4.8 Article

Cystatin B increases autophagic flux by sustaining proteolytic activity of cathepsin B and fuels glycolysis in pancreatic cancer: CSTB orchestrates autophagy and glycolysis in PDAC

Journal

CLINICAL AND TRANSLATIONAL MEDICINE
Volume 12, Issue 12, Pages -

Publisher

JOHN WILEY & SONS LTD
DOI: 10.1002/ctm2.1126

Keywords

autophagy; biomarker; cystatin B; cystatin C; cathepsin B; glycolysis; pancreatic ductal adenocarcinoma

Funding

  1. National Natural Science Foundation of China [81470894, 81802316, 81871906, 82073326, 81902387]
  2. Shanghai Rising-Star Program [KY2019460]
  3. Shanghai Municipal Education Commission-Gaofeng Clinical Medicine [RC20200037]
  4. Medical-Engineering Cross Foundation of Shanghai Jiao Tong University [ZH2018ZDA01]
  5. Shanghai Collaborative Innovation Center for Translational Medicine [TM201903]
  6. Shanghai Sailing Program [19YF1431100]

Ask authors/readers for more resources

This study found that autophagy and glycolysis play important roles in tumorigenesis of pancreatic ductal adenocarcinoma (PDAC). Cystatin B (CSTB) acts as a key player in orchestrating these processes to ensure energy supply in PDAC cells.
BackgroundBoth autophagy and glycolysis are essential for pancreatic ductal adenocarcinoma (PDAC) survival due to desmoplasia. We investigated whether targeting a hub gene which participates in both processes could be an efficient strategy for PDAC treatment. MethodsThe expression pattern of glycolysis signatures (GS) and autophagy signatures (AS) and their correlation with cystatin B (CSTB) in PDAC were analysed. It was discovered how CSTB affected the growth, glycolysis, and autophagy of PDAC cells. We assessed competitive binding to cathepsin B (CTSB) between CSTB and cystatin C (CSTC) via immunoprecipitation (IP) and immunofluorescence (IF). Chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR) and luciferase reporter gene assays were used to unveil the mechanism underlying CSTB upregulation. The expression pattern of CSTB was examined in clinical samples and KrasG12D/+, Trp53R172H/+, Pdx1-Cre (KPC) mice. ResultsGS and AS were enriched and closely associated in PDAC tissues. CSTB increased autophagic flux and provided substrates for glycolysis. CSTB knockdown attenuated the proliferation of PDAC cells and patient-derived xenografts. The liquid chromatography-tandem mass spectrometry assay indicated CSTB interacted with CTSB and contributed to the proteolytic activity of CTSB in lysosomes. IF and IP assays demonstrated that CSTB competed with CSTC to bind to CTSB. Mutation of the key sites of CSTB abolished the interaction between CSTB and CTSB. CSTB was highly expressed in PDAC due to H3K27acetylation and SP1 expression. High expression of CSTB in PDAC was observed in tissue microarray and patients' serum samples. ConclusionsOur work demonstrated the tumorigenic roles of autophagy and glycolysis in PDAC. CSTB is a key role in orchestrating these processes to ensure energy supply of PDAC cells.

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