4.8 Article

PELP1/SRC-3-dependent regulation of metabolic PFKFB kinases drives therapy resistant ER+ breast cancer

Journal

ONCOGENE
Volume 40, Issue 25, Pages 4384-4397

Publisher

SPRINGERNATURE
DOI: 10.1038/s41388-021-01871-w

Keywords

-

Funding

  1. NIH [R01 CA236948, R01 CA229697, F32 CA210340, T32 HL007741, U54 CA224076, R01 CA248158-01, R01 AG069727-01]
  2. ACS Institutional Research Grant [124166-IRG-58-001-52-IRG5]
  3. University of Minnesota Masonic Cancer Center
  4. Tickle Family Land Grant Endowed Chair in Breast Cancer Research
  5. National Center for Advancing Translational Sciences of the NIH [UL1TR000114]
  6. Department of Defense [W81XWH-14-1-0417]

Ask authors/readers for more resources

Recurrence of metastatic breast cancer in women with luminal (ER+) breast cancer is a significant health burden, with contributing factors including the maintenance and expansion of breast cancer stem cells. The study demonstrates that cytoplasmic complexes composed of steroid receptor co-activators PELP1 and SRC-3 modulate breast cancer stem cell expansion by upregulating metabolic target genes PFKFB3 and PFKFB4. This suggests a potential new approach to overcoming endocrine and chemotherapy resistance in ER+ tumor cell populations.
Recurrence of metastatic breast cancer stemming from acquired endocrine and chemotherapy resistance remains a health burden for women with luminal (ER+) breast cancer. Disseminated ER+ tumor cells can remain viable but quiescent for years to decades. Contributing factors to metastatic spread include the maintenance and expansion of breast cancer stem cells (CSCs). Breast CSCs frequently exist as a minority population in therapy resistant tumors. In this study, we show that cytoplasmic complexes composed of steroid receptor (SR) co-activators, PELP1 and SRC-3, modulate breast CSC expansion through upregulation of the HIF-activated metabolic target genes PFKFB3 and PFKFB4. Seahorse metabolic assays demonstrated that cytoplasmic PELP1 influences cellular metabolism by increasing both glycolysis and mitochondrial respiration. PELP1 interacts with PFKFB3 and PFKFB4 proteins, and inhibition of PFKFB3 and PFKFB4 kinase activity blocks PELP1-induced tumorspheres and protein-protein interactions with SRC-3. PFKFB4 knockdown inhibited in vivo emergence of circulating tumor cell (CTC) populations in mammary intraductal (MIND) models. Application of PFKFB inhibitors in combination with ER targeted therapies blocked tumorsphere formation in multiple models of advanced breast cancer including tamoxifen (TamR) and paclitaxel (TaxR) resistant models, murine tumor cells, and ER+ patient-derived organoids (PDxO). Together, our data suggest that PELP1, SRC-3, and PFKFBs cooperate to drive ER+ tumor cell populations that include CSCs and CTCs. Identifying non-ER pharmacological targets offers a useful approach to blocking metastatic escape from standard of care ER/estrogen (E2)-targeted strategies to overcome endocrine and chemotherapy resistance.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available