Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Brain glucose metabolism, including glycolysis, the pentose phosphate pathway, and glycogen turnover, produces ATP for energetic support and provides the precursors for the synthesis of biological macromolecules. Although glucose metabolism in neurons and astrocytes has been extensively studied, the glucose metabolism of microglia and oligodendrocytes, and their interactions with neurons and astrocytes, remain critical to understand brain function. Brain regions with heterogeneous cell composition and cell-type-specific profiles of glucose metabolism suggest that metabolic networks within the brain are complex. Signal transduction proteins including those in the Wnt, GSK-3β, PI3K-AKT, and AMPK pathways are involved in regulating these networks. Additionally, glycolytic enzymes and metabolites, such as hexokinase 2, acetyl-CoA, and enolase 2, are implicated in the modulation of cellular function, microglial activation, glycation, and acetylation of biomolecules. Given these extensive networks, glucose metabolism dysfunction in the whole brain or specific cell types is strongly associated with neurologic pathology including ischemic brain injury and neurodegenerative disorders. This review characterizes the glucose metabolism networks of the brain based on molecular signaling and cellular and regional interactions, and elucidates glucose metabolism-based mechanisms of neurological diseases and therapeutic approaches that may ameliorate metabolic abnormalities in those diseases.

Free full text 


Logo of nihpaLink to Publisher's site
Prog Neurobiol. Author manuscript; available in PMC 2022 Sep 1.
Published in final edited form as:
PMCID: PMC8380002
NIHMSID: NIHMS1714855
PMID: 34118354

Glucose metabolic crosstalk and regulation in brain function and diseases

Shuai Zhang, PhD,1 Brittany Bolduc Lachance, D.O.,2 Mark P. Mattson, PhD,3 and Xiaofeng Jia, MD, PhD, FCCM1,4,5,6,7

Abstract

Brain glucose metabolism, including glycolysis, the pentose phosphate pathway, and glycogen turnover, produces ATP for energetic support and provides the precursors for the synthesis of biological macromolecules. Although glucose metabolism in neurons and astrocytes has been extensively studied, the glucose metabolism of microglia and oligodendrocytes, and their interactions with neurons and astrocytes, remain critical to understand brain function. Brain regions with heterogeneous cell composition and cell-type-specific profiles of glucose metabolism suggest that metabolic networks within the brain are complex. Signal transduction proteins including those in the Wnt, GSK-3β, PI3K-AKT, and AMPK pathways are involved in regulating these networks. Additionally, glycolytic enzymes and metabolites, such as hexokinase 2, acetyl-CoA, and enolase 2, are implicated in the modulation of cellular function, microglial activation, glycation, and acetylation of biomolecules. Given these extensive networks, glucose metabolism dysfunction in the whole brain or specific cell types is strongly associated with neurologic pathology including ischemic brain injury and neurodegenerative disorders. This review characterizes the glucose metabolism networks of the brain based on molecular signaling and cellular and regional interactions, and elucidates glucose metabolism-based mechanisms of neurological diseases and therapeutic approaches that may ameliorate metabolic abnormalities in those diseases.

Keywords: glucose metabolism, brain, glycolysis, signal regulation, neurological disease

1. Introduction

Glucose is the primary energy source of the brain, which accounts for about 20% of whole-body glucose consumption (Jais et al., 2016; Zhang et al., 2014). Alternate fuel sources supporting the brain include ketone bodies (acetoacetate, and β-hydroxybutyrate), lactate, fatty acids, and amino acids (valine, glutamine, leucine, isoleucine), which are primarily used in settings of limited glucose availability, such as fasting, starvation, and extended exercise (Fig. 1) (Bowman et al., 2019; Camandola and Mattson, 2017; Qi et al., 2019; Schönfeld and Reiser, 2017). Brain glucose metabolism can produce adenosine triphosphate (ATP) to fulfill complex neurological functions, including neuronal signaling transmission (action potentials, synaptic transmission, glutamate cycling, etc.), which consumes 70% of the brain’s energy, and non-signaling activities (resting potentials, axonal transport, mitochondrial proton leak, oligonucleotide turnover, actin cytoskeleton remodeling, etc.), using 30% of ATP produced from brain glucose metabolism (Fig. 1). Moreover, the carbon in the glucose structure can be transmitted to metabolites, such as pyruvate and glyceraldehyde-3-phosphate, which contribute to the synthesis of nucleic acids, fatty acids, and amino acids (Dienel, 2019). Glucose metabolism is also involved in oxidative stress modulation (NADH/NAD+ and NADP+/NADPH) (Fig. 1) (Dienel, 2019; Yu et al., 2018). A growing literature base shows that glucose metabolites and metabolism-related enzymes, such as hexokinase, acetyl-CoA, and glycolytic enzyme glyceradehyde-3-phosphate dehydrogenase, can also directly participate in cellular signaling and functional regulation (Li et al., 2018; Nakajima et al., 2015; Sivanand et al., 2018). For instance, lactate acts as a messenger modulating multiple molecular targets (hydroxycarboxylic acid receptor 1, acid-sensing ion channel, NADH/NAD+ ratio, ATP-sensitive potassium channel, etc.) (Barros, 2013). In addition, the reduced ratio of NADH and NAD+ resulting from glucose metabolism in microglia can suppress the inflammatory cascade (Shen et al., 2017). Therefore, glucose metabolites not only support ATP production and supply the carbon for macromolecule synthesis, but also modulate various functions of neurons and glial cells.

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0001.jpg

Energetic fuels supporting the ATP-dependent and ATP-independent actions in the brain. Glucose is the most important fuel for the brain to fulfill complex neurological functions including neuronal signal transmission (action potential, calcium activities, synaptic transmission, and glutamate cycling), and non-signaling activities (axonal transport, resting potential, and cytoskeleton remodeling). Additionally, glucose metabolism provides the carbon for the synthesis of nucleic acids, fatty acids, and amino acids. Glucose metabolites, such as NADPH, are involved in the regulation of inflammation and redox reaction. Aside from glucose, alternative fuels such as ketone bodies, lactate, fatty acids, and amino acids, are used by the brain, particularly in response to stress, to maintain brain functions.

Glucose entrance into the brain is controlled by a family of specific glucose transporters (GLUTs) via facilitated diffusion, and it has been demonstrated that serum glucose levels can be five-fold higher than levels in the brain (Hwang et al., 2017; Hwang et al., 2019). GLUT distribution in the vascular endothelial cells (ECs) of the blood-brain barrier (GLUT1), neurons (GLUT3,4,6,8), astrocytes (GLUT1,2), and microglia (GLUT 1, 3, 4, 5, 6, 8, 9, 10, 12, and 13) contribute to glucose transport from blood to cells in the brain (Fig. 2) (Wang et al., 2019b; Zhang et al., 2014), and GLUT-mediated glucose control is pivotal for cerebral metabolism.

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0002.jpg

Glucose transporter (GLUT) expression in brain cells and the role in glucose or fluorodeoxyglucose F18 (18F-FDG) transport into cells. (A) Distinct expression of GLUTs in vascular endothelial cells, neurons, astrocytes, and microglia. (B) 18F-FDG is transported into brain tissue by GLUTs and is phosphorylated to 18F-FDG-6-phosphate by HK. FDG is not further metabolized but remains trapped and can be detected in cells for evaluating brain glucose uptake. TCA, tricarboxylic acid; HK, hexokinase; G6P, glucose 6-phosphate.

The hypothalamus and pituitary play important roles maintaining blood glucose homeostasis, with some involvement of the cortex and striatum as well (Cheah and Amiel, 2012; Ter Horst et al., 2018). The glucose-sensing neurons of the brain, parasympathetic (vagus) and sympathetic nerves, and peripheral organs, such as the liver, muscles, intestines, and pancreas, work together to regulate whole-body glucose metabolism (Duncan et al., 2019; Soty et al., 2017; Steinbusch et al., 2015; Tan et al., 2020a). Additionally, insulin receptors are widely expressed in the cerebral cortex, hippocampus, hypothalamus, thalamus, choroid plexus and cerebellum (Fernandez and Torres-Alemán, 2012). Therefore, the brain plays an important role in regulating peripheral glucose metabolism. In turn, systemic metabolic disorders caused by high fat diets can decrease brain glucose uptake by downregulating GLUT1 expression on the vascular endothelial cells. This response can be countered by the compensatory production of vascular endothelial growth factor, which helps to block obesity-associated neurodegeneration (Jais et al., 2016). Ultimately, brain glucose metabolism and systemic glucose metabolism work in concert, and both brain and systemic metabolic disorders can contribute to the development of neurological disease.

Disturbances of glucose metabolism occur in the early stages of neurological disorders, catastrophically in stroke, and more subtly in Alzheimer’s disease (AD) and Parkinson’s disease (PD) (Camandola and Mattson, 2017; Putzu et al., 2018). In the early reperfusion in a rat model of stroke, elevated glucose levels are observed in the infarct areas (Zhang et al., 2016). In the brain of AD patients and AD mouse models, glucose utilization is reduced, particularly in the brain regions most affected by the disease (Camandola and Mattson, 2017; Zhou et al., 2018b). Other evidence suggests that impaired glucose metabolism or disruption of cerebral insulin action contributes to the degeneration of neurons in AD (Camandola and Mattson, 2017; Kleinridders et al., 2014; Yan et al., 2020). Advances in methods for analyzing cell-type-specific features of glucose metabolism indicate that the metabolic alterations of glial cells strongly influence neuronal metabolism and regulate neurological function (Barros et al., 2018b), suggesting that metabolic coupling between neurons and glia is important for normal brain function and that alterations in such coupling may contribute to the pathogenesis of neurodegenerative disorders.

The present review provides an overview of the important technologies for studying brain glucose metabolism and illustrates progress and remaining challenges regarding the role of glucose metabolism in neurological diseases. We highlight the heterogeneity of glucose metabolism among different cell populations in the brain and how such cell type-specific features of glucose metabolism may affect neuronal vulnerability. We then consider the intercellular metabolic networks involved in brain glucose metabolism. Understanding these networks and their involvement in intercellular signaling may reveal novel approaches for preventing and treating neurological disorders.

2. Glucose metabolism in the brain

2.1. Glucose metabolism for ATP generation

Cellular glucose is metabolized in the processes of glycolysis (including lactate production and oxidative phosphorylation), the pentose phosphate pathway (PPP), and glycogenesis (Fig. 3), which are critical to support cell function and energy storage (Mulukutla et al., 2016). Under the presence of adequate oxygen, glucose enters into the oxidative phosphorylation pathway, while 10–15% of glucose is metabolized into carbon dioxide and water via an oxygen-independent process termed aerobic glycolysis (Shannon et al., 2016; Vaishnavi et al., 2010). Aerobic glycolysis is involved in multiple functions including biosynthesis, synaptic plasticity, synapse formation, and neurite growth (Goyal et al., 2014; Shannon et al., 2016).

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0003.jpg

Pathways of glucose metabolism for ATP production and synthesis of biological compounds. The glycolytic (oxygen-independent) and mitochondrial tricarboxylic acid (TCA) cycle (oxygen dependent) pathways contribute to ATP generation for energetic support in the brain. Additionally, glucose-6-P can enter into the pentose phosphate pathway (PPP) for producing NADPH, regulating redox reaction, and providing a source of nucleotides. Glucose can be stored primarily in astrocytes as glycogen in response to stress. Additionally, glucose metabolites 3-phosphoglycerate and pyruvate are implicated in the synthesis of amino acids, directly or indirectly, via the TCA cycle. GA3P contributes to the synthesis of fatty acids and nucleic acids. The H(1–6) and C(1–6) atoms in glucose can be traced to different metabolites. C(1–3) atoms in glucose enter into pyruvate and C(1–5) and access the PPP pathway. H atoms are widely distributed in nucleic acids, fatty acids, and amino acids. HK, hexokinase; GPI, glucose-6-phosphate isomerase; PFK, phosphofructokinase; DHAP, dihydroxyacetone phosphate; GA3P, glyceraldehyde-3-phosphate; G6PDH, glucose-6-phosphate dehydrogenase; 6-PGL, 6-phosphoglucono-d-lactone; PGM, phosphoglucomutase; GS, glycogen synthase; DE, debranching enzyme; GP, glycogen phosphorylase; LDH, lactate dehydrogenase.

2.2. Glucose metabolism in different brain regions

Positron emission tomography (PET) has identified areas of low glucose metabolism (amygdala, hippocampus, enthorinal cortex, parahippocampus, ventral diecncephalon, temporal poles), medium glucose metabolism (cerebellum, thalamus, caudate, nucleus accumbens, globus pallidus), and high glucose metabolism (putamen, remaining cortical regions) in the adult human brain (>18 years) (Tomasi et al., 2017). In resting human brains (20–33 years old), aerobic glycolysis is used at the highest levels in the prefrontal cortices, medial and lateral parietal cortex, and lowest levels in the cerebellum and medial temporal lobes (Vaishnavi et al., 2010). Brain aerobic glycolysis increases in childhood, reaching the peak at 5 years old, and gradually declines to lower levels at a mean age of 21, which persists with aging (Goyal et al., 2014). Loss of brain aerobic glycolysis is related to human aging and Alzheimer’s disease (Goyal et al., 2017; Vlassenko and Raichle, 2015).

In addition to regional differences in the levels of glucose metabolism, it has also been noted that different regions of the brain may rely on different mechanisms of glucose metabolism. In rodents, the PPP is the primary mechanism of glucose metabolism in oligodendrocytes of the thalamus with low ATP/adenosine diphosphate (ADP) ratios, while glycolysis is the primary mechanism in the cortex and hippocampus with high ATP/ADP ratios (Kleinridders et al., 2018). Additionally, fasting promotes lactate production in the cortex and hippocampus, which does not occur in the thalamus, hypothalamus, or amygdala (Kleinridders et al., 2018).

In awake rats, 80% of energy expenditure in white matter is used for non-signaling processes, while 70% of energy expenditure in gray matter is utilized for signaling processes (Yu et al., 2018). A higher activity rate of ATPase in the gray matter, compared to white matter, is conducive to this energetic support (Hyder and Rothman, 2017). Ultimately, different cell types with distinct characteristics of glucose metabolism contribute to metabolic heterogeneity in specific brain regions.

2.3. Glucose metabolism in brain cells

The mammalian brain has a complex anatomical structure, multiple cell types, and a multitude of signaling pathways that regulate neuronal network activity and neuroplasticity. The main types of cells in the brain are neurons, astrocytes, oligodendrocytes, and microglia (Allen and Lyons, 2018). Moreover, there are subpopulations of neurons and glial cells that exhibit distinct morphological, molecular, and functional features (Davie et al., 2018; Mu et al., 2019). Various cell types and their cellular interconnections form intricate neural networks with complex mechanisms of glucose metabolism.

2.3.1. Glucose metabolism in neurons and astrocytes

In the resting, awake rat, excitatory neurons, inhibitory neurons, and glial cells utilize 27%, 22%, and 47% of energy respectively for non-signaling processes, such as housekeeping and resting potentials (Yu et al., 2018). The energetic demands of specific cellular processes are estimated at 50% for synaptic transmission, 10–15% for action potentials, and 9–18% for other signaling-related processes, such as calcium responses and glutamate/GABA recycling in neurons (Yu et al., 2018).

Glucose metabolism in neurons and astrocytes exhibits a compartmentalized pattern, which may be related to different enzymatic actions. Neurons have higher levels of hexokinase expression than astrocytes, therefore glucose is taken up preferentially by neurons (Lundgaard et al., 2015). Astrocytes can come in contact with vasculature via their endfeet, facilitating their glucose uptake. Additionally, glucose-6-phosphatase-β, specifically expressed in the endoplasmic reticulum (ER) of astrocytes, cooperates with glucose-6-phosphate translocase, transferring glucose-6-phosphate from the cytosol to the ER, and allowing for the rapid uptake of glucose by astrocytes and delivery of glucose from astrocytes to neurons (Muller et al., 2018; Pellerin, 2018). Glucose primarily undergoes anaerobic glycolysis in astrocytes due to higher levels of lactate dehydrogenase (LDH) compared to neurons. In contrast, neurons utilize oxidative phosphorylation as primary glucose metabolism (Halim et al., 2010). Additionally, mitochondrial complex I in neurons can conjugate with complex III to form supercomplexes that allow for mitochondrial respiration, while most mitochondrial complex I is free in astrocytes, blocking glucose oxidative phosphorylation (Lopez-Fabuel et al., 2016). The different enzymatic activity of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB) in neurons and astrocytes also contributes to the cellular diversity of glucose metabolism. PFKFB has four isoforms, and isoform 3 (PFKFB3) has the highest enzymatic activity contributing to the generation of fructose-2,6-bisphosphate. Although the gene expression of PFKFB3 in neurons and astrocytes is similar, neurons have lower levels of available PFKFB3 than astrocytes due to rapid neuronal degradation of the enzyme (Bolaños et al., 2010). As a result of this difference, neurons undergo spontaneous apoptosis in the setting of mitochondrial dysfunction due to insufficient ATP production. In contrast, astrocytes can maintain energy production by rapid glycolysis via the upregulation of PFKFB, making them more resistant to mitochondrial dysfunction (Almeida et al., 2001; Bolaños et al., 2010). The PPP in neurons has an antioxidant function due to the production of NADPH(H+), a required factor for reduced glutathione generation (Bolaños et al., 2010). Overexpression of PFKFB3 in neurons promotes oxidative stress and cellular injury, because the increase in glycolysis inhibits the PPP and the production of reduced glutathione (Herrero-Mendez et al., 2009).

In summary: (1) Astrocytes have a higher affinity for glucose uptake and rapid glucose processing than neurons; (2) Astrocytes primarily utilize anaerobic metabolism of glucose and produce lactate; (3) Neurons primarily utilize oxidative phosphorylation of glucose.

2.3.2. Astrocyte-neuron energy interaction

The astrocyte-neuron lactate shuttle (ANLS) hypothesis suggests that astrocytes convert more glucose to lactate after stimulation by glutamate released from neurons (Bélanger et al., 2011). The released lactate then serves as an energy source for the neighboring neurons (Fig. 4) (Bélanger et al., 2011). Genetically encoded fluorescence resonance energy transfer utilizing the lactate sensor Laconic, in combination with in vivo two-photon laser scanning microscopy, identified higher levels of lactate in astrocytes compared to neurons, validating the concept that this lactate gradient is important for a carrier-mediated lactate flux from astrocytes to neurons (Fig. 4) (Machler et al., 2016). However, the ANLS hypothesis is limited in its ability to explain the brain’s response to the metabolic challenge of activation. Under rapid stimulation, mitochondrial metabolism may not be the primary mechanism for neuronal ATP production, due to the number of enzymatic steps required for oxidative phosphorylation (Díaz-García et al., 2017). Rather, in settings of high activity, neurons have increased glucose consumption and produce lactate for energy demand predominantly via glycolysis, which does not depend on external lactate from astrocytes (Díaz-García et al., 2017; Patel et al., 2014).

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0004.jpg

Astrocyte-neuron-oligodendrocyte energy metabolism interactions. Glucose in neurons is primarily used for ATP production and is metabolized into lactate in astrocytes. The glucose transporters (GLUTs) mediate the transportation of glucose into cells. GLUT1, located in cerebral endothelial cells, transits glucose from the blood into the brain tissue. GLUT1 also mediates the entrance of glucose into astrocytes and oligodendrocytes. Glucose is transported into neurons via GLUT3. Monocarboxylate transporters (MCTs) and connexin (Cx) contribute to astrocyte-neuron-oligodendrocyte lactate transport. MCT1 and MCT4 in astrocytes release lactate, which diffuses into neurons due to the lactate gradient and is actively taken up by neurons via the MCT2. Additionally, MCT1 in the myelin cooperates with MCT2 in the axon to translate the lactate from oligodendrocytes to neurons. Therefore, glucose and lactate, together with their transporters, contribute to the astrocyte-neuron-oligodendrocyte energy metabolism interactions. TCA, tricarboxylic acid; PPP, pentose phosphate pathway; LDH, lactate dehydrogenase.

To better understand the role of lactate in the brain, it is important to understand lactate metabolism and transport between neurons and astrocytes. The LDH-1 isoform, primarily expressed in neurons, oxidizes lactate to pyruvate, while the LDH-5 isoform is expressed in astrocytes and promotes the conversion of pyruvate to lactate (Fig. 4). Despite their distinct distribution, LDH-1 and LDH-5 do not directly modulate the lactate flow between neurons and astrocytes (Bak and Schousboe, 2017; Machler et al., 2016). Connexin 43 and 30, the primary astroglial gap-junction proteins, allow for intercellular trafficking of energetic metabolites among astrocytes, blood vessels, and neurons (Rouach et al., 2008). Monocarboxylate transporters (MCTs) are a family of transmembrane proteins (Perez-Escuredo et al., 2016). In the brain, MCT1 is expressed exclusively in the vascular endothelial cells, ependymocytes, oligodendrocytes, and astrocytes. MCT2 is expressed on neuronal membranes (Suzuki et al., 2011). MCT4 receptors are specifically expressed in astrocytes. MCT1, MCT2, and MCT4 contribute to the intracellular flow of lactate, pyruvate, and ketone bodies with a varying affinity (MCT2 > MCT1 >> MCT4) across plasma membranes according to concentration gradients (Perez-Escuredo et al., 2016). Inhibition of either MCT1, MCT2, or MCT4 expression in the hippocampus leads to severely impaired long-term memory formation in rats. Providing exogenous lactate can rescue this memory impairment in MCT1 or MCT4 deficient rats, but not under MCT2 inhibition, which suggests the flow of lactate from glial cells to neurons is a critical function (Suzuki et al., 2011).

In summary, the astrocytes near blood vessels can obtain glucose and convert it to lactate, which can then be shuttled into neurons for energy production.

2.3.3. Glycogen metabolism in astrocytes

Glycogen granules are distributed in the cell bodies of astrocytes, where they are concentrated near adjacent axons and dendritic spines (Cali et al., 2016). In brain regions with high neural activities, astrocytes have increased glycogen stores. By using ESG1A9 and IV58B6 to label glycogen in the mouse brain, distinct brain regions show different glycogen storage, with high levels in the hippocampus, striatum, the outer layer of the cortex, the cerebellar molecular layer, and the thalamic reticular nuclei, whereas glycogen levels are relatively low in the hypothalamus and most regions of the thalamus (Oe et al., 2016).

It has been proposed that glycogen in astrocytes provides a critical source of glucose during periods of food deprivation and stress (Dienel, 2019). Glycogen phosphorylase is the rate-limiting enzyme in glycogen metabolism (Mathieu et al., 2016). Lactate derived from astrocytic glycogen can be transported into neurons via MCT2 and can support the maintenance of ATP production in the brain (Matsui et al., 2017).

Glycogen metabolism may also play roles in synaptic plasticity, learning, and memory (Alberini et al., 2018; Cali et al., 2016; Hackett et al., 2016). Astrocytic glycogen mobilization is important for long-term memory formation, and blocking glycogen metabolism leads to memory impairment (Suzuki et al., 2011). Hippocampal astrocyte to neuron lactate transport is mediated by MCT1 and MCT4. Glycogen breakdown is regulated by 3’,5’-cyclic adenosine monophosphate (cAMP) and Ca2+, and supports astrocytic potassium and glutamate uptake and glutamine synthesis. Disruption of glycogen metabolism in astrocytes may occur in epilepsy, AD, and type 2 diabetes (Bak et al., 2018). Glycogen-derived energy is required for the elimination of glutamate and potassium ions from the synaptic gap. The blockage of glycogen metabolism increases the levels of synaptic extracellular potassium and glutamate, which can lead to hyperexcitability and can stimulate migraine headaches (Kilic et al., 2018). In contrast, excessive accumulation of non-mobilizable glycogen can cause Lafora disease (Nitschke et al., 2018).

2.3.4. Oligodendroglia

Four types of oligodendroglia have been identified based on morphology and are primarily distributed in the cerebral white matter (Perez-Cerda et al., 2015). Single-cell analysis has further revealed divergent subpopulations of oligodendrocytes in the juvenile and adult mouse brain (Marques et al., 2016; Zeisel et al., 2015). Oligodendroglia are specialized glial cells that wrap axons with a multilamellar lipid structure called myelin and play pivotal roles in the functional maintenance and survival of neurons. In addition to their function in accelerating action potential propagation along the axon, oligodendrocytes are essential for energetic support to neurons (Philips and Rothstein, 2017). Oligodendrocytes have both a high rate of mitochondrial glucose metabolism and lactate production. These cells also have high PPP activities and use approximately 10–15% of glucose in the PPP compared with glycolysis (Amaral et al., 2016a; Amaral et al., 2016b). The energy metabolites produced by oligodendrocytes may contribute to lipid synthesis, trophic support to axons, and myelin protection (Amaral et al., 2016b).

Oligodendrocytes transfer energy materials, such as pyruvate and lactate, to neurons through noncompacted regions of myelin and MCTs, which facilitate the fast delivery of energy metabolites to neurons for ATP synthesis (Fig. 4) (Philips and Rothstein, 2017). MCT1 in oligodendrocytes and MCT2 in neurons work together to transport lactate from oligodendrocytes to neurons, and downregulation of MCT1 results in axonal injury and neuronal loss (Fig. 4) (Lee et al., 2012; Morrison et al., 2013). In the corpus callosum, neuron function is evaluated by detecting compound action potentials, and the absence of glucose abolishes these compound action potentials. Thus, MCT1 and GLUT1 are vital for providing the lactate and glucose derived from oligodendrocytes for axonal survival (Fig. 4) (Meyer et al., 2018).

In addition to these transporters, gap junctions comprised of connexins (Cx) are also important for the intercellular delivery of ions, regulatory molecules, and metabolites, including glucose and lactate (Philips and Rothstein, 2017). Cx30 and Cx43 are located in astrocytes, and Cx32 and Cx47 are in oligodendrocytes. The coupling of Cx47-Cx43 and Cx32-Cx30 form channels for ionic and chemical delivery between oligodendroglia and astrocytes, respectively (Fasciani et al., 2018; Orthmann-Murphy et al., 2007). Some evidence suggests that gap junctions, including Cx30, Cx43, Cx32, and Cx47, contribute to energy metabolite transport from astrocytes to oligodendroglia (Morrison et al., 2013; Nave and Werner, 2014). The metabolic interactions between neurons and oligodendrocytes serve to maintain neuronal function.

2.3.5. Microglia

Microglia account for 10–15% of cells in the brain where they play important roles in innate immune responses and neuroplasticity. Microglial densities are heterogeneous across brain regions, with the highest densities in the cortex, striatum, corpus callosum, hippocampus, SVZ, and rostral migratory stream (RMS), intermediate densities in the olfactory bulb, thalamus, hypothalamus, midbrain and brainstem, and the lowest density in the cerebellum (Fig. 5) (Stratoulias et al., 2019; Tan et al., 2020b; Yang et al., 2013). The distinct subpopulations of microglia, defined by varying gene expressions, have unique functional characteristics (Stratoulias et al., 2019). Most microglia express ionized calcium-binding adaptor molecule 1 (Iba1) and CD11b, although some Iba1/Chemokine (CCR) and fractalkine (CX3CR1+) microglia are observed in the SVZ and the adjacent RMS, which terminates in the olfactory bulb. These Iba1/CX3CR1+ microglia are involved in promoting neurogenesis (Fig. 5) (Stratoulias et al., 2019). Microglia in the resting state is characterized by a ramified morphology. Upon stimulation, activated microglia change their morphology, with amplified soma and retraction of ramified processes (Yang et al., 2013). The activated microglia have dynamic phenotypes, including the pro- or anti-inflammatory activities at different stages of disease, and it remains difficult to define the status of individual microglia (Colonna and Butovsky, 2017).

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0005.jpg

Microglia in the brain and glucose metabolism. (A, B) The distinct cellular structure, function, and density of microglia across different mouse brain areas. CX3CR1 positive microglia, which have less ramification and are primarily distributed in the olfactory bulb, RMS, and SVZ, support neurogenesis. In contrast, Iba1 and CD11b positive microglia, which have obvious ramification, are widely distributed in the whole brain and account for the clearance and neurotransmission. Microglial density gradually increases from the cerebellum posteriorly, forward to the cortex. (C) Glucose in resting microglia primarily enters into mitochondrial oxidative phosphorylation. In contrast, activated microglia primarily use glucose for lactate production. RMS: rostral migratory stream. SVZ: subventricular zone.

Glucose is the main energy source for microglial survival and function. Oxygen-glucose deprivation can lead to microglial death (Eyo et al., 2013; Kacimi et al., 2011). Multiple glucose transporters, including GLUT1, 3, 4, 5, 6, 8, 9, 10, 12, 13, and high hexokinase 2 (HK2) expression offer sufficient glucose influx for glial cells (Kalsbeek et al., 2016; Wang et al., 2019b). GLUT1 has the highest expression in microglia and predominately contributes to glucose uptake, particularly under inflammatory conditions (Wang et al., 2019b). Microglia also use glutamine as an energy substrate, with high expression of glutamine receptors SLC1A5 and SLC38A1 (Liu et al., 2018).

Microglial energy metabolism is dependent upon the degree of microglial reactivity. Quiescent microglia primarily rely on oxidative phosphorylation of glucose for energy supply, while stimulated microglia transition from oxidative phosphorylation to glycolysis (Fig. 5) (Baik et al., 2019). Despite less ATP generation in glycolysis than mitochondrial respiration, the rate of lactate production is faster than that of oxidative phosphorylation, which enables a rapid supply for energy-intensive processes such as proliferation, migration, and secretion (Cheng et al., 2014; Peek et al., 2017). AMP-activated protein kinase (AMPK), a sensor for AMP and ADP, inhibits mTOR phosphorylation and subsequently suppresses the expression of HIF-1α, the master transcriptional regulator of glycolysis (Cheng et al., 2014). The inhibition of the mTOR pathway dramatically reduces the inflammatory responses of microglia. Thus, cellular metabolic pathways (mTOR-HIF-1α pathway) can alter the functional phenotypes of microglia (Baik et al., 2019).

2.3.6. Cerebrovascular endothelial cells (CECs)

ECs have proliferative and migratory abilities after activation by a growth factor, such as vascular endothelial growth factor (VEGF). In recent years, EC metabolism and metabolic relationships with other cell types have played a pivotal role in health and diseases (Eelen et al., 2018). Glycolysis, rather than oxidative phosphorylation, is the major glucose metabolism pathway for ATP production in these cells, which is beneficial for oxygen transfer to surrounding cells (Eelen et al., 2018). Compared with ECs in other tissues, capillary ECs in the brain have more mitochondrial contents (Oldendorf et al., 1977). When facing shear stress, oxidative phosphorylation in CECs becomes the major metabolic pathway, due to the increased expression of enzymes in the Krebs cycle and downregulated glycolytic genes (Cucullo et al., 2011). A recent study about single-cell transcriptome atlas of murine ECs characterizes the ECs from multiple organs, and CECs have more expression of transporters of glucose (Slc2a1), amino acids (Slc3a2, Slc7a5), and fatty acids than ECs from other tissues (Kalucka et al., 2020). The systems of tight junctions and material transportation in CECs are critical for maintaining the blood-brain barrier function, and glucose metabolism in CECs is involved in their modulation (Kalucka et al., 2020; Zhang et al., 2014). Low GLUT1 levels block angiogenesis and diminish the brain microvasculature network (Tang et al., 2017). Patients with GLUT1 deficiency syndrome have low brain glucose levels and cerebral disorders with seizures, developmental delay, and motor disturbance (Wang et al., 2005). The phosphatase and tensin homolog/protein kinase B (AKT) pathway in CECs increases monocarboxylic acid transporter 1 expression to promote lactate transport across the CECs, which is vital for maintaining lactate homeostasis contributing to hippocampal neurogenesis and cognitive function (Wang et al., 2019a).

2.4. Signal modulation of glucose metabolism

Multiple signal molecules are involved in regulating brain glucose metabolism (Fig. 6). Wnt3a-mediated Wnt signaling activation enhances glucose metabolism by increasing HK activity and elevating glucose uptake and glycolytic rates in neurons, which is linked with the activation of the AKT and AMP-activated protein kinase (AMPK) pathways (Cisternas et al., 2016; Cisternas et al., 2019). Wnt3a ligand binding to the Frizzled receptor also inhibits glycogen synthase kinase-3β (GSK-3β) activity, and thereby activates AMPK, enhancing energetic effects (acetyl-coenzyme A carboxylase, 6-phos-phofructo-2-kinase) and mediating autophagy in hippocampal neurons (Mulukutla et al., 2016; Ríos et al., 2018). Additionally, Wnt signaling activation via Wnt5a stimulates nitric oxide-mediated glucose metabolism in neurons. The canonical and non-canonical Wnt signals are all involved in the modulation of neuronal glucose metabolism (Cisternas and Inestrosa, 2017).

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0006.jpg

Signaling pathways that modulate cerebral glucose metabolism. Frizzled receptors located in the cellular membrane can conjugate with Wnt ligands to activate the GSK-3β/AMPK/GLUTs pathway or GSK-3β/AMPK/TSC/mTOR pathway, which regulate glucose uptake and metabolism. Moreover, insulin can activate the PI3K/AKT pathway to modulate glucose metabolism. LRPs assist both frizzled receptors and insulin receptors to bind to their ligands. LRP, lipoprotein receptor-related protein; NO, nitric oxide; GSK-3β, glycogen synthase kinase; AMPK, AMP-activated protein kinase; GLUT, glucose transporter; TSC, Tuberous sclerosis; Rheb, Ras homolog enriched in the brain; mTOR, mammalian target of rapamycin; Hif-1α, hypoxia-inducible factor 1α.

Peripheral insulin can enter the brain through the choroid plexus and cerebrospinal fluid, or directly via vascular endothelial cells (Duarte et al., 2018). Moreover, the adult brain may also produce insulin via the gene Insulin 2, which exists in brain cells such as hippocampal neurons (Mehran et al., 2012). Insulin is found in the hippocampus, cortex, anterior olfactory nucleus, cerebellar Purkinje neurons, amygdala, and other discrete nuclei, and insulin receptors are expressed in neurons and glial cells, indicating the role of insulin signaling in the regulation of brain function (Kleinridders et al., 2014; Mehran et al., 2012; Soto et al., 2019). Insulin signaling in the hippocampus is important for maintaining spatial learning and memory, partly via the GLUT4-mediated glucose uptake (Pearson-Leary et al., 2018; Soto et al., 2019). Insulin signaling in the hypothalamic proopiomelanocortin astrocytes is involved in controlling cerebral glucose uptake (García-Cáceres et al., 2016). Insulin binds to receptors in the brain and primarily modulates two signaling cascades, including the phosphoinositide 3-kinase (PI3K) pathway controlling metabolism and the mitogen-activated protein kinase pathway, regulating mitochondrial function, proliferation, and growth (Kullmann et al., 2020). Enhanced insulin signaling can activate the PI3K-AKT pathway and stimulate glucose uptake by elevating the levels of GLUT3 and GLUT4 in the membrane (Gabbouj et al., 2019; Liu et al., 2015). In the mouse hippocampus, AKT1 and AKT3 are expressed in neurons, while AKT2 is primarily expressed in astrocytes, which may be implicated in insulin-dependent glucose metabolic regulation in neurons, microglia, and astrocytes (Gabbouj et al., 2019; Levenga et al., 2017).

2.5. Genetic factors influencing brain glucose metabolism

Genetic influences on brain glucose metabolism are vital for affecting brain function and neurological diseases. By comparing the brain glucose metabolism differences in elderly twins, it was discovered that the glucose metabolism in some brain regions, specifically the right and left parietal lobes and left temporal lobe, is affected more by genetics than by environment (Watanabe et al., 2016). The genes modulating the expression of glucose metabolism enzymes and glucose transporters are the primary genetic factors affecting brain glucose metabolism. These key genes include pyruvate dehydrogenase kinase 3 and pyruvate dehydrogenase phosphatase catalytic subunit 1, which modulate glycolytic pathways, and GLUT1 deficiency syndrome, which causes a neurodevelopmental disorder (Goyal et al., 2014; Tang et al., 2019). Genome-wide association studies have further discovered many genes involved in regulating neuronal development and brain glucose metabolism, such as potassium ion channels, synaptic transmission and plasticity, neuropeptides and their receptors, and RNA-binding proteins, such as Fox-1 homolog 1. (Casanovas et al., 2020; Goyal et al., 2014; Kong et al., 2018). In addition, the Dopamine D4 receptor gene with the 7-repeat (7R) allele may promote age-independent brain glucose metabolism in the cerebellum, inferior temporal cortex, and striatum, suggesting that carriers of the 7R allele have a stronger ability to resist age-induced brain glucose metabolism decline (Volkow et al., 2013). Better understandings of the genetic factors affecting brain glucose metabolism are needed and may be beneficial for diagnosis and targeted treatment of brain diseases.

2.6. Glucose metabolism for non-energetic processes

2.6.1. Glucose metabolites for synthesis of biological compounds

In addition to carbon dioxide and ATP, glucose as the carbon and hydrogen carrier (C1–6) is involved in the synthesis of amino acids (C1–3 and H1, 2/6 for glutamine, alanine, glycine, and serine), lipid acids (C1,2 and H1,2,3/4,5,6), nucleotides (C1–5 and H2,4,5,6), NAPDH (H1/3), NADH (H4), glycogen, neurotransmitters, and neuromodulators (acetylcholine, glutamate, γ-aminobutyric acid, aspartate, and acetylcholine), glycolipids, and glycoproteins (Fig. 3) (Camandola and Mattson, 2017; Dienel, 2019), which has been verified by the tracking of carbon or hydrogen atoms transferred from glucose to other materials (Lane and Fan, 2015; Zhang et al., 2019a). These products further support the cellular function and synthesis of biological macromolecules. Impaired glucose oxidation inhibits the production of glutamate, a key neurotransmitter, and causes altered neuronal network activity, highlighting the interaction of glucose metabolism and neuronal excitability (Jakkamsetti et al., 2019).

2.6.2. Glycation

Glycation is a spontaneous, non-enzymatic reaction involving the attachment of sugar, such as glucose or glucose metabolites, to a protein or lipid and resulting in advanced glycation end-products (AGEs). Nε-(carboxymethyl)lysine, pyrraline, Nε-(carboxyethyl)lysine, methylglyoxal-lysine dimer, and glyoxal-lysine dimer are examples of frequently occurring AGEs (Kaur et al., 2016). The glycating agent methylglyoxal (MGO) is generated from the decomposition of the glyceraldehyde 3-phosphate (GA3P) and dihydroxiacetone phosphate (DHAP) derived from glycolytic metabolites, and is catabolized by the glutathione (GSH)-dependent glyoxalase and NADPH-mediated aldose reductase systems (Fig. 7) (Vicente Miranda et al., 2016). MGO can react with the cysteine, arginine, and lysine of proteins and can change the structural conformation, impairing protein function (Polykretis et al., 2020). AGEs and the irreversible glycation process have been implicated in stroke and neurodegenerative diseases such as PD and AD (Ng et al., 2019; Sharma et al., 2019; Vicente Miranda et al., 2016; Vicente Miranda et al., 2017).

An external file that holds a picture, illustration, etc.
Object name is nihms-1714855-f0007.jpg

Glucose metabolism-mediated signal regulation in neurological disease. Disease-related gene mutation, aging, or environmental toxins can cause dysfunction of glucose metabolism in the brain, which leads to neuronal injury by activating inflammation or promoting oxidative stress and the accumulation of glycated toxic proteins (such as α-synuclein, Aβ), impairing neuronal function. Aβ, amyloid beta; GLUT, glucose transporter; PPP, pentose phosphate pathway; DHAP, dihydroxyacetone phosphate; GA3P, glyceraldehyde-3-phosphate; GSH, glutathione.

A recent study demonstrated that MGO can glycate superoxide dismutase 1 and increase cellular oxidative stress (Polykretis et al., 2020), thus highlighting the important fact that glycation is not only involved in the aggregation of disease-related toxic proteins, but also influences cellular functions, such as antioxidative capacity. Aside from MGO, other glycating agents may play unknown roles in brain function and diseases. Aging and diabetes are currently considered the primary factors contributing to glycation and AGE generation, and studies suggest that enhancing the glyoxalase and aldose reductase systems and manipulating levels of NADPH and GSH are potential strategies for suppressing glycation-mediated neurotoxicity.

2.6.3. Non-metabolic functions of glycolytic enzymes and metabolites

In addition to maintaining energetic homeostasis, glucose metabolism molecules, such as intermediates and metabolic enzymes, also exhibit non-metabolic roles in regulating cellular activities, including the modification of proteins and DNA. Dysfunction in these non-metabolic processes has been implicated in disease processes such as malignancy and immune responses (Ghosh-Choudhary et al., 2020; Snaebjornsson and Schulze, 2018). For example, the pyruvate kinase isoform M2 activates signal transducer and activator of transcription 3 and focal adhesion kinase signals and plays a crucial role in decreasing stroke injury by enhancing neurogenesis (Chen et al., 2018a).

HK2, a key glycolytic enzyme, plays a role in microglial-mediated neuroinflammation. In hypoxic conditions, HK2 is up-regulated, leading to the accumulation of acetyl coenzyme A (acetyl-CoA) and promoting transcription and expression of interleukin-1β. Blocking HK2 is protective against ischemic neurological injury (Li et al., 2018). In addition, activation of AKT was shown to facilitate the binding of HK2 to the mitochondrial membrane, maintaining structural and functional integrity of mitochondria and reducing apoptosis (Li et al., 2019).

Acetyl-CoA is implicated in histone acetylation and gene modification (Sivanand et al., 2018). Acetyl-CoA synthetase 2, which generates acetyl-CoA, can act on chromatin and directly modulate histone acetylation in hippocampal neurons, which has been shown to subsequently regulate memory-related neuronal genes and affect long-term spatial memory in mice. This study demonstrated a connection between glucose metabolism, gene modulation, and neural function (Mews et al., 2017). Inhibition of acetyl-CoA carboxylase 1 can increase the levels of acetyl-CoA in neurons and cause acetylation of histone H3K9, enhancing memory in the aging brain (Currais et al., 2019) and making acetyl-CoA and its metabolic enzymes an interesting target involving molecular acetylation, memory, and aging.

Glycolytic enzymes have also been implicated in neurotransmission. Glutamate neurotransmission in astrocytes can be modulated by the binding of serine racemase to the glycolytic enzyme glyceradehyde-3-phosphate dehydrogenase (GAPDH), which decreases the amount of available D-serine for N-Methyl-D-aspartic acid (NMDA) receptor activation (Suzuki et al., 2015). GAPDH can also translocate into the nucleus and interact with poly (ADP-ribose) polymerase-1, causing overactivation in settings of oxidative stress and exacerbating stroke injury (Nakajima et al., 2015). GAPDH nuclear translocation, aggregation, and binding to toxic proteins, such as β-amyloid peptides, tau protein, and α-synuclein, have been linked to neuronal death in neurodegenerative diseases such as AD and PD (Gerszon and Rodacka, 2018). Further, nitric oxide-mediated GADPH nitrosylation has been shown to promote Rheb degradation and decreases mTOR activation. Inhibition of this GAPDH nitrosylation exerted antidepressant effects (Harraz et al., 2016).

Enolase 2 (ENO2), also known as γ-enolase or neuron-specific enolase, is a glycolytic enzyme located in neuronal cytosol and synaptic plasma membranes. Blocking this enzyme with an anti-ENO2 antibody was shown to promote the production of hydrogen peroxide and cause brain cell death via a non-apoptotic pathway (Yamamoto et al., 2015), suggesting a neuroprotective effect of ENO2. Additionally, the role of ENO2 in the PI3K/AKT and MAPK/ERK pathways has been shown to activate inflammation, influencing cell survival, and has been shown to have neurotrophic functions, such as neurite regeneration (Hafner et al., 2012; Haque et al., 2018; Haque et al., 2016).

Understanding the non-metabolic functions of glucose metabolites and enzymes is critical to fully comprehend the networks between glucose metabolism and neuronal function and may allow for targeted modulation of metabolism, neurotransmission, cellular survival, and neuronal functions.

3. Methods for evaluating brain glucose metabolism

3.1. Positron emission tomography (PET)

Fluorodeoxyglucose F18 (18F-FDG) is transported into brain tissue by GLUTs and is phosphorylated 18F-FDG-6-phosphate by HK. Because FDG is not further metabolized, it is then trapped in cells and can be measured for evaluating brain glucose uptake (Fig. 2B) (Izuishi et al., 2014). Thus, FDG-PET directly reflects cerebral glucose uptake, but not glucose metabolism. Several parameters impacting FDG transport between blood and brain may lead to changes in glucose uptake under pathological conditions (Backes et al., 2011; Walberer et al., 2012; Wang et al., 2020).

Advancements in PET imaging have led to the development of new PET probes, which can be combined for studying the correlation of glucose metabolism and other factors in disease, such as (18F) 2-tert-butyl-4-chloro-5–2H-pyridazin-3-one (mitochondrial labeling) and FDG-PET, 123Ioflupane brain single-photon emission computed tomography (dopamine transporter) and FDG-PET, and fluorine-18-l-dihydroxyphenylalanine-PET (reflecting the dopamine distribution) and FDG-PET (Huber et al., 2020; Ruppert et al., 2020; Terada et al., 2020).

3.2. Magnetic resonance spectroscopy (MRS)

Radioactive isotopes to track metabolism are widely used for studying metabolic pathways and rates of transport within the brain, including oxygen consumption (17O, 13C, 1H), glucose consumption (13C, 1H, 19F), creatine kinase/ATPase flux (31P), neurotransmitter flux (13C, 1H), and pH (1H) (Hwang et al., 2019; Hyder and Rothman, 2017). The application of specific radioisotopes can trace the fate of single or multiple atoms of the glucose structure and metabolites. For example, glycogen can be detected with the specific C1 glycogen peak after the movement of the 13C from glucose to glycogen (DiNuzzo, 2013). Intravenous administration of 13C-labelled nutrients indicates the turnover flux of lactate is the highest of all metabolites and contributes to the Krebs cycle intermediates (Hui et al., 2017). Recently, 1H-MRS has been demonstrated as a powerful tool monitor the metabolism of 13C-enriched substrates in brain (Dehghani et al., 2020).

3.3. Microdialysis

Microdialysis is an invasive measure of glucose metabolism from directly within the brain. A cannula is implanted into the target region of the brain, and a microdialysis probe is then inserted through the cannula to collect samples of metabolic substrates from the interstitial fluid among brain cells via perfusion of artificial cerebrospinal fluid, allowing for the detection of real-time changes in small molecules in the brain interstitium (Le Douce et al., 2020). The collected samples are separated by high-performance liquid chromatography and determined with mass spectrometry, biochemical reactions, immunoassays (radioimmunoassay and enzyme linked immunosorbent assay), microchip electrophoresis, and microfabricated fluidic systems (Davies et al., 2000; Sandlin et al., 2005; Saylor and Lunte, 2018). The enzyme-based microelectrodes can be used for real-time measurements of cerebral interstitial fluid (Barros et al., 2018a), but this technology has low spatial and temporal resolution and may cause local brain disruption. Currently, microdialysis is widely used to detect the levels of glucose, lactate, pyruvate, and insulin in the brain to investigate the role of cerebral metabolism in brain function and disease. Some studies indicate that lactate in the hippocampal interstitial fluid supports memory function (Harris et al., 2016). Conversely, increased glucose or lactate levels in the hippocampal interstitial fluid were related with elevated amyloid-beta or tau levels, which may contribute to the formation of amyloid plaques and neurofibrillary tangles (Harris et al., 2016; Holth et al., 2019; Macauley et al., 2015; Roh et al., 2012; Stanley et al., 2016). Larger molecules, such as proteins, cytokines, neuropeptides, and monoclonal antibodies, can also be analyzed via microdialysis (Jadhav et al., 2016; Perkins et al., 2021).

High-resolution electrochemical biosensors may be used as an alternative method to detect glucose, lactate, and glutamate levels within the brain (Naylor et al., 2012), making microdialysis a promising and valuable technique for metabolomics studies (Hammarlund-Udenaes, 2017). Limitations of microdialysis include localized tissue injury caused by the inserted probes (200–500 μm in external diameter), difficulty with precise targeting of small target nuclei, localized sampling that may not represent the brain as a whole, and levels of intracellular metabolites below the limits of detection. Utilizing specialized perfusion fluid, such as dexamethasone, an anti-inflammatory drug, may decrease local lesion burden without marked effects on dopamine terminals, tyrosine hydroxylase, or the dopamine transporter after probe implantation in the striatum of rats (Jaquins-Gerstl and Michael, 2020; Nesbitt et al., 2015).

3.4. Imaging methods for metabolic detection

3.4.1. Spectroscopic imaging

Fourier transform infrared (FTIR) spectroscopy can detect biochemical materials, including nucleic acids, proteins, lipids, and carbohydrates within cells, tissues, and even extracellular vesicles. The unique spectrum of the materials is based on the wavelength and quantity of infrared radiation (Su and Lee, 2020). FTIR imaging can simultaneously detect the changes of lipid and protein contents, such as Aβ protein and Lewy bodies, in the brain (Ali et al., 2018; Araki et al., 2015). The glycogen and lactate in the frozen slice of the brain can be detected by FTIR spectroscopic imaging based on the intensity at 1152 and 1125 cm−1 respectively (Hackett et al., 2016). This approach can be used to study spatiotemporal alterations of metabolites in the brain.

Deuterium-labeled glucose given to cells or animals can be used for tracking the carbon–deuterium bonds in macromolecules with multichannel stimulated Raman scattering microscopy. The specific Raman peaks of carbon-deuterium bonds can distinguish various types of glucose-derived macromolecules (Shen et al., 2019b). This microscopy technique can elucidate the metabolic fate of glucose in diverging biosynthesis pathways, which may be superior to other glucose-imaging tools, such as PET and fluorescence, which are primarily used for visualizing the earlier processes of glucose metabolism (Zhang et al., 2019a).

3.4.2. Autoradiography

Quantitative autoradiography can achieve a high degree of spatial localization of target materials via the radioactive element, which is useful in evaluating cerebral blood flow, glucose metabolism, and protein changes (Saré et al., 2019; Schmidt and Smith, 2005). 2-Deoxy-D-glucose (2DG) is an analogue of glucose and is used for the measurement of the rates of glucose metabolism in vivo. After intravenous injection of [14C]-2DG in animals, brain sections were exposed to autoradiographic film for several days. The autoradiographic images provided a quantitative metabolic map of the brain (Le Douce et al., 2020; Lundgaard et al., 2015). 18F-FDG can also be used for autoradiographic images to assess the glucose uptake levels in different brain areas (Putzu et al., 2018). Therefore, autoradiography presents a method for measuring regional rates of metabolites and understanding glucose metabolism-based brain actions and diseases.

3.4.3. Mass spectrometry imaging

Mass spectrometry imaging can provide more information about glucose metabolic fate than PET imaging (Kleinridders et al., 2018). Mass spectrometry imaging enables imaging of the spatial distribution of multiple molecules such as metabolites, glycans, peptides, and lipids without labeling. This is achieved by detecting the mass-to-charge value of the ionized molecules on the surface of the sample with a mass spectrometer and mapping a construction of the values distributed on the sample’s surface using computational software (Buchberger et al., 2018; Luan et al., 2019). Matrix-assisted laser desorption/ionization, desorption electrospray ionization, and nanostructure-initiator techniques are used for the qualitative analysis of metabolites (Luan et al., 2019). This chemical analysis tool can visualize the spatial distributions of glucose metabolites and characterize region-specific metabolism in the brain. It is also a powerful tool for researching the metabolic crosstalk between the various brain regions. However, without the chromatographic assistant, mass spectrometry imaging is limited in its analytical depth. The use of a laser capture microdissection system coupled with liquid chromatography mass spectrometry (LC-MS) can enhance the analysis of regions of interest for further molecular identification (Dewez et al., 2020; Dilillo et al., 2017).

3.4.4. Biological imaging

Metabolic imaging based on the histochemistry of metabolic enzymes, such as glucose-6-phosphate dehydrogenase (G6PD), LDH, and GAPDH, is a powerful tool for investigating metabolic configurations, specifically allowing for the metabolic characterization of individual cells within the tissues (Miller et al., 2017). According to the catalytic activity of metabolic enzymes at key nodes of glucose metabolism (Fig. 3), other biological functions of these enzymes, such as the redox balance and cell signaling events, can be determined via the enzyme-dependent metabolic imaging method. NADH autofluorescence has a short lifetime of ~400 ps by two-photon excitation. Upon enzymatic binding to NADH, this fluorescence lifetime extends to 2000 ps. Thus, the lifetime of NADH can be used to evaluate the ratio of glycolysis to mitochondrial oxidative phosphorylation (Bernier et al., 2020). Biosensors are sensitive and selective detectors that can be used for observing and quantifying specific biomolecules. Förster resonance energy transfer-based biosensors are exploited for the detection of DNA, miRNA, proteins, and enzymes (Wu et al., 2020; Zhang et al., 2019b). A C1-type d-glucose-conjugated fluorescent probe has competitive uptake capacity for glucose and is employed for glucose uptake imaging (Cheng et al., 2020b). Some genetically encoded metabolite sensors, including the iGlucoSnFR-TS (glucose sensor), Frex family (NADH sensors), LigA-cpVenus (NAD+ sensor), Peredox, RexYFP, SoNar (NAD+/NADH ratio sensors), iNap family (NADPH sensors), and Apollo-NADP+ (NADP+ sensor), are used for the detection of cerebral glucose metabolism (Díaz-García et al., 2019; Zhang et al., 2018b). These metabolic enzymes and biosensor-enabled imaging methods can advance the understanding of the features and functions of glucose metabolism in the brain.

3.5. Metabolomics assays

Metabolomics can provide systematic information on metabolites and their crosstalk in brain function and disease (Luan et al., 2019). Hundreds of metabolites in brain tissue or cerebrospinal fluid can be detected with nuclear magnetic resonance (NMR), gas chromatography-mass spectrometry (GC-MS), LC-MS, ion mobility systems, and capillary electrophoresis (An et al., 2018; Wishart, 2019). GC-MS can detect volatile and non-polar compounds, as well as some polar metabolites (neurotransmitters, polyunsaturated fatty acids, amines, and amino acids), while polar chemicals, non-volatile, or thermo-unstable compounds can be determined by LC-MS (Luan et al., 2019). Compared with MS, NMR is less sensitive to chemicals, but retains chemical integrity without chromatographic separation or ionization and identifies compounds with hydrogen chemical shift fingerprints (Wishart, 2019). Additionally, mass spectrometry imaging techniques can be used to select areas of interest in tissue samples for metabolomics analysis, which provides the spatial distributions of metabolites (Johnson et al., 2016). Metabolomics is suitable for understanding the chemical composition of the brain and unveiling metabolic signals (Gonzalez-Riano et al., 2016). With further development of MS sensitivity, coverage, speed, and single-cell technique, the high-throughput of metabolomics enables large scale metabolome profiling, real-time determination, and spatial resolution (single cell, organelle, and intracellular and extracellular metabolites) (Zampieri et al., 2017). Challenges still exist in the validation of metabolites, biological interpretation of novel metabolites, and comprehensive informatics for metabolomic analyses (Cui et al., 2018; Johnson et al., 2016). Currently, many metabolomics libraries (such as AMDIS, MASCOT, XCMS) and metabolite identification databases serve for data analysis (Kusonmano et al., 2016). The Human Metabolome Database (www.hmdb.ca) and MetaboAnalyst (http://metaboanalyst.ca) are updated and provide comprehensive information about the biological function of metabolites, metabolic pathways and their integration with other omics data (Chong et al., 2018; Wishart et al., 2018). New informatics strategies, such as the metabolic reaction network-based recursive algorithm, are major drivers in overcoming some of the challenges presented with metabolomic analysis (Domingo-Almenara et al., 2019; Shen et al., 2019a). The integration of metabolomics, together with the other omics, including genomics, transcriptomics, and proteomics, will be promising for gaining new insights into the mechanisms underlying neurological disorders and finding novel methods for diagnosis and evaluation of therapeutic efficacy in disease.

3.6. Metabolic endpoint measurements

Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) are important parameters reflecting glycolysis levels. Normally, cells have low ECAR and high OCR due to the advantage of oxidative phosphorylation. The addition of glucose, as a new substrate, causes an elevation of ECAR and decrease of OCR, reflecting a shift away from oxidative phosphorylation and toward glycolysis. Mitochondrial inhibition with oligomycin (ATP synthase inhibitor) and rotenone (mitochondrial electron transport chain blocker by inhibiting complex I) increases ECAR and decreases OCR (Klinge, 2020; Mookerjee et al., 2017). Mitochondrial phosphate/oxygen ratios indicate the generated ATP per oxygen atom consumption, and suggest the oxidation of pyruvate plus malate (2.73), oxidation of succinate (1.64), and complete oxidation of glucose (2.79) (Hinkle, 2005; Mookerjee et al., 2017). Many materials, such as ATP, ADP, oxygen, multiple ions, and metabolic substrates can be quantified and subsequently employed to evaluate the metabolic status, though it remains challenging to precisely understand complex metabolic networks and modulation with the current metabolic parameters.

4. Glucose metabolism in aging brain

Aging is a common risk factor for many neurological diseases. Multiple changes in glucose metabolism-related factors, such as glucose transport, mitochondrial function, DNA repair, and neurotrophic factors, contribute to aging in the brain (Camandola and Mattson, 2017). Aging neurons primarily rely on glucose oxidation via glycolysis, rather than astrocytic lactate support, as an energy source, and the enzymes of glycogen metabolism increase with aging in both astrocytes and neurons (Drulis-Fajdasz et al., 2018). However, the ability of neurons to use ketone bodies as an energy source may be maintained even when glucose availability is reduced (Courchesne-Loyer et al., 2017). Brain glucose uptake declines with normal aging in a greater proportion to oxygen consumption, which leads to a reduction in aerobic glycolysis that is most obvious in brain areas that utilize more aerobic glycolysis in youth (Goyal et al., 2017). Electron microscopy has demonstrated that myelinated fibers in the aging mouse brain have longer and thicker mitochondria, which was associated with lower ATP levels and increased production of reactive oxygen species (ROS) (Stahon et al., 2016). Aging-related metabolic alterations in the fly Drosophila melanogaster brain were recently characterized by single-cell transcriptome sequencing, which revealed changes in cell composition (a relative increase of glia), more prominent aging in glia versus neurons, decreased mitochondrial turnover, a decline in oxidative phosphorylation levels, and unique changes of ribosomal gene expression (Davie et al., 2018). Ultimately, dysfunction of glucose metabolism related to aging may be vital for the occurrence and progression of neurological diseases.

5. Glucose metabolism in neurologic diseases

Glucose metabolism plays a central role in maintaining brain cell viability and function and is also involved in the progress of multiple neurological diseases (Table 1). Chemical manipulation of the glucose metabolism pathway is promising for the development of new drugs to treat these diseases. Chemicals regulating glucose transportation or metabolism (Table 2) may become tools for experimental research and have the potential for clinical application.

Table 1

Altered brain glucose metabolism in neurological diseases

DiseasesBrain glucose metabolismFDG uptake in PETReferences
Focal ischemic strokeInfarct area: hypometabolismInfarct area: reduction, Peri-ischemic region: elevation(Walberer et al., 2012; Yuan et al., 2013)
CA-induced global brain ischemiaCortex: hypometabolism HK I and II levels reductionWhole brain reduction, particularly in the occipital cortex, putamen, and white matter(Hahn et al., 2014; Li et al., 2015; Putzu et al., 2018)
Alzheimer’s diseaseHypometabolism in injured brain regions
Reduction of glycolysis and PPP; Cortex: glucose transport reduction with decreased levels of GLUT1 and GLUT3; Microglia: oxidative phosphorylation decrease, aerobic glycolysis increase
Reduction in bilateral temporal lobes, precuneus, parahippocampal gyrus, posterior cingulate gyrus, parietal lobe, temporoparietal and posterior cingulate regions(Baik et al., 2019; Butterfield and Halliwell, 2019; Dave et al., 2020; Demarest et al., 2020; Gordon et al., 2018; Shivamurthy et al., 2015; Szablewski, 2017; Winkler et al., 2015)
Dementia with Lewy bodiesHypometabolism in injured brain regionsReduction in occipital cortex, precuneus, temporal and parietal cerebral cortexes, parietotemporal, posterior cingulate, and frontal association cortices(Dave et al., 2020; Ishii, 2014; Shivamurthy et al., 2015)
Frontotemporal dementiaHypometabolism in injured brain regionsReduction in the frontal lobe, anterior temporal lobe, anterior cingulate gyrus, ventromedial frontopolar cortex, bilateral prefrontal areas, and subcortical structures, including the basal ganglia (putamen and globus pallidus) and medial thalamic regions(Dave et al., 2020; Ishii, 2014; Shivamurthy et al., 2015)
Dementia vascular dementiaHypometabolism in injured brain regionsReduction in focal cortical and subcortical, deep gray nuclei, cerebellum, primary cortexes, middle temporal gyrus, and anterior cingulate cortex(Shivamurthy et al., 2015)
Parkinson’s diseaseHypometabolism
Reduction of pyruvate kinase 2 activity; Glycolysis impairment; Mitochondrial metabolic impairment
Reduction in parietal cortex, putamen, caudate nucleus and inferior parietal lobule(Bogetofte et al., 2019; Firbank et al., 2017; Knight et al., 2014; Liu et al., 2016; Meles et al., 2017; Ruppert et al., 2020)
Huntington’s diseaseHypometabolism and hypermetabolism
Mitochondrial complex II inhibition
Creatine kinase reduction
GLUT3 expression reduction
Reduction in striatum
Elevation in inferior parietal and temporal lobules, dentate nucleus, anterior cingulate, and motor regions of the cerebellum
(Gaura et al., 2017; López-Mora et al., 2016) (Duan et al., 2014; Solís-Maldonado et al., 2018)

FDG: fluorodeoxyglucose; PET: positron emission tomography; CA: cardiac arrest; HK: hexokinase; PPP: pentose phosphate pathway; GLUT: glucose transporters

Table 2

Chemicals regulating cerebral glucose metabolism

ChemicalsFunctionReferences
Cytochalasin BGLUT1-4 inhibitor(Díaz-García et al., 2017; Kapoor et al., 2016)
Stf31GLUT1 inhibitor(Wang et al., 2019b)
PhloretinGLUT-1 inhibitor(Kilic et al., 2018)
OxamateLDH inhibitor(Abraham et al., 2018)
CarbenoxoloneConnexin 30 and connexin 43 gap junction inhibitor(Yin et al., 2018)
α-cyano-4-hydroxy-cinnamateMCT1,2,4 inhibition, mitochondrial pyruvate carrier inhibition(Matsui et al., 2017; Perez-Escuredo et al., 2016)
AR-C155858MCT1 and MCT2 inhibition(Le Douce et al., 2020; Perez-Escuredo et al., 2016)
4,4’-dibenzamidostilbene-2,2’-disulphonateMCT2 inhibitor(Perez-Escuredo et al., 2016)
7-(N-benzyl-N-methylamino)-2-oxo-2H-chromene-3-carboxylic acidMCT1 inhibitor (inhibiting lactate uptake)(Perez-Escuredo et al., 2016)
5-nitro-2-(3-phenylpropylamino)-benzoateMCT1 and MCT4 inhibitor(Perez-Escuredo et al., 2016)
AZD3965MCT1 and MCT2 inhibitor(Perez-Escuredo et al., 2016)
1,4-dideoxy-1,4-imino-D-arabinitolGlycogen phosphorylation inhibitor (inhibiting glycogen synthesis)(Kilic et al., 2018; Matsui et al., 2017)
IsofagomineGlycogen phosphorylase inhibitor(Choi et al., 2019)
GintoninActivating lysophosphatidic acid receptors (promoting glycogen synthesis)(Choi et al., 2019)
6-amino nicotinamideGlucose-6-phosphate dehydrogenase inhibitor (PPP inhibition)(Tu et al., 2019)
DehydroepiandrosteroneGlucose-6-phosphate dehydrogenase inhibitor (PPP inhibition)(Tu et al., 2019)
AP-III-a4Enolase inhibitor(Jung et al., 2013)

GLUT, glucose transporter; MCT, monocarboxylate transporter; PPP, pentose phosphate pathway.

5.1. Ischemic brain injury

5.1.1. Glucose metabolism changes in ischemic brain

Synaptic dysfunction is the earliest consequence of cerebral ischemia and may occur within seconds, resulting from aberrant transmitter release at the synapse, dysfunction of ATP-dependent sodium and calcium ion-motive ATPases, and calcium ion accumulation within synaptic terminals (Hofmeijer and van Putten, 2012). In focal ischemic stroke, neurons in the ischemic core die rapidly by necrosis, and more distant neurons may undergo delayed death by apoptosis. Thus, brain areas rich with neurons and high in metabolic activity suffer most from ischemic injury (Schaafsma et al., 2003). Cellular glucose deprivation is an important pathological mechanism of ischemic brain lesions.

After cardiac arrest (CA) exposure, brain glucose metabolism decreased in the cortex at 2, 4, 24, 48 h, and even 5 weeks after the return of spontaneous circulation (ROSC), and HK I and II levels reduced in injured brain areas (Hahn et al., 2014; Li et al., 2015; Putzu et al., 2018), suggesting the decreased capacity of brain glucose metabolism in injured neural cells. PET scan at 3h post-ROSC indicated that a high forebrain-to-hindbrain ratio of 18F-FDG uptake predicted good neurological outcomes after CA (Kim et al., 2019). In a rat model of stroke, FDG uptake is reduced in the infarct area, while it is elevated in the peri-ischemic region 2h after reperfusion (Walberer et al., 2012; Yuan et al., 2013). The cells in the peri-ischemic region may use aerobic glycolysis to support energy needs in hypoxic conditions (Yuan et al., 2013).

Multiple factors, such as cerebral blood flow and oxygen availability, can influence brain glucose metabolism. After CA, the blood flow and oxygen metabolism in the whole brain are reduced during the first week after ROSC, most prominently in the occipital cortex, putamen, and white matter (Hahn et al., 2014). Oxygen remains critical for brain glucose metabolism in the glycolytic pathway, and it has been demonstrated that reperfusion with hyperoxia (pressure of arterial oxygen ≥ 300 mmHg) after CA is associated with poor functional outcomes (Richards et al., 2007; Roberts et al., 2018). Additionally, hypothermia exerts neuroprotection in ischemic brain injury (He et al., 2019; Zhen et al., 2013) by preserving the ATP levels and decreasing the glucose metabolism flux into lactate (Erecinska et al., 2003).

Our understanding of the relationship between brain glucose uptake and metabolism changes with post-ischemia or post-stroke outcomes remains limited, and these changes and outcomes are further influenced by complex factors such as age, brain region, and time from injury.

5.1.2. Glucose metabolism in glial cells after brain ischemia

Microglia have a stronger capacity for energy production via non-oxidative glucose metabolism compared to neurons and astrocytes, improving their survival in peri-infarct areas (Backes et al., 2016). However, hyperglycolysis within the penumbra after brain ischemia has been shown to exacerbate brain injury by activating microglia and promoting ROS production. Inhibiting HK2, a key glycolytic enzyme, can suppress microglial activation and decrease ischemic brain injury (Li et al., 2018). It has also been shown that fractalkine receptor chemokine (C-X3-C motif) ligand 1 (CX3CL1) signaling in microglia, which modulates metabolism by increasing oxidative phosphorylation and reducing glycolysis after CA, supports the expression of anti-inflammatory genes and results in reduced brain injury (Lauro et al., 2019). In the early recovery after oxygen-glucose deprivation exposure, the increased glucose mobilization via the PPP and tricarboxylic acid cycle in surviving astrocytes contributes to energetic support and recovery (Amaral et al., 2010). Brain ischemia also triggers MCT1 expression in oligodendrocytes, particularly in the peri-infarct striatum, which may allow for lactate transportation from astrocytes to produce sufficient energy (Zhou et al., 2018a).

5.1.3. CECs in brain ischemia

CECs are highly associated with stroke injury and treatment. Targeting tight junctions or transporters in CECs is promising for regulating endothelial homeostasis and function in stroke (Abdullahi et al., 2018). Decreasing the loss of tight junction proteins in stroke can alleviate brain lesion size via inhibition of autophagy (Kim et al., 2020; Zhang et al., 2018a). miR-34a can reduce the mitochondrial oxidative phosphorylation, ATP production, and cytochrome c levels in CECs, which contribute to the destruction of the blood-brain barrier (Bukeirat et al., 2016). Knockout of miR-34a can markedly decrease the permeability of the blood-brain barrier and improve stroke outcomes (Hu et al., 2020). Targeting some miRNAs, such as miR-34a, miR-15a miR-26b, and miR-212/132, has the potential to reduce ischemic brain injury associated with stroke by modulating CECs (Burek et al., 2019; Ren et al., 2019; Shen and Ma, 2020). miR-126 derived from the exosomes of CECs has neurorestorative effects in stroke mice (Venkat et al., 2019). Transcriptional profiles of CECs in stroke provide more information for understanding the role of CECs in ischemia and may suggest some novel targets for modulating CECs for stroke treatment (Munji et al., 2019).

5.1.4. Glucose metabolism modulation for decreasing ischemic brain injury

Glucose metabolism regulation is a promising strategy in defense against ischemic injury. NADPH, a metabolic byproduct of PPP, has antioxidant abilities and reduces intracellular oxidative stress. Intravenous administration of NAD+ and NADPH has demonstrated neuroprotective effects in ischemic stroke (Huang et al., 2018; Li et al., 2016). Increasing the PPP flux by ischemic preconditioning, enhancing G6PD activity, increasing TP53-induced glycolysis and apoptosis regulator (TIGAR) expression, PFKFB3 inhibition, or suppressing oxygen-sensing prolyl hydroxylase domain proteins can decrease neuronal apoptosis and protect against ischemic brain injury (Burmistrova et al., 2019; Cao et al., 2017; Li et al., 2014; Quaegebeur et al., 2016). However, NADPH also mediates the ROS generation caused by NADPH oxidase (NOX). Thus, the cooperation of NOX inhibition and NADPH may better alleviate ischemic brain damage (Qin et al., 2017).

The glutathione glycosylase system (GSH) is responsible for MGO elimination. Inhibition of the GSH results in MGO accumulation, leading to increased glycation, including occludin glycation, which was shown to increase blood-brain barrier permeability, increase oxidative stress, and exacerbate the post-stroke injury (Wang et al., 2016b). It has been demonstrated that N-acetylcysteine (NAC) can decrease glycation-mediated stroke risk by enhancing the GSH-dependent MGO elimination (Wang et al., 2018). Activation of glutamate oxaloacetate transaminase by protein kinase C epsilon or isoflavone reduces ischemic stroke lesions by metabolizing neurotoxic glutamate and promoting the tricarboxylic acid cycle (Khanna et al., 2017; Rink et al., 2017; Xu et al., 2020). Resveratrol, a known activator of sirtuin 1, stimulates the glycolytic rate and enhances ischemic tolerance and neuroprotection in the ischemic penumbra (Koronowski et al., 2017). The inhibition of the abnormal astrocytic connexin-43 opening with carbenoxolone (Table 2) has neuroprotective effects in cerebral ischemia/reperfusion injury as well (Yin et al., 2018). TIGAR-mediated metabolic modulation in astrocytes suppresses the inflammatory injury to neurons after ischemia/reperfusion (Chen et al., 2018b). Targeting the cerebral glucose metabolism is promising for neuroprotection in ischemic brain injury.

5.2. Alzheimer’s disease

5.2.1. Hypometabolism in AD brain

In AD patients with cognitive decline, the accumulated amyloid-β (Aβ) peptide and hyperphosphorylated tau proteins in the brain contribute to dysfunction and loss of synapses and neuronal death. FDG-PET studies have consistently demonstrated reduced cerebral glucose utilization in AD which are correlated with Aβ and tau pathologies. Aβ aggregates induce oxidative stress, which impairs the enzymes involved in glucose metabolism and decreases glucose utilization for ATP biosynthesis (Butterfield and Halliwell, 2019; Gordon et al., 2018). In the brain of female AD mice, glucose-6-phosphate is accumulated and thereby inhibits hexokinase activity in glycolysis and the PPP. Cortical synaptic mitochondrial complex I function is reduced, causing glucose hypometabolism, energy failure, and neuronal death (Demarest et al., 2020).

Disturbance of glucose transport occurs in AD, with decreased levels of GLUT1 and GLUT3, particularly in the cerebral cortex (Szablewski, 2017; Winkler et al., 2015). Low glycolytic rates in the AD brain lead to higher glucose levels, which can down-regulate GLUT3 (An et al., 2018). In a Drosophila model of adult-onset AD, genetic overexpression of glucose transporters in neurons or stimulation of glucose uptake by metformin increased survival of the flys (Niccoli et al., 2016). These findings suggest that dysfunction of glycolytic enzymes and glucose transport contribute to hypometabolism in regions of Aβ aggregation and may be linked to phenotype severity.

5.2.2. Dysfunction of glucose metabolism in AD

It has been proposed that multiple pathogenic factors, including genetic mutation, aging, and environmental toxins, impair glucose metabolism and contribute to neuronal degeneration in AD patients (Fig. 7). This hypothesis has been supported with evidence such as altered thiamine metabolism and cerebral insulin resistance in AD patients (Chen and Zhong, 2013). Inhibition of insulin-mediated brain PI3K/AKT pathway may block tau dephosphorylation and exacerbate neuronal degeneration (Duarte et al., 2018; Kleinridders et al., 2014). The ε4 allele of apolipoprotein E (APOE4) is a predominant genetic risk factor for AD, which affects brain insulin signaling and glucose and amyloid metabolism by inducing deficits in glycolysis, glucose uptake, and mitochondrial respiration (Emrani et al., 2020; Keeney et al., 2015; Wu et al., 2018). In AD patients with APOE4, more tau accumulation and brain atrophy are observed in the medial temporal lobe, causing memory impairment (Emrani et al., 2020). Dysfunction of glucose metabolism has been observed in the early phase of AD. Embryonic neurons derived from the 3xTg-AD mouse hippocampus have significant mitochondrial respiration reduction (Yao et al., 2009). Glucose uptake in the amygdala, entorhinal cortex, and hippocampus is decreased in the early stage of 3xTg-AD female mice (6- to 7-month old) without senile plaque formation, and this has been linked to impaired synthesis of L-serine and D-serine. L-serine is produced from the glycolytic intermediate 3-phosphoglycerate and is the precursor of D-serine, which is involved in regulating synaptic plasticity (Le Douce et al., 2020).

Mitochondrial dysfunction, reflected by reduced [18F]BCPP-EF uptake, has been found in the parahippocampus and may precede glycolysis-related hypometabolism in the early stages of AD (Terada et al., 2020). Cerebral β-amyloidosis leads to an increase in Ca2+ levels in synapses and mitochondria and has been linked to plaque deposition and neuronal death (Calvo-Rodriguez et al., 2020). Pentatricopeptide repeat-containing protein 1 (PTCD1) in mitochondria is required for ATP production via oxidative phosphorylation. Gene PTCD1 variant is found in AD patients and disrupts neuronal energy homeostasis, suggesting that the AD-related gene alteration may cause dysfunction of glucose metabolism and further contribute to impaired synaptic plasticity and behavioral deficits in the early phases of AD (Fleck et al., 2019).

Metabolic intermediate deficiencies have also been associated with cognitive impairment in AD. O-linked N-acetylglucosamine (O-GlcNAc) is produced via the glucose metabolism pathway, and aging has shown a decline of this metabolite in the hippocampus of mice ages 18–22 months (Wheatley et al., 2019). O-GlcNAc deficiency was shown to promote the production of hyperphosphorylated tau and amyloidogenic Aβ-peptides, resulting in memory impairment in mice (Pinho et al., 2018; Wang et al., 2016a). Increasing O-GlcNAc levels in the hippocampus can enhance fear memory development in young adult mice and rescue the impairments of spatial learning and memory in aging mice (Wheatley et al., 2019). Increased lactate levels in the hippocampal interstitial fluid are related to memory decline in AD mice (Harris et al., 2016). Additionally, the elevation of plasma, but not cerebrospinal fluid, lactate levels are highly associated with Aβ aggregation (Kavanagh et al., 2019). More studies are required to clarify whether disturbances of specific steps in glucose metabolism are directly related to etiologies and the natural history of AD in the absence of this gene mutation.

5.2.3. Dysfunction of microglial glucose metabolism in AD

Dysfunction of glucose metabolism in microglia may occur in AD. The metabolism of microglia is shifted from oxidative phosphorylation to aerobic glycolysis during Aβ-induced inflammatory activation, boosting microglial glycolysis, increasing interferon-γ and Aβ phagocytosis (Baik et al., 2019). Rutin, a natural flavonoid in many foods, can promote the microglial metabolic transition from anaerobic glycolysis to oxidative phosphorylation, enhancing ATP production and Aβ phagocytosis (Pan et al., 2019). Thus, maintaining oxidative phosphorylation in microglia has the potential for improving brain injury in AD. A variant of the microglial surface receptor triggering receptor expressed on myeloid cells 2 (TREM2), has been identified in AD patients and is linked to disruption of energy metabolism through mTOR signaling (Ulland et al., 2017), further suggesting that abnormal metabolism in microglia occurs in AD and modulation of microglial metabolism may be a potential therapeutic strategy.

5.2.4. Targeting impaired glucose metabolism for AD therapy

Peripheral metabolism disorders, such as glucose intolerance, have been correlated with AD progression (Griffith et al., 2019). Insulin resistance is a risk factor for age-related cognitive impairment and AD. Accordingly, interventions that enhance insulin sensitivity such as regular exercise and intermittent fasting may reduce the risk of AD (de Cabo and Mattson, 2019; Mattson, 2012). Drugs that enhance insulin sensitivity also have the potential to be beneficial for patients with mild cognitive impairment and AD, although this remains to be established. Administration of metformin reverses Aβ-induced metabolic defects and decreases protein aggregation in neurons (Teo et al., 2019). Another approach is to circumvent an impairment in neuronal glucose utilization by either modifying food intake so as to include regular periods in a ketogenic state with either intermittent fasting or ketogenic diets (Cunnane et al., 2020). Alternatively, supplementing the diet with a ketone ester has proven effective in lessening Aβ and tau pathologies and improving cognitive function in mouse models of AD (Cheng et al., 2020a; Kashiwaya et al., 2013). Mitochondrial metabolic remodeling has emerged as an area of focus for potential AD diagnosis and treatment (Peng et al., 2020; Teo et al., 2019). Increasing glucose uptake via chemicals, such as metformin, has been shown to improve AD symptoms (Niccoli et al., 2016). Activating Wnt signaling by andrographolide or lithium ions can enhance glucose metabolism in cortical and hippocampal neurons and delay the neuropathological progression in AD mice (Fig. 6) (Cisternas et al., 2019). L-serine and cilostazol have been shown to improve cognitive function in AD patients and mice by regulating glucose metabolism and enhancing glucose utilization (Le Douce et al., 2020; Lee et al., 2019). A deeper understanding of the links between glucose metabolism and AD remains critical for advancements in diagnosis, prevention, and therapies.

5.3. Parkinson’s disease

5.3.1. Hypometabolism in PD brain

Intraneuronal accumulation of the misfolded protein a-synuclein is a classic feature of PD pathology. The dysfunction and death of dopaminergic neurons in the substantia nigra pars compacta inhibit dopaminergic input to the striatum, causing motor dysfunction with bradykinesia, rigidity, and resting tremor, and nonmotor symptoms, including cognitive, sleep, mood, and olfactory dysfunction (Kalia and Lang, 2015). In newly diagnosed PD patients, reduced FDG in the parietal cortex is associated with cognitive decline (Firbank et al., 2017). Hypometabolism in the caudate nucleus and inferior parietal lobule has been associated with PD (Meles et al., 2017). Dopamine depletion in the putamen is followed by reduced 18F-FDG uptake in PD patients (Ruppert et al., 2020). Ultimately, hypometabolism in multiple brain regions occurs as the disease progresses.

5.3.2. Mutant genes in PD impair mitochondria metabolism

Gene mutations of PARK6 (PINK1), PARK2 (parkin), and PARK7 (DJ-1), are linked to familial PD and the proteins encoded by these genes are each important for mitochondrial homeostasis. Parkin can regulate glucose metabolism via the ubiquitination of pyruvate kinase 2, decreasing enzymatic activity (Liu et al., 2016). Knockout of the PARK2 gene impairs glycolysis in neurons (Bogetofte et al., 2019). These links suggest that mitochondrial metabolic impairment caused by these gene mutations may mediate neuronal degeneration in PD (Knight et al., 2014).

5.3.3. Modulation of glucose metabolism for PD treatment

A recent clinical trial showed that the glucagon-like 1 peptide analog exenatide improves motor function in patients with PD (Athauda et al., 2017). Exenatide is a drug used to treat patients with diabetes. It improves insulin sensitivity and also has direct neuroprotective effects (Li et al., 2009). Enhancing neuronal glycolysis is another potential therapeutic approach for PD. Glucose phosphate isomerase 1, an initial enzyme in glycolysis, mediates genetic modification of α-synuclein misfolding by modulating the DAF-16/FOXO pathway, resulting in neuroprotection (Knight et al., 2014). This study suggests a key role of energy-metabolism-associated genes in modifying α-synuclein-induced neurodegenerative processes. In postmortem human PD brains, down-regulation of PPP enzymes and reduced NADPH are found in the putamen and cerebellum (Dunn et al., 2014). Upregulation of G6PD, a rate-limiting enzyme of the PPP, in microglia promotes lipopolysaccharide-induced dopaminergic neurodegeneration by amplifying the inflammatory response, which suggests that targeting G6PD may be promising for PD interventions (Tu et al., 2019). The activation of NADPH oxidase, a superoxide-producing enzyme, causes dopaminergic neurodegeneration and impairments of learning, memory, and motor functions in PD, which can be blocked by apocynin, resulting in neuroprotection via suppression of oxidative stress and neuroinflammation (Hou et al., 2019). Enhancing phosphoglycerate kinase 1 activity with terazosin promotes glycolysis and ATP generation, which can slow neuronal loss and PD progression (Cai et al., 2019). Modulating brain metabolism is an important strategy for PD therapy.

5.4. Huntington’s disease

5.4.1. Hypometabolism in striatum

Huntington’s disease (HD) is a dominantly inherited neurodegenerative disease involving the loss of striatal neurons and progressive chorea, dementia, and psychological disturbance. The Huntingtin gene mutation, with a CAG repeat expansion, is predominantly expressed in the medium spiny neurons of the striatum (Ross et al., 2017). Hypometabolism in the striatum is related to disease severity in patients with early symptomatic HD (Gaura et al., 2017; López-Mora et al., 2016). In contrast, hypermetabolism in some brain regions (inferior parietal and temporal lobules, dentate nucleus, anterior cingulate, and motor regions of the cerebellum) is found to be relevant to more serious hyperkinetic symptoms (Gaura et al., 2017). In preclinical HD gene carriers, elevated glucose metabolism in the thalamus may be a compensatory mechanism for early lesions in the striatum (Feigin et al., 2007). Early metabolic changes in HD may cause later neurodegenerative progress and pose interesting targets for early therapies.

5.4.2. Astrocyte-derived metabolic abnormalities in HD

The early pathological cascade of HD is characterized by a failure in brain energy metabolism. Mutant huntingtin protein inhibits mitochondrial complex II function, disrupts Ca2+ buffering capacity in mitochondria, increases oxidative stress, impairs mitochondrial bioenergetics, trafficking, and dynamics, and reduces creatine kinase (Duan et al., 2014). Decreased GLUT3 expression in neurons causes energy decline in HD mice (Solís-Maldonado et al., 2018). While mutant huntingtin protein does not directly alter glucose metabolism in neurons, it does result in abnormal neuron-astrocyte signaling (Boussicault et al., 2014; Hamilton et al., 2015). Coculture of neurons with astrocytes with mutant huntingtin increases oxidative stress in neurons and reduces neuronal glucose uptake, suggesting the role of neuron-astrocyte signaling transduction in the early metabolic changes of the HD brain (Boussicault et al., 2014). In the striatum, astrocytes account for about 80–90% of cells. The low glucose environment in the brain of HD model animals forces astrocytes in the striatum to use endogenous fatty acids for energy support. Metabolism of fatty acids requires more oxygen than glycolysis and produces greater ROS, which causes initial impairment of neurons in the striatum (Polyzos et al., 2019). Reducing mutant huntington protein in astrocytes slows HD progression in the mouse HD model (Wood et al., 2019). This evidence suggests that astrocyte-derived metabolic abnormalities may be a direct factor causing neuronal toxicity in the early stage of HD.

5.4.3. Modulation of glucose metabolism for HD treatment

Reduced GLUT3 level is observed in in vitro and mouse HD models (Covarrubias-Pinto et al., 2015). Increasing GLUT3 expression and supplementing ascorbic acid stimulated neuronal lactate uptake and improved metabolic imbalance in an HD model (Solís-Maldonado et al., 2018). Manganese ion supplementation promotes glucose uptake in HD-model cells by enhancing insulin/IGF receptor-induced AKT signal (Bryan et al., 2020). Increasing glucose uptake and utilization is an important strategy for HD treatment.

6. Concluding remarks

The brain is composed of various specialized cell populations with distinct metabolic profiles, which contribute to region-specific metabolic features. Brain region-specific impairments of glucose metabolism have been characterized in ischemic brain injury, AD, PD, and HD. Ultimately, the development of a precise cell atlas of the brain and corresponding metabolic mapping will be critical for understanding the metabolism-based physiology and pathology of brain function.

Evidence has indicated the features of glucose metabolism related to neuron–glial interactions are vital for understanding neuropathological processes (Afridi et al., 2020; Boussicault et al., 2014; Morken et al., 2014). Therefore, more investigations may further elucidate the interactions among various types of cells in the brain and potential modulation for therapies in neurological disorders.

Cerebral glucose metabolism can be modulated via multiple signal pathways. The molecules in glucose metabolic pathways are involved in non-energetic functions, such as the antioxidant capacity of metabolites derived from the PPP and metabolism-based microglial activation. The role of glucose metabolism in determining cellular fate includes energetic and non-energetic regulation of cerebral function and neurological disease.

Thus, glucose metabolic crosstalk among different regions, cells, and signal molecules in the brain is central to cerebral function, and disruption of these metabolic networks is involved in neurological disease.

Highlights

  • Specific features of brain glucose metabolism (GM) are identified in different brain regions and cell types.

  • Intercellular GM crosstalk together with signal molecules contribute to the energetic and non-energetic function in brain.

  • Brain GM dysfunction is associated with neurologic pathology including ischemic brain injury and neurodegenerative disorders.

  • Targeting GM is promising for treating neurological diseases.

Acknowledgments:

The work was supported by R01HL118084 and R01NS110387 from the National Institutes of Health (NIH) (both to X Jia). XJ was partially supported by NIH RO1NS117102 (to X Jia).

Footnotes

Conflicts of Interests: The authors declare no conflict of interests. The founding sponsors had no role in the writing of this review.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • Abdullahi W, Tripathi D, Ronaldson PT, 2018. Blood-brain barrier dysfunction in ischemic stroke: targeting tight junctions and transporters for vascular protection. Am J Physiol Cell Physiol 315, C343–c356. [Europe PMC free article] [Abstract] [Google Scholar]
  • Abraham MA, Rasti M, Bauer PV, Lam TKT, 2018. Leptin enhances hypothalamic lactate dehydrogenase A (LDHA)-dependent glucose sensing to lower glucose production in high-fat-fed rats. The Journal of biological chemistry 293, 4159–4166. [Europe PMC free article] [Abstract] [Google Scholar]
  • Afridi R, Kim JH, Rahman MH, Suk K, 2020. Metabolic Regulation of Glial Phenotypes: Implications in Neuron-Glia Interactions and Neurological Disorders. Frontiers in cellular neuroscience 14, 20. [Europe PMC free article] [Abstract] [Google Scholar]
  • Alberini CM, Cruz E, Descalzi G, Bessières B, Gao V, 2018. Astrocyte glycogen and lactate: New insights into learning and memory mechanisms. Glia 66, 1244–1262. [Europe PMC free article] [Abstract] [Google Scholar]
  • Allen NJ, Lyons DA, 2018. Glia as architects of central nervous system formation and function. Science 362, 181–185. [Europe PMC free article] [Abstract] [Google Scholar]
  • Almeida A, Almeida J, Bolaños JP, Moncada S, 2001. Different responses of astrocytes and neurons to nitric oxide: the role of glycolytically generated ATP in astrocyte protection. Proc Natl Acad Sci U S A 98, 15294–15299. [Europe PMC free article] [Abstract] [Google Scholar]
  • Amaral AI, Hadera MG, Tavares JM, Kotter MR, Sonnewald U, 2016a. Characterization of glucose-related metabolic pathways in differentiated rat oligodendrocyte lineage cells. Glia 64, 21–34. [Europe PMC free article] [Abstract] [Google Scholar]
  • Amaral AI, Tavares JM, Sonnewald U, Kotter MR, 2016b. Oligodendrocytes: Development, Physiology and Glucose Metabolism. Advances in neurobiology 13, 275–294. [Abstract] [Google Scholar]
  • Amaral AI, Teixeira AP, Martens S, Bernal V, Sousa MF, Alves PM, 2010. Metabolic alterations induced by ischemia in primary cultures of astrocytes: merging 13C NMR spectroscopy and metabolic flux analysis. Journal of neurochemistry 113, 735–748. [Abstract] [Google Scholar]
  • An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, Chia CW, Egan JM, Ferrucci L, Troncoso J, Levey AI, Lah J, Seyfried NT, Legido-Quigley C, O’Brien R, Thambisetty M, 2018. Evidence for brain glucose dysregulation in Alzheimer’s disease. Alzheimers Dement 14, 318–329. [Europe PMC free article] [Abstract] [Google Scholar]
  • Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, Hibbert S, Budnik N, Zampedri L, Dickson J, Li Y, Aviles-Olmos I, Warner TT, Limousin P, Lees AJ, Greig NH, Tebbs S, Foltynie T, 2017. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet (London, England) 390, 1664–1675. [Europe PMC free article] [Abstract] [Google Scholar]
  • Backes H, Walberer M, Endepols H, Neumaier B, Graf R, Wienhard K, Mies G, 2011. Whiskers area as extracerebral reference tissue for quantification of rat brain metabolism using (18)F-FDG PET: application to focal cerebral ischemia. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 52, 1252–1260. [Abstract] [Google Scholar]
  • Backes H, Walberer M, Ladwig A, Rueger MA, Neumaier B, Endepols H, Hoehn M, Fink GR, Schroeter M, Graf R, 2016. Glucose consumption of inflammatory cells masks metabolic deficits in the brain. NeuroImage 128, 54–62. [Europe PMC free article] [Abstract] [Google Scholar]
  • Baik SH, Kang S, Lee W, Choi H, Chung S, Kim JI, Mook-Jung I, 2019. A Breakdown in Metabolic Reprogramming Causes Microglia Dysfunction in Alzheimer’s Disease. Cell Metab 30, 493–507.e496. [Abstract] [Google Scholar]
  • Bak LK, Schousboe A, 2017. Misconceptions regarding basic thermodynamics and enzyme kinetics have led to erroneous conclusions regarding the metabolic importance of lactate dehydrogenase isoenzyme expression. Journal of neuroscience research 95, 2098–2102. [Abstract] [Google Scholar]
  • Bak LK, Walls AB, Schousboe A, Waagepetersen HS, 2018. Astrocytic glycogen metabolism in the healthy and diseased brain. The Journal of biological chemistry 293, 7108–7116. [Europe PMC free article] [Abstract] [Google Scholar]
  • Barros LF, 2013. Metabolic signaling by lactate in the brain. Trends in neurosciences 36, 396–404. [Abstract] [Google Scholar]
  • Barros LF, Bolaños JP, Bonvento G, Bouzier-Sore AK, Brown A, Hirrlinger J, Kasparov S, Kirchhoff F, Murphy AN, Pellerin L, Robinson MB, Weber B, 2018a. Current technical approaches to brain energy metabolism. Glia 66, 1138–1159. [Europe PMC free article] [Abstract] [Google Scholar]
  • Barros LF, Brown A, Swanson RA, 2018b. Glia in brain energy metabolism: A perspective. Glia 66, 1134–1137. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bélanger M, Allaman I, Magistretti PJ, 2011. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metab 14, 724–738. [Abstract] [Google Scholar]
  • Bernier LP, York EM, Kamyabi A, Choi HB, Weilinger NL, MacVicar BA, 2020. Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nature communications 11, 1559. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bogetofte H, Jensen P, Ryding M, Schmidt SI, Okarmus J, Ritter L, Worm CS, Hohnholt MC, Azevedo C, Roybon L, Bak LK, Waagepetersen H, Ryan BJ, Wade-Martins R, Larsen MR, Meyer M, 2019. PARK2 Mutation Causes Metabolic Disturbances and Impaired Survival of Human iPSC-Derived Neurons. Frontiers in cellular neuroscience 13, 297. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bolaños JP, Almeida A, Moncada S, 2010. Glycolysis: a bioenergetic or a survival pathway? Trends Biochem Sci 35, 145–149. [Abstract] [Google Scholar]
  • Boussicault L, Hérard AS, Calingasan N, Petit F, Malgorn C, Merienne N, Jan C, Gaillard MC, Lerchundi R, Barros LF, Escartin C, Delzescaux T, Mariani J, Hantraye P, Beal MF, Brouillet E, Véga C, Bonvento G, 2014. Impaired brain energy metabolism in the BACHD mouse model of Huntington’s disease: critical role of astrocyte-neuron interactions. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 34, 1500–1510. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bowman CE, Scafidi J, Scafidi S, 2019. Metabolic perturbations after pediatric TBI: It’s not just about glucose. Experimental neurology 316, 74–84. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bryan MR, Nordham KD, Rose DIR, O’Brien MT, Joshi P, Foshage AM, Gonçalves FM, Nitin R, Uhouse MA, Aschner M, Bowman AB, 2020. Manganese Acts upon Insulin/IGF Receptors to Phosphorylate AKT and Increase Glucose Uptake in Huntington’s Disease Cells. Molecular neurobiology 57, 1570–1593. [Europe PMC free article] [Abstract] [Google Scholar]
  • Buchberger AR, DeLaney K, Johnson J, Li L, 2018. Mass Spectrometry Imaging: A Review of Emerging Advancements and Future Insights. Analytical chemistry 90, 240–265. [Europe PMC free article] [Abstract] [Google Scholar]
  • Bukeirat M, Sarkar SN, Hu H, Quintana DD, Simpkins JW, Ren X, 2016. MiR-34a regulates blood-brain barrier permeability and mitochondrial function by targeting cytochrome c. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 36, 387–392. [Europe PMC free article] [Abstract] [Google Scholar]
  • Burek M, König A, Lang M, Fiedler J, Oerter S, Roewer N, Bohnert M, Thal SC, Blecharz-Lang KG, Woitzik J, Thum T, Förster CY, 2019. Hypoxia-Induced MicroRNA-212/132 Alter Blood-Brain Barrier Integrity Through Inhibition of Tight Junction-Associated Proteins in Human and Mouse Brain Microvascular Endothelial Cells. Translational stroke research 10, 672–683. [Europe PMC free article] [Abstract] [Google Scholar]
  • Burmistrova O, Olias-Arjona A, Lapresa R, Jimenez-Blasco D, Eremeeva T, Shishov D, Romanov S, Zakurdaeva K, Almeida A, Fedichev PO, Bolaños JP, 2019. Targeting PFKFB3 alleviates cerebral ischemia-reperfusion injury in mice. Scientific reports 9, 11670. [Europe PMC free article] [Abstract] [Google Scholar]
  • Butterfield DA, Halliwell B, 2019. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nature reviews. Neuroscience 20, 148–160. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cai R, Zhang Y, Simmering JE, Schultz JL, Li Y, Fernandez-Carasa I, Consiglio A, Raya A, Polgreen PM, Narayanan NS, Yuan Y, Chen Z, Su W, Han Y, Zhao C, Gao L, Ji X, Welsh MJ, Liu L, 2019. Enhancing glycolysis attenuates Parkinson’s disease progression in models and clinical databases. The Journal of clinical investigation 129, 4539–4549. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cali C, Baghabra J, Boges DJ, Holst GR, Kreshuk A, Hamprecht FA, Srinivasan M, Lehvaslaiho H, Magistretti PJ, 2016. Three-dimensional immersive virtual reality for studying cellular compartments in 3D models from EM preparations of neural tissues. The Journal of comparative neurology 524, 23–38. [Europe PMC free article] [Abstract] [Google Scholar]
  • Calvo-Rodriguez M, Hou SS, Snyder AC, Kharitonova EK, Russ AN, Das S, Fan Z, Muzikansky A, Garcia-Alloza M, Serrano-Pozo A, Hudry E, Bacskai BJ, 2020. Increased mitochondrial calcium levels associated with neuronal death in a mouse model of Alzheimer’s disease. Nature communications 11, 2146. [Europe PMC free article] [Abstract] [Google Scholar]
  • Camandola S, Mattson MP, 2017. Brain metabolism in health, aging, and neurodegeneration. The EMBO journal 36, 1474–1492. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cao L, Zhang D, Chen J, Qin YY, Sheng R, Feng X, Chen Z, Ding Y, Li M, Qin ZH, 2017. G6PD plays a neuroprotective role in brain ischemia through promoting pentose phosphate pathway. Free radical biology & medicine 112, 433–444. [Abstract] [Google Scholar]
  • Casanovas S, Schlichtholz L, Mühlbauer S, Dewi S, Schüle M, Strand D, Strand S, Zografidou L, Winter J, 2020. Rbfox1 Is Expressed in the Mouse Brain in the Form of Multiple Transcript Variants and Contains Functional E Boxes in Its Alternative Promoters. Front Mol Neurosci 13, 66. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cheah YS, Amiel SA, 2012. Metabolic neuroimaging of the brain in diabetes mellitus and hypoglycaemia. Nature reviews. Endocrinology 8, 588–597. [Abstract] [Google Scholar]
  • Chen D, Wei L, Liu ZR, Yang JJ, Gu X, Wei ZZ, Liu LP, Yu SP, 2018a. Pyruvate Kinase M2 Increases Angiogenesis, Neurogenesis, and Functional Recovery Mediated by Upregulation of STAT3 and Focal Adhesion Kinase Activities After Ischemic Stroke in Adult Mice. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics 15, 770–784. [Europe PMC free article] [Abstract] [Google Scholar]
  • Chen J, Zhang DM, Feng X, Wang J, Qin YY, Zhang T, Huang Q, Sheng R, Chen Z, Li M, Qin ZH, 2018b. TIGAR inhibits ischemia/reperfusion-induced inflammatory response of astrocytes. Neuropharmacology 131, 377–388. [Abstract] [Google Scholar]
  • Chen Z, Zhong C, 2013. Decoding Alzheimer’s disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Progress in neurobiology 108, 21–43. [Abstract] [Google Scholar]
  • Cheng A, Wang J, Ghena N, Zhao Q, Perone I, King TM, Veech RL, Gorospe M, Wan R, Mattson MP, 2020a. SIRT3 Haploinsufficiency Aggravates Loss of GABAergic Interneurons and Neuronal Network Hyperexcitability in an Alzheimer’s Disease Model. The Journal of neuroscience : the official journal of the Society for Neuroscience 40, 694–709. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos-Bourboulis EJ, Martens JH, Rao NA, Aghajanirefah A, Manjeri GR, Li Y, Ifrim DC, Arts RJ, van der Veer BM, Deen PM, Logie C, O’Neill LA, Willems P, van de Veerdonk FL, van der Meer JW, Ng A, Joosten LA, Wijmenga C, Stunnenberg HG, Xavier RJ, Netea MG, 2014. mTOR- and HIF-1alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345, 1250684. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cheng Y, Shabir G, Li X, Fang L, Xu L, Zhang H, Li E, 2020b. Development of a deep-red fluorescent glucose-conjugated bioprobe for in vivo tumor targeting. Chemical communications (Cambridge, England) 56, 1070–1073. [Abstract] [Google Scholar]
  • Choi SH, Kim HJ, Cho HJ, Park SD, Lee NE, Hwang SH, Cho IH, Hwang H, Rhim H, Kim HC, Nah SY, 2019. Gintonin, a Ginseng-Derived Exogenous Lysophosphatidic Acid Receptor Ligand, Protects Astrocytes from Hypoxic and Re-oxygenation Stresses Through Stimulation of Astrocytic Glycogenolysis. Molecular neurobiology 56, 3280–3294. [Abstract] [Google Scholar]
  • Chong J, Soufan O, Li C, Caraus I, Li S, Bourque G, Wishart DS, Xia J, 2018. MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic acids research 46, W486–w494. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cisternas P, Inestrosa NC, 2017. Brain glucose metabolism: Role of Wnt signaling in the metabolic impairment in Alzheimer’s disease. Neuroscience and biobehavioral reviews 80, 316–328. [Abstract] [Google Scholar]
  • Cisternas P, Salazar P, Silva-Alvarez C, Barros LF, Inestrosa NC, 2016. Activation of Wnt Signaling in Cortical Neurons Enhances Glucose Utilization through Glycolysis. The Journal of biological chemistry 291, 25950–25964. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cisternas P, Zolezzi JM, Martinez M, Torres VI, Wong GW, Inestrosa NC, 2019. Wnt-induced activation of glucose metabolism mediates the in vivo neuroprotective roles of Wnt signaling in Alzheimer disease. Journal of neurochemistry 149, 54–72. [Europe PMC free article] [Abstract] [Google Scholar]
  • Colonna M, Butovsky O, 2017. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 35, 441–468. [Europe PMC free article] [Abstract] [Google Scholar]
  • Courchesne-Loyer A, Croteau E, Castellano CA, St-Pierre V, Hennebelle M, Cunnane SC, 2017. Inverse relationship between brain glucose and ketone metabolism in adults during short-term moderate dietary ketosis: A dual tracer quantitative positron emission tomography study. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 37, 2485–2493. [Europe PMC free article] [Abstract] [Google Scholar]
  • Covarrubias-Pinto A, Moll P, Solís-Maldonado M, Acuña AI, Riveros A, Miró MP, Papic E, Beltrán FA, Cepeda C, Concha II, Brauchi S, Castro MA, 2015. Beyond the redox imbalance: Oxidative stress contributes to an impaired GLUT3 modulation in Huntington’s disease. Free radical biology & medicine 89, 1085–1096. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cucullo L, Hossain M, Puvenna V, Marchi N, Janigro D, 2011. The role of shear stress in Blood-Brain Barrier endothelial physiology. BMC Neurosci 12, 40. [Europe PMC free article] [Abstract] [Google Scholar]
  • Cui L, Lu H, Lee YH, 2018. Challenges and emergent solutions for LC-MS/MS based untargeted metabolomics in diseases. Mass Spectrom Rev 37, 772–792. [Abstract] [Google Scholar]
  • Cunnane SC, Trushina E, Morland C, Prigione A, Casadesus G, Andrews ZB, Beal MF, Bergersen LH, Brinton RD, de la Monte S, Eckert A, Harvey J, Jeggo R, Jhamandas JH, Kann O, la Cour CM, Martin WF, Mithieux G, Moreira PI, Murphy MP, Nave KA, Nuriel T, Oliet SHR, Saudou F, Mattson MP, Swerdlow RH, Millan MJ, 2020. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov 19, 609–633. [Europe PMC free article] [Abstract] [Google Scholar]
  • Currais A, Huang L, Goldberg J, Petrascheck M, Ates G, Pinto-Duarte A, Shokhirev MN, Schubert D, Maher P, 2019. Elevating acetyl-CoA levels reduces aspects of brain aging. eLife 8. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dave A, Hansen N, Downey R, Johnson C, 2020. FDG-PET Imaging of Dementia and Neurodegenerative Disease. Semin Ultrasound CT MR 41, 562–571. [Abstract] [Google Scholar]
  • Davie K, Janssens J, Koldere D, De Waegeneer M, Pech U, Kreft Ł, Aibar S, Makhzami S, Christiaens V, Bravo González-Blas C, Poovathingal S, Hulselmans G, Spanier KI, Moerman T, Vanspauwen B, Geurs S, Voet T, Lammertyn J, Thienpont B, Liu S, Konstantinides N, Fiers M, Verstreken P, Aerts S, 2018. A Single-Cell Transcriptome Atlas of the Aging Drosophila Brain. Cell 174, 982–998.e920. [Europe PMC free article] [Abstract] [Google Scholar]
  • Davies MI, Cooper JD, Desmond SS, Lunte CE, Lunte SM, 2000. Analytical considerations for microdialysis sampling. Advanced drug delivery reviews 45, 169–188. [Abstract] [Google Scholar]
  • de Cabo R, Mattson MP, 2019. Effects of Intermittent Fasting on Health, Aging, and Disease. The New England journal of medicine 381, 2541–2551. [Abstract] [Google Scholar]
  • Dehghani M, Zhang S, Kumaragamage C, Rosa-Neto P, Near J, 2020. Dynamic (1) H-MRS for detection of (13) C-labeled glucose metabolism in the human brain at 3T. Magnetic resonance in medicine 84, 1140–1151. [Abstract] [Google Scholar]
  • Demarest TG, Varma VR, Estrada D, Babbar M, Basu S, Mahajan UV, Moaddel R, Croteau DL, Thambisetty M, Mattson MP, Bohr VA, 2020. Biological sex and DNA repair deficiency drive Alzheimer’s disease via systemic metabolic remodeling and brain mitochondrial dysfunction. Acta neuropathologica. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dewez F, Oejten J, Henkel C, Hebeler R, Neuweger H, De Pauw E, Heeren RMA, Balluff B, 2020. MS Imaging-Guided Microproteomics for Spatial Omics on a Single Instrument. Proteomics 20, e1900369. [Abstract] [Google Scholar]
  • Díaz-García CM, Lahmann C, Martínez-François JR, Li B, Koveal D, Nathwani N, Rahman M, Keller JP, Marvin JS, Looger LL, Yellen G, 2019. Quantitative in vivo imaging of neuronal glucose concentrations with a genetically encoded fluorescence lifetime sensor. Journal of neuroscience research 97, 946–960. [Europe PMC free article] [Abstract] [Google Scholar]
  • Díaz-García CM, Mongeon R, Lahmann C, Koveal D, Zucker H, Yellen G, 2017. Neuronal Stimulation Triggers Neuronal Glycolysis and Not Lactate Uptake. Cell Metab 26, 361–374.e364. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dienel GA, 2019. Brain Glucose Metabolism: Integration of Energetics with Function. Physiological reviews 99, 949–1045. [Abstract] [Google Scholar]
  • Dilillo M, Pellegrini D, Ait-Belkacem R, de Graaf EL, Caleo M, McDonnell LA, 2017. Mass Spectrometry Imaging, Laser Capture Microdissection, and LC-MS/MS of the Same Tissue Section. Journal of proteome research 16, 2993–3001. [Abstract] [Google Scholar]
  • DiNuzzo M, 2013. Kinetic analysis of glycogen turnover: relevance to human brain 13C-NMR spectroscopy. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 33, 1540–1548. [Europe PMC free article] [Abstract] [Google Scholar]
  • Domingo-Almenara X, Montenegro-Burke JR, Guijas C, Majumder EL, Benton HP, Siuzdak G, 2019. Autonomous METLIN-Guided In-source Fragment Annotation for Untargeted Metabolomics. Analytical chemistry 91, 3246–3253. [Europe PMC free article] [Abstract] [Google Scholar]
  • Drulis-Fajdasz D, Gizak A, Wójtowicz T, Wiśniewski JR, Rakus D, 2018. Aging-associated changes in hippocampal glycogen metabolism in mice. Evidence for and against astrocyte-to-neuron lactate shuttle. Glia 66, 1481–1495. [Europe PMC free article] [Abstract] [Google Scholar]
  • Duan W, Jiang M, Jin J, 2014. Metabolism in HD: still a relevant mechanism? Movement disorders : official journal of the Movement Disorder Society 29, 1366–1374. [Europe PMC free article] [Abstract] [Google Scholar]
  • Duarte AI, Santos MS, Oliveira CR, Moreira PI, 2018. Brain insulin signalling, glucose metabolism and females’ reproductive aging: A dangerous triad in Alzheimer’s disease. Neuropharmacology 136, 223–242. [Abstract] [Google Scholar]
  • Duncan A, Heyer MP, Ishikawa M, Caligiuri SPB, Liu XA, Chen Z, Micioni Di Bonaventura MV, Elayouby KS, Ables JL, Howe WM, Bali P, Fillinger C, Williams M, O’Connor RM, Wang Z, Lu Q, Kamenecka TM, Ma’ayan A, O’Neill HC, Ibanez-Tallon I, Geurts AM, Kenny PJ, 2019. Habenular TCF7L2 links nicotine addiction to diabetes. Nature 574, 372–377. [Europe PMC free article] [Abstract] [Google Scholar]
  • Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, Hargreaves IP, Pope S, Holton JL, Lees A, Heales SJ, Bandopadhyay R, 2014. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiology of aging 35, 1111–1115. [Europe PMC free article] [Abstract] [Google Scholar]
  • Eelen G, de Zeeuw P, Treps L, Harjes U, Wong BW, Carmeliet P, 2018. Endothelial Cell Metabolism. Physiological reviews 98, 3–58. [Europe PMC free article] [Abstract] [Google Scholar]
  • Emrani S, Arain HA, DeMarshall C, Nuriel T, 2020. APOE4 is associated with cognitive and pathological heterogeneity in patients with Alzheimer’s disease: a systematic review. Alzheimers Res Ther 12, 141. [Europe PMC free article] [Abstract] [Google Scholar]
  • Erecinska M, Thoresen M, Silver IA, 2003. Effects of hypothermia on energy metabolism in Mammalian central nervous system. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 23, 513–530. [Abstract] [Google Scholar]
  • Eyo UB, Miner SA, Ahlers KE, Wu LJ, Dailey ME, 2013. P2X7 receptor activation regulates microglial cell death during oxygen-glucose deprivation. Neuropharmacology 73, 311–319. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fasciani I, Pluta P, Gonzalez-Nieto D, Martinez-Montero P, Molano J, Paino CL, 2018. Directional coupling of oligodendrocyte connexin-47 and astrocyte connexin-43 gap junctions. Glia 66, 2340–2352. [Abstract] [Google Scholar]
  • Feigin A, Tang C, Ma Y, Mattis P, Zgaljardic D, Guttman M, Paulsen JS, Dhawan V, Eidelberg D, 2007. Thalamic metabolism and symptom onset in preclinical Huntington’s disease. Brain : a journal of neurology 130, 2858–2867. [Europe PMC free article] [Abstract] [Google Scholar]
  • Fernandez AM, Torres-Alemán I, 2012. The many faces of insulin-like peptide signalling in the brain. Nature reviews. Neuroscience 13, 225–239. [Abstract] [Google Scholar]
  • Firbank MJ, Yarnall AJ, Lawson RA, Duncan GW, Khoo TK, Petrides GS, O’Brien JT, Barker RA, Maxwell RJ, Brooks DJ, Burn DJ, 2017. Cerebral glucose metabolism and cognition in newly diagnosed Parkinson’s disease: ICICLE-PD study. Journal of neurology, neurosurgery, and psychiatry 88, 310–316. [Abstract] [Google Scholar]
  • Fleck D, Phu L, Verschueren E, Hinkle T, Reichelt M, Bhangale T, Haley B, Wang Y, Graham R, Kirkpatrick DS, Sheng M, Bingol B, 2019. PTCD1 Is Required for Mitochondrial Oxidative-Phosphorylation: Possible Genetic Association with Alzheimer’s Disease. The Journal of neuroscience : the official journal of the Society for Neuroscience 39, 4636–4656. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gabbouj S, Ryhänen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, Martiskainen H, Tanila H, Haapasalo A, Hiltunen M, Natunen T, 2019. Altered Insulin Signaling in Alzheimer’s Disease Brain - Special Emphasis on PI3K-Akt Pathway. Frontiers in neuroscience 13, 629. [Europe PMC free article] [Abstract] [Google Scholar]
  • García-Cáceres C, Quarta C, Varela L, Gao Y, Gruber T, Legutko B, Jastroch M, Johansson P, Ninkovic J, Yi CX, Le Thuc O, Szigeti-Buck K, Cai W, Meyer CW, Pfluger PT, Fernandez AM, Luquet S, Woods SC, Torres-Alemán I, Kahn CR, Götz M, Horvath TL, Tschöp MH, 2016. Astrocytic Insulin Signaling Couples Brain Glucose Uptake with Nutrient Availability. Cell 166, 867–880. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gaura V, Lavisse S, Payoux P, Goldman S, Verny C, Krystkowiak P, Damier P, Supiot F, Bachoud-Levi AC, Remy P, 2017. Association Between Motor Symptoms and Brain Metabolism in Early Huntington Disease. JAMA neurology 74, 1088–1096. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gerszon J, Rodacka A, 2018. Oxidatively modified glyceraldehyde-3-phosphate dehydrogenase in neurodegenerative processes and the role of low molecular weight compounds in counteracting its aggregation and nuclear translocation. Ageing research reviews 48, 21–31. [Abstract] [Google Scholar]
  • Ghosh-Choudhary S, Liu J, Finkel T, 2020. Metabolic Regulation of Cell Fate and Function. Trends in cell biology 30, 201–212. [Europe PMC free article] [Abstract] [Google Scholar]
  • Gonzalez-Riano C, Garcia A, Barbas C, 2016. Metabolomics studies in brain tissue: A review. J Pharm Biomed Anal 130, 141–168. [Abstract] [Google Scholar]
  • Gordon BA, Blazey TM, Su Y, Hari-Raj A, Dincer A, Flores S, Christensen J, McDade E, Wang G, Xiong C, Cairns NJ, Hassenstab J, Marcus DS, Fagan AM, Jack CR Jr., Hornbeck RC, Paumier KL, Ances BM, Berman SB, Brickman AM, Cash DM, Chhatwal JP, Correia S, Förster S, Fox NC, Graff-Radford NR, la Fougère C, Levin J, Masters CL, Rossor MN, Salloway S, Saykin AJ, Schofield PR, Thompson PM, Weiner MM, Holtzman DM, Raichle ME, Morris JC, Bateman RJ, Benzinger TLS, 2018. Spatial patterns of neuroimaging biomarker change in individuals from families with autosomal dominant Alzheimer’s disease: a longitudinal study. The Lancet. Neurology 17, 241–250. [Europe PMC free article] [Abstract] [Google Scholar]
  • Goyal MS, Hawrylycz M, Miller JA, Snyder AZ, Raichle ME, 2014. Aerobic glycolysis in the human brain is associated with development and neotenous gene expression. Cell Metab 19, 49–57. [Europe PMC free article] [Abstract] [Google Scholar]
  • Goyal MS, Vlassenko AG, Blazey TM, Su Y, Couture LE, Durbin TJ, Bateman RJ, Benzinger TL, Morris JC, Raichle ME, 2017. Loss of Brain Aerobic Glycolysis in Normal Human Aging. Cell Metab 26, 353–360.e353. [Europe PMC free article] [Abstract] [Google Scholar]
  • Griffith CM, Macklin LN, Cai Y, Sharp AA, Yan XX, Reagan LP, Strader AD, Rose GM, Patrylo PR, 2019. Impaired Glucose Tolerance and Reduced Plasma Insulin Precede Decreased AKT Phosphorylation and GLUT3 Translocation in the Hippocampus of Old 3xTg-AD Mice. Journal of Alzheimer’s disease : JAD 68, 809–837. [Abstract] [Google Scholar]
  • Hackett MJ, Sylvain NJ, Hou H, Caine S, Alaverdashvili M, Pushie MJ, Kelly ME, 2016. Concurrent Glycogen and Lactate Imaging with FTIR Spectroscopy To Spatially Localize Metabolic Parameters of the Glial Response Following Brain Ischemia. Analytical chemistry 88, 10949–10956. [Abstract] [Google Scholar]
  • Hafner A, Obermajer N, Kos J, 2012. γ-Enolase C-terminal peptide promotes cell survival and neurite outgrowth by activation of the PI3K/Akt and MAPK/ERK signalling pathways. The Biochemical journal 443, 439–450. [Abstract] [Google Scholar]
  • Hahn DK, Geocadin RG, Greer DM, 2014. Quality of evidence in studies evaluating neuroimaging for neurologic prognostication in adult patients resuscitated from cardiac arrest. Resuscitation 85, 165–172. [Abstract] [Google Scholar]
  • Halim ND, McFate T, Mohyeldin A, Okagaki P, Korotchkina LG, Patel MS, Jeoung NH, Harris RA, Schell MJ, Verma A, 2010. Phosphorylation status of pyruvate dehydrogenase distinguishes metabolic phenotypes of cultured rat brain astrocytes and neurons. Glia 58, 1168–1176. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hamilton J, Pellman JJ, Brustovetsky T, Harris RA, Brustovetsky N, 2015. Oxidative metabolism in YAC128 mouse model of Huntington’s disease. Human molecular genetics 24, 4862–4878. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hammarlund-Udenaes M, 2017. Microdialysis as an Important Technique in Systems Pharmacology-a Historical and Methodological Review. The AAPS journal 19, 1294–1303. [Abstract] [Google Scholar]
  • Haque A, Polcyn R, Matzelle D, Banik NL, 2018. New Insights into the Role of Neuron-Specific Enolase in Neuro-Inflammation, Neurodegeneration, and Neuroprotection. Brain sciences 8. [Europe PMC free article] [Abstract] [Google Scholar]
  • Haque A, Ray SK, Cox A, Banik NL, 2016. Neuron specific enolase: a promising therapeutic target in acute spinal cord injury. Metabolic brain disease 31, 487–495. [Europe PMC free article] [Abstract] [Google Scholar]
  • Harraz MM, Tyagi R, Cortés P, Snyder SH, 2016. Antidepressant action of ketamine via mTOR is mediated by inhibition of nitrergic Rheb degradation. Molecular psychiatry 21, 313–319. [Europe PMC free article] [Abstract] [Google Scholar]
  • Harris RA, Tindale L, Lone A, Singh O, Macauley SL, Stanley M, Holtzman DM, Bartha R, Cumming RC, 2016. Aerobic Glycolysis in the Frontal Cortex Correlates with Memory Performance in Wild-Type Mice But Not the APP/PS1 Mouse Model of Cerebral Amyloidosis. The Journal of neuroscience : the official journal of the Society for Neuroscience 36, 1871–1878. [Europe PMC free article] [Abstract] [Google Scholar]
  • He J, Lu H, Young L, Deng R, Callow D, Tong S, Jia X, 2019. Real-time quantitative monitoring of cerebral blood flow by laser speckle contrast imaging after cardiac arrest with targeted temperature management. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 39, 1161–1171. [Europe PMC free article] [Abstract] [Google Scholar]
  • Herrero-Mendez A, Almeida A, Fernandez E, Maestre C, Moncada S, Bolanos JP, 2009. The bioenergetic and antioxidant status of neurons is controlled by continuous degradation of a key glycolytic enzyme by APC/C-Cdh1. Nature cell biology 11, 747–752. [Abstract] [Google Scholar]
  • Hinkle PC, 2005. P/O ratios of mitochondrial oxidative phosphorylation. Biochimica et biophysica acta 1706, 1–11. [Abstract] [Google Scholar]
  • Hofmeijer J, van Putten MJ, 2012. Ischemic cerebral damage: an appraisal of synaptic failure. Stroke 43, 607–615. [Abstract] [Google Scholar]
  • Holth JK, Fritschi SK, Wang C, Pedersen NP, Cirrito JR, Mahan TE, Finn MB, Manis M, Geerling JC, Fuller PM, Lucey BP, Holtzman DM, 2019. The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science (New York, N.Y.) 363, 880–884. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hou L, Sun F, Huang R, Sun W, Zhang D, Wang Q, 2019. Inhibition of NADPH oxidase by apocynin prevents learning and memory deficits in a mouse Parkinson’s disease model. Redox biology 22, 101134. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hu H, Hone EA, Provencher EAP, Sprowls SA, Farooqi I, Corbin DR, Sarkar SN, Hollander JM, Lockman PR, Simpkins JW, Ren X, 2020. MiR-34a Interacts with Cytochrome c and Shapes Stroke Outcomes. Scientific reports 10, 3233. [Europe PMC free article] [Abstract] [Google Scholar]
  • Huang Q, Sun M, Li M, Zhang D, Han F, Wu JC, Fukunaga K, Chen Z, Qin ZH, 2018. Combination of NAD(+) and NADPH Offers Greater Neuroprotection in Ischemic Stroke Models by Relieving Metabolic Stress. Molecular neurobiology 55, 6063–6075. [Abstract] [Google Scholar]
  • Huber M, Beyer L, Prix C, Schönecker S, Palleis C, Rauchmann BS, Morbelli S, Chincarini A, Bruffaerts R, Vandenberghe R, Van Laere K, Kramberger MG, Trost M, Grmek M, Garibotto V, Nicastro N, Frisoni GB, Lemstra AW, van der Zande J, Pilotto A, Padovani A, Garcia-Ptacek S, Savitcheva I, Ochoa-Figueroa MA, Davidsson A, Camacho V, Peira E, Arnaldi D, Bauckneht M, Pardini M, Sambuceti G, Vöglein J, Schnabel J, Unterrainer M, Perneczky R, Pogarell O, Buerger K, Catak C, Bartenstein P, Cumming P, Ewers M, Danek A, Levin J, Aarsland D, Nobili F, Rominger A, Brendel M, 2020. Metabolic Correlates of Dopaminergic Loss in Dementia with Lewy Bodies. Movement disorders : official journal of the Movement Disorder Society 35, 595–605. [Abstract] [Google Scholar]
  • Hui S, Ghergurovich JM, Morscher RJ, Jang C, Teng X, Lu W, Esparza LA, Reya T, Le Z, Yanxiang Guo J, White E, Rabinowitz JD, 2017. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hwang JJ, Jiang L, Hamza M, Sanchez Rangel E, Dai F, Belfort-DeAguiar R, Parikh L, Koo BB, Rothman DL, Mason G, Sherwin RS, 2017. Blunted rise in brain glucose levels during hyperglycemia in adults with obesity and T2DM. JCI insight 2. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hwang JJ, Jiang L, Sanchez Rangel E, Fan X, Ding Y, Lam W, Leventhal J, Dai F, Rothman DL, Mason GF, Sherwin RS, 2019. Glycemic Variability and Brain Glucose Levels in Type 1 Diabetes. Diabetes 68, 163–171. [Europe PMC free article] [Abstract] [Google Scholar]
  • Hyder F, Rothman DL, 2017. Advances in Imaging Brain Metabolism. Annual review of biomedical engineering 19, 485–515. [Abstract] [Google Scholar]
  • Ishii K, 2014. PET approaches for diagnosis of dementia. AJNR Am J Neuroradiol 35, 2030–2038. [Europe PMC free article] [Abstract] [Google Scholar]
  • Izuishi K, Yamamoto Y, Mori H, Kameyama R, Fujihara S, Masaki T, Suzuki Y, 2014. Molecular mechanisms of [18F]fluorodeoxyglucose accumulation in liver cancer. Oncology reports 31, 701–706. [Abstract] [Google Scholar]
  • Jadhav SB, Khaowroongrueng V, Derendorf H, 2016. Microdialysis of Large Molecules. Journal of pharmaceutical sciences 105, 3233–3242. [Abstract] [Google Scholar]
  • Jais A, Solas M, Backes H, Chaurasia B, Kleinridders A, Theurich S, Mauer J, Steculorum SM, Hampel B, Goldau J, Alber J, Forster CY, Eming SA, Schwaninger M, Ferrara N, Karsenty G, Bruning JC, 2016. Myeloid-Cell-Derived VEGF Maintains Brain Glucose Uptake and Limits Cognitive Impairment in Obesity. Cell 165, 882–895. [Abstract] [Google Scholar]
  • Jakkamsetti V, Marin-Valencia I, Ma Q, Good LB, Terrill T, Rajasekaran K, Pichumani K, Khemtong C, Hooshyar MA, Sundarrajan C, Patel MS, Bachoo RM, Malloy CR, Pascual JM, 2019. Brain metabolism modulates neuronal excitability in a mouse model of pyruvate dehydrogenase deficiency. Science translational medicine 11. [Europe PMC free article] [Abstract] [Google Scholar]
  • Jaquins-Gerstl A, Michael AC, 2020. Dexamethasone-Enhanced Microdialysis and Penetration Injury. Frontiers in bioengineering and biotechnology 8, 602266. [Europe PMC free article] [Abstract] [Google Scholar]
  • Johnson CH, Ivanisevic J, Siuzdak G, 2016. Metabolomics: beyond biomarkers and towards mechanisms. Nature reviews. Molecular cell biology 17, 451–459. [Europe PMC free article] [Abstract] [Google Scholar]
  • Jung DW, Kim WH, Park SH, Lee J, Kim J, Su D, Ha HH, Chang YT, Williams DR, 2013. A unique small molecule inhibitor of enolase clarifies its role in fundamental biological processes. ACS chemical biology 8, 1271–1282. [Abstract] [Google Scholar]
  • Kacimi R, Giffard RG, Yenari MA, 2011. Endotoxin-activated microglia injure brain derived endothelial cells via NF-kappaB, JAK-STAT and JNK stress kinase pathways. Journal of inflammation (London, England) 8, 7. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kalia LV, Lang AE, 2015. Parkinson’s disease. Lancet (London, England) 386, 896–912. [Abstract] [Google Scholar]
  • Kalsbeek MJ, Mulder L, Yi CX, 2016. Microglia energy metabolism in metabolic disorder. Molecular and cellular endocrinology 438, 27–35. [Abstract] [Google Scholar]
  • Kalucka J, de Rooij L, Goveia J, Rohlenova K, Dumas SJ, Meta E, Conchinha NV, Taverna F, Teuwen LA, Veys K, García-Caballero M, Khan S, Geldhof V, Sokol L, Chen R, Treps L, Borri M, de Zeeuw P, Dubois C, Karakach TK, Falkenberg KD, Parys M, Yin X, Vinckier S, Du Y, Fenton RA, Schoonjans L, Dewerchin M, Eelen G, Thienpont B, Lin L, Bolund L, Li X, Luo Y, Carmeliet P, 2020. Single-Cell Transcriptome Atlas of Murine Endothelial Cells. Cell 180, 764–779.e720. [Abstract] [Google Scholar]
  • Kapoor K, Finer-Moore JS, Pedersen BP, Caboni L, Waight A, Hillig RC, Bringmann P, Heisler I, Müller T, Siebeneicher H, Stroud RM, 2016. Mechanism of inhibition of human glucose transporter GLUT1 is conserved between cytochalasin B and phenylalanine amides. Proc Natl Acad Sci U S A 113, 4711–4716. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kashiwaya Y, Bergman C, Lee JH, Wan R, King MT, Mughal MR, Okun E, Clarke K, Mattson MP, Veech RL, 2013. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer’s disease. Neurobiology of aging 34, 1530–1539. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kaur H, Kamalov M, Brimble MA, 2016. Chemical Synthesis of Peptides Containing Site-Specific Advanced Glycation Endproducts. Accounts of chemical research 49, 2199–2208. [Abstract] [Google Scholar]
  • Kavanagh K, Day SM, Pait MC, Mortiz WR, Newgard CB, Ilkayeva O, McClain DA, Macauley SL, 2019. Type-2-Diabetes Alters CSF but Not Plasma Metabolomic and AD Risk Profiles in Vervet Monkeys. Frontiers in neuroscience 13, 843. [Europe PMC free article] [Abstract] [Google Scholar]
  • Keeney JT, Ibrahimi S, Zhao L, 2015. Human ApoE Isoforms Differentially Modulate Glucose and Amyloid Metabolic Pathways in Female Brain: Evidence of the Mechanism of Neuroprotection by ApoE2 and Implications for Alzheimer’s Disease Prevention and Early Intervention. Journal of Alzheimer’s disease : JAD 48, 411–424. [Europe PMC free article] [Abstract] [Google Scholar]
  • Khanna S, Stewart R, Gnyawali S, Harris H, Balch M, Spieldenner J, Sen CK, Rink C, 2017. Phytoestrogen isoflavone intervention to engage the neuroprotective effect of glutamate oxaloacetate transaminase against stroke. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 31, 4533–4544. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kilic K, Karatas H, Dönmez-Demir B, Eren-Kocak E, Gursoy-Ozdemir Y, Can A, Petit JM, Magistretti PJ, Dalkara T, 2018. Inadequate brain glycogen or sleep increases spreading depression susceptibility. Annals of neurology 83, 61–73. [Abstract] [Google Scholar]
  • Kim D, Yoon HJ, Lee WJ, Woo SH, Kim BS, 2019. Prognostic value of (18)F-FDG brain PET as an early indicator of neurological outcomes in a rat model of post-cardiac arrest syndrome. Scientific reports 9, 14798. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kim KA, Kim D, Kim JH, Shin YJ, Kim ES, Akram M, Kim EH, Majid A, Baek SH, Bae ON, 2020. Autophagy-mediated occludin degradation contributes to blood-brain barrier disruption during ischemia in bEnd.3 brain endothelial cells and rat ischemic stroke models. Fluids Barriers CNS 17, 21. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kleinridders A, Ferris HA, Cai W, Kahn CR, 2014. Insulin action in brain regulates systemic metabolism and brain function. Diabetes 63, 2232–2243. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kleinridders A, Ferris HA, Reyzer ML, Rath M, Soto M, Manier ML, Spraggins J, Yang Z, Stanton RC, Caprioli RM, Kahn CR, 2018. Regional differences in brain glucose metabolism determined by imaging mass spectrometry. Molecular metabolism 12, 113–121. [Europe PMC free article] [Abstract] [Google Scholar]
  • Klinge CM, 2020. Estrogenic control of mitochondrial function. Redox biology 31, 101435. [Europe PMC free article] [Abstract] [Google Scholar]
  • Knight AL, Yan X, Hamamichi S, Ajjuri RR, Mazzulli JR, Zhang MW, Daigle JG, Zhang S, Borom AR, Roberts LR, Lee SK, DeLeon SM, Viollet-Djelassi C, Krainc D, O’Donnell JM, Caldwell KA, Caldwell GA, 2014. The glycolytic enzyme, GPI, is a functionally conserved modifier of dopaminergic neurodegeneration in Parkinson’s models. Cell Metab 20, 145–157. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kong LL, Miao D, Tan L, Liu SL, Li JQ, Cao XP, Tan L, 2018. Genome-wide association study identifies RBFOX1 locus influencing brain glucose metabolism. Ann Transl Med 6, 436. [Europe PMC free article] [Abstract] [Google Scholar]
  • Koronowski KB, Khoury N, Saul I, Loris ZB, Cohan CH, Stradecki-Cohan HM, Dave KR, Young JI, Perez-Pinzon MA, 2017. Neuronal SIRT1 (Silent Information Regulator 2 Homologue 1) Regulates Glycolysis and Mediates Resveratrol-Induced Ischemic Tolerance. Stroke 48, 3117–3125. [Europe PMC free article] [Abstract] [Google Scholar]
  • Kullmann S, Kleinridders A, Small DM, Fritsche A, Häring HU, Preissl H, Heni M, 2020. Central nervous pathways of insulin action in the control of metabolism and food intake. Lancet Diabetes Endocrinol 8, 524–534. [Abstract] [Google Scholar]
  • Kusonmano K, Vongsangnak W, Chumnanpuen P, 2016. Informatics for Metabolomics. Advances in experimental medicine and biology 939, 91–115. [Abstract] [Google Scholar]
  • Lane AN, Fan TW, 2015. Regulation of mammalian nucleotide metabolism and biosynthesis. Nucleic acids research 43, 2466–2485. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lauro C, Chece G, Monaco L, Antonangeli F, Peruzzi G, Rinaldo S, Paone A, Cutruzzolà F, Limatola C, 2019. Fractalkine Modulates Microglia Metabolism in Brain Ischemia. Frontiers in cellular neuroscience 13, 414. [Europe PMC free article] [Abstract] [Google Scholar]
  • Le Douce J, Maugard M, Veran J, Matos M, Jégo P, Vigneron PA, Faivre E, Toussay X, Vandenberghe M, Balbastre Y, Piquet J, Guiot E, Tran NT, Taverna M, Marinesco S, Koyanagi A, Furuya S, Gaudin-Guérif M, Goutal S, Ghettas A, Pruvost A, Bemelmans AP, Gaillard MC, Cambon K, Stimmer L, Sazdovitch V, Duyckaerts C, Knott G, Hérard AS, Delzescaux T, Hantraye P, Brouillet E, Cauli B, Oliet SHR, Panatier A, Bonvento G, 2020. Impairment of Glycolysis-Derived l-Serine Production in Astrocytes Contributes to Cognitive Deficits in Alzheimer’s Disease. Cell Metab 31, 503–517.e508. [Abstract] [Google Scholar]
  • Lee JY, Lee H, Yoo HB, Choi JS, Jung HY, Yoon EJ, Kim H, Jung YH, Lee HY, Kim YK, 2019. Efficacy of Cilostazol Administration in Alzheimer’s Disease Patients with White Matter Lesions: A Positron-Emission Tomography Study. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics 16, 394–403. [Europe PMC free article] [Abstract] [Google Scholar]
  • Lee Y, Morrison BM, Li Y, Lengacher S, Farah MH, Hoffman PN, Liu Y, Tsingalia A, Jin L, Zhang PW, Pellerin L, Magistretti PJ, Rothstein JD, 2012. Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature 487, 443–448. [Europe PMC free article] [Abstract] [Google Scholar]
  • Levenga J, Wong H, Milstead RA, Keller BN, LaPlante LE, Hoeffer CA, 2017. AKT isoforms have distinct hippocampal expression and roles in synaptic plasticity. eLife 6. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li M, Sun M, Cao L, Gu JH, Ge J, Chen J, Han R, Qin YY, Zhou ZP, Ding Y, Qin ZH, 2014. A TIGAR-regulated metabolic pathway is critical for protection of brain ischemia. The Journal of neuroscience : the official journal of the Society for Neuroscience 34, 7458–7471. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li M, Zhou ZP, Sun M, Cao L, Chen J, Qin YY, Gu JH, Han F, Sheng R, Wu JC, Ding Y, Qin ZH, 2016. Reduced Nicotinamide Adenine Dinucleotide Phosphate, a Pentose Phosphate Pathway Product, Might Be a Novel Drug Candidate for Ischemic Stroke. Stroke 47, 187–195. [Abstract] [Google Scholar]
  • Li Y, Lu B, Sheng L, Zhu Z, Sun H, Zhou Y, Yang Y, Xue D, Chen W, Tian X, Du Y, Yan M, Zhu W, Xing F, Li K, Lin S, Qiu P, Su X, Huang Y, Yan G, Yin W, 2018. Hexokinase 2-dependent hyperglycolysis driving microglial activation contributes to ischemic brain injury. Journal of neurochemistry 144, 186–200. [Abstract] [Google Scholar]
  • Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, Powers K, Shen H, Egan JM, Sambamurti K, Brossi A, Lahiri DK, Mattson MP, Hoffer BJ, Wang Y, Greig NH, 2009. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci U S A 106, 1285–1290. [Europe PMC free article] [Abstract] [Google Scholar]
  • Li Y, Yang Y, Zhao Y, Zhang J, Liu B, Jiao S, Zhang X, 2019. Astragaloside IV reduces neuronal apoptosis and parthanatos in ischemic injury by preserving mitochondrial hexokinase-II. Free radical biology & medicine 131, 251–263. [Abstract] [Google Scholar]
  • Li YQ, Liao XX, Lu JH, Liu R, Hu CL, Dai G, Zhang XS, Shi XC, Li X, 2015. Assessing the early changes of cerebral glucose metabolism via dynamic (18)FDG-PET/CT during cardiac arrest. Metabolic brain disease 30, 969–977. [Abstract] [Google Scholar]
  • Liu CC, Hu J, Tsai CW, Yue M, Melrose HL, Kanekiyo T, Bu G, 2015. Neuronal LRP1 regulates glucose metabolism and insulin signaling in the brain . The Journal of neuroscience : the official journal of the Society for Neuroscience 35, 5851–5859. [Europe PMC free article] [Abstract] [Google Scholar]
  • Liu K, Li F, Han H, Chen Y, Mao Z, Luo J, Zhao Y, Zheng B, Gu W, Zhao W, 2016. Parkin Regulates the Activity of Pyruvate Kinase M2. The Journal of biological chemistry 291, 10307–10317. [Europe PMC free article] [Abstract] [Google Scholar]
  • Liu Y, Li M, Zhang Z, Ye Y, Zhou J, 2018. Role of microglia-neuron interactions in diabetic encephalopathy. Ageing research reviews 42, 28–39. [Abstract] [Google Scholar]
  • Lopez-Fabuel I, Le Douce J, Logan A, James AM, Bonvento G, Murphy MP, Almeida A, Bolaños JP, 2016. Complex I assembly into supercomplexes determines differential mitochondrial ROS production in neurons and astrocytes. Proc Natl Acad Sci U S A 113, 13063–13068. [Europe PMC free article] [Abstract] [Google Scholar]
  • López-Mora DA, Camacho V, Pérez-Pérez J, Martínez-Horta S, Fernández A, Sampedro F, Montes A, Lozano-Martínez GA, Gómez-Anson B, Kulisevsky J, Carrió I, 2016. Striatal hypometabolism in premanifest and manifest Huntington’s disease patients. European journal of nuclear medicine and molecular imaging 43, 2183–2189. [Abstract] [Google Scholar]
  • Luan H, Wang X, Cai Z, 2019. Mass spectrometry-based metabolomics: Targeting the crosstalk between gut microbiota and brain in neurodegenerative disorders. Mass Spectrom Rev 38, 22–33. [Abstract] [Google Scholar]
  • Lundgaard I, Li B, Xie L, Kang H, Sanggaard S, Haswell JD, Sun W, Goldman S, Blekot S, Nielsen M, Takano T, Deane R, Nedergaard M, 2015. Direct neuronal glucose uptake heralds activity-dependent increases in cerebral metabolism. Nature communications 6, 6807. [Europe PMC free article] [Abstract] [Google Scholar]
  • Macauley SL, Stanley M, Caesar EE, Yamada SA, Raichle ME, Perez R, Mahan TE, Sutphen CL, Holtzman DM, 2015. Hyperglycemia modulates extracellular amyloid-β concentrations and neuronal activity in vivo. The Journal of clinical investigation 125, 2463–2467. [Europe PMC free article] [Abstract] [Google Scholar]
  • Machler P, Wyss MT, Elsayed M, Stobart J, Gutierrez R, von Faber-Castell A, Kaelin V, Zuend M, San Martin A, Romero-Gomez I, Baeza-Lehnert F, Lengacher S, Schneider BL, Aebischer P, Magistretti PJ, Barros LF, Weber B, 2016. In Vivo Evidence for a Lactate Gradient from Astrocytes to Neurons. Cell Metab 23, 94–102. [Abstract] [Google Scholar]
  • Marques S, Zeisel A, Codeluppi S, van Bruggen D, Mendanha Falcao A, Xiao L, Li H, Haring M, Hochgerner H, Romanov RA, Gyllborg D, Munoz Manchado A, La Manno G, Lonnerberg P, Floriddia EM, Rezayee F, Ernfors P, Arenas E, Hjerling-Leffler J, Harkany T, Richardson WD, Linnarsson S, Castelo-Branco G, 2016. Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science (New York, N.Y.) 352, 1326–1329. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mathieu C, Li de la Sierra-Gallay I, Duval R, Xu X, Cocaign A, Léger T, Woffendin G, Camadro JM, Etchebest C, Haouz A, Dupret JM, Rodrigues-Lima F, 2016. Insights into Brain Glycogen Metabolism: THE STRUCTURE OF HUMAN BRAIN GLYCOGEN PHOSPHORYLASE. The Journal of biological chemistry 291, 18072–18083. [Europe PMC free article] [Abstract] [Google Scholar]
  • Matsui T, Omuro H, Liu YF, Soya M, Shima T, McEwen BS, Soya H, 2017. Astrocytic glycogen-derived lactate fuels the brain during exhaustive exercise to maintain endurance capacity. Proc Natl Acad Sci U S A 114, 6358–6363. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mattson MP, 2012. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab 16, 706–722. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mehran AE, Templeman NM, Brigidi GS, Lim GE, Chu KY, Hu X, Botezelli JD, Asadi A, Hoffman BG, Kieffer TJ, Bamji SX, Clee SM, Johnson JD, 2012. Hyperinsulinemia drives diet-induced obesity independently of brain insulin production. Cell Metab 16, 723–737. [Abstract] [Google Scholar]
  • Meles SK, Teune LK, de Jong BM, Dierckx RA, Leenders KL, 2017. Metabolic Imaging in Parkinson Disease. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 58, 23–28. [Abstract] [Google Scholar]
  • Mews P, Donahue G, Drake AM, Luczak V, Abel T, Berger SL, 2017. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature 546, 381–386. [Europe PMC free article] [Abstract] [Google Scholar]
  • Meyer N, Richter N, Fan Z, Siemonsmeier G, Pivneva T, Jordan P, Steinhauser C, Semtner M, Nolte C, Kettenmann H, 2018. Oligodendrocytes in the Mouse Corpus Callosum Maintain Axonal Function by Delivery of Glucose. Cell reports 22, 2383–2394. [Abstract] [Google Scholar]
  • Miller A, Nagy C, Knapp B, Laengle J, Ponweiser E, Groeger M, Starkl P, Bergmann M, Wagner O, Haschemi A, 2017. Exploring Metabolic Configurations of Single Cells within Complex Tissue Microenvironments. Cell Metab 26, 788–800.e786. [Abstract] [Google Scholar]
  • Mookerjee SA, Gerencser AA, Nicholls DG, Brand MD, 2017. Quantifying intracellular rates of glycolytic and oxidative ATP production and consumption using extracellular flux measurements. The Journal of biological chemistry 292, 7189–7207. [Europe PMC free article] [Abstract] [Google Scholar]
  • Morken TS, Brekke E, Håberg A, Widerøe M, Brubakk AM, Sonnewald U, 2014. Altered astrocyte-neuronal interactions after hypoxia-ischemia in the neonatal brain in female and male rats. Stroke 45, 2777–2785. [Abstract] [Google Scholar]
  • Morrison BM, Lee Y, Rothstein JD, 2013. Oligodendroglia: metabolic supporters of axons. Trends in cell biology 23, 644–651. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mu Q, Chen Y, Wang J, 2019. Deciphering Brain Complexity Using Single-cell Sequencing. Genomics, proteomics & bioinformatics 17, 344–366. [Europe PMC free article] [Abstract] [Google Scholar]
  • Muller MS, Fouyssac M, Taylor CW, 2018. Effective Glucose Uptake by Human Astrocytes Requires Its Sequestration in the Endoplasmic Reticulum by Glucose-6-Phosphatase-beta. Current biology : CB 28, 3481–3486.e3484. [Europe PMC free article] [Abstract] [Google Scholar]
  • Mulukutla BC, Yongky A, Le T, Mashek DG, Hu WS, 2016. Regulation of Glucose Metabolism - A Perspective From Cell Bioprocessing. Trends in biotechnology 34, 638–651. [Abstract] [Google Scholar]
  • Munji RN, Soung AL, Weiner GA, Sohet F, Semple BD, Trivedi A, Gimlin K, Kotoda M, Korai M, Aydin S, Batugal A, Cabangcala AC, Schupp PG, Oldham MC, Hashimoto T, Noble-Haeusslein LJ, Daneman R, 2019. Profiling the mouse brain endothelial transcriptome in health and disease models reveals a core blood-brain barrier dysfunction module. Nat Neurosci 22, 1892–1902. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nakajima H, Kubo T, Ihara H, Hikida T, Danjo T, Nakatsuji M, Shahani N, Itakura M, Ono Y, Azuma YT, Inui T, Kamiya A, Sawa A, Takeuchi T, 2015. Nuclear-translocated Glyceraldehyde-3-phosphate Dehydrogenase Promotes Poly(ADP-ribose) Polymerase-1 Activation during Oxidative/Nitrosative Stress in Stroke. The Journal of biological chemistry 290, 14493–14503. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nave KA, Werner HB, 2014. Myelination of the nervous system: mechanisms and functions. Annual review of cell and developmental biology 30, 503–533. [Abstract] [Google Scholar]
  • Naylor E, Aillon DV, Barrett BS, Wilson GS, Johnson DA, Johnson DA, Harmon HP, Gabbert S, Petillo PA, 2012. Lactate as a biomarker for sleep. Sleep 35, 1209–1222. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nesbitt KM, Varner EL, Jaquins-Gerstl A, Michael AC, 2015. Microdialysis in the rat striatum: effects of 24 h dexamethasone retrodialysis on evoked dopamine release and penetration injury. ACS chemical neuroscience 6, 163–173. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ng J, Kaur H, Collier T, Chang K, Brooks AES, Allison JR, Brimble MA, Hickey A, Birch NP, 2019. Site-specific glycation of Aβ1–42 affects fibril formation and is neurotoxic. The Journal of biological chemistry 294, 8806–8818. [Europe PMC free article] [Abstract] [Google Scholar]
  • Niccoli T, Cabecinha M, Tillmann A, Kerr F, Wong CT, Cardenes D, Vincent AJ, Bettedi L, Li L, Grönke S, Dols J, Partridge L, 2016. Increased Glucose Transport into Neurons Rescues Aβ Toxicity in Drosophila. Current biology : CB 26, 2291–2300. [Europe PMC free article] [Abstract] [Google Scholar]
  • Nitschke F, Ahonen SJ, Nitschke S, Mitra S, Minassian BA, 2018. Lafora disease - from pathogenesis to treatment strategies. Nature reviews. Neurology 14, 606–617. [Europe PMC free article] [Abstract] [Google Scholar]
  • Oe Y, Baba O, Ashida H, Nakamura KC, 2016. Glycogen distribution in the microwave-fixed mouse brain reveals heterogeneous astrocytic patterns. Glia 64, 1532–1545. [Europe PMC free article] [Abstract] [Google Scholar]
  • Oldendorf WH, Cornford ME, Brown WJ, 1977. The large apparent work capability of the blood-brain barrier: a study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat. Annals of neurology 1, 409–417. [Abstract] [Google Scholar]
  • Orthmann-Murphy JL, Freidin M, Fischer E, Scherer SS, Abrams CK, 2007. Two distinct heterotypic channels mediate gap junction coupling between astrocyte and oligodendrocyte connexins. The Journal of neuroscience : the official journal of the Society for Neuroscience 27, 13949–13957. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pan RY, Ma J, Kong XX, Wang XF, Li SS, Qi XL, Yan YH, Cheng J, Liu Q, Jin W, Tan CH, Yuan Z, 2019. Sodium rutin ameliorates Alzheimer’s disease-like pathology by enhancing microglial amyloid-β clearance. Science advances 5, eaau6328. [Europe PMC free article] [Abstract] [Google Scholar]
  • Patel AB, Lai JC, Chowdhury GM, Hyder F, Rothman DL, Shulman RG, Behar KL, 2014. Direct evidence for activity-dependent glucose phosphorylation in neurons with implications for the astrocyte-to-neuron lactate shuttle. Proc Natl Acad Sci U S A 111, 5385–5390. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pearson-Leary J, Jahagirdar V, Sage J, McNay EC, 2018. Insulin modulates hippocampally-mediated spatial working memory via glucose transporter-4. Behav Brain Res 338, 32–39. [Europe PMC free article] [Abstract] [Google Scholar]
  • Peek CB, Levine DC, Cedernaes J, Taguchi A, Kobayashi Y, Tsai SJ, Bonar NA, McNulty MR, Ramsey KM, Bass J, 2017. Circadian Clock Interaction with HIF1α Mediates Oxygenic Metabolism and Anaerobic Glycolysis in Skeletal Muscle. Cell Metab 25, 86–92. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pellerin L, 2018. Neuroenergetics: Astrocytes Have a Sweet Spot for Glucose. Current biology : CB 28, R1258–r1260. [Abstract] [Google Scholar]
  • Peng Y, Gao P, Shi L, Chen L, Liu J, Long J, 2020. Central and Peripheral Metabolic Defects Contribute to the Pathogenesis of Alzheimer’s Disease: Targeting Mitochondria for Diagnosis and Prevention. Antioxidants & redox signaling 32, 1188–1236. [Europe PMC free article] [Abstract] [Google Scholar]
  • Perez-Cerda F, Sanchez-Gomez MV, Matute C, 2015. Pio del Rio Hortega and the discovery of the oligodendrocytes. Frontiers in neuroanatomy 9, 92. [Europe PMC free article] [Abstract] [Google Scholar]
  • Perez-Escuredo J, Van Hee VF, Sboarina M, Falces J, Payen VL, Pellerin L, Sonveaux P, 2016. Monocarboxylate transporters in the brain and in cancer. Biochimica et biophysica acta 1863, 2481–2497. [Europe PMC free article] [Abstract] [Google Scholar]
  • Perkins AE, Piazza MK, Vore AS, Deak MM, Varlinskaya EI, Deak T, 2021. Assessment of neuroinflammation in the aging hippocampus using large-molecule microdialysis: Sex differences and role of purinergic receptors. Brain, behavior, and immunity 91, 546–555. [Europe PMC free article] [Abstract] [Google Scholar]
  • Philips T, Rothstein JD, 2017. Oligodendroglia: metabolic supporters of neurons. The Journal of clinical investigation 127, 3271–3280. [Europe PMC free article] [Abstract] [Google Scholar]
  • Pinho TS, Verde DM, Correia SC, Cardoso SM, Moreira PI, 2018. O-GlcNAcylation and neuronal energy status: Implications for Alzheimer’s disease. Ageing research reviews 46, 32–41. [Abstract] [Google Scholar]
  • Polykretis P, Luchinat E, Boscaro F, Banci L, 2020. Methylglyoxal interaction with superoxide dismutase 1. Redox biology 30, 101421. [Europe PMC free article] [Abstract] [Google Scholar]
  • Polyzos AA, Lee DY, Datta R, Hauser M, Budworth H, Holt A, Mihalik S, Goldschmidt P, Frankel K, Trego K, Bennett MJ, Vockley J, Xu K, Gratton E, McMurray CT, 2019. Metabolic Reprogramming in Astrocytes Distinguishes Region-Specific Neuronal Susceptibility in Huntington Mice. Cell Metab 29, 1258–1273.e1211. [Europe PMC free article] [Abstract] [Google Scholar]
  • Putzu A, Valtorta S, Di Grigoli G, Haenggi M, Belloli S, Malgaroli A, Gemma M, Landoni G, Beretta L, Moresco RM, 2018. Regional Differences in Cerebral Glucose Metabolism After Cardiac Arrest and Resuscitation in Rats Using [(18)F]FDG Positron Emission Tomography and Autoradiography. Neurocritical care 28, 370–378. [Abstract] [Google Scholar]
  • Qi G, Mi Y, Yin F, 2019. Cellular Specificity and Inter-cellular Coordination in the Brain Bioenergetic System: Implications for Aging and Neurodegeneration. Frontiers in physiology 10, 1531. [Europe PMC free article] [Abstract] [Google Scholar]
  • Qin YY, Li M, Feng X, Wang J, Cao L, Shen XK, Chen J, Sun M, Sheng R, Han F, Qin ZH, 2017. Combined NADPH and the NOX inhibitor apocynin provides greater anti-inflammatory and neuroprotective effects in a mouse model of stroke. Free radical biology & medicine 104, 333–345. [Abstract] [Google Scholar]
  • Quaegebeur A, Segura I, Schmieder R, Verdegem D, Decimo I, Bifari F, Dresselaers T, Eelen G, Ghosh D, Davidson SM, Schoors S, Broekaert D, Cruys B, Govaerts K, De Legher C, Bouché A, Schoonjans L, Ramer MS, Hung G, Bossaert G, Cleveland DW, Himmelreich U, Voets T, Lemmens R, Bennett CF, Robberecht W, De Bock K, Dewerchin M, Ghesquière B, Fendt SM, Carmeliet P, 2016. Deletion or Inhibition of the Oxygen Sensor PHD1 Protects against Ischemic Stroke via Reprogramming of Neuronal Metabolism. Cell Metab 23, 280–291. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ren X, Engler-Chiurazzi EB, Russell AE, Sarkar SN, Rellick SL, Lewis S, Corbin D, Clapper J, Simpkins JW, 2019. MiR-34a and stroke: Assessment of non-modifiable biological risk factors in cerebral ischemia. Neurochemistry international 127, 73–79. [Europe PMC free article] [Abstract] [Google Scholar]
  • Richards EM, Fiskum G, Rosenthal RE, Hopkins I, McKenna MC, 2007. Hyperoxic reperfusion after global ischemia decreases hippocampal energy metabolism. Stroke 38, 1578–1584. [Europe PMC free article] [Abstract] [Google Scholar]
  • Rink C, Gnyawali S, Stewart R, Teplitsky S, Harris H, Roy S, Sen CK, Khanna S, 2017. Glutamate oxaloacetate transaminase enables anaplerotic refilling of TCA cycle intermediates in stroke-affected brain. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 31, 1709–1718. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ríos JA, Godoy JA, Inestrosa NC, 2018. Wnt3a ligand facilitates autophagy in hippocampal neurons by modulating a novel GSK-3β-AMPK axis. Cell communication and signaling : CCS 16, 15. [Europe PMC free article] [Abstract] [Google Scholar]
  • Roberts BW, Kilgannon JH, Hunter BR, Puskarich MA, Pierce L, Donnino M, Leary M, Kline JA, Jones AE, Shapiro NI, Abella BS, Trzeciak S, 2018. Association Between Early Hyperoxia Exposure After Resuscitation From Cardiac Arrest and Neurological Disability: Prospective Multicenter Protocol-Directed Cohort Study. Circulation 137, 2114–2124. [Europe PMC free article] [Abstract] [Google Scholar]
  • Roh JH, Huang Y, Bero AW, Kasten T, Stewart FR, Bateman RJ, Holtzman DM, 2012. Disruption of the sleep-wake cycle and diurnal fluctuation of β-amyloid in mice with Alzheimer’s disease pathology. Science translational medicine 4, 150ra122. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ross CA, Kronenbuerger M, Duan W, Margolis RL, 2017. Mechanisms underlying neurodegeneration in Huntington disease: applications to novel disease-modifying therapies. Handbook of clinical neurology 144, 15–28. [Abstract] [Google Scholar]
  • Rouach N, Koulakoff A, Abudara V, Willecke K, Giaume C, 2008. Astroglial metabolic networks sustain hippocampal synaptic transmission. Science (New York, N.Y.) 322, 1551–1555. [Abstract] [Google Scholar]
  • Ruppert MC, Greuel A, Tahmasian M, Schwartz F, Stürmer S, Maier F, Hammes J, Tittgemeyer M, Timmermann L, van Eimeren T, Drzezga A, Eggers C, 2020. Network degeneration in Parkinson’s disease: multimodal imaging of nigro-striato-cortical dysfunction. Brain : a journal of neurology 143, 944–959. [Abstract] [Google Scholar]
  • Sandlin ZD, Shou M, Shackman JG, Kennedy RT, 2005. Microfluidic electrophoresis chip coupled to microdialysis for in vivo monitoring of amino acid neurotransmitters. Analytical chemistry 77, 7702–7708. [Abstract] [Google Scholar]
  • Saré RM, Torossian A, Rosenheck M, Huang T, Beebe Smith C, 2019. Quantitative Autoradiographic Method for Determination of Regional Rates of Cerebral Protein Synthesis In Vivo. J Vis Exp. [Abstract] [Google Scholar]
  • Saylor RA, Lunte SM, 2018. PDMS/glass hybrid device with a reusable carbon electrode for on-line monitoring of catecholamines using microdialysis sampling coupled to microchip electrophoresis with electrochemical detection. Electrophoresis 39, 462–469. [Europe PMC free article] [Abstract] [Google Scholar]
  • Schaafsma A, de Jong BM, Bams JL, Haaxma-Reiche H, Pruim J, Zijlstra JG, 2003. Cerebral perfusion and metabolism in resuscitated patients with severe post-hypoxic encephalopathy. Journal of the neurological sciences 210, 23–30. [Abstract] [Google Scholar]
  • Schmidt KC, Smith CB, 2005. Resolution, sensitivity and precision with autoradiography and small animal positron emission tomography: implications for functional brain imaging in animal research. Nucl Med Biol 32, 719–725. [Abstract] [Google Scholar]
  • Schönfeld P, Reiser G, 2017. Brain energy metabolism spurns fatty acids as fuel due to their inherent mitotoxicity and potential capacity to unleash neurodegeneration. Neurochemistry international 109, 68–77. [Abstract] [Google Scholar]
  • Shannon BJ, Vaishnavi SN, Vlassenko AG, Shimony JS, Rutlin J, Raichle ME, 2016. Brain aerobic glycolysis and motor adaptation learning. Proc Natl Acad Sci U S A 113, E3782–3791. [Europe PMC free article] [Abstract] [Google Scholar]
  • Sharma N, Rao SP, Kalivendi SV, 2019. The deglycase activity of DJ-1 mitigates α-synuclein glycation and aggregation in dopaminergic cells: Role of oxidative stress mediated downregulation of DJ-1 in Parkinson’s disease. Free radical biology & medicine 135, 28–37. [Abstract] [Google Scholar]
  • Shen G, Ma Q, 2020. MicroRNAs in the Blood-Brain Barrier in Hypoxic-Ischemic Brain Injury. Curr Neuropharmacol 18, 1180–1186. [Europe PMC free article] [Abstract] [Google Scholar]
  • Shen X, Wang R, Xiong X, Yin Y, Cai Y, Ma Z, Liu N, Zhu ZJ, 2019a. Metabolic reaction network-based recursive metabolite annotation for untargeted metabolomics. Nature communications 10, 1516. [Europe PMC free article] [Abstract] [Google Scholar]
  • Shen Y, Hu F, Min W, 2019b. Raman Imaging of Small Biomolecules. Annual review of biophysics 48, 347–369. [Abstract] [Google Scholar]
  • Shen Y, Kapfhamer D, Minnella AM, Kim JE, Won SJ, Chen Y, Huang Y, Low LH, Massa SM, Swanson RA, 2017. Bioenergetic state regulates innate inflammatory responses through the transcriptional co-repressor CtBP. Nature communications 8, 624. [Europe PMC free article] [Abstract] [Google Scholar]
  • Shivamurthy VK, Tahari AK, Marcus C, Subramaniam RM, 2015. Brain FDG PET and the diagnosis of dementia. AJR Am J Roentgenol 204, W76–85. [Abstract] [Google Scholar]
  • Sivanand S, Viney I, Wellen KE, 2018. Spatiotemporal Control of Acetyl-CoA Metabolism in Chromatin Regulation. Trends Biochem Sci 43, 61–74. [Europe PMC free article] [Abstract] [Google Scholar]
  • Snaebjornsson MT, Schulze A, 2018. Non-canonical functions of enzymes facilitate cross-talk between cell metabolic and regulatory pathways. Experimental & molecular medicine 50, 34. [Europe PMC free article] [Abstract] [Google Scholar]
  • Solís-Maldonado M, Miró MP, Acuña AI, Covarrubias-Pinto A, Loaiza A, Mayorga G, Beltrán FA, Cepeda C, Levine MS, Concha II, Bátiz LF, Carrasco MA, Castro MA, 2018. Altered lactate metabolism in Huntington’s disease is dependent on GLUT3 expression. CNS neuroscience & therapeutics 24, 343–352. [Europe PMC free article] [Abstract] [Google Scholar]
  • Soto M, Cai W, Konishi M, Kahn CR, 2019. Insulin signaling in the hippocampus and amygdala regulates metabolism and neurobehavior. Proc Natl Acad Sci U S A 116, 6379–6384. [Europe PMC free article] [Abstract] [Google Scholar]
  • Soty M, Gautier-Stein A, Rajas F, Mithieux G, 2017. Gut-Brain Glucose Signaling in Energy Homeostasis. Cell Metab 25, 1231–1242. [Abstract] [Google Scholar]
  • Stahon KE, Bastian C, Griffith S, Kidd GJ, Brunet S, Baltan S, 2016. Age-Related Changes in Axonal and Mitochondrial Ultrastructure and Function in White Matter. The Journal of neuroscience : the official journal of the Society for Neuroscience 36, 9990–10001. [Europe PMC free article] [Abstract] [Google Scholar]
  • Stanley M, Macauley SL, Caesar EE, Koscal LJ, Moritz W, Robinson GO, Roh J, Keyser J, Jiang H, Holtzman DM, 2016. The Effects of Peripheral and Central High Insulin on Brain Insulin Signaling and Amyloid-β in Young and Old APP/PS1 Mice. The Journal of neuroscience : the official journal of the Society for Neuroscience 36, 11704–11715. [Europe PMC free article] [Abstract] [Google Scholar]
  • Steinbusch L, Labouèbe G, Thorens B, 2015. Brain glucose sensing in homeostatic and hedonic regulation. Trends in endocrinology and metabolism: TEM 26, 455–466. [Abstract] [Google Scholar]
  • Stratoulias V, Venero JL, Tremblay M, Joseph B, 2019. Microglial subtypes: diversity within the microglial community. The EMBO journal 38, e101997. [Europe PMC free article] [Abstract] [Google Scholar]
  • Su KY, Lee WL, 2020. Fourier Transform Infrared Spectroscopy as a Cancer Screening and Diagnostic Tool: A Review and Prospects. Cancers 12. [Europe PMC free article] [Abstract] [Google Scholar]
  • Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti PJ, Alberini CM, 2011. Astrocyte-neuron lactate transport is required for long-term memory formation. Cell 144, 810–823. [Europe PMC free article] [Abstract] [Google Scholar]
  • Suzuki M, Sasabe J, Miyoshi Y, Kuwasako K, Muto Y, Hamase K, Matsuoka M, Imanishi N, Aiso S, 2015. Glycolytic flux controls D-serine synthesis through glyceraldehyde-3-phosphate dehydrogenase in astrocytes. Proc Natl Acad Sci U S A 112, E2217–2224. [Europe PMC free article] [Abstract] [Google Scholar]
  • Szablewski L, 2017. Glucose Transporters in Brain: In Health and in Alzheimer’s Disease. Journal of Alzheimer’s disease : JAD 55, 1307–1320. [Abstract] [Google Scholar]
  • Tan HE, Sisti AC, Jin H, Vignovich M, Villavicencio M, Tsang KS, Goffer Y, Zuker CS, 2020a. The gut-brain axis mediates sugar preference. Nature 580, 511–516. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tan YL, Yuan Y, Tian L, 2020b. Microglial regional heterogeneity and its role in the brain. Molecular psychiatry 25, 351–367. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tang M, Gao G, Rueda CB, Yu H, Thibodeaux DN, Awano T, Engelstad KM, Sanchez-Quintero MJ, Yang H, Li F, Li H, Su Q, Shetler KE, Jones L, Seo R, McConathy J, Hillman EM, Noebels JL, De Vivo DC, Monani UR, 2017. Brain microvasculature defects and Glut1 deficiency syndrome averted by early repletion of the glucose transporter-1 protein. Nature communications 8, 14152. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tang M, Park SH, De Vivo DC, Monani UR, 2019. Therapeutic strategies for glucose transporter 1 deficiency syndrome. Annals of clinical and translational neurology 6, 1923–1932. [Europe PMC free article] [Abstract] [Google Scholar]
  • Teo E, Ravi S, Barardo D, Kim HS, Fong S, Cazenave-Gassiot A, Tan TY, Ching J, Kovalik JP, Wenk MR, Gunawan R, Moore PK, Halliwell B, Tolwinski N, Gruber J, 2019. Metabolic stress is a primary pathogenic event in transgenic Caenorhabditis elegans expressing pan-neuronal human amyloid beta. eLife 8. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ter Horst KW, Lammers NM, Trinko R, Opland DM, Figee M, Ackermans MT, Booij J, van den Munckhof P, Schuurman PR, Fliers E, Denys D, DiLeone RJ, la Fleur SE, Serlie MJ, 2018. Striatal dopamine regulates systemic glucose metabolism in humans and mice. Science translational medicine 10. [Abstract] [Google Scholar]
  • Terada T, Obi T, Bunai T, Matsudaira T, Yoshikawa E, Ando I, Futatsubashi M, Tsukada H, Ouchi Y, 2020. In vivo mitochondrial and glycolytic impairments in patients with Alzheimer disease. Neurology 94, e1592–e1604. [Abstract] [Google Scholar]
  • Tomasi DG, Shokri-Kojori E, Wiers CE, Kim SW, Demiral SB, Cabrera EA, Lindgren E, Miller G, Wang GJ, Volkow ND, 2017. Dynamic brain glucose metabolism identifies anti-correlated cortical-cerebellar networks at rest. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 37, 3659–3670. [Europe PMC free article] [Abstract] [Google Scholar]
  • Tu D, Gao Y, Yang R, Guan T, Hong JS, Gao HM, 2019. The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration. Journal of neuroinflammation 16, 255. [Europe PMC free article] [Abstract] [Google Scholar]
  • Ulland TK, Song WM, Huang SC, Ulrich JD, Sergushichev A, Beatty WL, Loboda AA, Zhou Y, Cairns NJ, Kambal A, Loginicheva E, Gilfillan S, Cella M, Virgin HW, Unanue ER, Wang Y, Artyomov MN, Holtzman DM, Colonna M, 2017. TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s Disease. Cell 170, 649–663.e613. [Europe PMC free article] [Abstract] [Google Scholar]
  • Vaishnavi SN, Vlassenko AG, Rundle MM, Snyder AZ, Mintun MA, Raichle ME, 2010. Regional aerobic glycolysis in the human brain. Proc Natl Acad Sci U S A 107, 17757–17762. [Europe PMC free article] [Abstract] [Google Scholar]
  • Venkat P, Cui C, Chopp M, Zacharek A, Wang F, Landschoot-Ward J, Shen Y, Chen J, 2019. MiR-126 Mediates Brain Endothelial Cell Exosome Treatment-Induced Neurorestorative Effects After Stroke in Type 2 Diabetes Mellitus Mice. Stroke 50, 2865–2874. [Europe PMC free article] [Abstract] [Google Scholar]
  • Vicente Miranda H, El-Agnaf OM, Outeiro TF, 2016. Glycation in Parkinson’s disease and Alzheimer’s disease. Movement disorders : official journal of the Movement Disorder Society 31, 782–790. [Abstract] [Google Scholar]
  • Vicente Miranda H, Szego É M, Oliveira LMA, Breda C, Darendelioglu E, de Oliveira RM, Ferreira DG, Gomes MA, Rott R, Oliveira M, Munari F, Enguita FJ, Simões T, Rodrigues EF, Heinrich M, Martins IC, Zamolo I, Riess O, Cordeiro C, Ponces-Freire A, Lashuel HA, Santos NC, Lopes LV, Xiang W, Jovin TM, Penque D, Engelender S, Zweckstetter M, Klucken J, Giorgini F, Quintas A, Outeiro TF, 2017. Glycation potentiates α-synuclein-associated neurodegeneration in synucleinopathies. Brain : a journal of neurology 140, 1399–1419. [Abstract] [Google Scholar]
  • Vlassenko AG, Raichle ME, 2015. Brain aerobic glycolysis functions and Alzheimer’s disease. Clin Transl Imaging 3, 27–37. [Europe PMC free article] [Abstract] [Google Scholar]
  • Volkow ND, Tomasi D, Wang GJ, Telang F, Fowler JS, Goldstein RZ, Klein N, Wong C, Swanson JM, Shumay E, 2013. Association between dopamine D4 receptor polymorphism and age related changes in brain glucose metabolism. PLoS One 8, e63492. [Europe PMC free article] [Abstract] [Google Scholar]
  • Walberer M, Backes H, Rueger MA, Neumaier B, Endepols H, Hoehn M, Fink GR, Schroeter M, Graf R, 2012. Potential of early [(18)F]-2-fluoro-2-deoxy-D-glucose positron emission tomography for identifying hypoperfusion and predicting fate of tissue in a rat embolic stroke model. Stroke 43, 193–198. [Abstract] [Google Scholar]
  • Wang AC, Jensen EH, Rexach JE, Vinters HV, Hsieh-Wilson LC, 2016a. Loss of O-GlcNAc glycosylation in forebrain excitatory neurons induces neurodegeneration. Proc Natl Acad Sci U S A 113, 15120–15125. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang B, Aw TY, Stokes KY, 2016b. The protection conferred against ischemia-reperfusion injury in the diabetic brain by N-acetylcysteine is associated with decreased dicarbonyl stress. Free radical biology & medicine 96, 89–98. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang B, Yee Aw T, Stokes KY, 2018. N-acetylcysteine attenuates systemic platelet activation and cerebral vessel thrombosis in diabetes. Redox biology 14, 218–228. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang D, Pascual JM, Yang H, Engelstad K, Jhung S, Sun RP, De Vivo DC, 2005. Glut-1 deficiency syndrome: clinical, genetic, and therapeutic aspects. Annals of neurology 57, 111–118. [Abstract] [Google Scholar]
  • Wang J, Cui Y, Yu Z, Wang W, Cheng X, Ji W, Guo S, Zhou Q, Wu N, Chen Y, Chen Y, Song X, Jiang H, Wang Y, Lan Y, Zhou B, Mao L, Li J, Yang H, Guo W, Yang X, 2019a. Brain Endothelial Cells Maintain Lactate Homeostasis and Control Adult Hippocampal Neurogenesis. Cell Stem Cell 25, 754–767.e759. [Abstract] [Google Scholar]
  • Wang L, Pavlou S, Du X, Bhuckory M, Xu H, Chen M, 2019b. Glucose transporter 1 critically controls microglial activation through facilitating glycolysis. Molecular neurodegeneration 14, 2. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wang Z, Mascarenhas C, Jia X, 2020. Positron Emission Tomography After Ischemic Brain Injury: Current Challenges and Future Developments. Translational stroke research 11, 628–642. [Europe PMC free article] [Abstract] [Google Scholar]
  • Watanabe S, Kato H, Shimosegawa E, Hatazawa J, 2016. Genetic and Environmental Influences on Regional Brain Uptake of 18F-FDG: A PET Study on Monozygotic and Dizygotic Twins. Journal of nuclear medicine : official publication, Society of Nuclear Medicine 57, 392–397. [Abstract] [Google Scholar]
  • Wheatley EG, Albarran E, White CW 3rd, Bieri G, Sanchez-Diaz C, Pratt K, Snethlage CE, Ding JB, Villeda SA, 2019. Neuronal O-GlcNAcylation Improves Cognitive Function in the Aged Mouse Brain. Current biology : CB 29, 3359–3369.e3354. [Europe PMC free article] [Abstract] [Google Scholar]
  • Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D, Sengillo JD, Hillman S, Kong P, Nelson AR, Sullivan JS, Zhao Z, Meiselman HJ, Wendy RB, Soto J, Abel ED, Makshanoff J, Zuniga E, De Vivo DC, Zlokovic BV, 2015. GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction and degeneration. Nat Neurosci 18, 521–530. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wishart DS, 2019. Metabolomics for Investigating Physiological and Pathophysiological Processes. Physiological reviews 99, 1819–1875. [Abstract] [Google Scholar]
  • Wishart DS, Feunang YD, Marcu A, Guo AC, Liang K, Vázquez-Fresno R, Sajed T, Johnson D, Li C, Karu N, Sayeeda Z, Lo E, Assempour N, Berjanskii M, Singhal S, Arndt D, Liang Y, Badran H, Grant J, Serra-Cayuela A, Liu Y, Mandal R, Neveu V, Pon A, Knox C, Wilson M, Manach C, Scalbert A, 2018. HMDB 4.0: the human metabolome database for 2018. Nucleic acids research 46, D608–d617. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wood TE, Barry J, Yang Z, Cepeda C, Levine MS, Gray M, 2019. Mutant huntingtin reduction in astrocytes slows disease progression in the BACHD conditional Huntington’s disease mouse model. Human molecular genetics 28, 487–500. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu L, Huang C, Emery BP, Sedgwick AC, Bull SD, He XP, Tian H, Yoon J, Sessler JL, James TD, 2020. Förster resonance energy transfer (FRET)-based small-molecule sensors and imaging agents. Chemical Society reviews 49, 5110–5139. [Europe PMC free article] [Abstract] [Google Scholar]
  • Wu L, Zhang X, Zhao L, 2018. Human ApoE Isoforms Differentially Modulate Brain Glucose and Ketone Body Metabolism: Implications for Alzheimer’s Disease Risk Reduction and Early Intervention. The Journal of neuroscience : the official journal of the Society for Neuroscience 38, 6665–6681. [Europe PMC free article] [Abstract] [Google Scholar]
  • Xu J, Khoury N, Jackson CW, Escobar I, Stegelmann SD, Dave KR, Perez-Pinzon MA, 2020. Ischemic Neuroprotectant PKCε Restores Mitochondrial Glutamate Oxaloacetate Transaminase in the Neuronal NADH Shuttle after Ischemic Injury. Translational stroke research 11, 418–432. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yamamoto Y, Koma H, Yagami T, 2015. Hydrogen peroxide mediated the neurotoxicity of an antibody against plasmalemmal neuronspecific enolase in primary cortical neurons. Neurotoxicology 49, 86–93. [Abstract] [Google Scholar]
  • Yan X, Hu Y, Wang B, Wang S, Zhang X, 2020. Metabolic Dysregulation Contributes to the Progression of Alzheimer’s Disease. Frontiers in neuroscience 14, 530219. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yang TT, Lin C, Hsu CT, Wang TF, Ke FY, Kuo YM, 2013. Differential distribution and activation of microglia in the brain of male C57BL/6J mice. Brain structure & function 218, 1051–1060. [Abstract] [Google Scholar]
  • Yao J, Irwin RW, Zhao L, Nilsen J, Hamilton RT, Brinton RD, 2009. Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 106, 14670–14675. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yin X, Feng L, Ma D, Yin P, Wang X, Hou S, Hao Y, Zhang J, Xin M, Feng J, 2018. Roles of astrocytic connexin-43, hemichannels, and gap junctions in oxygen-glucose deprivation/reperfusion injury induced neuroinflammation and the possible regulatory mechanisms of salvianolic acid B and carbenoxolone. Journal of neuroinflammation 15, 97. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yu Y, Herman P, Rothman DL, Agarwal D, Hyder F, 2018. Evaluating the gray and white matter energy budgets of human brain function. Journal of cerebral blood flow and metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism 38, 1339–1353. [Europe PMC free article] [Abstract] [Google Scholar]
  • Yuan H, Frank JE, Hong Y, An H, Eldeniz C, Nie J, Bunevicius A, Shen D, Lin W, 2013. Spatiotemporal uptake characteristics of [18]F-2-fluoro-2-deoxy-D-glucose in a rat middle cerebral artery occlusion model. Stroke 44, 2292–2299. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zampieri M, Sekar K, Zamboni N, Sauer U, 2017. Frontiers of high-throughput metabolomics. Current opinion in chemical biology 36, 15–23. [Abstract] [Google Scholar]
  • Zeisel A, Munoz-Manchado AB, Codeluppi S, Lonnerberg P, La Manno G, Jureus A, Marques S, Munguba H, He L, Betsholtz C, Rolny C, Castelo-Branco G, Hjerling-Leffler J, Linnarsson S, 2015. Brain structure. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science (New York, N.Y.) 347, 1138–1142. [Abstract] [Google Scholar]
  • Zhang L, Shi L, Shen Y, Miao Y, Wei M, Qian N, Liu Y, Min W, 2019a. Spectral tracing of deuterium for imaging glucose metabolism. Nature biomedical engineering 3, 402–413. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhang S, An Q, Wang T, Gao S, Zhou G, 2018a. Autophagy- and MMP-2/9-mediated Reduction and Redistribution of ZO-1 Contribute to Hyperglycemia-increased Blood-Brain Barrier Permeability During Early Reperfusion in Stroke. Neuroscience 377, 126–137. [Abstract] [Google Scholar]
  • Zhang S, Song X-Y, Xia C-Y, Ai Q-D, Chen J, Chu S-F, He W-B, Chen N-H, 2016. Effects of cerebral glucose levels in infarct areas on stroke injury mediated by blood glucose changes. RSC Advances 6, 93815–93825. [Google Scholar]
  • Zhang S, Zuo W, Guo XF, He WB, Chen NH, 2014. Cerebral glucose transporter: the possible therapeutic target for ischemic stroke. Neurochemistry international 70, 22–29. [Abstract] [Google Scholar]
  • Zhang X, Hu Y, Yang X, Tang Y, Han S, Kang A, Deng H, Chi Y, Zhu D, Lu Y, 2019b. FÖrster resonance energy transfer (FRET)-based biosensors for biological applications. Biosensors & bioelectronics 138, 111314. [Abstract] [Google Scholar]
  • Zhang Z, Chen W, Zhao Y, Yang Y, 2018b. Spatiotemporal Imaging of Cellular Energy Metabolism with Genetically-Encoded Fluorescent Sensors in Brain. Neuroscience bulletin 34, 875–886. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhen G, Endres C, Jia X, 2013. Cerebral glucose metabolism with therapeutic hypothermia after cardiac arrest. In: The XXVIth International Symposium on Cerebral Blood Flow, Metabolism and Function & XIth International Conference on Quantification of Brain Function with PET: Shanghai, China. pp. 733–734. [Google Scholar]
  • Zhou P, Guan T, Jiang Z, Namaka M, Huang QJ, Kong JM, 2018a. Monocarboxylate transporter 1 and the vulnerability of oligodendrocyte lineage cells to metabolic stresses. CNS neuroscience & therapeutics 24, 126–134. [Europe PMC free article] [Abstract] [Google Scholar]
  • Zhou Q, Zheng H, Chen J, Li C, Du Y, Xia H, Gao H, 2018b. Metabolic fate of glucose in the brain of APP/PS1 transgenic mice at 10 months of age: a (13)C NMR metabolomic study. Metabolic brain disease 33, 1661–1668. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/107546560
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/107546560

Article citations


Go to all (35) article citations

Funding 


Funders who supported this work.

NHLBI (3)

NHLBI NIH HHS (1)

NINDS (3)

NINDS NIH HHS (2)

National Institutes of Health (3)