Skip to main content
Article
Cbl negatively regulates nlrp3 inflammasome activation through glut1-dependent glycolysis inhibition
International Journal of Molecular Sciences
  • Hsin Chung Lin, National Defense Medical Center Taiwan
  • Yu Jen Chen, Mackay Memorial Hospital Taiwan
  • Yau Huei Wei, Changhua Christian Hospital Taiwan
  • Yu Ting Chuang, Mackay Medical College
  • Su Heng Hsieh, Mackay Medical College
  • Jing Yu Hsieh, Mackay Medical College
  • Yi Lin Hsieh, Mackay Medical College
  • David M. Ojcius, University of the Pacific
  • Kuo Yang Huang, National Defense Medical Center Taiwan
  • I. Che Chung, Chang Gung University
  • Sheng Ning Yuan, Chang Gung University
  • Yu Sun Chang, Chang Gung University
  • Lih Chyang Chen, Mackay Medical College
ORCiD
David M. Ojcius: 0000-0003-1461-4495
Department
Biomedical Sciences
Document Type
Article
DOI
10.3390/ijms21145104
Publication Date
7-2-2020
Abstract

Activation of the nod-like receptor 3 (NLRP3) inflammasomes is crucial for immune defense, but improper and excessive activation causes inflammatory diseases. We previously reported that Cbl plays a pivotal role in suppressing NLRP3 inflammasome activation by inhibiting Pyk2-mediated apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization. Here, we showed that Cbl dampened NLRP3 inflammasome activation by inhibiting glycolysis, as demonstrated with Cbl knockout cells and treatment with the Cbl inhibitor hydrocotarnine. We revealed that the inhibition of Cbl promoted caspase-1 cleavage and interleukin (IL)-1β secretion through a glycolysis-dependent mechanism. Inhibiting Cbl increased cellular glucose uptake, glycolytic capacity, and mitochondrial oxidative phosphorylation capacity. Upon NLRP3 inflammasome activation, inhibiting Cbl increased glycolysis-dependent activation of mitochondrial respiration and increased the production of reactive oxygen species, which contributes to NLRP3 inflammasome activation and IL-1β secretion. Mechanistically, inhibiting Cbl increased surface expression of glucose transporter 1 (GLUT1) protein through post-transcriptional regulation, which increased cellular glucose uptake and consequently raised glycolytic capacity, and in turn enhanced NLRP3 inflammasome activation. Together, our findings provide new insights into the role of Cbl in NLRP3 inflammasome regulation through GLUT1 downregulation. We also show that a novel Cbl inhibitor, hydrocortanine, increased NLRP3 inflammasome activity via its effect on glycolysis.

Creative Commons License
Creative Commons Attribution-NonCommercial-No Derivative Works 4.0 International
Citation Information
Hsin Chung Lin, Yu Jen Chen, Yau Huei Wei, Yu Ting Chuang, et al.. "Cbl negatively regulates nlrp3 inflammasome activation through glut1-dependent glycolysis inhibition" International Journal of Molecular Sciences Vol. 21 Iss. 14 (2020) p. 1 - 16 ISSN: 1661-6596
Available at: http://works.bepress.com/david-ojcius/333/