4.8 Article

ApoC1 promotes the metastasis of clear cell renal cell carcinoma via activation of STAT3

Journal

ONCOGENE
Volume 39, Issue 39, Pages 6203-6217

Publisher

SPRINGERNATURE
DOI: 10.1038/s41388-020-01428-3

Keywords

-

Funding

  1. National Natural Science Foundation of China [81702887, 81272473]
  2. Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province [2020E10021]
  3. Key Medical Discipline of Zhejiang Province [2018-2-3]
  4. Key Medical Discipline of Hangzhou City [2017-51-07]
  5. Zhejiang Provincial Foundation of Natural Science [LY19H310004]
  6. Hangzhou Major Science and Technology Project [20172016A01]
  7. High-level Talents Coming Back from Abroad Innovation and Entrepreneurship Program in Hangzhou, Scientific and Technological Developing Scheme of Hangzhou City [20191203B49]
  8. Science Research Foundation of Zhejiang Health Bureau [2020RC026]
  9. Teachers Research Fund of Zhejiang University City College [J-19006]

Ask authors/readers for more resources

Clear cell renal cell carcinoma (ccRCC) is the most common renal cancer and frequently diagnosed at an advanced stage. It is prone to develop unpredictable metastases even with proper treatment. Antiangiogenic therapy is the most effective medical treatment for metastatic ccRCC. Thus, exploration of novel approaches to inhibit angiogenesis and metastasis may potentially lead to a better therapeutic option for ccRCC. Among all the types of cancer, renal cancer samples exhibited the maximum upregulation of ApoC1 as referred to in the Oncomine database. The expression of ApoC1 was increased accompanied by ccRCC progression. A high level of ApoC1 was closely related to poor survival time in ccRCC patients. Furthermore, ApoC1 was over-expressed in the highly invasive ccRCC cells as compared to that in the low-invasive ccRCC cells. Besides, ApoC1 promoted metastasis of ccRCC cells via EMT pathway, whereas depletion of ApoC1 alleviated these effects. ApoC1 as a novel pro-metastatic factor facilitates the activation of STAT3 and enhances the metastasis of ccRCC cells. Meanwhile, ApoC1 in the exosomes were transferred from the ccRCC cells to the vascular endothelial cells and promoted metastasis of the ccRCC cells via activating STAT3. Finally, the metastatic potential of the ccRCC cells driven by ApoC1 was suppressed by DPP-4 inhibition. Our study not only identifies a novel ApoC1-STAT3 pathway in ccRCC metastasis but also provides direction for the exploration of novel strategies to predict and treat metastatic ccRCC in the future.

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