File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)

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

TitlePMI-controlled mannose metabolism and glycosylation determines tissue tolerance and virus fitness
Authors
Issue Date8-Mar-2024
PublisherTaylor and Francis Group
Citation
Environmental Communication: A Journal of Nature and Culture, 2024, v. 15, n. 1 How to Cite?
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.


Persistent Identifierhttp://hdl.handle.net/10722/347331
ISSN
2023 Impact Factor: 3.0
2023 SCImago Journal Rankings: 0.952

 

DC FieldValueLanguage
dc.contributor.authorLiang, Ronghui-
dc.contributor.authorYe, Zi Wei-
dc.contributor.authorQin, Zhenzhi-
dc.contributor.authorXie, Yubin-
dc.contributor.authorYang, Xiaomeng-
dc.contributor.authorSun, Haoran-
dc.contributor.authorDu, Qiaohui-
dc.contributor.authorLuo, Peng-
dc.contributor.authorTang, Kaiming-
dc.contributor.authorHu, Bodan-
dc.contributor.authorCao, Jianli-
dc.contributor.authorWong, Xavier Hoi-Leong-
dc.contributor.authorLing, Guang-Sheng-
dc.contributor.authorChu, Hin-
dc.contributor.authorShen, Jiangang-
dc.contributor.authorYin, Feifei-
dc.contributor.authorJin, Dong-Yan-
dc.contributor.authorChan, Jasper Fuk-Woo-
dc.contributor.authorYuen, Kwok-Yung-
dc.contributor.authorYuan, Shuofeng-
dc.date.accessioned2024-09-21T00:31:02Z-
dc.date.available2024-09-21T00:31:02Z-
dc.date.issued2024-03-08-
dc.identifier.citationEnvironmental Communication: A Journal of Nature and Culture, 2024, v. 15, n. 1-
dc.identifier.issn1752-4032-
dc.identifier.urihttp://hdl.handle.net/10722/347331-
dc.description.abstract<p>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.</p>-
dc.languageeng-
dc.publisherTaylor and Francis Group-
dc.relation.ispartofEnvironmental Communication: A Journal of Nature and Culture-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.titlePMI-controlled mannose metabolism and glycosylation determines tissue tolerance and virus fitness-
dc.typeArticle-
dc.identifier.doi10.1038/s41467-024-46415-4-
dc.identifier.scopuseid_2-s2.0-85187117958-
dc.identifier.volume15-
dc.identifier.issue1-
dc.identifier.eissn1752-4040-
dc.identifier.issnl1752-4032-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats