Your browser doesn't support javascript.
loading
Downregulation of the glucose transporter GLUT 1 in the cerebral microvasculature contributes to postoperative neurocognitive disorders in aged mice.
Chen, Ying; Joo, Jin; Chu, John Man-Tak; Chang, Raymond Chuen-Chung; Wong, Gordon Tin-Chun.
Afiliación
  • Chen Y; Department of Anaesthesiology, LKS Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Room K424, 4Th Floor, K Block, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China.
  • Joo J; Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, L4-49, Laboratory Block, Faculty of Medicine Building, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China.
  • Chu JM; Department of Anesthesiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
  • Chang RC; Department of Anaesthesiology, LKS Faculty of Medicine, Queen Mary Hospital, The University of Hong Kong, Room K424, 4Th Floor, K Block, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China.
  • Wong GT; Department of Anaesthesia and Pain Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seocho-Gu, Seoul, 06591, Korea.
J Neuroinflammation ; 20(1): 237, 2023 Oct 19.
Article en En | MEDLINE | ID: mdl-37858199
ABSTRACT

INTRODUCTION:

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.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Barrera Hematoencefálica / Trastornos del Conocimiento Límite: Animals Idioma: En Revista: J Neuroinflammation Asunto de la revista: NEUROLOGIA Año: 2023 Tipo del documento: Article País de afiliación: China

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Barrera Hematoencefálica / Trastornos del Conocimiento Límite: Animals Idioma: En Revista: J Neuroinflammation Asunto de la revista: NEUROLOGIA Año: 2023 Tipo del documento: Article País de afiliación: China