4.8 Article

Integration of metabolomics and expression of enolase-phosphatase 1 links to hepatocellular carcinoma progression

Journal

THERANOSTICS
Volume 9, Issue 12, Pages 3639-3652

Publisher

IVYSPRING INT PUBL
DOI: 10.7150/thno.31693

Keywords

hepatocellular carcinoma; enolase-phosphatase 1; AKT; omics technologies

Funding

  1. National Natural Science Foundation of China [81872377]
  2. Tianjin Natural Science Foundation of China [16JCQNJC09600, 18JCYBJC25600]
  3. Science and Technology Development Foundation of Henan Province [172102310103]
  4. Henan Provincial Medical Science and Technology Project [2018020480]

Ask authors/readers for more resources

Reprogramming of cellular metabolism is one of the hallmarks for cancer, in which tumor cells rewire their metabolic fluxes to generate sufficient energy and biosynthetic intermediates. Therefore, elucidating the correlation between cellular metabolism and hepatocellular carcinoma (HCC) progression may provide insights into novel approaches to cancer therapy. Methods: We assembled an integrated pathway-level metabolic profiling by mining metabolomic, transcriptomic and proteomic data of three HCC cell lines with increasing metastatic potentials. Immunohistochemical staining was performed in a tissue microarray from 185 HCC clinical specimens. Kaplan-Meier survival and Cox regression analyses were applied to test the association between gene expression and survival outcome. In vitro assays were conducted to investigate the functional role of enolase-phosphatase 1 (ENOPH1) in HCC malignant behaviors. Reversed genetics analysis was performed to determine the function of ENOPH 1 in HCC metastasis. An intrahepatic mouse model further confirmed the role of ENOPH1 in metastasis. Results: We have determined that HCC cell metastasis is associated with alterations in metabolite levels and expressions of metabolic enzymes in the cysteine/methionine metabolism pathway, and show that one of metabolic enzymes, enolase-phosphatase 1 (ENOPH1), is persistently upregulated with an increase in metastatic potential. The upregulation of ENOPH1 expression was observed as an independent prognostic factor for HCC patients. ENOPH1 overexpression promoted cell migration and invasion, whereas ENOPH 1 downregulation inhibited cell migration and invasion. Furthermore, an enhanced phosphorylation of AKT with ENOPH 1 upregulation was observed. ENOPH 1-mediated malignant capacity in HCC cells can be rescued by an AKT inhibitor. Conclusion: Taken together, our findings illustrate that ENOPH1 promotes HCC progression and could serve as a novel biomarker and therapeutic target for HCC.

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