Home > Research > Publications & Outputs > A Systematic Review of Glucose Transport Altera...

Links

Text available via DOI:

View graph of relations

A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Published

Standard

A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease. / Kyrtata, N.; Emsley, H.C.A.; Sparasci, O. et al.
In: Frontiers in Neuroscience, Vol. 15, 626636, 20.05.2021.

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Harvard

Kyrtata, N, Emsley, HCA, Sparasci, O, Parkes, LM & Dickie, BR 2021, 'A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease', Frontiers in Neuroscience, vol. 15, 626636. https://doi.org/10.3389/fnins.2021.626636

APA

Kyrtata, N., Emsley, H. C. A., Sparasci, O., Parkes, L. M., & Dickie, B. R. (2021). A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease. Frontiers in Neuroscience, 15, Article 626636. https://doi.org/10.3389/fnins.2021.626636

Vancouver

Kyrtata N, Emsley HCA, Sparasci O, Parkes LM, Dickie BR. A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease. Frontiers in Neuroscience. 2021 May 20;15:626636. doi: 10.3389/fnins.2021.626636

Author

Kyrtata, N. ; Emsley, H.C.A. ; Sparasci, O. et al. / A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease. In: Frontiers in Neuroscience. 2021 ; Vol. 15.

Bibtex

@article{c231c1cc075c4a8d9682e0e90edf398f,
title = "A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease",
abstract = "Introduction: Alzheimer's disease (AD) is characterized by cerebral glucose hypometabolism. Hypometabolism may be partly due to reduced glucose transport at the blood-brain barrier (BBB) and across astrocytic and neuronal cell membranes. Glucose transporters (GLUTs) are integral membrane proteins responsible for moving glucose from the bloodstream to parenchymal cells where it is metabolized, and evidence indicates vascular and non-vascular GLUTs are altered in AD brains, a process which could starve the brain of glucose and accelerate cognitive decline. Here we review the literature on glucose transport alterations in AD from human and rodent studies. Methods: Literature published between 1st January 1946 and 1st November 2020 within EMBASE and MEDLINE databases was searched for the terms “glucose transporters” AND “Alzheimer's disease”. Human and rodent studies were included while reviews, letters, and in-vitro studies were excluded. Results: Forty-three studies fitting the inclusion criteria were identified, covering human (23 studies) and rodent (20 studies). Post-mortem studies showed consistent reductions in GLUT1 and GLUT3 in the hippocampus and cortex of AD brains, areas of the brain closely associated with AD pathology. Tracer studies in rodent models of AD and human AD also exhibit reduced uptake of glucose and glucose-analogs into the brain, supporting these findings. Longitudinal rodent studies clearly indicate that changes in GLUT1 and GLUT3 only occur after amyloid-β pathology is present, and several studies indicate amyloid-β itself may be responsible for GLUT changes. Furthermore, evidence from human and rodent studies suggest GLUT depletion has severe effects on brain function. A small number of studies show GLUT2 and GLUT12 are increased in AD. Anti-diabetic medications improved glucose transport capacity in AD subjects. Conclusions: GLUT1 and GLUT3 are reduced in hippocampal and cortical regions in patients and rodent models of AD, and may be caused by high levels of amyloid-β in these regions. GLUT3 reductions appear to precede the onset of clinical symptoms. GLUT2 and GLUT12 appear to increase and may have a compensatory role. Repurposing anti-diabetic drugs to modify glucose transport shows promising results in human studies of AD. {\textcopyright} Copyright {\textcopyright} 2021 Kyrtata, Emsley, Sparasci, Parkes and Dickie.",
keywords = "Alzheimer's disease, blood-brain barrier, glucose transporters, GLUT 1, GLUT 3, amyloid beta protein, glucose transporter, glucose transporter 1, glucose transporter 2, glucose transporter 3, GLUT12 protein, liraglutide, placebo, unclassified drug, Alzheimer disease, blood brain barrier, brain cortex, brain function, brain level, brain metabolism, glucose metabolism, glucose transport, hippocampus, human, insulin resistance, neuropathology, nonhuman, protein depletion, Review, systematic review",
author = "N. Kyrtata and H.C.A. Emsley and O. Sparasci and L.M. Parkes and B.R. Dickie",
year = "2021",
month = may,
day = "20",
doi = "10.3389/fnins.2021.626636",
language = "English",
volume = "15",
journal = "Frontiers in Neuroscience",
issn = "1662-453X",
publisher = "Frontiers Media S.A.",

}

RIS

TY - JOUR

T1 - A Systematic Review of Glucose Transport Alterations in Alzheimer's Disease

AU - Kyrtata, N.

AU - Emsley, H.C.A.

AU - Sparasci, O.

AU - Parkes, L.M.

AU - Dickie, B.R.

PY - 2021/5/20

Y1 - 2021/5/20

N2 - Introduction: Alzheimer's disease (AD) is characterized by cerebral glucose hypometabolism. Hypometabolism may be partly due to reduced glucose transport at the blood-brain barrier (BBB) and across astrocytic and neuronal cell membranes. Glucose transporters (GLUTs) are integral membrane proteins responsible for moving glucose from the bloodstream to parenchymal cells where it is metabolized, and evidence indicates vascular and non-vascular GLUTs are altered in AD brains, a process which could starve the brain of glucose and accelerate cognitive decline. Here we review the literature on glucose transport alterations in AD from human and rodent studies. Methods: Literature published between 1st January 1946 and 1st November 2020 within EMBASE and MEDLINE databases was searched for the terms “glucose transporters” AND “Alzheimer's disease”. Human and rodent studies were included while reviews, letters, and in-vitro studies were excluded. Results: Forty-three studies fitting the inclusion criteria were identified, covering human (23 studies) and rodent (20 studies). Post-mortem studies showed consistent reductions in GLUT1 and GLUT3 in the hippocampus and cortex of AD brains, areas of the brain closely associated with AD pathology. Tracer studies in rodent models of AD and human AD also exhibit reduced uptake of glucose and glucose-analogs into the brain, supporting these findings. Longitudinal rodent studies clearly indicate that changes in GLUT1 and GLUT3 only occur after amyloid-β pathology is present, and several studies indicate amyloid-β itself may be responsible for GLUT changes. Furthermore, evidence from human and rodent studies suggest GLUT depletion has severe effects on brain function. A small number of studies show GLUT2 and GLUT12 are increased in AD. Anti-diabetic medications improved glucose transport capacity in AD subjects. Conclusions: GLUT1 and GLUT3 are reduced in hippocampal and cortical regions in patients and rodent models of AD, and may be caused by high levels of amyloid-β in these regions. GLUT3 reductions appear to precede the onset of clinical symptoms. GLUT2 and GLUT12 appear to increase and may have a compensatory role. Repurposing anti-diabetic drugs to modify glucose transport shows promising results in human studies of AD. © Copyright © 2021 Kyrtata, Emsley, Sparasci, Parkes and Dickie.

AB - Introduction: Alzheimer's disease (AD) is characterized by cerebral glucose hypometabolism. Hypometabolism may be partly due to reduced glucose transport at the blood-brain barrier (BBB) and across astrocytic and neuronal cell membranes. Glucose transporters (GLUTs) are integral membrane proteins responsible for moving glucose from the bloodstream to parenchymal cells where it is metabolized, and evidence indicates vascular and non-vascular GLUTs are altered in AD brains, a process which could starve the brain of glucose and accelerate cognitive decline. Here we review the literature on glucose transport alterations in AD from human and rodent studies. Methods: Literature published between 1st January 1946 and 1st November 2020 within EMBASE and MEDLINE databases was searched for the terms “glucose transporters” AND “Alzheimer's disease”. Human and rodent studies were included while reviews, letters, and in-vitro studies were excluded. Results: Forty-three studies fitting the inclusion criteria were identified, covering human (23 studies) and rodent (20 studies). Post-mortem studies showed consistent reductions in GLUT1 and GLUT3 in the hippocampus and cortex of AD brains, areas of the brain closely associated with AD pathology. Tracer studies in rodent models of AD and human AD also exhibit reduced uptake of glucose and glucose-analogs into the brain, supporting these findings. Longitudinal rodent studies clearly indicate that changes in GLUT1 and GLUT3 only occur after amyloid-β pathology is present, and several studies indicate amyloid-β itself may be responsible for GLUT changes. Furthermore, evidence from human and rodent studies suggest GLUT depletion has severe effects on brain function. A small number of studies show GLUT2 and GLUT12 are increased in AD. Anti-diabetic medications improved glucose transport capacity in AD subjects. Conclusions: GLUT1 and GLUT3 are reduced in hippocampal and cortical regions in patients and rodent models of AD, and may be caused by high levels of amyloid-β in these regions. GLUT3 reductions appear to precede the onset of clinical symptoms. GLUT2 and GLUT12 appear to increase and may have a compensatory role. Repurposing anti-diabetic drugs to modify glucose transport shows promising results in human studies of AD. © Copyright © 2021 Kyrtata, Emsley, Sparasci, Parkes and Dickie.

KW - Alzheimer's disease

KW - blood-brain barrier

KW - glucose transporters

KW - GLUT 1

KW - GLUT 3

KW - amyloid beta protein

KW - glucose transporter

KW - glucose transporter 1

KW - glucose transporter 2

KW - glucose transporter 3

KW - GLUT12 protein

KW - liraglutide

KW - placebo

KW - unclassified drug

KW - Alzheimer disease

KW - blood brain barrier

KW - brain cortex

KW - brain function

KW - brain level

KW - brain metabolism

KW - glucose metabolism

KW - glucose transport

KW - hippocampus

KW - human

KW - insulin resistance

KW - neuropathology

KW - nonhuman

KW - protein depletion

KW - Review

KW - systematic review

U2 - 10.3389/fnins.2021.626636

DO - 10.3389/fnins.2021.626636

M3 - Journal article

VL - 15

JO - Frontiers in Neuroscience

JF - Frontiers in Neuroscience

SN - 1662-453X

M1 - 626636

ER -