File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Downregulation of the glucose transporter GLUT 1 in the cerebral microvasculature contributes to postoperative neurocognitive disorders in aged mice

TitleDownregulation of the glucose transporter GLUT 1 in the cerebral microvasculature contributes to postoperative neurocognitive disorders in aged mice
Authors
KeywordsAging
Blood–brain barrier
Glucose metabolism
GLUT1
Postoperative neurocognitive disorders
Issue Date1-Dec-2023
PublisherBioMed Central
Citation
Journal of Neuroinflammation, 2023, v. 20, n. 1 How to Cite?
AbstractIntroduction: Glucose transporter 1 (GLUT1) is essential for glucose transport into the brain and is predominantly expressed in the cerebral microvasculature. Downregulation of GLUT1 precedes the development of cognitive impairment in neurodegenerative conditions. Surgical trauma induces blood–brain barrier (BBB) disruption, neuroinflammation, neuronal mitochondria dysfunction, and acute cognitive impairment. We hypothesized that surgery reduces the expression of GLUT1 in the BBB that in turn disrupts its integrity and contributes to metabolic dysregulation in the brain that culminates in postoperative cognitive impairment. Methodology: Using an abdominal surgery model in aged WT mice, we assessed the perioperative changes in cognitive performance, tight junction proteins expression, GLUT1 expression, and the associated metabolic effects in the hippocampus. Thereafter, we evaluated the effects of these parameters in aged mice with conditional overexpression of GLUT1, and then again in aged mice with conditional overexpression of GLUT1 with or without prior exposure to the GLUT1 inhibitor ST-31. Results: We showed a significant decline in cognitive performance, along with GLUT1 reduction and diminished glucose metabolism, especially in the ATP level in the postoperative mice compared with controls. Overexpression of GLUT1 expression alleviated postoperative cognitive decline and improved metabolic profiles, especially in adenosine, but did not directly restore ATP generation to control levels. GLUT1 inhibition ameliorated the postoperative beneficial effects of GLUT1 overexpression. Conclusions: Surgery-induced GLUT1 reduction significantly contributes to postoperative cognitive deficits in aged mice by affecting glucose metabolism in the brain. It indicates the potential of targeting GLUT1 to ameliorate perioperative neurocognitive disorders.
Persistent Identifierhttp://hdl.handle.net/10722/348623

 

DC FieldValueLanguage
dc.contributor.authorChen, Ying-
dc.contributor.authorJoo, Jin-
dc.contributor.authorChu, John Man Tak-
dc.contributor.authorChang, Raymond Chuen Chung-
dc.contributor.authorWong, Gordon Tin Chun-
dc.date.accessioned2024-10-11T00:30:54Z-
dc.date.available2024-10-11T00:30:54Z-
dc.date.issued2023-12-01-
dc.identifier.citationJournal of Neuroinflammation, 2023, v. 20, n. 1-
dc.identifier.urihttp://hdl.handle.net/10722/348623-
dc.description.abstractIntroduction: Glucose transporter 1 (GLUT1) is essential for glucose transport into the brain and is predominantly expressed in the cerebral microvasculature. Downregulation of GLUT1 precedes the development of cognitive impairment in neurodegenerative conditions. Surgical trauma induces blood–brain barrier (BBB) disruption, neuroinflammation, neuronal mitochondria dysfunction, and acute cognitive impairment. We hypothesized that surgery reduces the expression of GLUT1 in the BBB that in turn disrupts its integrity and contributes to metabolic dysregulation in the brain that culminates in postoperative cognitive impairment. Methodology: Using an abdominal surgery model in aged WT mice, we assessed the perioperative changes in cognitive performance, tight junction proteins expression, GLUT1 expression, and the associated metabolic effects in the hippocampus. Thereafter, we evaluated the effects of these parameters in aged mice with conditional overexpression of GLUT1, and then again in aged mice with conditional overexpression of GLUT1 with or without prior exposure to the GLUT1 inhibitor ST-31. Results: We showed a significant decline in cognitive performance, along with GLUT1 reduction and diminished glucose metabolism, especially in the ATP level in the postoperative mice compared with controls. Overexpression of GLUT1 expression alleviated postoperative cognitive decline and improved metabolic profiles, especially in adenosine, but did not directly restore ATP generation to control levels. GLUT1 inhibition ameliorated the postoperative beneficial effects of GLUT1 overexpression. Conclusions: Surgery-induced GLUT1 reduction significantly contributes to postoperative cognitive deficits in aged mice by affecting glucose metabolism in the brain. It indicates the potential of targeting GLUT1 to ameliorate perioperative neurocognitive disorders.-
dc.languageeng-
dc.publisherBioMed Central-
dc.relation.ispartofJournal of Neuroinflammation-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectAging-
dc.subjectBlood–brain barrier-
dc.subjectGlucose metabolism-
dc.subjectGLUT1-
dc.subjectPostoperative neurocognitive disorders-
dc.titleDownregulation of the glucose transporter GLUT 1 in the cerebral microvasculature contributes to postoperative neurocognitive disorders in aged mice -
dc.typeArticle-
dc.description.naturepublished_or_final_version-
dc.identifier.doi10.1186/s12974-023-02905-8-
dc.identifier.pmid37858199-
dc.identifier.scopuseid_2-s2.0-85174488859-
dc.identifier.volume20-
dc.identifier.issue1-
dc.identifier.eissn1742-2094-
dc.identifier.issnl1742-2094-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats