Nature Communications (Mar 2024)

PMI-controlled mannose metabolism and glycosylation determines tissue tolerance and virus fitness

  • Ronghui Liang,
  • Zi-Wei Ye,
  • Zhenzhi Qin,
  • Yubin Xie,
  • Xiaomeng Yang,
  • Haoran Sun,
  • Qiaohui Du,
  • Peng Luo,
  • Kaiming Tang,
  • Bodan Hu,
  • Jianli Cao,
  • Xavier Hoi-Leong Wong,
  • Guang-Sheng Ling,
  • Hin Chu,
  • Jiangang Shen,
  • Feifei Yin,
  • Dong-Yan Jin,
  • Jasper Fuk-Woo Chan,
  • Kwok-Yung Yuen,
  • Shuofeng Yuan

DOI
https://doi.org/10.1038/s41467-024-46415-4
Journal volume & issue
Vol. 15, no. 1
pp. 1 – 15

Abstract

Read online

Abstract Host survival depends on the elimination of virus and mitigation of tissue damage. Herein, we report the modulation of D-mannose flux rewires the virus-triggered immunometabolic response cascade and reduces tissue damage. Safe and inexpensive D-mannose can compete with glucose for the same transporter and hexokinase. Such competitions suppress glycolysis, reduce mitochondrial reactive-oxygen-species and succinate-mediated hypoxia-inducible factor-1α, and thus reduce virus-induced proinflammatory cytokine production. The combinatorial treatment by D-mannose and antiviral monotherapy exhibits in vivo synergy despite delayed antiviral treatment in mouse model of virus infections. Phosphomannose isomerase (PMI) knockout cells are viable, whereas addition of D-mannose to the PMI knockout cells blocks cell proliferation, indicating that PMI activity determines the beneficial effect of D-mannose. PMI inhibition suppress a panel of virus replication via affecting host and viral surface protein glycosylation. However, D-mannose does not suppress PMI activity or virus fitness. Taken together, PMI-centered therapeutic strategy clears virus infection while D-mannose treatment reprograms glycolysis for control of collateral damage.