4.7 Article

Kynurenine 3-monooxygenase upregulates pluripotent genes through beta-catenin and promotes triple-negative breast cancer progression

Journal

EBIOMEDICINE
Volume 54, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.ebiom.2020.102717

Keywords

KM0; Triple negative breast cancer; beta-catenin

Funding

  1. Taiwan Clinical Oncology Research Foundation
  2. Yen Tjing Ling Medical Foundation [CI-107-10, CI-108-15]
  3. Ministry of Science and Technology, Taiwan [MOST 104-2628-B-075-001-MY3]
  4. Yang-Ming Branch of Taipei City Hospital [10701-62-030, 10801-62-071]
  5. Taipei Veterans General Hospital [V106C-101, V107C-025]
  6. Ministry of Health and Welfare, Executive Yuan, Taiwan [MOHW107-TDU-B-212-112015]
  7. Taipei Veterans General Hospital
  8. National Taiwan-University Hospital

Ask authors/readers for more resources

Background: Triple-negative breast cancer (TNBC) is aggressive and has a poor prognosis. Kynurenine 3-monooxygenase (KMO), a crucial kynurenine metabolic enzyme, is involved in inflammation, immune response and tumorigenesis. We aimed to study the role of KMO in TNBC. Methods: KMO alteration and expression data from public databases were analyzed. KMO expression levels in TNBC samples were analyzed using immunohistochemistry. Knockdown of KMO in TNBC cells was achieved by RNAi and CRISPR/Cas9. KMO functions were examined by MTT, colony-forming, transwell migration/invasion, and mammosphere assays. The molecular events were analyzed by cDNA microarrays, Western blot, quantitative real-time PCR and luciferase reporter assays. Tumor growth and metastasis were detected by orthotopic xenograft and tail vein metastasis mouse models, respectively. Findings: KMO was amplified and associated with worse survival in breast cancer patients. KMO expression levels were higher in TNBC tumors compared to adjacent normal mammary tissues. In vitro ectopic KMO expression increased cell growth, colony and mammosphere formation, migration, invasion as well as mesenchymal marker expression levels in TNBC cells. In addition, KMO increased pluripotent gene expression levels and promoter activities in vitro. Mechanistically, KMO was associated with beta-catenin and prevented beta-catenin degradation, thereby enhancing the transcription of pluripotent genes. KMO knockdown suppressed tumor growth and the expression levels of beta-catenin, CD44 and Nanog. Furthermore, mutant KMO (known with suppressed enzymatic activity) could still promote TNBC cell migration/invasion. Importantly, mice bearing CRISPR KMO-knockdown TNBC tumors showed decreased lung metastasis and prolonged survival. Interpretation: KMO regulates pluripotent genes via beta-catenin and plays an oncogenic role in TNBC progression. (C) 2020 Published by Elsevier B.V.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available