and metabolites of cancer cells. These findings contribute to a better understanding of the role of SOX2 in prostate cancer and suggest its potential as a biomarker and pharmacologic target in clinical settings.
New strategies are needed to predict and overcome metastatic progression and therapy resistance in prostate cancer. One potential clinical target is the stem cell transcription factor SOX2, which has a critical role in prostate development and cancer. We thus investigated the impact of SOX2 expression on patient outcomes and its function within prostate cancer cells. Analyses of SOX2 expression among a case-control cohort of 1028 annotated tumor specimens demonstrated that SOX2 expression confers a more rapid time to metastasis and decreased patient survival after biochemical recurrence. SOX2 ChIP-Seq analyses revealed SOX2-binding sites within prostate cancer cells which differ significantly from canonical embryonic SOX2 gene targets, and prostate-specific SOX2 gene targets are associated with multiple oncogenic pathways. Interestingly, phenotypic and gene expression analyses after CRISPR-mediated deletion of SOX2 in castration-resistant prostate cancer cells, as well as ectopic SOX2 expression in androgen-sensitive prostate cancer cells, demonstrated that SOX2 promotes changes in multiple metabolic pathways and metabolites. SOX2 expression in prostate cancer cell lines confers increased glycolysis and glycolytic capacity, as well as increased basal and maximal oxidative respiration and increased spare respiratory capacity. Further, SOX2 expression was associated with increased quantities of mitochondria, and metabolomic analyses revealed SOX2-associated changes in the metabolism of purines, pyrimidines, amino acids and sugars, and the pentose phosphate pathway. Analyses of SOX2 gene targets with central functions metabolism (CERK, ECHS1, HS6SDT1, LPCAT4, PFKP, SLC16A3, SLC46A1, and TST) document significant expression correlation with SOX2 among RNA-Seq datasets derived from patient tumors and metastases. These data support a key role for SOX2 in metabolic reprogramming of prostate cancer cells and reveal new mechanisms to understand how SOX2 enables metastatic progression, lineage plasticity, and therapy resistance. Further, our data suggest clinical opportunities to exploit SOX2 as a biomarker for staging and imaging, as well as a potential pharmacologic target.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据