4.5 Article

Mutant p53 Attenuates Oxidative Phosphorylation and Facilitates Cancer Stemness through Downregulating miR-200c-PCK2 Axis in Basal-Like Breast Cancer

Journal

MOLECULAR CANCER RESEARCH
Volume 19, Issue 11, Pages 1900-1916

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1541-7786.MCR-21-0098

Keywords

-

Funding

  1. Ministry of Science and Technology [105-2320-B-009-004, 106-2320-B-009-002, 107-2628-B-009-002, 108-2628-B-009-002, 109-2628-B-009-004, 109-2311-B-009-002, 109-2314-B-001-002, 109-2314-B-001-008, 107-2320-B-009-006-MY2, 109-2320-B-009-002]
  2. Smart Platform of Dynamic Systems Biology for Therapeutic Development from The Featured Areas Research Center Program within the Ministry of Education (MOE) in Taiwan
  3. Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B) from The Featured Areas Research Center Program within the Ministry of Education (MOE) in Taiwan
  4. Kaohsiung Medical University [KMU-TC108A04-0]

Ask authors/readers for more resources

miR-200c acts as a tumor suppressor miRNA that regulates epithelial phenotype and cancer stemness. p53 deficiency downregulates miR-200c expression, leading to breast cancer progression. CRISPR-mediated knockout of miR-200c induces metabolic changes similar to those caused by p53 mutations and impairs stemness induced by miR-200c deficiency.
miR-200c is a tumor suppressor miRNA that plays a critical role in regulating epithelial phenotype and cancer stemness. p53 deficiency downregulates the expression of miR-200c and leads to epithelial-mesenchymal transition (EMT) and stemness phenotype, which contributes to the progression of breast cancers. In this study, we demonstrated that CRISPR-mediated knockout (KO) of miR-200c induces metabolic features similar to the metabolic rewiring caused by p53 hot-spot mutations, and that impairing this metabolic reprogramming interferes with miR-200c deficiency-induced stemness and transformation. Moreover, restoring miR-200c expression compromised EMT, stem-cell properties, and the Warburg effect caused by p53 mutations, suggesting that mutant p53 (MTp53) induces EMT-associated phenotypes and metabolic reprogramming by downregulating miR-200c. Mechanistically, decreased expression of PCK2 was observed in miR-200c- and p53-deficient mammary epithelial cells, and forced expression of miR-200c restored PCK2 in p53 mutant-expressing cells. Reduced PCK2 expression not only led to attenuated oxidative phosphorylation (OXPHOS) and increased stemness in normal mammary epithelial cells but also compromised the enhanced OXPHOS and suppression of cancer stemness exerted by miR-200c in p53 mutation-bearing basal-like breast cancer (BLBC) cells. Clinically, PCK2 expression is negatively associated with EMT markers and is downregulated in basal-like subtype and cases with low miR-200c expression or p53 mutation. Notably, low expression of PCK2 is associated with poor overall survival (OS) in patients with breast cancer.

Authors

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

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available