Next Article in Journal
Muscarinic Receptor Antagonism and TRPM3 Activation as Stimulators of Mitochondrial Function and Axonal Repair in Diabetic Sensorimotor Polyneuropathy
Previous Article in Journal
Quercetin as an Anti-Diabetic Agent in Rodents—Is It Worth Testing in Humans?
Previous Article in Special Issue
Transporter-Mediated Interactions Between Uremic Toxins and Drugs: A Hidden Driver of Toxicity in Chronic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Human Glucose Transporters in Health and Selected Neurodegenerative Diseases

by
Leszek Szablewski
Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
Int. J. Mol. Sci. 2025, 26(15), 7392; https://doi.org/10.3390/ijms26157392 (registering DOI)
Submission received: 30 June 2025 / Revised: 25 July 2025 / Accepted: 28 July 2025 / Published: 31 July 2025
(This article belongs to the Special Issue Transporters in Health and Disease)

Abstract

Glucose is the main source of energy and the source of carbon for the biosynthesis of several molecules, such as neurotransmitters, for most mammalian cells. Therefore, the transport of glucose into cells is very important. There are described three distinct families of glucose transporters: facilitative glucose transporters (GLUTs), sodium-dependent glucose cotransporters (SGLTs), and a uniporter, the SWEET protein. Impaired function and/or expression of these transporters due to, for example, mutations in their genes, may cause severe diseases. Associations with the impaired function of glucose transporters have been described in the case of neurodegenerative diseases (NDs) such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, GLUT1-deficiency syndrome, stroke, and traumatic brain injury. Changes in the presence of glucose transporters may be a cause of NDs, and they may be the effect of NDs. On the other hand, in many cases of neurodegenerative diseases, changes in the expression of glucose transporters may be a targeted therapy in the treatment of patients with these diseases.

1. Introduction

In most mammalian cells, glucose plays an important role as a source of energy for cellular functions and as a source of carbon for the biosynthesis of several biomolecules. For the appropriate delivery of glucose to the parenchyma, it must overcome four barriers. These barriers are associated with the absorption of glucose by the small intestine into the blood; its hepatic storage and release to the circulation; traversing the vascular endothelium for delivering sugar to parenchymal cells, for example, in the case of the blood–brain barrier (BBB); and reabsorption of glucose by the kidney back into the blood. The presence of these four cellular barriers means that glucose must be delivered from one compartment to another, from extracellular space to intracellular space and from intracellular compartment to extracellular compartment [1]. However, although the transport of glucose between components is necessary, it is difficult. This is due to the properties of glucose and the cell membrane. Glucose is a polar, hydrophilic molecule, whereas the cell membrane is made up of a lipid bilayer that is lipophilic and hydrophobic. Therefore, the cell membrane is impermeable to several cations (K+, Na+, Ca2+), anions (Cl, HCO3), hydrophilic molecules, as well as glucose. Therefore, to undergo diffusion through the lipid bilayer, glucose needs specific carrier proteins.
The transport of glucose across the cell membrane is caused by so-called “glucose transporters”, which transport glucose and several other substrates. In mammals, including humans, glucose is transported by a specific saturable transport process involving members of three different classes of glucose transporters: facilitative glucose transporters (GLUT proteins, SLC2 genes), sodium-dependent glucose symporters (secondary active transport, SGLT proteins, SLC5 genes), and a new class of glucose uniporters (SWEET proteins, SLC50 genes) [2], each with different kinetic properties. Most cells express a variety of glucose transporters. The expression of glucose transporters in different tissues depends on the specific metabolic requirements. Disturbances in the function and/or expression of these transport proteins cause not only neurodegenerative diseases [3,4,5,6,7], but also several other human diseases, such as cancers [8], renal and heart diseases [9,10], and several others.

2. Characteristics of Human Glucose Transporters

Glucose transporters belong to the major facilitator superfamily (MFS). In the MFS, the sugar porter family is the largest group. These porters are detected in bacteria, archaea, and eukaryotes [11,12]. MFS contains 74 families of membrane transporters, each of which transports specific substrates. To date, more than 10,000 members have been sequenced. Proteins belonging to the MFS are ubiquitously expressed. They are also highly conserved from bacteria to humans. Glucose transporters exhibit different substrate specificities, kinetic properties, and tissue expression profiles.

2.1. The Human GLUT Transporters

In 1948, LeFevre suggested that for the transport of glucose across the lipid bilayer, a specific component within the cellular plasma membrane is necessary [13]. The human GLUT1 protein was purified in 1977 from erythrocytes [14], and the first GLUT protein to be isolated was cloned from HepG2 cell lines in 1985. The genetic and protein sequence of GLUT1 was performed in 1985 [15].

Structure of GLUT Proteins

All GLUT proteins contain 12 hydrophobic membranes spanning α-helical transmembrane (TM) domains, which are connected by the hydrophilic loop between TM6 and TM7 of the protein [16,17,18]. These glucose transporters contain a short intracellular N-terminal segment and a single site for glycosylation on the exofacial end. This site may be located in the large loop between TM1 and TM2 (first extracellular loop) or between TM9 and TM10 (fifth extracellular loop) [19]. Sequences of the GLUT family are better conserved in the putative TM regions, while in the loops between the TM domains and the C- and N-termini, sequences are more divergent. Among members of the GLUT proteins, the sequences are 14–63% identical and 30–79% conserved [20]. In all members of GLUTs are several highly conserved sequences, such as PMY in TM4, QQLSGIN in TM7, GPGPIP/TW in TM10, PESPRY/FLL in loop 6, GRR in loop 8, and VPETKG in the terminal tail. There are also 18 conserved glycine residues. Performed observations revealed that glycine residues in helices 1, 2, 4, 5, 7, 8, and 10 play an important role in the structure of these helices [20].
GLUT proteins are encoded by the solute-linked carrier family 2, subfamily A gene family [19,21,22]. In humans, 14 members of the GLUT family have been identified that are categorized into three classes according to sequence similarity [16,17,19,20,23,24]. Class I GLUTs include GLUT1–GLUT4 and GLUT14. These glucose transporters are 48–63% identical in humans. Specific residues for Class I GLUTs are the following: Glutamine in α-helical structure 5 (QL motif corresponding with Q161 in GLUT1) plays a role in the identification of glucose [25]. The STS (serine–threonine–serine) motif in extracellular loop 7 is involved in the conformational change during the transport process [26]. The highly conserved motif GPXXXP in TM10 is important for the binding of the two inhibitors cytochalasin B and forskolin, which affect the transport of glucose [27]. A QLS motif in TM7 is present in GLUT proteins that transport glucose but not fructose. This motif is associated with the selectivity of glucose and fructose [28]. Class II GLUTs comprise GLUT5, GLUT7, GLUT9, and GLUT11. These transporters are 36–40% identical. The lack of tryptophan residue following the conserved GPXXXP motif in TM10, corresponding with tryptophan 388 in GLUT1, and the absence of the QLS motif, are characteristic sequences for Class II proteins [28]. Class III GLUTs comprise GLUT6, GLUT8, GLUT10, GLUT12, and GLUT13 (HMIT). These transporters are only 19–41% identical. Many of the motifs detected in the other classes are conserved, such as PESPR in TM6, GRR in loops 2 and 8, PETKGR in TM12, arginine and glutamate residues in loops 4 and 10, and tryptophan residue after the GPXXXP motif in TM10 [21]. GLUTs of Class III do not contain the QLS motif, which is unique to the Class I GLUTs [22] (Figure 1). For more details, see [7].
All GLUT proteins are facilitative transporters, except HMIT, encoded by the SLC2A13 gene, which is an H+/myo-inositol symporter [29]. In dependence on GLUT proteins, these transporters may play different roles (Table 1).
Pseudogenes
There are described four pseudogenes of the SLC2A family [17,19]:
  • SLC2A3P1 (alias GLUT6 or GLUT3 pseudogene) located on chromosome 5q35. It is a retroposon of SLC2A3.
  • SLC2A3P2 (alias GLUT3 pseudogene 2) located on chromosome 1p31.3. It is a retroposon of SLC2A3.
  • SLC2A3P4 (alias GLUT3 pseudogene 4) located on chromosome 8q21.3. It is a retroposon of SLC2A3.
  • SLC2AXP1 is located on chromosome 2q11.2. It contains internal stop sequences.

2.2. The Human Sodium-Dependent Glucose Cotransporters

The role of sodium in glucose transport was proposed in 1958 by Riklis and Quastel [115]. In 1962, Crane presented a hypothesis on sodium/glucose cotransport [116]. Crane also introduced the term “secondary active transport” (indirect active transport), because the hydrolysis of ATP is indirectly associated with the transport of glucose via the electrochemical gradient. According to the suggestion of Crane, the sodium-dependent glucose cotransporter contains two binding sites: one for glucose and one for sodium [117].

Structure of Sodium-Dependent Glucose Cotransporters

The sodium-dependent glucose cotransporters, also known as sodium/glucose cotransporters, are energy-dependent and function as active transporters. They transport glucose against the glucose concentration gradient, and they are dependent on sodium cotransport. One molecule of glucose is transported with one or two sodium ions. Sodium ions are transported down a sodium concentration gradient [5,118]. The sodium/glucose cotransporters belong to the gene family (SLC5A), the SGLTs or sodium/substrate symporters family (SSSF), that contains more than 450 members [119,120,121]. SLC5A genes encode for proteins of 580–718 amino acid residues with a predicted mass of 60–80 kDa. There are differences between the structures of the genes. In eight genes, the coding sequences are in 14–15 exons (SLC5A1, SLC5A2, SLC5A4SLC5A6, and SLC5A9SLC5A11). The coding sequence for SLC5A3 and for SLC5A7 are present in exons 1 and 8, respectively. In the SLC5A3 and SLC5A9SLC5A11 genes, there is alternative splicing. Performed investigations suggest that all of these symporters contain 14 TM α-helices (TMHs). The sodium/iodide symporter (NIS) and SMCT1 lack TMH14 [122]. Both the N- and C-termini are located on the extracellular side of the plasma membrane [2] (Figure 2). There are variable numbers for N-linked glycosylation. However, SGLTs are highly glycosylated membrane proteins; glycosylation is not required for their function. In humans, there are 12 genes that encode these symporters, which are expressed in different tissues. All of these genes code sodium/glucose cotransporters, except for SGLT3, encoded by the SLC5A4 gene, which plays a role as a glucose sensor [123]. Six genes of this family encode cotransporters for solute, such as glucose, myo-inositol, and iodide; one is a Na+/Cl choline cotransporter and another is a glucose-activated ion channel [121]. In the dependence on sodium-dependent glucose cotransporter proteins, these transporters may play different roles (Table 2).

2.3. The Human SWEET Transporter

SWEET proteins are involved in the transport of mono- and disaccharides across the vacuolar and plasma membranes. These transporters have seven predicted TM domains with two internal triple-helix bundles (THBs) connected by an inversion linker helix (TM4), creating the translocation pathway (3 + 1 + 3 configuration). This class of transporters was first identified in Arabidopsis [131]. In A. thaliana, up to two dozen SWEETs have been identified, whereas animals usually have only one SWEET, except Caenorhabditis elegans, in which seven genes encoding SWEETs were found [42]. Human SWEET (RAG1AP1) is encoded by the SLC50A1 gene located on chromosome 1q22 [156]. It contains 221 amino acids with a molecular weight of 25 kDa. SWEET1 does not stimulate glucose uptake, but instead mediates a weak efflux. Expression of human SWEET1 in HEK293T cells reveals its localization in the Golgi complex. Its higher expression was detected in the oviduct, epididymis, and intestine. In the mouse mammary gland, its expression was induced during lactation. It is suggested that in the basolateral membrane of enterocytes, SWEET may be associated with the exit of glucose from the cell into the blood in patients with Fanconi Bickel Syndrome [157].

3. Association of Human Glucose Transporters with Neurodegenerative Diseases

The brain consumes approximately 20% of the energy intake of the body. Nutrients and small molecules are exchanged by the brain and blood at the blood–brain barrier (BBB). The brain contains various transport systems for acquiring substances such as amino acids, lactate, ketone bodies, lipids, and cofactors for functions of neurons. Glucose plays an important role in the production of energy and as a source of carbon for biogenesis in neurons. Glucose is taken up from blood by glucose transporters, which are expressed in several cell types. As an alternative source of energy for neurons, lactate may be used [158,159], as well as ketones, by the capillaries of the brain and by astrocytes [160,161]. As mentioned earlier, 10 GLUTs and 10 sodium-dependent symporters are expressed in the CNS [3]. The expression of the SWEET1 protein in the human brain was not detected. The human brain expresses many transporters, but only a small number have been characterized [162]. However, in the human brain various isoforms of GLUTs and sodium-dependent cotransporters exist; a major role in fueling energy requirements is played by GLUT1 and GLUT3 [70].

3.1. Expression of Glucose Transporters in Alzheimer’s Disease

Alzheimer’s disease (AD) is a progressive neurodegenerative disease associated with severe and progressive impairment of cognitive function [163], memory loss, and language problems [164]. It may also include non-cognitive dysfunction (executive) that may be followed by behavioral disorders such as agitation, aggressiveness, and depression [163,164,165]. There are over 100 types of dementia, but the most well-known dementia is AD, which accounts for 50–75% of all cases of dementia [166]. Many observations have revealed that diabetes mellitus increases susceptibility to AD [167]; therefore, it has been suggested to use a new term for AD: type 3 diabetes mellitus (T3DM) [168,169,170,171]. Clinically, there are two types of AD [172]. About 90–95% of AD patients are aged ≥ 65 years and are diagnosed with “late onset” or “sporadic AD” (sAD) [173]. In 5–10% of patients, the disease is caused by a rare genetic mutation. These patients are diagnosed with “early onset” or “familial AD” (fAD). In fAD, symptoms of disease are detected in younger persons—those in their thirties, forties, or fifties [173,174,175,176]. Symptoms detected in fAD are caused by mutations in three genes: amyloid precursor protein (APP), presenilin-1 (PS-1), and presenilin-2 (PS-2). It is suggested also there are other mutations, but they are yet unknown [172]. In the case of sAD, the genetics are more complex [177]. Obtained results in performed observations showed that the epsilon 4 allele of the Apolipoprotein E 4 (ApoE-ε4) gene is a significant risk factor for the development of sAD. Two copies of the ApoE-ε4 gene increase the risk of AD 12-fold, and one copy increases the risk of AD 4-fold [178]. The mentioned gene is carried by 50–60% of individuals. It is suggested that other factors also influence the risk of developing AD [179].
The development of AD is associated with abnormal protein aggregation [180,181]. The proteinopathy symptoms observed in AD are associated with two types of misfolded protein in the brain: the neurofibrillary tangles (NFTs), containing hyperphosphorylated tau, and cellular plaques, with aggregated amyloid β (Aβ) peptides [182,183]. Impairment of synaptic functions begins the pathogenesis of AD. This pathology is caused by the accumulation of Aβ produced from the APP. APP is a transmembrane protein expressed in many tissues, including the CNS. The main sites of its concentration are the synapses of neurons, where it plays a role as a cell surface receptor. It is involved in the regulation of synapse formation, neural plasticity, antimicrobial activity, and export of iron [167,184]. APP is processed in different pathways. The first pathway, in which 90% of APP is processed, is the non-amyloidogenic (non-plaque-forming) pathway. The remaining 10% of APP is processed by the amyloidogenic pathway. In this process is involved γ-secretase. In fAD, its role is changed due to mutation in the APP, PS1, and PS2 genes, as well as the inheritance ApoE-ε4, resulting in the increased production of Aβ42, a neurotic isoform. Aβ peptides aggregate into oligomers and create fibrils in amyloid plaques, which block the signaling pathway and connection, causing cell death. Insulin plays an important role in the metabolism of APP. It regulates the balance between anabolism and catabolism of Aβ [167,184,185,186,187]. The tau protein is a neuronal microtubule-associated protein expressed in axons [188]. In the human brain are detected six isoforms of this protein caused by alternative splicing [189]. Tau is involved in the assembly and stability of microtubules, which are involved in processes such as morphogenesis, cell division, and intracellular trafficking. In neurons, tau is associated with synapses and nuclei [190]. The activity of tau is regulated by its phosphorylation. This protein contains more than 85 phosphorylated or phosphorylable sites, about 80 Ser/Thr and 5 Tyr phosphorylation sites [191,192]. The neuronal homeostasis is associated with kinases and phosphatases, which regulate the balance between tau phosphorylation and dephosphorylation. An imbalance between these enzymes may cause hyperphosphorylation of tau. If tau is phosphorylated at more than 30 sites, it may be involved in the development of Alzheimer’s diseases and NFT [188]. In the brains of patients with AD, tau protein is three times more phosphorylated than tau phosphorylation in brains of healthy subjects [193]. For more details, see [167].
In addition to Aβ and tau, AD is also a metabolic disease associated with reduced cerebral glucose metabolism, brain insulin resistance, and age-induced mitochondrial dysfunction [194,195]. It is suggested that glucose hypometabolism is a prominent feature of the brains of AD patients [196]. In sAD, the synthesis of ATP from the metabolism of glucose is decreased by 50%. Also observed is a tendency to decrease the synthesis of ATP throughout the progression of the disease [173,197]. Decreased uptake of glucose and glucose hypometabolism in specific brain areas are associated with extensive synaptic loss in advanced stages of AD [129,198,199]. It is suggested that hypometabolism in AD may be due to decreased glucose transport in the brain by glucose transporters and/or impaired brain metabolism [129]. Decreased glucose transport in the human AD-affected brain is observed in the most metabolically active brain regions such as the cortex, hippocampus, and cerebral microvessels [200,201,202]. Similar results were obtained also in animal studies [129]. The postmortem investigations of samples of the brains of AD patients revealed decreased levels of GLUT1 and GLUT3 proteins, mainly in the cerebral cortex, with significant loss of GLUT3 [203,204,205]. As mentioned earlier, GLUT1 and GLUT3 play a major role in glucose uptake by the brain; however, other GLUT proteins expressed in the brain also play a role in glucose transport in the brain. Decreased levels of GLUT1 and GLUT3 were observed in the hippocampal and cortical regions of AD patients [206] and in the frontal cortex [207]. Results obtained in another study also showed decreased levels of GLUT1 and GLUT3 in the brains of AD patients. Based on these results, the authors conclude that decreased levels of GLUT3 are related to the severity of AD pathology and the expression of AD symptoms [208]. Decreased levels of GLUT1 and GLUT3 were confirmed in animal models of AD [129,206,209]. Obtained results revealed also that decreased levels of GLUT1 and GLUT3 were correlated with decreased O-GlcNAcylation and abnormal hyperphosphorylation of tau [207,210]. Based on these results, it is suggested that deficiency of GLUT1 and GLUT3 in AD may impair brain glucose uptake/metabolism, causing neurodegeneration by the downregulation of O-GlcNAcylation and hyperphosphorylation of tau [207,210,211]. Decreased levels of the transcription factor hypoxia-inducible factor 1 (HIF-1) also were found [207]. HIF-1 is a major regulator of GLUT1 and GLUT3, and it is important in normal glucose metabolism [212]. Based on the obtained results, the authors suggest that a decrease in the mentioned glucose transporters may be due to downregulation of HIF-1 in the AD brain [207]. There are also other suggested mechanisms. For example, it is suggested that glucose hypometabolism caused by decreased levels of GLUT1 and GLUT3 contributes to abnormal tau hyperphosphorylation and/or neurofibrillary degeneration by hexosamine biosynthesis pathway downregulation [213]. Another mechanism associated with decreased levels of GLUT3, it is postulated that the promoter of human GLUT3 contains three potential cAMP response element (CRE)-like dements: CRE1, CRE2, and CRE3. Increased expression of GLUT3 may be associated with the overexpression of CRE-binding protein (CREB) or the activation of cAMP-dependent protein kinase. In the AD brain, decreased full-length CRB was detected, whereas truncation of CREB was increased. In the human brain, truncation is involved in the activation of calpain 1, which proteolyzes CREB, generating a truncated CREB. Truncated CREB has less activity to stimulate the expression of GLUT3. Performed investigations revealed in the human brain a positive correlation of GLUT3 with full-length CREB and a negative correlation with the activation of calpain 1. Overactivation of calpain 1 due to calcium and the overloaded proteolysis of CREB resulted in decreased expression of GLUT3. The effect is impaired glucose uptake and metabolism in the AD-affected brain [214]. Correlation between decreased levels of AD and Aβ deposition was confirmed in animal studies [215]. Also investigated was the expression of other glucose transporters in the AD brain. Obtained results revealed markedly increased (~1.5-fold) levels of GLUT2, whereas no change was detected in the level of GLUT4 [207]. Researchers observed that the increased level of GLUT2 in the brain is correlated to a similar extent with the activation of astrocytes. Therefore, it is suggested that the increased level of GLUT2 is correlated with the activation of astrocytes in AD [207]. In the frontal cortex of AD patients was detected a significantly high level of GLUT12 [216] and in the brain of mouse models of AD [217]. Unfortunately, there is insufficient information on this issue. Of note, in a Drosophila model of Aβ toxicity, increased glucose transport into neurons rescues Aβ toxicity [196].

3.2. Expression of Glucose Transporters in Parkinson’s Disease

Parkinson’s disease (PD), also known as paralysis agitans, is a common neurological disease in middle-aged and elderly people. It is the second most common neurodegenerative disease. In PD patients are observed resting tremor, bradykinesia, muscle rigidity, and postural instability [218]. The primary pathological change in PD is the degeneration and death of dopaminergic (DO) neurons, causing their loss in the substantia nigra (SN) pars compacta [219], and decreasing activity of the dopamine transmitter system in the nigrostriatal area. Closely associated with the viability of dopaminergic neurons are Lewy bodies, which are formed by the aggregation of α-synuclein (α-Syn). The cause of neuronal degeneration remains unclear; however, there are suggested to be multiple factors rather than a single cause, such as oxidative stress, mitochondrial dysfunction, neuroinflammation, and protein misfolding [220]. Most PD cases are sporadic (sPD), caused by a combination of genetic and environmental risk factors. Only 5% to 10% of PD may be due to a single pathogenic mutation (single gene) [221], and only about 15% are caused by heritable familial mutations [222]. There are suggestions that PD may be associated with changes in glucose metabolism in the frontal lobe and caudatum putamen [223]. Disorders of glucose metabolism resulting in energy metabolism failure may also be the cause of PD [224,225,226]. Unfortunately, the literature on the glucose transporters in PD is very scarce. Human induced pluripotent stem cells (hiPSCs) derived from fibroblasts of PD patients, and their differentiation product, neural precursor cells (hNPCs), were used in observations to expand the etiology of sPD. These cells were used as a human cellular model of sPD. Obtained results revealed that the levels of GLUT1 mRNA and GLUT3 mRNA were not altered in sPD hNPCs. Also, GLUT1 and GLUT3 proteins were not affected in these cells. The expression of the SLC2A4 gene was significantly decreased, but the SLC2A2 and SLC2A4 genes were weakly expressed. Therefore, their role in glucose metabolism is questionable [227]. Experiments performed on a mouse model of PD revealed that density and localization of Glut1 were unaltered in these animals. The authors suggest that, although in PD there are metabolic disturbances, the level of Glut1 is not changed following dopaminergic neurodegeneration [228]. On the other hand, decreased levels of Glut1 in the striatum of a PD model in mice were reported [229,230]. Associations between glucose metabolism and Parkinson’s disease need further investigation.

3.3. Expression of Glucose Transporters in Huntington’s Disease

Huntington’s disease (HD) is a progressive, neurodegenerative disease caused by a CAG expansion in exon 1 of the huntingtin (HTT) gene, resulting in the formation of a prolonged polyglutamine (polyQ) tract in the N-terminal region, which leads to the production of a toxic mutant huntingtin protein (mHTT) [231]. PolyQ stretches of more than 36 residues produce a gain of function in mHTT, affecting the healthy function of cellular machinery, resulting in neurotoxicity and detrimental cell lethality [232]. In healthy individuals, the CAG region of HTT contains 11–35 trinucleotide repeats [6]. A fundamental pathogenesis of HD is abnormal accumulation of mHTT in the neurons. The misfolded and aggregated toxic protein propagates and spreads in a prion-like fashion [233]. The length of the CAG repeat inversely correlates with age of onset in patients with Huntington’s disease [234]. Mutations in the HTT gene cause the loss of critically important brain neurons such as medium spiny neurons in the striatum, a subcritical part of the forebrain. Disturbances of energy homeostasis in AD are associated with impaired glucose transporters, abnormal functions of glycolytic enzymes, etc. [235]. It was found that intracellular glucose levels in mHTT cells are between 0.8 and 1.5 mM. This is a 2.5-fold decrease in intracellular glucose levels in these cells, which may indicate a change in glucose metabolism. This suggestion was confirmed in a further study [236]. In HD patients, decreased uptake of striatal glucose was observed [237] in the caudate, putamen, and thalamic regions [238]. Reduced uptake of glucose was positively correlated with disease severity [238]. A loss in uptake of glucose restricted to the frontal and inferior parietal cortex was observed in newly diagnosed patients. A global loss of glucose uptake was detected in patients with symptoms of HD lasting longer than 5 years. In symptomatic patients and carriers of asymptomatic mHTT was observed a decreased uptake of glucose in the caudate nucleus and putamen that was correlated with the number of CAG repeat lengths [239]. Postmortem investigations of brain samples of patients with grade 3 HD revealed a decreased expression of GLUT1 and GLUT3 by three- and four-fold, respectively. On the other hand, similar observations in patients with earlier stages of HD (grade 1) revealed no changed expression of these GLUT proteins [240]. Results obtained from experiments performed on animal models of HD revealed a decreased uptake of glucose in the Huntington’s disease cortex caused by the loss of glucose transporters’ expression [241]. Results obtained in observations performed on animal models of HD may suggest that impaired glucose metabolism in patients with HD may be associated with impaired expression of the insulin gene [242]. In HD patients is observed not only decreased uptake of glucose, but also, increased uptake of lactate. This observation suggests that HD causes decreased levels of energy [243]. Mitochondrial metabolism disorders are observed in patients with HD and in animal models of this disease [244]. For example, there was observed in the postmortem brain samples of HD patients a loss of pyruvate dehydrogenase (PDH) and oxyglutarate dehydrogenase (OGDH), important enzymes in the Krebs cycle [245]. The N-terminal of mHTT interacts with mitochondrial membranes, resulting in the inhibition of respiratory complex II. Impaired mitochondrial electron transport causes the overproduction of reactive oxygen species (ROS) and decreased production of ATP [246]. Damage due to oxidative stress decreases the expression of GLUT3, causing stimulation of lactate uptake and inhibition of glucose uptake [247]. As mentioned above, in HD patients, the onset of HD is correlated with the copy number of the SLC2A3 gene [234]. It was also observed that three copies of the SLC2A3 gene may have a neuroprotective effect, and between the copy number of this gene and the level of GLUT3 expression is the linear correlation. These results, as well as those obtained in the Drosophila HD model, may suggest that an increased number of SLC2A3 genes ameliorates HD pathologies. Because the progression of HD and the uptake of glucose by neurons mainly depends on GLUT3, it may have therapeutic ramifications. Observations performed on the Drosophila HD model revealed that an increased dosage of Glut1 also ameliorates HD phenotypes in these flies [234]. The beneficial role of increased levels of glucose transporters in HD pathology was described also by other authors [235,248]. However, disturbances in the function and expression of glucose transporters may contribute to the pathophysiology observed in HD; these transporters, in particular GLUT1 and GLUT3, may be involved with targeted therapy in the treatment of HD. It should also be investigated whether there is a correlation between HD and dysfunction of other glucose transporters. This problem needs further investigation.

3.4. Expression of Glucose Transporter in GLUT1 Deficiency Syndrome

GLUT1 deficiency syndrome (GLUT1-DS) is a rare genetic metabolic disease inherited in an autosomal recessive manner. It was first described as an early-onset childhood epileptic neuropathy [249]. Description of additional cases associated with mutations in the SLC2A1 gene caused the phenotype spectrum to be expanded by epileptic encephalopathy with different types of seizures, movement disorders, and paroxysmal events of non-epileptic origin [250,251]. GLUT1-DS accounts for around 1% of idiopathic generalized epilepsies and approximately 10% of early-onset absence of epilepsies [252]. This neurological disease is associated with decreased expression and/or function of GLUT1 in the brain [249,251,253,254]. To date, there are more than 20 mutations described. In most cases, GLUT1-DS is due to heterozygous single-nucleotide variants (SNVs) in the SLC2A1 gene. These mutations cause complete or severe impairment of functionality and/or expression of GLUT1 in the brain [255]. The SNVs cause amino acid exchanges, exon deletions, and frame shifts, influencing the regulation of transcription or translation [256,257,258,259]. Clinical neurological symptoms depend on the type of mutation. Missense mutations are predominant in mild and moderate clinical categories [260,261]. Insertion causes the most severe phenotype of GLUT1-DS [262]. Heterozygous forms of the SLC2A1 gene, causing partial deficiency of GLUT1, are associated with intractable infantile epilepsy [262]. In these babies, the head is normal size, but they display mild chronic encephalopathy with infrequent seizures, spasticity, and ataxia. The lighter forms of GLUT1-DS are associated with hypoglycemia, clumsiness, speech delay, frequent perplexity, and loss of consciousness [263]. Results obtained in animal studies revealed that the recessive homozygous form is lethal [264,265]. This syndrome affects all ages. In patients with GLUT1-DS, other clinical manifestations are observed, such as myoclonic limb jerking with alternating staring and eye-rolling, unresponsiveness, head bobbing, generalized seizures, ataxia, spasticity dystonia, intermittent ataxia, periodic confusion, periodic weakness, and recurrent headaches. In these patients may be detected also specific, atypical manifestations of GLUT1-DS, such as intermittent ataxia, dystonia, migraine, etc. [250,251]. It is suggested that the pathologies observed in GLUT1-DS are due to an insufficient supply of glucose for neuronal activities [162]. For therapy of GLUT1-DS, ketonic or high-fat diets are prescribed to patients. Ketonic diets improve various, but not all, symptoms of GLUT1-DS. For example, this diet controls the seizures and other paroxysmal activities, but is less effective on cognitive functions [262]. In some patients, modified Atkins diets have been introduced [266]. Results obtained on animal models of GLUT1-DS are described in detail in [5].

3.5. Expression of Glucose Transporters in Stroke

Stroke is a devastating neurological disturbance that is the second cause of death worldwide. Performed observations revealed that more than 80% of stroke events are ischemic stroke, caused by restricted blood flow to a brain part. The most cases are due to occlusion as an effect of thrombosis, embolism, and/or arteriosclerosis [5]. Performed observations and experiments on animals revealed changes in the expression of glucose transporters and/or their mRNAs. In a rat model of median cerebral artery occlusion (MCAO), one hour after short-term MCAO, upregulation of GLUT1 mRNA was observed in the ipsilateral and contralateral brain cortex. Upregulation of GLUT1 in the investigated rats was detected in microvessels, astrocytes, and distinct populations of neurons. Upregulation of GLUT1 mRNA was detected in glial cells of the penumbra, but not in neurons, one day after MCAO [267]. It was found also that in rats with the median cerebral artery (MCA) occluded for 3 h, upregulation of GLUT1 protein and GLUT1 mRNA was observed at 12 h of reperfusion in an ipsilateral cortical area outside the core infract region [268]. Upregulation of GLUT1 protein and GLUT1 mRNA as an effect of ischemia was described also by other researchers [269,270]. Based on the obtained results, it is suggested that in the regulation of the expression of GLUT1/GLUT1 during stroke, heat shock protein (HSP) 70, hypoxia-inducible factor (HIF) 1, and insulin-like growth factor-1 (IGF-1) are involved [271,272]. During stroke, GLUT3 is also upregulated; however, its regulation differs in comparison with GLUT1. No changes in levels of GLUT3 mRNA were observed in the ipsilateral or contralateral forebrain throughout the first couple of hours after short-term MCAO in experimental rats [267]. The slightly increased expression of GLUT3 mRNA in neurons of the ipsilateral cortex was observed one day after MCAO. At this time, the expression of GLUT1 mRNA subsided [268]. Associations of different expressions of GLUT3 protein and GLUT3 mRNA with ischemia were described also by other authors [270]. The regulation of GLUT3 expression in response to energy depletion during ischemia was investigated on cultivated neurons and astrocytes. Obtained results revealed that during the onset of ischemia, GLUT3 is rapidly inserted into the plasma membrane, which may protect neuronal cells against death in response to ischemia [273]. It also was observed that during ischemia, in cultivated porcine brain cells, Sglt1-mediated uptake of glucose across the BBM was increased [5]. For more details, see [5].

3.6. Expression of Glucose Transporters in Traumatic Brain Injury

Traumatic brain injury (TBI) comprises “… focal and diffuse brain damage caused by different types of violation and brain concussion” [5]. It may cause mental and physical disabilities. Following brain injury, patients have been observed to have changes in cerebral D-glucose uptake and expression of cerebral glucose transporters. Decreased uptake of D-glucose was found in patients during the acute phase of TBI. This observation may suggest decreased metabolism (hypometabolism) [274]. In patients after severe brain trauma, cerebral glucose uptake was increased after 1 week [275]. In brain samples obtained from patients about 8 h after TBI upregulation of 55 kDa GLUT1 expression was observed, predominantly located in the endothelial cells of brain capillaries [276]. Experiments performed on animal models revealed that in the brains of rodents, no effects on the expression of 45 kDa GLUT1 were observed up to 2 days after traumatic injury. The increased expression was observed in the case of 55 kDa GLUT1 6 h and 2 days after trauma [277]. Based on these results and others, there is suggested to be posttranslational upregulation of GLUT1/GLUT1 after TBI [5]. It is suggested that in the regulation of GLUT1/GLUT1 expression, HIF-α is involved [5,278]. In the case of GLUT3, it was found in observations of animals that distinct upregulation of this glucose transporter was detected in the cerebral cortex and cerebellum 4 h after TBI [279]. Upregulation of GLUT3 was still detected two days after TBI [5]. Only very limited observations are available in the case of the expression and function of SGLT1/Sglt1 in the brain during TBI. Obtained results from animal studies suggest that upregulation of SGLT1/Sglt1 during TBI aggravates secondary tissue damage and clinical outcomes [5].

4. Therapeutic Strategies in Neurodegenerative Diseases

Epidemiological studies showed a potential link between diabetes mellitus and neurodegenerative diseases (NDs) such as AD, PD, HD, and amyotrophic lateral sclerosis (ALS) [280]. In these diseases are observed pathological features similar to those observed in DM, such as mitochondrial dysfunction, insulin resistance, impaired insulin signaling, oxidative stress, etc. There are also pathologies specific to particular NDs. Diabetic patients have an increased risk of developing NDs. However, in the case of association between DM and PD, there are different results. For example, case-control studies suggest a negative association between DM and PD [281,282]. On the other hand, meta-analyses of prospective cohort studies suggest an increased risk of PD development in diabetic patients [283,284]. Similarity in the pathological features detected in the mentioned diseases means that the therapeutic strategies for NDs are based on anti-diabetic drugs [285,286,287]. However, the exact mechanisms remain not fully known; these drugs may play a novel role in managing neurodegenerative diseases (Table 3). Most of these drugs show a beneficial effect on NDs. On the other hand, it must be added that some of these drugs only alleviate certain symptoms rather than providing a cure.

4.1. Incretins

Glucagon-like peptide-1 (GLP-1) is an endogenous multifunctional peptide secreted by intestinal L cells in response to food intake. It enhances insulin secretion by pancreatic β cells and regulates blood glucose, food intake, and body weight [324,325]. GLP-1 is also synthesized in the brain by hypothalamic neurons within the nucleus of solitary tract, intermediate reticular nucleus, pyriform cortex, and olfactory bulb [300]. GLP-1 receptors (GLP-1Rs) are mainly expressed in the intestine and the α and β cells of the pancreatic islets. GLP-1Rs are expressed also in several CNS regions, such as the hippocampus, cortex, hypothalamus, and other areas, which are associated with AD and PD [326,327]. GLP-1R triggers cyclic adenosine-monophosphate (cAMP)-dependent PI3K/AKT and PKA/MAPK pathway activation. These pathways modulate mitochondrial function, glucose homeostasis, and apoptosis, and they are associated with processes such as inflammation, satiety, memory, synaptic plasticity, neurogenesis, and stress response [328]. To date, most GLP-1R agonists, such as liraglutide, semaglutide, lixisenatide, albiglutide, and dulaglutide, are used in therapy in diabetic patients. Results obtained in investigations of GLP-1R performed through tests in vivo and in vitro in cell lines, animal models of NDs, and ND patients revealed neuroprotective properties [288,289,290,291,292,293,294,295,296,297]. Exendin-4 is a peptide agonist of GLP-1R that has been detected in the saliva of the Gila monster (Heloderma suspectum). The synthetic version of exendin-4 revealed a 53% amino acid sequence homology with human GLP-1 [329]. Treatment by exenatide halts the cognitive decline in Aβ-induced rat and mouse models of AD [297,298]. It improves degeneration of dopaminergic neurons and motor function in mouse and rat models of PD [289,299]. Exendin-4 decreases mitochondrial toxicity in an Aβ-induced AD mouse model [290], alleviates cognitive dysfunction, prevents amyloid-β accumulation, and protects mitochondrial function in 5xFAD mice [297]. Liraglutide ameliorates learning and memory impairment in various rat and mouse models of AD. It attenuates mutant huntingtin-induced neurotoxicity in human neuronal cells [292,293,294,295]. GLP-1R agonists may promote neuroprotection against ischemic stroke [296].
Glucose-dependent insulin-releasing polypeptide (GIP), also known as gastric inhibitory, is a gut hormone involved in insulin secretion in response to food intake. Its receptors (GIPRs) are expressed in the neurons of the olfactory bulb, hippocampus, cerebellum, cerebral cortex, amygdala, substantia nigra, thalamus, hypothalamus, and brainstem. These receptors play an important role in neurotransmission, neurogenesis, and neuroplasticity [330,331]. Several experiments revealed that GIPR agonists have neuroprotective properties in mouse models of AD and PD [300,301,302]. These agonists reduce the activation of chronic inflammatory response in the brain, oxidative stress, synapse loss, amyloid plaque burden, and DNA damage, and they reduce lipid peroxidation and α-synuclein levels, protecting dopaminergic neurons in the substantia nigra [303,304]. Novel GLP-1R and GIP receptor agonists are being developed as novel therapy. These dual-receptor agonists of GIP and GLP-1 showed promising effect in animal models of AD and PD [300,301]. They activate both GIP and GLP-1 receptors. Obtained results showed that dual-receptor agonists rescue or prevent spatial learning and memory dysfunction [305,306,307], reduce amyloid plaques and phosphorylated tau [305], upregulate p-PI3K and p-Akt growth factor kinases, and prevent overactivation of p-GSK3β [307]. Other investigations observed in animal models of PD that dual agonists can improve motor function, protect dopaminergic neurons, and rescue glial cell line-derived neurotrophic factor levels [332]. Recently, triple-receptor agonists have been developed, activating GLP-1, GIP, and glucagon (Gcg) receptors (GLP-1/GIP/Gcg receptors). In mouse models of AD, the observed effects of triple-receptor agonists were anti-apoptotic effects, reduced Aβ deposition and hyperphosphorylated hippocampal tau, protection from loss of synapses, and reduced inflammatory and oxidative stress [308,309,310]. In PD mice, the triple-receptor agonist protected against dopaminergic neuronal death, decreased α-synuclein accumulation, and prevented nigrostriatal neurodegeneration [311,312].

4.2. Dipeptidyl Peptidase-4 Inhibitors

Dipeptidyl peptidase-4 (DPP-4) inhibitors influence the enzymes causing the inactivation of incretin, such as GLP-1. The inhibition of DPP-4 increases levels of GLP-1 in the blood. In animal models of AD, linagliptin improves cognitive function and decreases tau phosphorylation and Aβ aggregation [313,314]. Linagliptin has a neuroprotective effect on human nerve cells, because it protects neurons from the effect of Aβ on mitochondrial damage, oxidative stress, and impairment of the insulin signaling pathway. Unfortunately, the positive effects of DPP-4 inhibitors in animal models of NDs were obtained with higher doses than the ones used in human treatments. Based on these results, it is difficult to extrapolate the results to humans. On the other hand, regarding data in humans, in individuals who receive DPP-4 inhibitors, the incidence of PD is significantly decreased [315,316].

4.3. Thiazolidinediones

Thiazolidinediones such as pioglitazone, rosiglitazone, and troglitazone increase insulin sensitivity by binding to the peroxisome proliferator-activated receptor gamma (PPARγ). Experimental and clinical studies revealed the neuroprotective role of this class of drugs. These drugs exert neuroprotective effects in animal models of AD and PD, significantly improving behavior and motor responses [317,318,319]. Rosiglitazone improves spatial memory, tested in the Morris water maze, and increases removal of Aβ deposits in AD mice [333]. There are conflicting results from clinical trials regarding the effect of rosiglitazone in the treatment of dementia. Some results indicate that rosiglitazone improves cognitive performance in AD patients [320], but other results do not confirm this effect of the drug [321].

4.4. Biguanides

Metformin, a drug that belongs in the drug class biguanides, is the main first-line medication for the treatment of type 2 diabetes. In animal models of PD, it decreases α-synuclein expression. In AD, metformin has a beneficial effect on cognitive function [322] by promoting neurogenesis and preventing amyloid deposition and tau phosphorylation [334,335]. The role of metformin in neurodegenerative diseases is not clear. It was found that metformin may stimulate the production of Aβ [336,337,338]. In diabetic patients treated with metformin is found an increased risk of cognitive impairment [337]. A meta-analysis that studied the effect of metformin on neurodegeneration revealed that this drug has no effect on NDs [323]. But in the case of PD, a significantly higher incidence of PD was observed in patients treated with metformin in comparison with non-metformin patients [323]. The associations between metformin and PD need further investigation [339]. Metformin also regulates the activity of astrocytes in PD [340].

5. Conclusions and Perspectives

Glucose transporters play an important role in human health. They are involved in transport not only of glucose, but also several other substances into and out of cells. Therefore, their expression is exactly controlled. Disturbances in their expression and/or function are associated with different diseases, including cancers and metabolic diseases. Changes in the expression of glucose transporters are observed also in neurodegenerative diseases such as Alzheimer’s disease, Huntington’s disease, stroke, etc. Therapy of these diseases is based on anti-diabetic drugs. The beneficial effects of these drugs, as well as potentially negative effects, have been described. Unfortunately, there is a lack of investigation of the associations between glucose transporters and used therapeutic strategies in NDs. Maybe this association will be helpful in the treatment of ND patients. Significant clinical data are required. Because our understanding of these associations remains limited, further research is necessary. Research is underway to investigate the mechanism underlying the neuroprotective effects of anti-diabetic drugs to create effective strategies for the prevention and treatment of NDs. The deficiency of insulin and its impaired signaling contributes to dysregulation in brain function. Activation of PI3K signaling can boost Insulin Degrading Enzyme, showing promise against NDs. To validate the use of anti-diabetic drugs such as those mentioned above for the treatment of NDs, significant clinical data are required. Unfortunately, our understanding of these therapies remains limited. Therapies should include measures that increase the insulin supply and enhance INSR responsiveness and insulin-targeted gene expression. Animal models of NDs, especially mice and rats, used in preclinical studies have provided substantial insights into NDs’ pathogenesis. There are differences between the results obtained in these animal studies and clinical presentations. Therefore, the use of non-human primate models in these investigations may enhance the translational relevance of preclinical findings and validate novel therapeutic strategies. It is also important to enhance collaborations between experimental researchers and clinical practitioners to ensure the effective translation of obtained results into specialized treatment and prevention methods.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Klipp, A.; De Bock, K.; Bilan, P.J.; Richter, E.A. Transcellular barriers to glucose delivery in the body. Annu. Rev. Physiol. 2024, 86, 149–173. [Google Scholar] [CrossRef]
  2. Wright, E.M. Glucose transport families SLC5 and SLC50. Mol. Asp. Med. 2013, 34, 183–196. [Google Scholar] [CrossRef]
  3. Szablewski, L. Glucose transporters in brain: In health and Alzheimer’s disease. J. Alzheimer’s Dis. 2017, 55, 1307–1320. [Google Scholar] [CrossRef]
  4. Głuchowska, K.; Pliszka, M.; Szablewski, L. Expression of glucose transporters in human neurodegenerative diseases. Biochem. Biophys. Res. Commun. 2021, 540, 8–15. [Google Scholar] [CrossRef]
  5. Kaepsel, H. Glucose transporters in brain in health and disease. Pflügers Archiv. Eur. J. Physiol. 2020, 472, 1299–1343. [Google Scholar] [CrossRef]
  6. Morea, V.; Bidollari, E.; Colotti, G.; Fiorillo, A.; Rosati, J.; De Filippis, L.; Squitieri, F.; Ilan, A. Glucose transportation in the brain and its impairment in Huntington disease: One more shade of the energetic metabolism failure. Amino Acids 2017, 49, 1147–1157. [Google Scholar] [CrossRef] [PubMed]
  7. Szablewski, L. Human Glucose Transporters in Health and Diseases; Cambridge Scholar Publishing: Newcastle upon Tyne, UK, 2019; ISBN 978-1-5275-3558-9. [Google Scholar]
  8. Szablewski, L. Expression of glucose transporters in cancers. Biochim. Biophys. Acta Rev. Canc. 2013, 1835, 164–169. [Google Scholar] [CrossRef] [PubMed]
  9. Szablewski, L. Distribution of glucose transporters in renal diseases. J. Biomed. Sci. 2017, 24, 64. [Google Scholar] [CrossRef]
  10. Szablewski, L. Glucose transporters in healthy heart and in cardiac disease. Int. J. Cardiol. 2017, 230, 70–75. [Google Scholar] [CrossRef] [PubMed]
  11. Pao, S.S.; Paulsen, I.T.; Saier, M.H., Jr. Major facilitator superfamily. Microbiol. Mol. Biol. Rev. 1998, 62, 1–34. [Google Scholar] [CrossRef]
  12. Saier, M.H., Jr. Families of transmembrane sugar transport protein. Mol. Microbiol. 2000, 35, 699–710. [Google Scholar] [CrossRef]
  13. LeFevre, P.G. Evidence of active transfer of certain non-electrolytes across the human red cell membrane. J. Gen. Physiol. 1948, 31, 505–527. [Google Scholar] [CrossRef]
  14. Kasahara, M.; Hinkle, P.C. Reconstitution and purification of the D-glucose transporter from human erythrocytes. J. Biol. Chem. 1997, 252, 7384–7390. [Google Scholar] [CrossRef]
  15. Mueckler, M.; Caruso, C.; Baldwin, S.A.; Panico, I.; Blench, I.; Morris, H.R. Sequence and structure of a human glucose transporter. Science 1985, 229, 941–945. [Google Scholar] [CrossRef]
  16. Joost, H.G.; Bell, G.I.; Best, J.D.; Birnbaum, M.J.; Charron, M.J.; Chen, Y.T.; Doege, H.; James, D.E.; Lodish, H.F.; Moley, K.H.; et al. Nomenclature of the GLUT/SLC2A family of sugar/polyol transport facilitators. Am. J. Physiol. Endocrinol. Metab. 2002, 282, E974–E976. [Google Scholar] [CrossRef] [PubMed]
  17. Joost, H.-G.; Thorens, B. The extend GLUT-family of sugar-polyol transport facilitators: Nomenclature, sequence characteristics, and potential function of its novel members. Mol. Membr. Biol. 2001, 18, 247–256. [Google Scholar] [CrossRef]
  18. Uldry, M.; Thorens, B. The SLC2 family of facilitative hexose and polyol transporters. Pflügers Arch. 2004, 447, 480–489. [Google Scholar] [CrossRef] [PubMed]
  19. Mueckler, M.; Thorens, B. The SLC2 (GLUT) family of membrane transporters. Mol. Asp. Med. 2013, 34, 121–138. [Google Scholar] [CrossRef]
  20. Zhao, F.-Q.; Keating, A.F. Functional properties and genomic of glucose transporters. Curr. Genom. 2007, 8, 113–128. [Google Scholar] [CrossRef]
  21. Long, W.; Cheeseman, C.I. Structure and functional insight into the GLUT family of membrane transporters. Cell Health Cytosk. 2015, 7, 167–183. [Google Scholar]
  22. Thorens, B.; Mueckler, M. Glucose transporters in the 21st Century. AJP Endocrinol. Metab. 2010, 298, E141–E145. [Google Scholar] [CrossRef] [PubMed]
  23. Scheepers, A.; Joost, H.G.; Schürmann, A. The glucose transporter families SGLT and GLUT: Molecular basis of normal and aberrant function. J. Parenter. Enter. Nutr. 2004, 28, 364–371. [Google Scholar] [CrossRef] [PubMed]
  24. Manolescu, A.R.; Witkowska, K.; Kinnaird, A.; Cessford, T.; Cheeseman, C. Facilitated hexose transporters: New perspectives on form and function. Physiology 2007, 22, 234–240. [Google Scholar] [CrossRef]
  25. Mueckler, M.; Weng, W.; Kruse, M. Glutamine 161 of GLUT1 glucose transporter is critical for transport activity and exofacial ligand binding. J. Biol. Chem. 1994, 269, 20533–20538. [Google Scholar] [CrossRef]
  26. Doege, H.; Schürmann, A.; Ohnimus, H.; Monser, V.; Holman, G.D.; Joost, H.G. Serine 294 and threonine 259 in the exofacial loop domain between helices 7 and 8 of glucose transporters (GLUT) are involved in the conformational alterations during the transport process. Biochem. J. 1998, 329, 289–297. [Google Scholar] [CrossRef]
  27. Garcia, J.C.; Strube, M.; Leingang, K.; Keller, K.; Mueckler, M.M. Amino acids substitutions at tryptophan 388 and tryptophan 412 of the Hep62 (Glut1) glucose transporter inhibit transport activity and targeting to the plasma membrane in Xenopus oocytes. J. Biol. Chem. 1992, 267, 7770–7776. [Google Scholar] [CrossRef]
  28. Seater, M.J.; De La Ru, S.A.; Porter, L.M.; Gould, G.W. QLS motif in transmembrane helix VII of the glucose transporter family interacts with C-1 position of D-glucose and is involved in substrate selection at the exofacial binding site. Biochemistry 1998, 37, 1322–1326. [Google Scholar] [CrossRef]
  29. Uldry, M.; Ibberson, M.; Horsiberger, J.-D.; Rieder, B.M.; Thorens, B. Identification of a mammalian H+-myoinositol symporter expressed predominantly in the brain. EMBO J. 2001, 20, 4467–4477. [Google Scholar] [CrossRef]
  30. Longo, N.; Elsas, L.J. Human glucose transporters. Adv. Pediatr. 1998, 45, 293–313. [Google Scholar] [CrossRef]
  31. Hruz, P.W.; Mueckler, M.M. Structural analysis of the GLUT1 facilitative glucose transporter (Review). Mol. Membr. Biol. 2001, 18, 183–193. [Google Scholar] [CrossRef] [PubMed]
  32. Maher, F.; Davies-Hill, T.M.; Lysko, P.G.; Henneberry, R.C.; Simpson, I.A. Expression of two glucose transporters GLUT1 and GLUT3 in cultured cerebral neurons: Evidence for neuron specific expression of GLUT3. Mol. Cell. Neurosci. 1991, 2, 351–360. [Google Scholar] [CrossRef]
  33. Maher, F.; Vannucci, S.J.; Simpson, I.A. Glucose transporter protein in brain. FASEB J. 1994, 8, 1003–1011. [Google Scholar] [CrossRef] [PubMed]
  34. Olson, A.L.; Pessin, J.E. Structure, function and regulation of the facilitative glucose transporter gene family. Annu. Rev. Nutr. 1996, 16, 235–256. [Google Scholar] [CrossRef]
  35. Pardridge, W.M.; Boado, R.J.; Farrel, C.R. Brain-type glucose transporter (GLUT-1) is selectively located to the blood-brain barrier. Studies with the quantitative Western blotting and in situ hybridization. J. Biol. Chem. 1990, 265, 18035–18040. [Google Scholar] [CrossRef]
  36. Kumagai, A.K.; Dwyer, K.J.; Pardridge, W.M. Differential glycosylation of the GLUT1 glucose transporter in brain capillaries and choroid plexus. Biochim. Biophys. Acta 1994, 1193, 24–30. [Google Scholar] [CrossRef]
  37. Takata, K.; Kasahara, M.; Ezaki, O.; Hirano, H. Erythrocyte/HepG2-type glucose transporter is concentrated in cells of blood-tissue barriers. Biochem. Biophys. Res. Commun. 1990, 173, 67–73. [Google Scholar] [CrossRef]
  38. Gould, G.W.; Bell, G.J. Facilitative glucose transporters: An expanding family. Trends Biochem. Sci. 1990, 15, 18–22. [Google Scholar] [CrossRef] [PubMed]
  39. Gould, G.W.; Holman, G.D. The glucose transporter family: Structure, function and tissue-specific expression. Biochem. J. 1993, 295, 329–341. [Google Scholar] [CrossRef]
  40. Korgun, E.T.; Demir, R.; Sedlmayer, P.; Desoue, G.; Arikan, G.; Puerstner, P.; Haeusler, M.; Dohr, G.; Skofitsch, G.; Hahn, T. Physiological leukocytosis during pregnancy with changes in glucose transporter expression of maternal peripheral blood granulocytes and monocytes. Am. J. Reprod. Immunol. 2002, 48, 110–116. [Google Scholar] [CrossRef]
  41. Sagun, K.C.; Cárcano, J.M.; Golde, D.W. Vitamin C enter mitochondria via facilitative glucose transporter 1 (Glut 1) and confers mitochondrial protection against oxidative injury. FASEB J. 2005, 19, 1657–1667. [Google Scholar]
  42. Deng, D.; Yan, N. GLUT, SGLT, and SWEET: Structural and mechanistic investigations of the glucose transporters. Protein Sci. 2016, 25, 546–558. [Google Scholar] [CrossRef]
  43. Jin, Q.; Agrawal, L.; VanHorn-Ali, Z.; Alkhatib, G. Infection of CD4+ lymphocytes by the glucose transporter GLUT1: Evidence using antibodies specific to the receptor’s large extracellular domain. Virology 2006, 349, 184–196. [Google Scholar] [CrossRef]
  44. Jones, K.S.; Fugo, K.; Petrow-Sadowski, C.; Huang, Y.; Bertolette, D.C.; Lisinski, I.; Cushman, S.W.; Jacobson, S.; Ruscetti, F.W. Human T-cell leukemia virus type 1 (HTLV-1) and HTLV-2 use different receptor complex to enter T cells. J. Virol. 2006, 80, 8291–8302. [Google Scholar] [CrossRef]
  45. Manel, N.; Kint, S.; Kim, F.J.; Taylor, N.; Sitbon, M.; Battini, J.J. GLUT-1 est le recepteur des retrovirus humains HTL. Méd. Sci. 2004, 20, 277–279. [Google Scholar]
  46. Wieczorke, R.; Dlugai, S.; Krampe, S.; Boles, E. Characterization of mammalian GLUT glucose transporters in a heterozygous yeast expression system. Cell Physiol. Biochem. 2003, 13, 123–134. [Google Scholar] [CrossRef]
  47. Uldry, M.; Ibberson, M.; Hosokawa, M.; Thorens, B. GLUT2 is a high affinity glucosamine transporter. FEBS Lett. 2002, 524, 199–203. [Google Scholar] [CrossRef] [PubMed]
  48. Fukumoto, H.; Seino, H.; Seino, Y.; Eddy, R.L.; Fukushima, Y.; Byers, M.G.; Shows, T.B.; Bell, G.I. Sequence, tissue distribution, and chromosomal localization of mRNA encoding a human glucose transporter-like protein. Proc. Natl. Acad. Sci. USA 1988, 85, 5435–5438. [Google Scholar] [CrossRef]
  49. Thorens, B.; Sarkar, H.K.; Lodish, H.F. Cloning and functional expression in bacteria of a novel glucose transporter present in liver, intestine, kidney, and beta-pancreatic islet cells. Cell 1988, 55, 281–290. [Google Scholar] [CrossRef] [PubMed]
  50. Kayano, T.; Fukumoto, H.; Eddy, R.I.; Fan, Y.S.; Byers, M.G.; Shows, T.B.; Bell, G.I. Evidence for a family of human glucose transporter-like proteins. Sequence and gene localization of a protein expressed in fetal skeletal muscle and other tissues. J. Biol. Chem. 1988, 263, 15245–15248. [Google Scholar] [CrossRef] [PubMed]
  51. Thorens, B.; Cheng, Z.Q.; Brown, D.; Lodish, H.F. Liver glucose transporter: A basolateral protein in hepatocytes and intestine and kidney cells. Am. J. Physiol. 1990, 259, C279–C285. [Google Scholar] [CrossRef]
  52. Thorens, B.; Lodish, H.F.; Brown, D. Differential localization of two glucose transporter isoforms in rat kidney. Am. J. Physiol. 1990, 259, C286–C294. [Google Scholar] [CrossRef]
  53. Dwyer, D.S.; Vannucci, S.J.; Simpson, I.A. Expression, regulation, and functional role of glucose transporters (GLUTs) in brain. Int. Rev. Neurobiol. 2002, 51, 159–188. [Google Scholar]
  54. Garcia, M.L.; Millan, C.; Balmaceda-Aguilera, C.; Castro, T.; Pastor, P.; Montecinos, H. Hypothalamic ependymal-glial cells express the glucose transporter 2 a protein involved in glucose sensing. J. Neurochem. 2003, 86, 709–724. [Google Scholar] [CrossRef] [PubMed]
  55. Young, J.K.; McKenzie, J.C. GLUT2 immunoreactivity in Gomori-positive astrocytes of hypothalamus. J. Histochem. Cytochem. 2004, 52, 1519–1524. [Google Scholar] [CrossRef]
  56. Leloup, C.; Arluison, M.; Lepetit, N.; Cartier, N.; Marfaing-Jallat, P.; Ferré, P.; Pénicaud, L. Glucose transporter 2 (GLUT2) expression in specific brain nuclei. Brain Res. 1994, 638, 221–225. [Google Scholar] [CrossRef] [PubMed]
  57. Piroli, G.G.; Grillo, C.A.; Reznikov, L.R.; Reagan, L.P. Expression and functional activities of glucose transporters in central nervous system. In Handbook of Neurochemistry and Molecular Neurobiology, 3rd ed.; Springer: Berlin/Heidelberg, Germany, 2007; pp. 388–404. [Google Scholar]
  58. Arluison, M.; Quinon, M.; Nguyen, P.; Thorens, B.; Leloup, C.; Penicaud, L. Distribution and anatomical localization of the glucose transporter 2 (GLUT2) in the adult brain—An immunohistochemical study. J. Chem. Neuroanat. 2004, 28, 117–136. [Google Scholar] [CrossRef]
  59. Marty, N.; Dallaporta, M.; Thorens, B. Brain glucose sensing, counterregulation, and energy homeostasis. Physiology 2007, 22, 241–251. [Google Scholar] [CrossRef]
  60. Conville, C.A.; Seater, M.J.; Jess, T.J.; Gould, G.W.; Thomas, H.M. Kinetic analysis of the liver-type (GLUT2) and brain type (GLUT3) glucose transporters in Xenopus oocytes; substrate specificities and effect of transport inhibitors. Biochem. J. 1993, 290, 701–706. [Google Scholar] [CrossRef]
  61. Jihnson, J.H.; Newgard, C.B.; Milburn, J.L.; Lodish, H.F.; Thorens, B. The high Km glucose transporter of islet Langerhans is functionally similar to the low affinity transporter of liver and has an identical primary sequence. J. Biol. Chem. 1990, 265, 6548–6551. [Google Scholar] [CrossRef]
  62. Nagamatsu, S.; Kornhauser, J.M.; Burant, C.F.; Seino, S.; Mayo, K.E.; Bell, G.J. Glucose transporter expression in brain. cDNA sequence of mouse GLUT3, the brain facilitative glucose transporter isoform, and identification of sites of expression by in-situ hybridization. J. Biol. Chem. 1992, 267, 467–472. [Google Scholar] [CrossRef] [PubMed]
  63. Leino, R.L.; Gerhart, D.Z.; van Bueren, A.M.; McCall, A.L.; Drewes, L.R. Ultrastructural localization of GLUT1 and GLUT3 glucose transporters in rat brain. J. Neurosci. Res. 1997, 49, 617–626. [Google Scholar] [CrossRef]
  64. Burant, C.F.; Davidson, N.O. GLUT3 glucose transporter isoform in rat testis: Localization, effect of diabetes mellitus, and comparison to human testis. Am. J. Physiol. 1994, 267, R1488–R1495. [Google Scholar] [CrossRef]
  65. Illsley, N.P. Glucose transporters in the human placenta. Placenta 2000, 21, 14–22. [Google Scholar] [CrossRef]
  66. Pantaleon, M.; Harvey, M.B.; Pascoe, W.S.; James, D.E.; Kaye, P.L. Glucose transporter GLUT3: Ontogeny, targeting and role in the mouse blastocyst. Proc. Natl. Acad. Sci. USA 1997, 94, 3795–3800. [Google Scholar] [CrossRef] [PubMed]
  67. Heijnen, H.F.G.; Oorschot, V.; Sixma, J.L.; Alat, J.W.; James, D.F. Trombin stimulates glucose transport in human platelets via the translocation of the glucose GLUT-3 from α-granules to the cell surface. J. Cell Biol. 1997, 138, 323–330. [Google Scholar] [CrossRef]
  68. Knott, R.M.; Robertson, M.; Muckensie, E.; Forrester, J.V. Regulation of glucose transporters (GLUT1 and GLUT3) in human endothelial cells. Biochem. J. 1996, 318, 313–317. [Google Scholar] [CrossRef]
  69. Leloup, C.; Arluison, M.; Kassis, N.; Lepetit, N.; Cartier, N.; Ferré, P.; Pénicaud, L. Discrete brain areas express the insulin-responsive glucose transporter GLUT4. Mol. Brain Res. 1996, 38, 45–53. [Google Scholar] [CrossRef]
  70. Vannucci, S.J.; Clark, R.R.; Koehler-Stec, E.; Li, K.; Smith, C.B.; Davies, P.; Maher, F.; Simpson, I.A. Glucose transporters expression in brain: Relationship to cerebral glucose utilization. Dev. Neurosci. 1998, 20, 369–379. [Google Scholar] [CrossRef] [PubMed]
  71. McEwen, B.S.; Reagan, L.P. Glucose transporter expression in the central nervous system: Relationship to the synaptic function. Eur. J. Pharmacol. 2004, 490, 13–24. [Google Scholar] [CrossRef] [PubMed]
  72. Medina, R.A.; Owen, G.I. Glucose transporters: Expression, regulation and cancer. Biol. Res. 2002, 35, 9–26. [Google Scholar] [CrossRef]
  73. Pascoe, W.; Inuaki, K.; Oka, Y.; Slot, J.; James, D.E. Differential targeting of facilitative glucose transporter in polarized epithelial cells. Am. J. Physiol. 1996, 271, C547–C554. [Google Scholar] [CrossRef]
  74. Sweeney, G.; Garg, R.R.; Ceddia, R.B.; Li, D.; Ishiki, M.; Somwar, R.; Foster, L.J.; Neilsen, P.O.; Prestwich, G.D.; Rudich, G.D.; et al. Intracellular delivery of phosphatidylionositol(3, 4, 5)-triphosphate causes incorporation of GLUT4 into the plasma membrane of muscle and fat cells without increasing glucose uptake. J. Biol. Chem. 2004, 279, 32233–32242. [Google Scholar] [CrossRef]
  75. Thong, F.S.L.; Dugani, C.B.; Klip, A. Turning signals on and off: GLUT4 traffic in the insulin-signaling highway. Physiology 2005, 20, 271–284. [Google Scholar] [CrossRef]
  76. Kasahara, T.; Kasahara, M. Characterization of the rat Glut4 glucose transporter expressed in the yeast Saccharomyces cerevisiae: Comparison with the Glut1 glucose transporter. Biochim. Biophys. Acta 1997, 1324, 111–119. [Google Scholar] [CrossRef]
  77. Drozdowski, L.A.; Thomas, A.B.R. Intestinal sugar transport. World J. Gastroenterol. 2006, 12, 1657–1670. [Google Scholar] [CrossRef]
  78. Davidson, N.O.; Hausman, A.M.; Ifkovits, C.A.; Buse, J.B.; Gould, G.W.; Burant, C.F.; Bell, G.I. Human intestinal glucose transporter expression and localization of GLUT5. Am. J. Physiol. 1992, 292, C795–C800. [Google Scholar] [CrossRef] [PubMed]
  79. Cheeseman, C. Fructose the odd man out. Why is the genomic control of intestinal GLUT5 expression different? J. Physiol. 2008, 586, 3563. [Google Scholar] [CrossRef]
  80. Burant, C.F.; Takeda, J.; Brat-Laroche, F.; Bell, G.J.; Davidson, N.O. Fructose transporter in human spermatozoa and small intestine is GLUT5. J. Biol. Chem. 1992, 267, 14523–14526. [Google Scholar] [CrossRef] [PubMed]
  81. Stuart, C.A.; Yin, D.; Howell, M.E.A.; Dykes, R.J.; Laffan, J.L.; Ferrando, A.A. Hexose transporter mRNA for GLUT4, GLUT5 and GLUT12 predominate in human muscle. Am. J. Physiol. Endocrinol. Metab. 2006, 291, E1067–E1073. [Google Scholar] [CrossRef] [PubMed]
  82. Payne, J.; Maher, F.; Simpson, I.; Mattice, L.; Davies, P. Glucose transporter Glut5 expression in microglial cells. Glia 1997, 21, 327–331. [Google Scholar] [CrossRef]
  83. Sasaki, A.; Horikoski, Y.; Yokoo, H.; Nakazano, Y.; Yamaguchi, H. Antiserum against human glucose transporter 5 is highly specific for microglia among cells of the mononuclear phagocyte system. Neurosci. Lett. 2003, 338, 17–20. [Google Scholar] [CrossRef] [PubMed]
  84. Douard, V.; Ferraris, R.P. Regulation of the fructose transporter GLUT5 in health and disease. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E227–E237. [Google Scholar] [CrossRef]
  85. Doege, H.; Bocianski, A.; Joost, H.G.; Schürmann, A. Activity and genomic organization of human glucose transporter 9 (GLUT9), a novel member of the family of sugar transport facilitators predominantly expressed in brain and leukocytes. Biochem. J. 2000, 350, 771–776. [Google Scholar] [CrossRef]
  86. Godoy, A.; Ulba, V.; Rodriquez, F.; Reinicke, K.; Yaňez, A.J.; Garcia Mde, L.; de los Angeles Garcia, M.; Medina, R.A.; Carrasco, M.; Barberis, S.; et al. Differential subcellular distribution of glucose transporters GLUT1-6 and GLUT9 in human cancer: Ultrastructural localization of GLUT1 and GLUT3 in breast tumor tissue. J. Cell. Physiol. 2006, 207, 614–627. [Google Scholar] [CrossRef]
  87. Li, Q.; Manolescu, A.; Ritzel, M.; Yao, S.; Slugoski, M.; Young, J.D.; Chen, X.-Z.; Cheeseman, C.I. Cloning and functional characterization of the human GLUT7 isoform SLC2A7 from the small intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 287, G236–G242. [Google Scholar] [CrossRef]
  88. Cheeseman, C. GLUT7: A new intestinal hexose transporter. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E238–E241. [Google Scholar] [CrossRef]
  89. Schürmann, A. Insight into the “odd” hexose transporters GLUT3, GLUT5, and GLUT7. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E225–E226. [Google Scholar] [CrossRef] [PubMed]
  90. Schmidt, S.; Joost, H.-G.; Schürmann, A. Glut8, the enigmatic intracellular hexose transporter. Am. J. Physiol. Endocrinol. Metab. 2009, 296, E614–E618. [Google Scholar] [CrossRef] [PubMed]
  91. Lisinski, Y.; Schürmann, A.; Joost, H.-G.; Cushman, S.W.; Al-Hasani, H. Targeting of GLUT6 (formerly GLUT9) and GLUT8 in rat adipose cells. Biochem. J. 2001, 358, 517–522. [Google Scholar] [CrossRef]
  92. Ibbersen, B.M.; Riederer, B.M.; Uldry, M.; Guhl, B.; Roth, J.; Thorens, B. Immunolocalization of GLUTX1 in the testis and to specific brain areas and vasopressin-containing neurons. Endocrinology 2002, 143, 276–284. [Google Scholar] [CrossRef]
  93. Henry, D.N.; Busik, J.V.; Botolin, D.; Grant, M.B.; Gorovits, N.; Charron, M. Glut8 expression in the blood retinal barrier. Investig. Ophtalmal. Vis. Sci. 2002, 43, 904. [Google Scholar]
  94. Goldman, N.A.; Katz, E.B.; Glen, A.S.; Weldon, R.H.; Jones, J.G.; Lynch, U.; Fezzari, M.J.; Runowicz, C.D.; Goldberg, G.L.; Charron, M.J. GLUT1 and GLUT8 in endometrium and endometrial adenocarcinoma. Modern Pathol. 2006, 19, 1429–1436. [Google Scholar] [CrossRef] [PubMed]
  95. Pinto, A.B.; Carayannopoulos, M.O.; Hoehn, A.; Dowd, L.; Moley, K.H. Glucose transporter 8 expression and translocation are critical for murine blastocyst survival. Biol. Reprod. 2002, 66, 1729–1733. [Google Scholar] [CrossRef]
  96. Tartaglia, L.A.; Weng, X. Nucleic Acid Molecules Encoding GLUTX and Uses Thereof. USPTO Patent Application No. 09/031,392, 24 August 1999. [Google Scholar]
  97. Phay, J.E.; Hussein, H.B.; Moley, J.F. Cloning and expression analyses of a novel member of the facilitative glucose transporter SLC2A9 (GLUT9). Genomics 2000, 66, 217–220. [Google Scholar] [CrossRef]
  98. Augustin, R.; Carayannopoulos, M.O.; Dowd, L.O.; Phay, J.E.; Moley, J.F.; Moley, K.H. Identification and characterization of human glucose transporter-like protein 9 (GLUT9): Alternative splicing alters trafficking. J. Biol. Chem. 2004, 279, 16229–16236. [Google Scholar] [CrossRef] [PubMed]
  99. Keembiyehetty, C.; Augustin, R.; Carayannopoulos, M.O.; Steer, S.; Manolescu, A.; Cheeseman, C.I.; Moley, K.H. Mouse glucose transporter 9 splice variants are expressed in adult liver and kidney and are up-regulated in diabetes. Mol. Endocrinol. 2006, 20, 686–697. [Google Scholar] [CrossRef]
  100. Mobasheri, A.; Dobson, H.; Mason, S.L.; Moley, J.F.; Moley, K.H. Expression of GLUT1 and GLUT9 facilitative glucose transporters in embryonic chondroblasts and mature chondrocytes in ovine articular cartilage. Cell Biol. Int. 2005, 29, 249–260. [Google Scholar] [CrossRef]
  101. Bibert, S.; Hess, S.K.; Firsov, D.; Thorens, B.; Geering, K.; Horisberger, J.D.; Bonny, O. Mouse GLUT9: Evidences for a urate uniporter. Am. J. Physiol. Renal. Physiol. 2009, 297, F612–F619. [Google Scholar] [CrossRef]
  102. Caulfield, M.J.; Monroe, P.B.; O’Neil, D.; Witkowska, K.; Charchar, F.J.; Doblado, M.; Evans, S.; Eyheramendy, S.; Onipinla, A.; Howard, P.; et al. SLC2A9 is a high capacity urate transporter in humans. PLoS Med. 2008, 5, e197. [Google Scholar] [CrossRef]
  103. Mc Vie-Wylie, A.J.; Lamson, D.R.; Chen, Y.T. Molecular cloning of a novel member of the GLUT family of Transporters, SLC2A10 (GLUT10), localized on chromosome 20q13.1: A candidate gene for NIDDM susceptibility. Genomics 2001, 72, 113–117. [Google Scholar] [CrossRef]
  104. Woods, I.S.; Trayhurn, P. Glucose transporters (GLUT and SGLT): Expressed families of sugar transport protein. Br. J. Nutr. 2003, 89, 3–9. [Google Scholar] [CrossRef]
  105. Dawson, P.A.; Mychaleckyj, J.C.; Fossey, S.C.; Mihic, S.J.; Bowden, B.W. Sequence and functional analysis of GLUT10: A glucose transporter in Type 2 diabetes-linked region of chromosome 20q12-13.1. Mol. Genet. Metab. 2001, 74, 186–199. [Google Scholar] [CrossRef]
  106. Doege, H.; Bocianski, A.; Scheepers, A.; Axer, H.; Eckel, J.; Joost, H.G.; Schürmann, A. Characterization of human glucose transporter (GLUT) 11 (encoded by SLC2A11), a novel sugar-transport facilitator specifically expressed in heart and skeletal muscle. Biochem. J. 2001, 359, 443–449. [Google Scholar] [CrossRef]
  107. Scheepers, A.; Schmidt, S.; Manolescu, A.; Cheeseman, C.I.; Bell, A.; Zahn, C.; Joost, H.-G.; Schürmann, A. Characterization of the human SLC2A11 (GLUT11) gene: Alternative promoter, usage, function, expression, and subcellular distribution of three isoforms, and lack of mouse orthologue. Mol. Membr. Biol. 2005, 22, 339–351. [Google Scholar] [CrossRef]
  108. Rogers, S.; Macheda, M.L.; Docherty, S.E.; Carty, M.D.; Henderson, M.A.; Soeller, W.C.; Gibbs, E.M.; James, D.E.; Best, J.D. Identification of a novel transporter-like protein-GLUT-12. Am. J. Physiol. Endocrinol. Metab. 2002, 283, E733–E738. [Google Scholar] [CrossRef]
  109. Stuart, C.A.; Howel, M.E.A.; Zhang, Y.; Yin, D. Insulin-stimulated translocation of GLUT12 parallels that of GLUT4 in normal muscle. J. Clin. Endocrinol. Metab. 2009, 94, 3535–3542. [Google Scholar] [CrossRef]
  110. Gude, N.M.; Stevenson, J.L.; Rogers, S.; Best, J.D.; Kalionis, B.; Huisman, M.A.; Erwich, J.J.H.M.; Timmer, A.; King, R.G. GLUT12 expression in human placenta in first trimester and term. Placenta 2003, 24, 566–570. [Google Scholar] [CrossRef] [PubMed]
  111. Augustin, R. The protein family of glucose transporters: It’s not only about glucose after all. IUBMB Life 2010, 62, 315–333. [Google Scholar] [CrossRef] [PubMed]
  112. Uldry, M.; Steiner, P.; Zurich, M.G.; Begun, P.; Hirling, H.; Dolci, W.; Thorens, B. Regulated exocytosis of an H+/myo-inositol symporter at synapses and growth cones. EMBO J. 2004, 23, 531–540. [Google Scholar] [CrossRef] [PubMed]
  113. Di Daniel, E.; Mok, M.H.; Mead, E.; Mutinelli, C.; Zambello, E.; Caberlotto, L.L.; Pell, T.J.; Langmead, C.J.; Shah, A.J.; Duddy, G.; et al. Evaluation of expression and function of the H+/myo-inositol transporter HMIT. BMC Cell Biol. 2009, 10, 54. [Google Scholar] [CrossRef]
  114. Wu, X.; Freeze, H.H. GLUT14, a duplication of GLUT3, is especially expressed in testis as alternative splice forms. Genomics 2002, 80, 553–557. [Google Scholar] [CrossRef]
  115. Riklis, E.; Quastel, J.H. Effects of cation on sugar absorption by isolated surviving guinea pig intestine. Can. J. Biochem. Physiol. 1958, 36, 347–362. [Google Scholar] [CrossRef] [PubMed]
  116. Crane, R.K. Hypothesis for mechanism of intestinal active transport of sugars. Fed. Proc. 1962, 21, 891–895. [Google Scholar] [PubMed]
  117. Crane, R.K. Na+-dependent transporter in the intestine and other animal sugars. Fed. Proc. 1965, 24, 1000–1006. [Google Scholar]
  118. Wright, E.M.; Loo, D.D.; Hirayama, B.A. Biology of human sodium glucose transporters. Physiol. Rev. 2011, 91, 733–794. [Google Scholar] [CrossRef]
  119. Wright, E.M.; Loo, D.D.F.; Hirayama, B.A.; Turk, E. Surprising versatility of Na+/glucose cotransporters: SLC5. Physiology 2004, 19, 371–376. [Google Scholar] [CrossRef]
  120. Wright, E.M. Renal Na+/glucose cotransporters. Am. J. Physiol. 2001, 280, F10–F18. [Google Scholar] [CrossRef]
  121. Wright, E.M.; Turk, E. The sodium/glucose cotransporter family SLC5. Pflügers Arch. Eur. J. Physiol. 2004, 447, 510–518. [Google Scholar] [CrossRef]
  122. Turk, E.; Wright, E.M. Membrane topology motifs in the SGLT cotransporter family. J. Membr. Biol. 1997, 159, 1–20. [Google Scholar] [CrossRef] [PubMed]
  123. Bianche, L.; Diez-Sampedro, A. A single amino acid change converts the sugar sensor SGLT3 into a sugar transporter. PLoS ONE 2010, 5, e10241. [Google Scholar] [CrossRef]
  124. Turk, E.; Martin, M.G.; Wright, E.M. Structure of the human Na+/glucose cotransporter gene SGLT1. J. Biol. Chem. 1994, 269, 15204–15209. [Google Scholar] [CrossRef] [PubMed]
  125. Panayotova-Heiermann, M.; Loo, D.D.F.; Kong, C.T.; Lever, J.E.; Wright, E.M. Sugar binding to Na+/glucose cotransporters is determined by the C-terminal half of the protein. J. Biol. Chem. 1996, 271, 10029–10034. [Google Scholar] [CrossRef]
  126. Zhou, L.; Cryan, E.V.; D’Andrea, M.R.; Belkowski, S.; Conway, B.R.; Demarest, K.T. Human cardiomyocytes express high level of Na+/glucose cotransporter 1 (SGLT1). J. Cell. Biochem. 2003, 90, 339–346. [Google Scholar] [CrossRef] [PubMed]
  127. Zhao, F.-Q.; Zheng, Y.C.; Wall, E.H.; McFadden, T.B. Cloning and expression of bovine sodium/glucose cotransporters. J. Diary Sci. 2005, 88, 182–194. [Google Scholar] [CrossRef]
  128. Elfeber, K.; Koehler, A.; Lutzenburg, M.; Osswald, C.; Galla, H.J.; Witte, O.W.; Koepsell, H. Localization of the Na+-D-glucose cotransporter SGLT1 in the blood-brain barrier. Histochem. Cell Biol. 2004, 121, 201–207. [Google Scholar] [CrossRef]
  129. Shah, K.; DeSilva, S.; Abbruscato, T. The role of glucose transporters in brain disease: Diabetes and Alzheimer’s disease. Int. J. Mol. Sci. 2012, 13, 12629–12655. [Google Scholar] [CrossRef]
  130. Venula, S.; Roderer, K.; Yang, T.; Bhat, G.J.; Thekkumkara, T.J.; Abbruscato, T.J. A functional role of sodium-dependent glucose transporter across the blood-brain barrier during oxygen glucose deprivation. J. Pharmacol. Exp. Therapeut. 2009, 328, 487–495. [Google Scholar] [CrossRef]
  131. Chen, L.Q.; Hou, B.H.; Lalonde, S.; Takanaga, H.; Hartung, M.L.; Qu, X.O.; Guo, W.-J.; Kim, J.-G.; Underwood, W.; Chaudhuri, B.; et al. Sugar transporters for intracellular exchange and nutrition of pathogens. Nature 2010, 468, 527–532. [Google Scholar] [CrossRef] [PubMed]
  132. Zeuthen, T.; Meinild, A.K.; Lao, D.D.; Wright, E.M.; Klaerke, D.A. Isotonic transport by the Na+-glucose cotransporter SGLT1 from humans and rabbit. J. Physiol. 2001, 531, 631–644. [Google Scholar] [CrossRef]
  133. Loo, D.D.F.; Wright, E.M.; Zeuthen, T. Water pumps. J. Physiol. 2002, 42, 53–60. [Google Scholar] [CrossRef]
  134. Leung, D.W.; Loo, D.D.F.; Hirayama, B.A.; Zeuthen, T.; Wright, E.M. Urea transport by cotransporters. J. Physiol. 2000, 528, 251–257. [Google Scholar] [CrossRef]
  135. Panayotova-Heiermann, M.; Wright, E.M. Mapping the urea channel through the rabbit Na+/glucose cotransporter SGLT1. J. Physiol. 2001, 535, 419–425. [Google Scholar] [CrossRef]
  136. Wright, E.M.; Loo, D.D.F.; Turk, E.; Hirayama, B.A. Sodium cotransporters. Curr. Opin. Cell Biol. 1996, 8, 468–473. [Google Scholar] [CrossRef]
  137. Wells, R.G.; Kamai, Y.; Pajor, A.M.; Turk, E.; Wright, E.M.; Hediger, M.A. The cloning of a human kidney cDNA with similarity to the sodium/glucose cotransporter. Am. J. Physiol. 1992, 263, F459–F465. [Google Scholar] [CrossRef] [PubMed]
  138. Wells, R.G.; Mohandas, T.K.; Hediger, M.A. Localization of the Na+/glucose cotransporter gene SGLT2 to human chromosome 16 close to the centromere. Genomics 1993, 17, 787–789. [Google Scholar] [CrossRef] [PubMed]
  139. Kong, C.T.; Yet, S.F.; Lever, J.E. Cloning and expression of a mammalian Na+/amino acid cotransporter with sequence similarity to Na+/glucose cotransporters. J. Biol. Chem. 1993, 268, 1509–1512. [Google Scholar] [CrossRef]
  140. Mackenzie, B.; Panayotova-Heiermann, M.; Loo, D.D.; Lever, J.E.; Wright, E.M. SAA1 is a low affinity Na+/glucose cotransporter and not an amino acid transporter. A reinterpretation. J. Biol. Chem. 1994, 269, 22488–22491. [Google Scholar] [CrossRef] [PubMed]
  141. Kanai, Y.; Lee, W.S.; You, G.; Brown, D.; Hediger, M.A. The human kidney low affinity Na+/glucose cotransporter SGLT2. Delineation of the major renal reabsorption mechanism for D-glucose. J. Clin. Investig. 1994, 93, 397–404. [Google Scholar] [CrossRef] [PubMed]
  142. Diez-Sampedro, A.; Eskanderi, S.; Wright, E.M.; Hiryama, B.A. Na+-to-sugar stoichiometry of SGLT3. Am. J. Physiol. Renal. Physiol. 2001, 280, F278–F282. [Google Scholar] [CrossRef]
  143. Kothinti, R.K.; Blodgett, A.B.; North, P.E.; Roman, R.J.; Tabatabai, N.M. A novel SGLT is expressed in the human kidney. Eur. J. Pharmacol. 2012, 690, 77–83. [Google Scholar] [CrossRef]
  144. Tazawa, S.; Yamamoto, T.; Fujikura, H.; Hiratochi, M.; Itoh, F.; Tomae, M.; Takemura, Y.; Maruyama, H.; Sugiyama, T.; Wakamatsu, A.; et al. SLC5A9/SGLT4, a new Na+-dependent glucose transporter, is an essential transporter for mannose and fructose. Life Sci. 2005, 76, 1039–1050. [Google Scholar] [CrossRef]
  145. Grempler, R.; Augustin, R.; Froehner, S.; Hildebrandt, T.; Simon, E.; Mark, M.; Eickelmann, P. Functional characterization of SGLT-5 as a novel kidney-specific sodium-dependent sugar transporter. FEBS Lett. 2012, 586, 248–253. [Google Scholar] [CrossRef]
  146. Augustin, R.; Mayoux, E. Mammalian sugar transporters. In Glucose Homeostasis, 1st ed.; Szablewski, L., Ed.; InTech: Rijeka, Croatia, 2014; pp. 3–36. [Google Scholar]
  147. Coady, M.J.; Wallendorf, B.; Cagan, D.G.; Lapointe, J.Y. Identification of a novel Na+/myo-inositol cotransporter. J. Biol. Chem. 2002, 299, 35219–35224. [Google Scholar] [CrossRef] [PubMed]
  148. Michaelis, T.; Helms, G.; Merboldt, K.D.; Hanicka, W.; Bruhn, H.; Frahm, J. Identification of Syllo-inositol in proton NMR spectra in human brain in vivo. NMR Biomed. 1993, 6, 105–109. [Google Scholar] [CrossRef]
  149. Tsai, L.J.; Hsiao, S.H.; Tsai, L.M.; Lin, C.Y.; Tsai, J.J.; Liou, D.M.; Lan, J.L. The sodium-dependent glucose cotransporter SLC5A11 as an autoimmune modifier gene in SLE. Tissue Antigens 2008, 71, 114–126. [Google Scholar] [CrossRef] [PubMed]
  150. Ho, H.T.B.; Dahlin, A.; Wang, J. Expression profiling of solute carrier gene families at the blood-CSF barrier. Front. Pharmacol. 2012, 3, 154. [Google Scholar] [CrossRef]
  151. De La Vieja, A.; Dohan, O.; Levy, O.; Carrasco, N. Molecular analysis of the sodium/iodide symporter: Impact on thyroid and extrathyroid pathophysiology. Physiol. Rev. 2000, 80, 1083–1105. [Google Scholar] [CrossRef] [PubMed]
  152. Van Sande, J.; Massart, C.; Beauwens, R.; Schouters, A.; Costaglia, S.; Dumont, J.E.; Wolff, J. Anion selectivity by the sodium iodide symporter. Endocrinology 2003, 144, 247–252. [Google Scholar] [CrossRef]
  153. Appansundaram, S.; Fergusson, S.M.; Blakely, R.D. Molecular cloning and characterization of a murine hemi-cholinum-3-sensitive choline transporter. Biochem. Soc. Trans. 2001, 29, 711–716. [Google Scholar] [CrossRef]
  154. Coady, M.J.; Wallendorf, B.; Bourgeois, F.; Charron, F.; Lapointe, J.Y. Establishing a definitive stoichiometry for the Na+/monocarboxylate cotransporter SMCT. Biophys. J. 2007, 93, 2325–2331. [Google Scholar] [CrossRef]
  155. Gopal, E.; Umapathy, N.S.; Martin, P.M.; Ananth, S.; Gnoma-Prakasom, J.P.; Becker, H.; Wagner, C.A.; Ganapathy, V.; Prasad, P.D. Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern in kidney. Biochim. Biophys. Acta 2012, 1768, 2690–2697. [Google Scholar] [CrossRef]
  156. Expression of SLC50A1 in Cancer–Summary the Human Protein Atlas. Available online: www.proteinatlas.org/ENSG00000169241-SLC50A1/pathology (accessed on 1 June 2025).
  157. Santer, R.; Hillebrand, G.; Steinmann, B.; Schaub, J. Intestinal glucose transport: Evidence for a membrane traffic-based pathway in humans. Gastroenterology 2003, 124, 34–39. [Google Scholar] [CrossRef] [PubMed]
  158. Larrabel, M.G. Lactate metabolism and its effect on glucose metabolism in an exercised tissue. J. Neurochem. 1995, 64, 1734–1741. [Google Scholar] [CrossRef]
  159. Magistretti, P.J.; Pellerin, L. Cellular mechanisms of brain energy metabolism and their relevance to functional brain imaging. Philos. Trans. R. Soc. Lond. 1999, 254, 1155–1163. [Google Scholar] [CrossRef]
  160. Goldstein, G.W. Relation of potassium transport to oxidative metabolism in isolated brain capillaries. J. Physiol. 1979, 286, 185–195. [Google Scholar] [CrossRef] [PubMed]
  161. Guzman, M.; Blazquez, C. Is there an astrocyte-neuron ketone body shuttle? Trends Endocrinol. Metab. 2001, 12, 169–173. [Google Scholar] [CrossRef] [PubMed]
  162. Nguyen, Y.T.K.; Ha, H.T.T.; Nguyen, T.H.; Nguyen, L.N. The role of SLC transporters for brain health and disease. Cell. Mol. Life. Sci. 2022, 79, 20. [Google Scholar] [CrossRef]
  163. Francis, P.T.; Palmer, A.M.; Snape, M.; Wilcock, G.K. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry 1999, 66, 137–147. [Google Scholar] [CrossRef]
  164. Lopez, O.L.; McDade, E.; Riverol, N.; Becker, J.T. Evaluation of the diagnostic criteria for degenerative and cognitive disorders. Curr. Opin. Neurol. 2011, 24, 532–541. [Google Scholar] [CrossRef]
  165. Marcelina de Naraneth, A. Type 2 diabetes mellitus in the pathophysiology of Alzheimer’s disease. Dement. Neuropsychol. 2017, 11, 105–113. [Google Scholar]
  166. Sun, Y.; Ma, C.; Sun, H.; Wang, H.; Peng, W.; Zhou, Z.; Wang, H.; Pi, C.; Shi, Y.; He, X. Metabolism: A novel shared link between diabetes mellitus and Alzheimer’s disease. J. Diabetes Res. 2020, 2020, 4981814. [Google Scholar] [CrossRef] [PubMed]
  167. Szablewski, L. Association between diabetes mellitus and neurodegenerative diseases. Int. J. Mol. Sci. 2025, 26, 542. [Google Scholar] [CrossRef]
  168. Rorbach-Dolata, A.; Piwowar, A. Neurometabolic evidence supporting the hypothesis of increased incidence of Type 3 diabetes mellitus in the 21st century. Biomed. Res. Int. 2019, 2019, 1435276. [Google Scholar] [CrossRef] [PubMed]
  169. Hamzé, R.; Delangre, E.; Tolu, S.; Moreau, M.; Janel, N.; Bailbé, D.; Movassat, J. Type 2 diabetes mellitus and Alzheimer’s disease: Shared molecular mechanisms and potential common therapeutic targets. Int. J. Mol. Sci. 2022, 23, 15287. [Google Scholar] [CrossRef] [PubMed]
  170. Madhusudhanan, J.; Suresh, G.; Devanathan, V. Neurodegeneration in type 2 diabetes: Alzheimer’s as a case study. Brain Behav. 2020, 10, e01577. [Google Scholar] [CrossRef]
  171. Diniz Pereira, J.; Gomes Fraga, V.; Morais Santos, A.L.; das Graças Carvalho, M.; Caramelli, P.; Braga Gomes, K. Alzheimer’s disease and type 2 diabetes mellitus, a systematic review of proteomic studies. J. Neurochem. 2021, 156, 753–776. [Google Scholar] [CrossRef]
  172. Chatterjee, S.; Mudher, A. Alzheimer’s disease and type 2 diabetes: A critical assessment of the shared pathological traits. Front. Neurosci. 2018, 12, 383. [Google Scholar] [CrossRef]
  173. Hoyer, S. The brain insulin signal transduction system and sporadic (type II) Alzheimer disease: An update. J. Neural Transm. 2002, 109, 341–360. [Google Scholar] [CrossRef]
  174. Chen, Y.; Deng, Y.; Zhang, B.; Gong, C.-X. Deregulation of brain insulin signaling in Alzheimer’s disease. Neurosci. Bull. 2014, 30, 282–294. [Google Scholar] [CrossRef]
  175. Cunnane, S.C.; Nugent, S.; Roy, M.; Courchesne-Loyer, A.; Croteau, E.; Tremblay, S.; Castellano, A.; Pifferi, F.; Bocti, C.; Paquet, N.; et al. Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition 2011, 27, 3–20. [Google Scholar] [CrossRef]
  176. De Strooper, B. Loss-of-function presenilin mutations in Alzheimer’s disease, talking point on the role of presenilin mutations in Alzheimer’s disease. EMBO Rep. 2007, 8, 141–146. [Google Scholar] [CrossRef]
  177. Doroszewska, J.; Prendecki, M.; Oczkowska, A.; Dezor, M.; Kozubski, W. Molecular basis of familial and sporadic Alzheimer’s disease. Curr. Alzheimer Res. 2016, 13, 952–963. [Google Scholar] [CrossRef]
  178. Bertram, L.; Tanzi, R.E. Genome-wide association studies in Alzheimer’s disease. Hum. Mol. Genet. 2009, 18, R137–R145. [Google Scholar] [CrossRef]
  179. Li, X.; Song, D.; Leng, S.X. Link between type 2 diabetes and Alzheimer’s disease from epidemiology to mechanism and treatment. Clin. Intern. Aging 2015, 10, 549–560. [Google Scholar] [CrossRef]
  180. Kisilevsky, R.; Raimondi, S.; Bellotti, V. Historical and current concepts of fibrillogenesis and in vivo amyloidogenesis: Implications of amyloid tissue targeting. Front. Mol. Biosci. 2016, 3, 17. [Google Scholar] [CrossRef]
  181. Ancarcona, M.; Winblad, B.; Monteiro, C.; Feans, C.; Powers, J.; Johansson, J.; Westmark, T.; Presto, J.; Kelly, J.W.; Institutet, K.; et al. Current and future treatment of amyloid diseases. J. Intern. Med. 2016, 280, 177–202. [Google Scholar] [CrossRef] [PubMed]
  182. Johnson, V.E.; Stewart, W.; Smith, D.H. Widespread τ and amyloid-β pathology many years after a single traumatic brain injury in humans. Brain Pathol. 2012, 22, 142–149. [Google Scholar] [CrossRef] [PubMed]
  183. Weller, J.; Budson, A. Current understanding of Alzheimer’s disease diagnosis and treatment. F1000Research 2018, 7, 1161. [Google Scholar] [CrossRef] [PubMed]
  184. Sędzikowska, A.; Szablewski, L. Insulin and insulin resistance in Alzheimer’s disease. Int. J. Mol. Sci. 2021, 22, 9987. [Google Scholar] [CrossRef]
  185. Walsh, D.M.; Klyubin, I.; Fadeeva, J.V.; Cullen, W.K.; Anwyl, R.; Wolfe, M.S.; Rowan, M.J.; Selkoe, D.J. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature 2002, 416, 535–539. [Google Scholar] [CrossRef]
  186. Berlanga-Acosta, J.; Guillén-Nieto, G.; Rodriguez-Rodriguez, N.; Bringas-Vegá, M.L.; Garcia-del-Barcó-Herrera, D.; Berlanga-Saez, J.O.; Garcia-Ojálvo, A.; Valdés-Sosa, M.J.; Valdés-Sosa, P.A. Insulin resistance at the crossroad of Alzheimer disease pathology: A review. Front. Endocrinol. 2020, 11, 560375. [Google Scholar] [CrossRef]
  187. Zhao, W.Q.; Lacor, P.N.; Chen, H.; Lambert, M.P.; Buon, M.J.; Krafft, G.A.; Klein, W.L. Insulin receptor dysfunction of neurotic oligomeric Aβ. J. Biol. Chem. 2009, 289, 18742–18753. [Google Scholar] [CrossRef] [PubMed]
  188. Tumminia, A.; Vinciguerra, F.; Parisi, M.; Frittitla, L. Type 2 diabetes mellitus and Alzheimer’s disease: Role of insulin signaling and therapeutic implications. Int. J. Mol. Sci. 2018, 19, 3306. [Google Scholar] [CrossRef]
  189. Caillet-Boudin, M.L.; Buee, L.; Sergrant, N.; Lefebvre, B. Regulation of human MAPT gene expression. Mol. Neurodegener. 2015, 10, 28. [Google Scholar] [CrossRef]
  190. Arendt, T.; Stieler, J.T.; Holzer, M. Tau and tauopathies. Brain Res. Bull. 2016, 126, 238–292. [Google Scholar] [CrossRef]
  191. Stoothoff, W.H.; Johnson, G.V. Tau phosphorylation, physiological and pathological consequences. Biochim. Biophys. Acta 2005, 1739, 280–297. [Google Scholar] [CrossRef]
  192. Chatterjee, S.; Sang, T.K.; Lawless, G.M.; Jackson, G.R. Dissociation of tau toxicity and phosphorylation: Role of GSK-3beta, MARK, and Cdk5 in Drosophila model. Hum. Mol. Genet. 2009, 18, 164–177. [Google Scholar] [CrossRef] [PubMed]
  193. Burillo, J.; Margués, P.; Jiménez, B.; González-Blanco, C.; Benito, M.; Guillén, C. Insulin resistance and diabetes mellitus in Alzheimer’s disease. Cells 2021, 10, 1236. [Google Scholar] [CrossRef]
  194. Van Der Velpen, V.; Teav, T.; Gallart-Ayala, H.; Mehl, F.; Konz, I.; Clark, C.; Oikonomidi, A.; Peratout, G.; Henry, H.; Delorenzi, M.; et al. Systemic and central nervous system metabolic alterations in Alzheimer’s disease. Alzheimer’s Res. Ther. 2019, 11, 93. [Google Scholar] [CrossRef]
  195. Mark, R.J.; Pang, Z.; Geddes, J.W.; Uchida, K.; Mattson, M.P. Amyloid β-peptide impairs glucose transport in hippocampal and cortical neurons: Involvement of membrane lipid peroxidation. J. Neurosci. 1997, 17, 1046–1054. [Google Scholar] [CrossRef] [PubMed]
  196. Niccoli, T.; Cabecinha, M.; Tillman, A.; Kerr, F.; Wong, C.T.; Cardenes, D.; Vincent, A.J.; Bettedi, L.; Li, L.; Gronke, S.; et al. Increased glucose transport into neurons rescues Aβ toxicity in Drosophila. Curr. Biol. 2016, 26, 2291–2300. [Google Scholar] [CrossRef] [PubMed]
  197. Hoyer, S. Oxidative energy metabolism in Alzheimer brain. Studies in early-onset and late-onset cases. Mol. Chem. Neuropathol. 1992, 16, 207–224. [Google Scholar] [CrossRef]
  198. DeKosky, S.T.; Scheff, S.W. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: Correlation with cognitive severity. Ann. Neurol. 1990, 27, 457–464. [Google Scholar] [CrossRef]
  199. Jia, Y.J.; Wang, N.; Zhang, Y.; Xue, D.; Lou, H.; Liu, X. Alteration in the function and expression of SLC and ABC transporters in the neurovascular unit in Alzheimer’s disease and the clinical significance. Aging Dis. 2020, 11, 390–404. [Google Scholar] [CrossRef]
  200. Jagust, W.J.; Seab, J.P.; Huesman, R.H.; Valk, P.E.; Mathis, C.A.; Reed, B.R.; Coxon, P.G.; Budinger, T.F. Diminished glucose transport in Alzheimer’s disease: Dynamic PET studies. J. Cereb. Blood Flow Metab. 1991, 11, 323–330. [Google Scholar] [CrossRef]
  201. Piert, M.; Koeppe, R.A.; Giordani, B.; Berent, S.; Kuhl, D.E. Diminished glucose transport and phosphorylation in Alzheimer’s disease determined by FDG-PET. J. Nucl. Med. 1996, 37, 201–208. [Google Scholar] [PubMed]
  202. Friedland, R.P.; Jagust, W.J.; Huesman, R.H.; Koss, E.; Knittel, B.; Mathis, C.A.; Ober, B.A.; Mazoyer, B.M.; Budinger, T.F. Regional cerebral glucose transport and utilization in Alzheimer’s disease. Neurology 1989, 39, 1427–1434. [Google Scholar] [CrossRef] [PubMed]
  203. Simpson, I.A.; Davies, P. Reduced glucose transporter concentrations in brain of patients with Alzheimer’s disease. Ann. Neurol. 1994, 36, 800–801. [Google Scholar] [CrossRef]
  204. Harik, S.I. Changes in the glucose transporter of brain capillaries. Can. J. Physiol. Pharmacol. 1992, 70 (Suppl. l), S113–S117. [Google Scholar] [CrossRef]
  205. Kalaria, R.N.; Harik, S.I. Reduced glucose transporter at the blood-brain barrier in cerebral cortex in Alzheimer’s disease. J. Neurochem. 1989, 53, 1083–1088. [Google Scholar] [CrossRef]
  206. Kyrtata, N.; Emsley, H.C.A.; Sparasci, O.; Parkes, L.M.; Dickie, B.R. A systemic review of glucose transport alterations in Alzheimer’s disease. Front. Neurosci. 2021, 15, 626636. [Google Scholar] [CrossRef]
  207. Liu, Y.; Liu, F.; Iqbal, K.; Grundke-Iqbal, I.; Gang, C.-X. Decreased glucose transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer’s disease. FEBS Lett. 2008, 582, 359–364. [Google Scholar] [CrossRef]
  208. An, Y.; Varma, V.R.; Varma, S.; Casanova, R.; Dammer, E.; Pletnikova, O.; Chia, C.W.; Egan, J.M.; Ferrucci, L.; Troncoso, J.; et al. Evidence for brain glucose dysregulation in Alzheimer’s disease. Alzheimer’s Dement. 2018, 14, 318–329. [Google Scholar] [CrossRef]
  209. Hendrix, R.D.; Ou, Y.; Davis, J.E.; Odle, A.K.; Groves, T.R.; Allen, A.R.; Childs, G.V.; Barger, S.W. Alzheimer amyloid-β-peptide disrupts membrane localization of glucose transporter 1 in astrocytes: Implications for glucose levels in brain and blood. Neurobiol. Aging 2021, 97, 73–88. [Google Scholar] [CrossRef] [PubMed]
  210. Deng, Y.; Li, B.; Liu, Y.; Iqbal, K.; Grundke-Iqbal, I.; Gong, C.-X. Dysregulation of insulin signaling, glucose transporters, O-GlcNAcylation, and phosphorylation of tau and neurofilaments in the brain. Am. J. Pathol. 2009, 175, 2089–2098. [Google Scholar] [CrossRef] [PubMed]
  211. Liu, Y.; Liu, F.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C.-X. Brain glucose transporters, O-GlcNAcylation and phosphorylation of tau in diabetes and Alzheimer’s disease. J. Neurochem. 2009, 111, 242–249. [Google Scholar] [CrossRef] [PubMed]
  212. Huang, Y.; Lei, L.; Liu, D.; Jovin, I.; Russel, R.; Johnson, R.S.; Di Lorenzo, A.; Giordano, F.J. Normal glucose uptake in the brain and heart requires an endothelial cell-specific HIF-1α-dependent function. Proc. Natl. Acad. Sci. USA 2012, 109, 17478–17483. [Google Scholar] [CrossRef] [PubMed]
  213. Liu, F.; Shi, J.; Tanimukai, H.; Gu, J.; Gu, J.; Grundke-Iqbal, I.; Iqbal, K.; Gong, C.X. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer’s disease. Brain 2009, 132, 1820–1832. [Google Scholar] [CrossRef]
  214. Jin, N.; Qian, W.; Yin, X.; Zhang, L.; Iqbal, K.; Grundke-Iqbal, I.; Gong, C.-X.; Liu, F. CREB regulates the expression of neuronal glucose transporter 3: A possible mechanism related to impaired brain glucose uptake in Alzheimer’s disease. Nucl. Acid. Res. 2013, 41, 3240–3256. [Google Scholar] [CrossRef]
  215. Hooijmans, C.R.; Graven, C.; Dederen, P.J.; Tanila, H.; van Groen, T.; Kiliaan, A.J. Amyloid beta deposition is related to decreased glucose transporter-1 levels and hippocampal atrophy in brain of aged APP/PS1 mice. Brain Res. 2007, 1181, 93–103. [Google Scholar] [CrossRef]
  216. Pujol-Gimenez, J.; Martisova, E.; Perez-Mediavilla, A.; Lostao, M.P.; Ramirez, M.J. Expression of glucose transporter GLUT12 in Alzheimer’s disease patients. J. Alzheimer’s Dis. 2014, 42, 97–101. [Google Scholar] [CrossRef]
  217. Gil-Iturbe, E.; Solas, M.; Cuadrado-Tejedo, M.; Garcia-Osta, A.; Escote, X.; Ramirez, M.J.; Lostao, M.P. GLUT12 expression in brain of mouse models of Alzheimer’s disease. Mol. Neurobiol. 2020, 57, 798–805. [Google Scholar] [CrossRef] [PubMed]
  218. Bloem, B.R.; Okun, M.S.; Klein, C. Parkinson’s disease. Lancet Lond. Engl. 2021, 397, 2284–2303. [Google Scholar] [CrossRef]
  219. Chase, T.N.; Oh, J.D.; Blanchet, P.J. Neostriatal mechanisms in Parkinson’s disease. Neurology 1998, 51 (Suppl. 2), S30–S35. [Google Scholar] [CrossRef]
  220. Chen, L.; Wang, C.; Qin, L.; Zhang, H. Parkinson’s disease and glucose metabolism impairment. Translat. Neurodegener. 2025, 14, 10. [Google Scholar] [CrossRef]
  221. Jia, F.; Fellner, A.; Kumar, K.R. Monogenic Parkinson’s disease: Genotype, phenotype, pathophysiology, and genetic testing. Genes 2022, 13, 471. [Google Scholar] [CrossRef]
  222. Inamdar, N.N.; Arulmozhi, D.K.; Tandon, A.; Bodhanker, S.L. Parkinson’s disease: Genetics and beyond. Curr. Neuropharmacol. 2007, 5, 99–113. [Google Scholar] [CrossRef] [PubMed]
  223. Xu, Y.; Wei, X.; Liu, X.; Liao, J.; Lin, J.; Zhu, C.; Meng, X.; Xie, D.; Chao, D.; Fenoy, A.J.; et al. Low cerebral glucose metabolism: A potential predictor for severity of vascular Parkinsonism and Parkinson’s disease. Aging Dis. 2015, 6, 426–436. [Google Scholar] [CrossRef]
  224. Dai, C.; Tai, C.; Zhao, L.; Liang, Y.; Liu, G.; Liu, H.; Zhong, Y.; Liu, Z.; Mo, L.; Liu, X.; et al. Glucose metabolism impairment in Parkinson’s disease. Brain Res. Bull. 2023, 199, 110672. [Google Scholar] [CrossRef]
  225. Sian-Hulsmann, J.; Rieder, P.; Michel, T.M. Metabolic dysfunction in Parkinson’s disease: Unraveling the glucose-lipid connection. Biomedicines 2024, 12, 2841. [Google Scholar] [CrossRef] [PubMed]
  226. Tanvir, A.; Jo, J.; Park, S.M. Targeting glucose metabolism: A novel therapeutic approach for Parkinson’s diseases. Cells 2024, 13, 1876. [Google Scholar] [CrossRef]
  227. Schmidt, S.; Stautner, C.; Vu, D.T.; Heinz, A.; Regensburger, M.; Karayel, O.; Trümbach, D.; Artati, A.; Kaltenhäuser, S.; Nassef, M.Z.; et al. A reversible state of hypometabolism in a human cellular model of sporadic Parkinson’s disease. Nat. Commun. 2023, 14, 7674. [Google Scholar] [CrossRef]
  228. Puchades, M.; Sogn, C.J.; Maehlen, J.; Bergsen, L.H.; Gundersen, V. Unaltered lactate and glucose transport levels in the MPTP mouse model of Parkinson’s disease. J. Parkinson’s Dis. 2013, 3, 371–385. [Google Scholar] [CrossRef] [PubMed]
  229. Sarkar, S.; Chigurupati, S.; Raymick, J.; Mann, D.; Bowyer, J.F.; Schmitt, T.; Beger, R.D.; Hanig, J.P.; Schmued, L.C.; Paule, M.G. Neuroprotective effect of the chemical chaperone, trehalose in a chronic MPTP-induced Parkinson’s disease mouse model. Neurotoxicology 2014, 44, 250–262. [Google Scholar] [CrossRef]
  230. Burks, S.; Raymick, J.; Robinson, B.; Hanig, J.; Sarkar, S. Neuroprotective effects of acetyl-1-carmitine (ALC) in a chronic MPTP-induced Parkinson’s disease mouse model: Endothelial and microglial effects. Neurosci. Lett. 2019, 703, 86–95. [Google Scholar] [CrossRef] [PubMed]
  231. Liu, H.; Chen, L.; Zhang, C.; Liu, C.; Li, Y.; Cheng, L.; Ouyang, Y.; Rutledge, C.; Anderson, J.; Wei, Z.; et al. Glymphatic influx and clearance are perturbed in Huntington’s disease. JCI Insight 2024, 9, 172286. [Google Scholar] [CrossRef]
  232. Andrew, S.E.; Goldberg, Y.P.; Kremer, B.; Telenius, H.; Theilmann, J.; Adam, S.; Starr, E.; Squitieri, F.; Lin, B.; Kalchman, M.A.; et al. The relationship between trinucleotide (CAG) repeat length and clinical features of Huntington’s disease. Nat. Genet. 1993, 4, 398–403. [Google Scholar] [CrossRef] [PubMed]
  233. Jucker, M.; Walker, L.C. Self-propagation of pathogenic protein aggregates in neurodegenerative diseases. Nature 2013, 501, 45–51. [Google Scholar] [CrossRef]
  234. Vittori, A.; Breda, C.; Repici, M.; Orth, M.; Roos, R.A.C.; Outeiro, T.F.; Giorgini, F.; Hollox, E.J.; The Registry Investigators of the European Huntington’s Disease Network. Copy- number variation of the neuronal glucose transporter gene SLC2A3 and age of onset in Huntington’s disease. Hum. Mol. Genet. 2014, 23, 3129–3137. [Google Scholar] [CrossRef]
  235. Chang, C.-P.; Wu, C.-W.; Chern, Y. Metabolic dysregulation in Huntington’s disease. Neuronal and glial perspectives. Neurobiol. Dis. 2024, 201, 106672. [Google Scholar] [CrossRef]
  236. Chaves, G.; Özel, R.E.; Rao, N.; Hadiprodjo, H.; Da Costa, Y.; Tokuno, Z.; Pourmand, N. Metabolic and transcriptomic analysis of Huntington’s disease model reveal changes in intracellular glucose levels and related genes. Heliyon 2017, 3, e00381. [Google Scholar] [CrossRef]
  237. Martin, W.R.; Clark, C.; Ammann, W.; Stossl, A.J.; Shtybel, W.; Hayden, M.R. Cortical glucose metabolism in Huntington disease. Neurology 1992, 42, 223–229. [Google Scholar] [CrossRef]
  238. Berent, S.; Giordari, B.; Lehtinen, S.; Markel, D.; Penney, J.B.; Buchtel, H.A.; Starosta-Rubinstein, S.; Hichwa, R.; Young, A.B. Positron emission tomography scan investigations of Huntington’s disease: Cerebral metabolic correlates of cognitive function. Ann. Neurol. 1988, 23, 541–546. [Google Scholar] [CrossRef]
  239. Antonini, A.; Leenders, K.L.; Eidelberg, D. [11C] Raclopride-PET studies of the Huntington’s disease rate of progression: Relevance of the trinucleotide repeat length. Ann. Neurol. 1998, 43, 253–255. [Google Scholar] [CrossRef]
  240. Gambino, W.C.; Brennan, W.A., Jr. Glucose transporter isoform expression in Huntington’s disease brain. J. Neurochem. 1994, 63, 1392–1397. [Google Scholar] [CrossRef]
  241. McDonald, T.S.; Lerskiatiphanich, T.; Woodruff, T.M.; McCombe, P.A.; Lee, J.D. Potential mechanisms to modify impaired glucose metabolism in neurodegenerative disorders. J. Cereb. Blood Flow Metab. 2023, 43, 26–43. [Google Scholar] [CrossRef] [PubMed]
  242. Andreassen, O.A.; Dedeoglu, A.; Stanojevic, V.; Hughes, D.B.; Browne, S.E.; Leech, C.A.; Ferrante, R.J.; Habener, J.F.; Beal, J.F.; Thomas, M.K. Huntington’s disease of the endocrine pancreas: Insulin deficiency and diabetes mellitus due to impaired insulin gene expression. Neurobiol. Dis. 2002, 11, 410–424. [Google Scholar] [CrossRef]
  243. Mochel, F.; Haller, R.G. Energy deficit in Huntington disease: Why it matters. J. Clin. Investig. 2011, 121, 493–499. [Google Scholar] [CrossRef]
  244. Guyot, M.C.; Hantraye, P.; Dolan, R.; Palfi, S.; Maziére, M.; Brouillet, E. Quantifiable bradykinesia, gait abnormalities and Huntington’s disease-like striatal lesions in rats chronically treated with 3-nitropropionic acid. Neuroscience 1997, 79, 45–56. [Google Scholar] [CrossRef] [PubMed]
  245. Dubinsky, J.M. Towards an understanding of energy impairment in Huntington’s disease brain. J. Huntingt. Dis. 2017, 6, 267–302. [Google Scholar] [CrossRef] [PubMed]
  246. Bossy-Wetzel, E.; Petrilli, A.; Knott, A.B. Mutant huntingtin and mitochondrial dysfunction. Trend. Neurosci. 2008, 31, 609–616. [Google Scholar] [CrossRef]
  247. Covarrubias-Pint, A.; Moll, P.; Solis-Maldonado, A.; Acuña, A.I.; Riveros, A.; Paz Miró, M.; Papic, E.; Beltrán, F.A.; Cepeda, C.; Concha, I.I.; et al. Beyond the redox imbalance: Oxidative stress contributes to an impaired GLUT3 modulation in Huntington’s disease. Free Rad. Biol. Med. 2015, 89, 1085–1096. [Google Scholar] [CrossRef]
  248. McClory, H.; Williams, D.; Sapp, E.; Gatune, L.W.; Wang, P.; DiFiglia, M.; Li, X. Glucose transporter 3 is a rab11-dependent trafficking cargo and its transport to the cell surface is reduced in neurons CAG140 Huntington’s mice. Acta Neuropathol. Commun. 2014, 2, 179. [Google Scholar] [CrossRef]
  249. De Vivo, D.C.; Trifiletti, R.R.; Jacobson, R.I.; Ronen, G.M.; Behmand, R.A.; Harik, S.I. Defective glucose transport across the blood-brain barrier as a cause of persistent hypoglycorrhachia, seizures, and developmental delay. N. Engl. J. Med. 1991, 325, 703–709. [Google Scholar] [CrossRef] [PubMed]
  250. Brockmann, K. The expanding phenotype of GLUT1-deficiency syndrome. Brain Develop. 2009, 31, 545–552. [Google Scholar] [CrossRef] [PubMed]
  251. De Giorgis, V.; Veggiotti, P. GLUT1 deficiency syndrome 2013: Current state of the art. Seizure 2013, 22, 803–811. [Google Scholar] [CrossRef]
  252. Arsov, T.; Mullen, S.A.; Damiano, J.A.; Lawrence, K.M.; Huh, L.L.; Nolan, M.; Young, H.; Touin, A.; Dahl, H.-H.M.; Bergovic, S.F.; et al. Early onset absence epilepsy: 1 in 10 cases is caused by GLUT1 deficiency. Epilepsia 2012, 53, e204–e207. [Google Scholar] [CrossRef] [PubMed]
  253. Benarroch, E.E. Brain glucose transporters. Implications for neurologic disease. Clin. Implic. Neurosci. Res. 2014, 85, 1374–1379. [Google Scholar]
  254. Patching, S.G. Glucose transporters at the blood-brain barrier: Function, regulation and gateways for drug delivery. Mol. Neurobiol. 2017, 54, 1046–1077. [Google Scholar] [CrossRef] [PubMed]
  255. Seider, G.; Alvarez, M.G.; Yeh, J.-J.; O’Driscoll, K.R.; Klepper, J.; Stump, T.S.; Wang, D.; Spiner, N.B.; Birnbaum, M.J.; De Vivo, D.C. GLUT-1 deficiency syndrome caused by haploinsufficiency of the blood-brain barrier hexose carrier. Nat. Genet. 1998, 18, 188–191. [Google Scholar] [CrossRef]
  256. Hashimoto, N.; Kagitani-Shimonok, K.; Sakai, N.; Otomo, T.; Tominaga, K.; Nabatame, S.; Mogami, Y.; Takahashi, Y.; Imai, K.; Yanagihara, K.; et al. SLC2A1 gene analysis of Japanese patients with glucose transporter 1 deficiency syndrome. J. Hum. Genet. 2011, 56, 846–851. [Google Scholar] [CrossRef]
  257. Leen, W.G.; Klepper, J.; Verbeek, M.M.; Leferink, M.; Hofste, T.; van Engelen, B.G.; Wevers, R.A.; Artur, T.; Bahi-Buisson, N.; Ballhausen, D.; et al. Glucose transporter-1 deficiency syndrome: The expanding clinical and genetic spectrum of a treatable disorder. Brain 2010, 133, 655–670. [Google Scholar] [CrossRef] [PubMed]
  258. Liu, Y.C.; Lee, J.W.; Bellows, S.T.; Damiano, J.A.; Mullen, S.A.; Berkovic, S.F.; Bahlo, M.; Scheffer, I.E.; Hildebrand, M.S.; Clinical, G. Evaluation of non-coding variation in GLUT1 deficiency. Dev. Med. Child Neurol. 2016, 58, 1295–1302. [Google Scholar] [CrossRef] [PubMed]
  259. Willemsen, M.A.; Vissers, L.E.; Verbeek, M.M.; Van Bon, B.W.; Geuer, S.; Gilissen, C.; Klepper, J.; Kwint, M.P.; Leen, W.G.; Pennings, M.; et al. Upstream SLC2A1 translocation initiation causes GLUT1 deficiency syndrome. Eur. J. Hum. Genet. 2017, 25, 771–774. [Google Scholar] [CrossRef] [PubMed]
  260. Klepper, J.; Scheffer, H.; Elsaid, M.F.; Kamsteeg, E.J.; Leferink, M.; Ben-Omran, T. Autosomal recessive inheritance of GLUT1 deficiency syndrome. Neuropediatrics 2009, 40, 207–210. [Google Scholar] [CrossRef]
  261. Rotstein, M.; Engelstad, K.; Yang, H.; Wang, D.; Levy, B.; Chung, W.K.; De Vivo, D.C. Glut1 deficiency: Inheritance pattern determined by haploinsufficiency. Am. Neurol. 2010, 68, 955–958. [Google Scholar] [CrossRef]
  262. Wang, D.; Pascual, J.M.; Yang, H.; Engelstad, K.; Ihung, S.; Sun, R.P.; De Vivo, D.C. Glut-1 deficiency syndrome: Clinical, genetic and therapeutic aspects. Ann. Neurol. 2005, 57, 111–118. [Google Scholar] [CrossRef]
  263. Klepper, J.; Akman, C.; Armeno, M.; Auvin, S.; Cervenko, M.; Cross, H.J.; De Giorgis, V.; Marina, M.; Engelstad, K.; Heussinger, N.; et al. Glut1 Deficiency Syndrome (Glut1DS): State of the art in 2020 and recommendations of the international Glut1DS study group. Epilepsia Open 2020, 5, 354–365. [Google Scholar] [CrossRef]
  264. Ohtsuki, S.; Kikkawa, T.; Hori, S.; Terasaki, T. Modulation and compensation of the mRNA expression of energy related transporters in the brain of glucose transporter 1-deficient mice. Biol. Pharm. Bull. 2006, 29, 1587–1591. [Google Scholar] [CrossRef]
  265. Wang, D.; Pascual, J.M.; Yang, H.; Engelstad, K.; Mao, X.; Chen, J.; Yoo, J.; Noebels, J.L.; De Vivo, D.C. A mouse model for Glut-1 haploinsufficiency. Hum. Mol. Genet. 2006, 15, 1169–1179. [Google Scholar] [CrossRef]
  266. Amolou, S.; Gras, D.; Ilea, A.; Greneche, M.D.; Francois, L.; Bellavoine, V.; Delanoe, C.; Auvin, S. Use of modified Atkins diet in glucose transporter type 1 deficiency syndrome. Des. Med. Child Neurol. 2016, 58, 1193–1199. [Google Scholar] [CrossRef] [PubMed]
  267. Lee, W.H.; Bondy, C.A. Ischemic injury induces brain glucose transporter gene expression. Endocrinology 1993, 133, 2540–2544. [Google Scholar] [CrossRef] [PubMed]
  268. Urabe, T.; Hattori, N.; Nagamatsu, S.; Sawa, H.; Mizuno, Y. Expression of glucose transporters in rat brain following transient focal ischemic injury. J. Neurochem. 1996, 67, 265–271. [Google Scholar] [CrossRef]
  269. Gerhart, D.Z.; Leino, R.L.; Taylor, W.E.; Borson, N.D.; Drewes, L.R. GLUT1 and GLUT3 gene expression in gerbil brain following brief ischemia: On in situ hybridization study. Brain Res. Mol. Brain Res. 1994, 25, 313–322. [Google Scholar] [CrossRef]
  270. Zhang, W.W.; Zhang, L.; Hou, W.K.; Xu, Y.X.; Hu, H.; Lou, F.C.; Zhang, Y.; Wang, Q. Dynamic expression of glucose transporters 1 and 3 in the brain of diabetic rats with cerebral ischemia reperfusion. Chin. Med. J. 2009, 122, 1996–2001. [Google Scholar] [PubMed]
  271. Chavez, J.C.; LaManna, J.C. Activation of hypoxia-inducible factor-1 in the rat cerebral cortex after transient global ischemia: Potential role of insulin-like growth factor-1. J. Neurosci. 2002, 22, 8922–8932. [Google Scholar] [CrossRef]
  272. McCall, A.L.; Van Bueren, A.M.; Nipper, V.; Moholt-Siebert, M.; Downes, H.; Lessov, N. Forebrain ischemia increases GLUT1 protein in brain microvessels and parenchyma. J. Cereb. Blood Flow Metab. 1996, 16, 69–76. [Google Scholar] [CrossRef]
  273. Weisova, P.; Concannon, C.G.; Devocelle, M.; Prehn, J.H.; Ward, M.W. Regulation of glucose transporter 3 surface expression by the AMP-activated protein kinase mediates tolerance to glutamate excitation in neurons. J. Neurosci. 2009, 29, 2997–3008. [Google Scholar] [CrossRef]
  274. Cruz, J. An additional therapeutic effect of adequate hyperventilation in severe acute brain trauma: Normalization of cerebral glucose uptake. J. Neurosurg. 1995, 82, 379–385. [Google Scholar] [CrossRef]
  275. Bergsneider, M.; Hovda, D.A.; Shalmon, E.; Kelly, D.F.; Vespa, P.M.; Martin, N.A.; Phelps, M.E.; McArthur, D.L.; Caron, M.J.; Kraus, J.F. Cerebral hyperglycolysis following severe traumatic brain injury in humans: A positron emission tomography study. J. Neurosurg. 1997, 86, 241–251. [Google Scholar] [CrossRef]
  276. Cornford, E.M.; Hyman, S.; Comford, M.E.; Caron, M.J. Glut1 glucose transporter activity in human brain injury. J. Neurotrauma 1996, 13, 523–536. [Google Scholar] [CrossRef] [PubMed]
  277. Umschweif, G.; Alexandrovich, A.G.; Trembovler, V.; Horowitz, M.; Shohami, E. Hypoxia-induces factor 1 is essential for spontaneous recovery from traumatic brain injury and is a key mediator of heat acclimation induced neuroprotection. J. Cereb. Blood Flow Metab. 2013, 33, 524–531. [Google Scholar] [CrossRef]
  278. Ding, J.Y.; Kreipke, C.W.; Speirs, S.L.; Schafer, P.; Schafer, S.; Rafols, J.A. Hypoxia-inducible factor-1 alpha signaling in aquaporin upregulation after traumatic brain injury. Neurosci. Lett. 2009, 453, 68–72. [Google Scholar] [CrossRef]
  279. Hamlin, G.P.; Cernak, I.; Wixey, J.A.; Vink, R. Increased expression of neuronal glucose transporter 3 but not glial glucose transporter 1 following severe diffuse traumatic brain injury in rats. J. Neurotrauma 2001, 18, 1011–1018. [Google Scholar] [CrossRef]
  280. Hu, L.; Wang, W.; Chen, X.; Bai, G.; Ma, L.; Yang, X.; Shu, Q.; Li, X. Prospects of antidiabetic drugs in the treatment of neurodegenerative disease. Brain-X 2024, 2, e52. [Google Scholar] [CrossRef]
  281. Cereda, E.; Barichella, M.; Pedrolli, C.; Klersy, C.; Cassani, E.; Caccialanza, R.; Pezzoli, G. Diabetes and risk of Parkinson’s disease: A systematic review and meta-analysis. Diabetes Care 2011, 34, 2614–2623. [Google Scholar] [CrossRef] [PubMed]
  282. Lu, L.; Fu, D.L.; Li, H.Q.; Liu, A.J.; Li, J.H.; Zheng, G.Q. Diabetes and risk of Parkinson’s disease: An updated meta-analysis of case-control studies. PLoS ONE 2014, 9, e85781. [Google Scholar] [CrossRef]
  283. Noyce, A.J.; Bestwick, J.P.; Silveira-Moriyama, L.; Hawkes, C.H.; Giovannoni, G.; Lees, A.J.; Schrag, A. Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann. Neurol. 2012, 72, 893–901. [Google Scholar] [CrossRef]
  284. Yue, X.; Li, H.; Yan, H.; Zhang, P.; Chang, L.; Li, T. Risk of Parkinson disease in diabetes mellitus: An updated meta-analysis of population-based cohort studies. Medicine 2016, 95, e3549. [Google Scholar] [CrossRef]
  285. Labandeira, C.M.; Fraga-Bau, A.; Ron, D.A.; Alvarez-Rodriguez, E.; Vincente-Alba, P.; Lago-Garma, J.; Rodriguez-Perez, A.I. Parkinson’s disease and diabetes mellitus: Common mechanisms and treatment repurposing. Neural Regen. Res. 2022, 17, 1652–1658. [Google Scholar] [CrossRef] [PubMed]
  286. Bednarz, K.; Siuda, J. Alzheimer’s disease and type 2 diabetes mellitus: Similarities in pathomechanisms lead to therapeutic opportunities. Neurol. Neurochir. Pol. 2021, 55, 418–428. [Google Scholar] [CrossRef] [PubMed]
  287. Patel, V.N.; Chorawala, M.R.; Shah, M.B.; Shah, K.C.; Dave, B.P.; Shah, M.P.; Patel, T.M. Emerging pathophysiological mechanisms linking diabetes mellitus and Alzheimer’s disease: An old wine in a new bottle. J. Alzheimer’s Dis. Rep. 2022, 6, 349–357. [Google Scholar] [CrossRef] [PubMed]
  288. Hölscher, C. Protective properties of GLP-1 and associated hormones in neurodegenerative disorders. Br. J. Pharmacol. 2022, 179, 695–714. [Google Scholar] [CrossRef]
  289. Li, Y.; Perry, T.; Kindy, M.S.; Harvey, B.K.; Tweedie, D.; Holloway, H.W.; Powers, K.; Shen, H.; Egan, J.M.; Sambamurti, K.; et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc. Natl. Acad. Sci. USA 2009, 106, 1285–1290. [Google Scholar] [CrossRef]
  290. Bertilsson, G.; Patrone, C.; Zachrison, O.; Andersson, A.; Dannaeus, K.; Heidrich, J.; Kortesmaa, J.; Mercer, A.; Nielsen, E.; Rönnholm, H.; et al. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J. Neurosci. Res. 2008, 86, 326–338. [Google Scholar] [CrossRef]
  291. Harkavyi, A.; Abuirmeileh, A.; Lever, R.; Kingsbury, A.E.; Biggs, C.S.; Whitton, P.S. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson’s disease. J. Neuroinflamm. 2008, 5, 19. [Google Scholar] [CrossRef]
  292. Qi, L.; Ke, L.; Liu, X.; Liao, L.; Ke, S.; Liu, X.; Wang, Y.; Lin, X.; Zhou, Y.; Wu, L.; et al. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulation tau hyperphosphorylation via the glycogen synthase kinase-3β pathway in an amyloid β protein induced Alzheimer disease mouse model. Eur. J. Pharmacol. 2016, 783, 23–32. [Google Scholar] [CrossRef]
  293. McClean, P.L.; Parthsarathy, V.; Faivre, E.; Hölscher, C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J. Neurosci. 2011, 31, 6587–6594. [Google Scholar] [CrossRef] [PubMed]
  294. Han, W.N.; Hölscher, C.; Yuan, L.; Yang, W.; Wang, X.-H.; Wu, M.-N.; Qi, J.-S. Liraglutide protects against amyloid-β protein-induced impairment of spatial learning and memory in rats. Neurobiol. Aging 2013, 34, 576–588. [Google Scholar] [CrossRef] [PubMed]
  295. Chang, C.C.; Lin, T.C.; Ho, H.L.; Kuo, C.-Y.; Li, H.-H.; Korolenko, T.A.; Chen, W.-J.; Lai, T.-J.; Ho, Y.-J.; Lin, C.-L. GLP-1 analogue liraglutide attenuates mutant huntingtin-induced neurotoxicity by restoration of neuronal insulin signaling. Int. J. Mol. Sci. 2018, 19, 2505. [Google Scholar] [CrossRef]
  296. Nizari, S.; Basalay, M.; Chapman, P.; Korte, N.; Korsak, A.; Christie, I.N.; Theparambil, S.M.; Davidson, S.M.; Reimann, F.; Trapp, S.; et al. Glucagon-like peptide-1 (GLP-1) receptor activation dilates cerebral arterioles, increases cerebral blood flow, and mediates remote (pre)conditioning neuroprotection against ischemic stroke. Basic Res. Cardiol. 2021, 116, 32. [Google Scholar] [CrossRef] [PubMed]
  297. An, J.; Zhou, Y.; Zhang, M.; Xie, Z.; Ke, S.; Liu, L.; Pan, X.; Chen, Z. Exenatide alleviates mitochondrial dysfunction and cognitive impairment in 5xFAD, mouse model of Alzheimer’s disease. Behav. Brain Res. 2019, 370, 111932. [Google Scholar] [CrossRef]
  298. Garabadu, D.; Verma, J. Extendin-4 attenuates brain mitochondrial toxicity through PI3K/Akt-independent pathway in amyloid (1–42)-induced cognitive deficit rats. Neurochem. Int. 2019, 128, 39–49. [Google Scholar] [CrossRef]
  299. Yu, H.Y.; Sun, T.; Wang, Z.; Li, H.; Xu, D.; An, J.; Wen, L.L.; Li, J.Y.; Li, W.; Feng, J. Exendin-4 and lingliptin attenuate neuroinflammation in a mouse model of Parkinson’s disease. Neural. Regen. 2023, 18, 1818–1826. [Google Scholar]
  300. Nowell, J.; Blunt, E.; Edison, P. Incretin and insulin targets for Alzheimer’s and Parkinson’s disease. Mol. Psychiatr. 2023, 28, 217–229. [Google Scholar] [CrossRef]
  301. Hölscher, C. Brain insulin resistance: Role in neurodegenerative disease and potential for targeting. Expert Opin. Investig. Drugs 2020, 29, 333–348. [Google Scholar] [CrossRef] [PubMed]
  302. Verma, M.K.; Goel, R.; Nandakumar, K.; Nemmani, K.V.S. Effect of D-Ala2GIP, a stable GIP receptor agonist on MPTP-induced neuronal impairments in mice. Eur. J. Pharmacol. 2017, 84, 38–45. [Google Scholar] [CrossRef] [PubMed]
  303. Duffy, A.M.; Hölscher, C. The incretin analogue D-Ala261P reduces plague load, astrogliosis and oxidative stress in an APP/PS1 mouse model of Alzheimer’s disease. Neuroscience 2013, 228, 294–300. [Google Scholar] [CrossRef] [PubMed]
  304. Li, Y.; Liu, W.; Li, L.; Hölscher, C. D-Ala2-GIP-glu-PAL is neuroprotective in a chronic Parkinson’s disease mouse model and increases BNDF expression while reducing neuroinflammation and lipid peroxidation. Eur. J. Pharm. 2017, 797, 162–172. [Google Scholar] [CrossRef]
  305. Panagaki, T.; Gengler, S.; Hölscher, C. The novel DA-CH3 dual incretin restores endoplasmic reticulum stress and autophagy impairments to attenuate Alzheimer-like pathology and cognitive decrements in the APPSWE/PS1ΔE9 mouse model. J. Alzheimer’s Dis. 2018, 66, 195–218. [Google Scholar] [CrossRef]
  306. Maskery, M.; Goulding, E.M.; Gengler, S.; Melchiorsen, J.U.; Rosenkilde, M.M.; Holscher, C. The dual GLP/GIP receptor agonist DA4-JC shows superior protective properties compared to GLP-1 analog Liraglutide in APP/PS1 mouse model of Alzheimer’s disease. Am. J. Alzheimer’s Dis. Other Demen. 2020, 35, 1533317520953041. [Google Scholar] [CrossRef]
  307. Cao, Y.; Hölscher, C.; Hu, M.-M.; Wang, T.; Zhao, F.; Bai, Y.; Zhang, J.; Wu, M.-N.; Qi, J.-S. DAS-CH, a novel GLP-GIP dual agonist, effectively ameliorates the cognitive impairments and pathology in the APP/PS1 mouse model of Alzheimer’s disease. Eur. J. Pharmacol. 2018, 827, 215–226. [Google Scholar] [CrossRef]
  308. Tai, J.; Liu, W.; Li, Y.; Hölscher, C. Neuroprotective effects of a triple GLP-1/GIP/glucagon receptor agonist in the APP/PS1 transgenic mouse model of Alzheimer’s disease. Brain Res. 2018, 1678, 64–74. [Google Scholar] [CrossRef]
  309. Li, T.; Jiao, J.J.; Hölscher, C.; Wu, M.N.; Zhang, J.; Tong, J.Q.; Dong, X.-F.; Qu, X.-S.; Cao, Y.; Cai, H.-Y.; et al. A novel GLP-1/GIP/Gcg triagonist reduces cognitive deficits and pathology in the 3xTg mouse model of Alzheimer’s disease. Hippocampus 2018, 28, 358–372. [Google Scholar] [CrossRef]
  310. Li, T.; Jiao, J.J.; Su, Q.; Hölscher, C.; Zhang, J.; Yan, X.D.; Zhao, H.-M.; Cai, H.-Y.; Qi, J.-S. A GLP-1/GIP/Gcg receptor trigonist improves memory behavior, as well as synaptic transmission, neuronal excitability and Ca2+ homeostasis in 3xTg-AD mice. Neuropharmacology 2020, 170, 108042. [Google Scholar] [CrossRef]
  311. Choi, I.Y.; Lee, J.S.; Kim, J.K.; Park, Y.J.; Lee, S.H.; Kim, Y.H.; Kim, S.J.; Kwon, S.C. Potent body weight loss and efficacy in a NASH animal model by a novel long-lasting GLP-1/Glucagon/GIP triple-agonist (HM15211). In Proceedings of the American Diabetes Association’s 77th Scientific Session, San Diego, CA, USA, 9–13 June 2017. [Google Scholar]
  312. Wonki, K.; Kim, J.A.; Lee, S.H.; Bae, S.; Choi, I.Y.; Kim, Y.H. 1810-P: Effect of HM15211, a novel long-acting GLP-1/GIP/Glucagon triple agonist in the neurodegenerative disease models. Diabetes 2019, 68, 1810-P. [Google Scholar] [CrossRef]
  313. Egan, M.F.; Kost, J.; Tariot, P.N.; Tariot, P.N.; Aisen, P.S.; Cummings, J.L.; Vellas, B.; Sur, C.; Michelson, D. Randomized trial of verubecestat for mild-to-moderate Alzheimer’s disease. N. Engl. J. Med. 2018, 378, 1691–1703. [Google Scholar] [CrossRef]
  314. Kosaraju, J.; Holsinger, R.M.; Guo, L.; Tam, K.Y. Linagliptin, a dipeptidyl peptidase-4 inhibitor, mitigates cognitive deficits and pathology in the 3xTg-AD mouse model of Alzheimer’s disease. Mol. Neurobiol. 2017, 54, 6074–6084. [Google Scholar] [CrossRef]
  315. Brauer, R.; Wei, L.; Ma, T.; Athuda, D.; Girges, C.; Vijiaratnam, N.; Auld, G.; Whittlesea, C.; Wong, I.; Foltynie, T. Diabetes medications and risk of Parkinson’s disease: A cohort study of patients with diabetes. Brain 2020, 143, 3067–3076. [Google Scholar] [CrossRef]
  316. Svennigsson, P.; Wirdefeldt, K.; Yin, L.; Fang, F.; Markaki, I.; Efendic, S.; Ludvigsson, J.F. Reduced incidence of Parkinson’s disease after dipeptidyl peptidase-4 inhibitors–A nationwide case-control study. Mov. Disord. 2016, 31, 1422–1423. [Google Scholar] [CrossRef]
  317. Justin, A.; Ananda Kumar, T.D.; Chinaswamy, M.; Kumar, B.R.P. Glitazones activate PGC-1α signaling via PPAR-γ a promising strategy for antiparkinsonism therapeutics. ACS Chem. Neurosci. 2021, 12, 2261–2272. [Google Scholar]
  318. Agarwal, S.; Yada, A.; Chaturvedi, R.K. Peroxisome proliferator activated receptors (PPARs) as therapeutic target in neurodegenerative disorders. Biochem. Biophys. Res. Commun. 2017, 483, 1166–1177. [Google Scholar] [CrossRef]
  319. Liu, J.; Wang, L.N.; Jia, J.P. Peroxisome proliferator-activated receptor-gamma agonists for Alzheimer’s disease and amnestic mild cognitive impairment: A systematic review and meta-analysis. Drugs Aging 2015, 32, 57–65. [Google Scholar] [CrossRef]
  320. Risner, M.E.; Saunders, A.M.; Altman, J.F.B.; Ormandy, G.C.; Craft, S.; Foley, I.M.; Zvartau-Hind, M.E.; Hosford, D.A.; Roses, A.D.; Rosiglitazone in Alzheimer’s Disease Study Group. Efficacy of rosiglitazone in a genetically defined population with mild-to moderate Alzheimer’s disease. Pharmacogenomics 2006, 6, 246–254. [Google Scholar] [CrossRef] [PubMed]
  321. Gold, M.; Alderton, C.; Zvartau-Hind, M.; Egginton, S.; Saunders, A.M.; Irizarry, M.; Craft, S.; Landreth, G.; Linnamägi, U.; Sawchak, S. Rosiglitazone, monotherapy in mild-to-moderate Alzheimer’s disease: Results from a randomized, double-blind, placebo-controlled phase III study. Dement. Geriatr. Cogn. Disord. 2010, 30, 131–146. [Google Scholar] [CrossRef]
  322. Sluggett, J.K.; Koponen, M.; Bell, J.S.; Taipale, H.; Tnaskanen, A.; Tiihonen, J.; Uusitupa, M.; Tolppanen, A.; Hartikainen, S. Metformin and risk of Alzheimer’s disease among community-dwelling people with diabetes: A randomized case-control study. J. Clin. Endocrinol. Metab. 2020, 105, e963–e972. [Google Scholar] [CrossRef]
  323. Ping, F.; Jiang, N.; Li, Y. Association between metformin and neurodegenerative diseases of observational studies: Systematic review and meta-analysis. BMJ Open Diab. Res. Care 2020, 8, e001370. [Google Scholar] [CrossRef]
  324. Müller, T.D.; Blüher, M.; Tschöp, M.H.; DiMarchi, R.D. Anti-obesity drug discovery: Advances and challenges. Nat. Rev. Drug Discov. 2022, 21, 201–223. [Google Scholar] [CrossRef]
  325. McLean, B.A.; Wong, C.K.; Campbell, J.E.; Hodson, D.J.; Trapp, S.; Drucker, D.J. Revisiting the complexity of GLP-1 action from sites of synthesis to receptor reaction. Endocr. Rev. 2021, 42, 101–132. [Google Scholar] [CrossRef]
  326. Heppner, K.M.; Kirigiti, M.; Secher, A.; Paulsen, S.J.; Buckingham, R.; Pyke, C.; Knudsen, L.B.; Vrang, N.; Grove, K.L. Expression and distribution of glucose-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain. Endocrinology 2015, 156, 255–267. [Google Scholar] [CrossRef]
  327. Farr, O.M.; Sofopoulos, M.; Tsoukas, M.A.; Dincer, F.; Thakkar, B.; Sahin-Efe, A.; Filippaios, A.; Bowers, J.; Srnka, A.; Gavrieli, A.; et al. GLP-1 receptors exist in the parietal cortex, hypothalamus and medulla of human brains and the GLP-1 analogue liraglutide alters brain activity related to highly desirable food cues in individuals with diabetes: A crossover randomized, placebo-controlled trial. Diabetologia 2016, 59, 954–965. [Google Scholar]
  328. Kim, D.S.; Choi, H.I.; Wang, Y.; Luo, Y.; Hoffer, B.J.; Greig, N.H. A new treatment strategy for Parkinson’s disease through the gut-brain axis: The glucagon-like peptide-1 receptor pathway. Cell Transplant. 2017, 26, 1560–1571. [Google Scholar] [CrossRef]
  329. Joint Formulary Committee. British National Formulary on Line; BMJ Group and Pharmaceutical Press: London, UK, 2020. [Google Scholar]
  330. Nyberg, J.; Anderson, M.F.; Alborn, A.; Stro, A.; Brederlau, A.; Illerskog, A.; Nilsson, O.; Kieffer, T.J.; Hietala, M.A.; Rickstein, A.; et al. Glucose-dependent insulinotropic polypeptide is expressed in adult hippocampus and induces progenitor cell proliferation. J. Neurosci. 2005, 25, 1816–1825. [Google Scholar] [CrossRef]
  331. Nyberg, J.; Jacobsson, C.; Andersen, M.F.; Eriksson, P.S. Immunohistochemical distribution of glucose-dependent insulinotropic polypeptide in the adult rat brain. J. Neurosci. 2007, 85, 2099–2119. [Google Scholar] [CrossRef]
  332. Feng, P.; Zhang, X.; Li, D.; Ji, C.; Yuan, Z.; Wang, R.; Xue, G.; Li, G.; Hölscher, C. Two dual GLP1/GIP receptor agonists are neuroprotective in the MPTP mouse model of Parkinson’s disease. Neuropharmacology 2018, 133, 385–394. [Google Scholar] [CrossRef]
  333. Li, H.; Wu, J.; Zhu, L.; Sha, L.; Yang, S.; Wei, J.; Ji, L.; Tang, X.; Mao, K.; Cao, L.; et al. Insulin degrading enzyme contributes to the pathology in a mixed model of Type 2 diabetes and Alzheimer’s disease: Possible mechanisms of IDE in T2D and AD. Biosci. Rep. 2018, 38, BSR20170862. [Google Scholar] [CrossRef]
  334. Ou, Z.; Kong, X.; Sun, X.; He, X.; Zhang, L.; Gong, Z.; Huang, J.; Xu, B.; Long, D.; Li, J.; et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 2018, 69, 351–363. [Google Scholar] [CrossRef]
  335. Farr, S.A.; Raesler, E.; Niehoff, M.L.; Roby, D.A.; Mckee, A.; Morley, J.E. Metformin improves learning and memory in the SAMP8 mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2019, 68, 1699–1710. [Google Scholar] [CrossRef]
  336. Biosa, A.; Outeiro, T.F.; Bubacco, L.; Bisaglia, M. Diabetes mellitus as a risk factor for Parkinson’s disease: A molecular point of view. Mol. Neurobiol. 2018, 55, 8754–8763. [Google Scholar] [CrossRef]
  337. Moore, E.M.; Mander, A.G.; Ames, D.; Kotowicz, M.A.; Carne, R.P.; Brodaty, H.; Woodward, M.; Boundy, K.; Ellis, K.A.; Bush, A.I.; et al. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care 2013, 36, 2981–2987. [Google Scholar] [CrossRef]
  338. Yaribeygi, H.; Ashrafizadeh, M.; Henney, N.C.; Sathyapalan, T.; Jamialahmadi, T.; Sabebkar, A. Neuromodulatory effects of anti-diabetes medications: A mechanistic review. Pharmacol. Res. 2020, 152, 104611. [Google Scholar] [CrossRef] [PubMed]
  339. Labandeira, C.M.; Fraga-Bau, A.; Ron, D.A.; Muñoz, A.; Alonso-Losada, G.; Koukoulis, A.; Romero-Lopez, J.; Rodriguez-Perez, A.I. Diabetes, insulin and new therapeutic strategies for Parkinson’s disease: Focus on glucagon-like peptide-1 receptor agonists. Front. Neuroendocrinol. 2021, 62, 100914. [Google Scholar] [CrossRef] [PubMed]
  340. Ryu, Y.; Go, J.; Park, H.; Choi, Y.; Jeong, Y.; Hyeon, J.; Rhee, M.; Geal, T.; Lee, C.; Kim, K. Metformin regulates astrocyte reactivity in Parkinson’s disease and normal aging. Neuropharmacology 2020, 175, 108173. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The N-linked glycosylation sites in Class I and Class II of glucose transporters (GLUTs) (upper panel) and in Class III (lower panel) [19].
Figure 1. The N-linked glycosylation sites in Class I and Class II of glucose transporters (GLUTs) (upper panel) and in Class III (lower panel) [19].
Ijms 26 07392 g001
Figure 2. Structure of the human sodium-dependent glucose cotransporter (SGLT) protein. Presented site of N-glycosylation is for SGLT1 [120].
Figure 2. Structure of the human sodium-dependent glucose cotransporter (SGLT) protein. Presented site of N-glycosylation is for SGLT1 [120].
Ijms 26 07392 g002
Table 1. General characteristics of human glucose transporters (GLUTs).
Table 1. General characteristics of human glucose transporters (GLUTs).
Glucose Transporter
Gene
Localization
Organ/Tissue of Transporter ExpressionTransported SubstratesReferences
GLUT1
SLC2A1
1p35-p31.3
The large-molecular-weight GLUT1 (55 kDa) is detected in the cerebral cortex microvessels; the low-molecular-weight isoform (45 kDa) is present in the microvessel-depleted membrane of the brain (neuronal/glial membranes) and synaptosomes; an intermediate isoform of this transport protein is detected in the choroid plexus. It is expressed in fetal cells, from oocyte to the blastocyst. This transporter transports glucose through the epithelial or epithelial-like barrier cells of the brain, eye, peripheral nerve, and placenta. These barriers express high levels of GLUT1. Its expression is detected also in the blood cells, such as erythrocytes, granulocytes, and lymphocytes, as well as in adipocytes, muscle cells, kidney cells, and colon. Expression of GLUT1 was also detected in mitochondria of the human kidney cells 293T.Glucose, galactose, mannose, glucosamine, the oxidized form of vitamin C.[19,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46]
GLUT2
SLC2A2
3q26.1-26.2.
It is expressed in the intestinal absorptive epithelial cells, hepatocytes, pancreatic β cells, and in the kidney cells. In the epithelial cells, GLUT2 is expressed in the basolateral membrane. In the brain, it is expressed in the brainstem, thalamus, cortex, hypothalamus, and hippocampus. Low levels of GLUT2 mRNA are detected in the nuclei of the brain, such as the nucleus tractus solitatius, the motor nucleus of the vagus, the paraventricular hypothalamic nucleus, the lateral hypothalamic area, the arcuate nucleus, and the olfactory bulbs, as well as in the neurons of limbic areas and related nuclei.Glucose, galactose, mannose, fructose, glucosamine.[17,20,30,42,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61]
GLUT3
SLC2A3
12p13.3
Its high expression is detected in the brain, especially in the neurons, while not in the glia. In the brain, its expression is detected in the hippocampus, gyrus, and temporal neocortex. It is also found in tissues and cells with high demand for glucose, such as spermatozoa, placenta, preimplantation embryos, fibroblasts, platelets, white blood cells, and retinal endothelial cells.Glucose, galactose.[7,16,17,19,30,32,33,57,62,63,64,65,66,67,68]
GLUT4
SLC2A4
17p13
Its preferential sites of expression are the skeletal muscle, cardiomyocytes, and adipose tissue. It is expressed also in the brain in the hypothalamus, cerebellum, cortex, and hippocampus.Glucose, dehydroascorbic acid, glucosamine.[20,57,69,70,71,72,73,74,75,76]
GLUT5
SLC2A5
1p36.2
At high levels, it is expressed in the apical membrane of enterocytes. GLUT5 mRNA levels increase with age, and its highest levels are found in the small intestine of adults. Its expression is found also in the small intestine (proximal region). GLUT5 mRNA is detected in the plasma membrane of mature spermatozoa, and GLUT5 mRNA and/or GLUT5 protein have been detected in the kidneys, adipose tissue, skeletal muscle, and in the brain. Its expression is also detected in the microglial cells of the brain, but only in microglia among cells of the mononuclear phagocyte system.Fructose.[16,17,19,20,30,33,50,56,57,77,78,79,80,81,82,83,84]
GLUT6
Formerly designated as GLUT9
SLC2A6
9q34
It is predominantly expressed in the brain, peripheral leukocytes, spleen, and germinal cells of the testis.Glucose.[16,17,20,85,86]
GLUT7
SLC2A7
1p36.22
Its primary expression is detected in the apical membrane of the small intestine (distal region) and colon. Its mRNA is detected in the testis and prostate.Glucose, fructose.[17,20,87,88,89]
GLUT8
Formerly designated as GLUTX1
SLC2A8
9q33.3
It is detected in endosomes, lysosomes, and membranes of the endoplasmic reticulum. GLUT8 mRNA is detected in the testis, cerebellum, adrenal gland, liver, spleen, and brown adipose tissue. In the brain, expression of GLUT8 is reported in the hippocampus, dente gyrus, amygdala, primary olfactory cortex, hypothalamic nuclei, and tractus solitaries in the blood–retinal barrier. It is also reported in the endometrium and endometrial adenocarcinoma and in blastocysts. It transports sugars or sugar derivatives through the intracellular membranes.Glucose.[16,19,20,57,90,91,92,93,94,95]
GLUT9
Formerly designated as GLUTX
SLC2A9
4p15.3-p16
Due to the use of alternative promoters, the gene encodes two isoforms of GLUT9: the major isoform, i.e., GLUT9 or GLUT9a, or an alternative splice variant of GLUT9 mRNA that codes the GLUT9ΔN or GLUT9b isoform. GLUT9 is expressed in the liver, kidneys, intestine, leukocytes, and chondrocytes, whereas GLUT9b is detected only in the liver and kidneys. GLUT9a is expressed in the basolateral membrane of the epithelial cells, while GLUT9b is localized to the apical pole.Urate, glucose, fructose.[19,20,96,97,98,99,100,101,102]
GLUT10
SLC2A10
20q13.1
It is detected in insulin-sensitive tissues such as the skeletal muscle and heart. GLUT10 mRNA is detected in the liver, pancreas, placenta, and the kidneys, as well as in human adipose tissue (omental and subcutaneous), in human preadipocyte cell strains (SGBS), and in 3T3-L1 adipocytes. Deoxy-D-glucose,
D-galactose.
[17,20,24,103,104,105]
GLUT11 Formerly designated as GLUT10
SLC2A11
22q11.2
In humans, three isoforms of GLUT11 are expressed. These isoforms are due to the separate exons (exons 1A, 1B, and 1C) of the SLC2A11 gene, which generate three variants of GLUT11 (GLUT11-A, GLUT11-B, and GLUT11-C). GLUT11-A is detected in the heart, skeletal muscle, and in the kidneys; GLUT11-B is expressed in the kidneys, adipose tissue, and the placenta; and GLUT11-C is expressed in the adipose tissue, heart, skeletal muscle, and pancreas. This transporter has no rodent ortholog.Fructose, glucose.[19,20,106,107]
GLUT12
Formerly designated
as GLUT8
SLC2A12
6q23.2
It is detected in the heart, skeletal muscle, adipose tissue, prostate glands, kidneys, small intestine, and chondrocytes. In the human placenta, different expressions of GLUT12 have been detected. In the first trimester, this glucose transporter is detected predominantly in the syncytiotrophoblast, in the lesser extent in the villous cytotrophoblast, and in extra-villous trophoblast cells. At term, GLUT12 is detected predominantly in the villous vascular smooth muscle and stromal cells, whereas in the syncytiotrophoblast, it is not detected.Glucose, galactose, fructose.[16,20,21,24,108,109,110]
GLUT13 (HMIT)
SLC2A13
22q12
GLUT13 (HMIT) is an H+/myo-inositol cotransporter. Its high expression is detected predominantly in the brain in the hippocampus, hypothalamus, cerebellum, and brainstem regions. In the brain, this transporter is expressed in neurons and in the glial cells. Low levels of GLUT13 are detected in the white and brown adipose tissue and in the kidneys. It uses a proton gradient that is a source of energy for movement of substrate.Myo-inositol, inositol-3-phosphate.[17,19,20,111,112,113]
GLUT14
SLC2A14
12p13.3
It is identified and has been cloned as a duplication of GLUT3. There are two alternatively spliced forms: the short form of GLUT14 (GLUT14-S) and the long form of GLUT14 (GLUT14-L). Both isoforms are detected in the human testis. The ortholog of GLUT14 is not detected in mice.Glucose, fructose.[114]
Table 2. General characteristics of the human sodium-dependent glucose cotransporters.
Table 2. General characteristics of the human sodium-dependent glucose cotransporters.
Glucose Cotransporter
Gene Localization
Organ/Tissue of Cotransporter ExpressionTransported SubstratesReferences
SGLT1
SLC5A1
22q12.3
It is expressed in the BBM of the mature enterocytes in the small intestine, trachea, prostate gland, heart, kidneys, mammary glands, liver, and in the endothelial cells of the luminal membrane of intracerebral capillaries. Its expression is detected in the cortical, pyramidal, and Purkinje neuronal cells of the brain; in neurons (hippocampal) and the cortical granule (pyramidal); and the BBB. It is suggested that in the brain, SGLT1 plays a role as a glucose receptor. Its mRNA is also detected in the uterus (cervix).Water molecules, urea, fluid across the intestine;
D-glucose;
D-galactose.
[2,20,77,119,120,121,122,124,125,126,127,128,129,130,131,132,133,134,135,136]
SGLT2
SLC5A2
16p11.2
Its predominant expression is detected on the apical membrane of renal convoluted proximal tubules (S1 and S2 segments). SGLT mRNA is detected also in the mammary glands, liver, lungs, intestine, skeletal muscle, and spleen. It is suggested also to have a role in the heart and brain as the receptor of glucose.Glucose.[2,20,119,120,137,138,139,140,141]
SGLT3
Alias SAAT1
SLC5A4
22q12.3
Its expression is detected in the kidneys, uterus, testis, intestinal autonomic nervous system, skeletal muscle, brain, the cholinergic neurons in the enteric nervous system, pancreas, lungs, and liver.Sodium absorption in kidneys.[20,119,121,131,142,143]
SGLT4
SLC5A9
1p32
Its expression is detected in the small intestine, kidneys, brain, liver, heart, uterus, and lungs. SGLT4 mRNA is detected in the testis, pancreas, and skeletal muscle. D-mannose,
D-fructose,
D-glucose.
[2,104,119,121,131,144]
SGLT5
SLC5A10
17p11.2
SGLT5 is expressed in the human kidney cortex, and its mRNA is detected in the left atrium of the heart, ovary, skin (foreskin), testis, and vas deferens. Its precise location on the chromosome remains unknown.Mannose, fructose,
α-methyl-D-glucopyanoside.
[121,131,145,146]
SGLT6
SMIT2
SLC5A11 16p12.1
SGLT6 protein and its mRNA are detected in the brain, heart, kidneys, skeletal muscle, spleen, liver, placenta, lungs, and leukocytes. The SLC5A11 gene reveals interaction with immune-related genes, and therefore, it is suggested that it may play a role as an autoimmune modifier gene.Myo-inositol, D-chiro-inositol,
D-glucose,
D-xylose.
[121,131,146,147,148,149]
SMIT1
SLC5A3
21q22.11
There are three transcripts, SMIT1-1, SMIT1-2, and SMIT1-3, caused by splicing within and distal to exon 2. Its expression is detected in the kidneys, choroid plexus, brain, placenta, pancreas, heart, skeletal muscle, and lungs. SMIT1 mRNA is found in the blood vessels, choroid plexus, and uterus (cervix).Myo-inositol,
L-fucose,
L-xylose.
[2,131,146,150]
NIS
SLC5A5
19p13.11
It is a sodium/iodide cotransporter mainly expressed in the thyroid as well as in the lactating breast, colon, stomach, and ovaries.I, ClO4, SCN, NO3, Br.[2,151,152]
SMVT
SLC5A6
2q12
It is detected in the brain, heart, kidneys, lungs, and placenta. SMVT is a multivitamin/sodium cotransporter.Vitamins such as pantothenic acid, biotin,
α-lipoic acid.
[2]
CHT1
SLC5A7
2q13
It is expressed in the central nervous system, spinal cord, and medulla.Choline.[2,153]
SMCT1
SLC5A8
12q23.1
Its expression is detected in the small intestine, kidneys, brain, retina, and muscle.Lactate, pyruvate, nicotinate.[2,154]
SMCT2
SLC5A12
11p14.2
Its expression is detected in the small intestine, kidneys, brain, retina, and muscle.Lactate, pyruvate, nicotinate.[2,154,155]
Table 3. Comparison and characteristics of medications used in pharmacologic treatment of NDs.
Table 3. Comparison and characteristics of medications used in pharmacologic treatment of NDs.
Anti-Diabetic
Drugs
Mechanism of Action and Effects [References]
IncretinsGlucagon-like peptide-1 (GLP-1) receptor agonists:
Liraglutide reveals neuroprotective properties in ND patients and ND animal models [288,289,290,291]. It ameliorates learning and memory impairment in animal models of AD. Liraglutide attenuates mutant huntingtin-induced neurotoxicity in human neuronal cells [292,293,294,295]. These agonists promote neuroprotection against ischemic stroke [296].

Exenatide halts cognitive decline in animal models of AD [297,298] and improves degeneration of dopaminergic neurons and motor function in PD animal models [289,299].
Exendin-4 decreases mitochondrial toxicity, alleviates cognitive dysfunction, prevents Aβ accumulation, and protects mitochondrial functions [297,298].

Glucose-dependent insulinotropic polypeptide (GIP) receptor agonists:
These agonists have neuroprotective properties in animal models of AD and PD [300,301,302] and reduce the activation of chronic inflammatory response in the brain, oxidative stress, synapse loss, amyloid plaque burden, and DNA damage. They reduce also lipid peroxidation and α-synuclein levels, protecting dopaminergic neurons in the substantia nigra [303,304].

Dual-receptor agonists of GLP-1 and GIP, which activate both GLP-1 and GIP receptors, show promising effects in animal models of AD and PD [300,301]. These novel agonists rescue or prevent spatial learning and memory dysfunction [305,306,307], reduce amyloid plaques and phosphorylated tau protein [305], and prevent overactivation of p-GSK3β. In animal models of PD, these agonists improve motor function and protect dopaminergic neurons.

Triple-receptor agonists activate GLP-1, GIP, and glucagon (Gcg) receptors. Treatment of a mouse model of AD with these agonists revealed anti-apoptotic effects and reduced Aβ deposition and hyperphosphorylated tau protein. These agonists protect against loss of synapses and reduce inflammatory symptoms and oxidative stress [308,309,310]. In PD mice, the triple-receptor agonist protects against dopaminergic neuronal death, decreases α-synuclein levels, and prevents nigrostriatal neurodegeneration [311,312].
Dipeptidyl Peptidase-4 InhibitorsLinagliptin improves cognitive function and decreases tau phosphorylation and Aβ aggregation in animal models of AD [313,314]. It has neuroprotective effects on human nerve cells caused by the protection of neurons from the effect of Aβ on mitochondrial damage, oxidative stress, and impaired insulin signaling. In humans who receive DPP-4 inhibitors, the incidence of PD is significantly decreased; however, high doses of inhibitors must be used [315,316].
ThiazolidinedionesThis class of drugs reveals neuroprotective effects in animal models of AD and PD, significantly improving behavior and motor responses [317,318,319]. Rosiglitazone improves spatial memory tested in the Morris water maze. Treatment of AD patients with rosiglitazone revealed different results in the case of dementia. Some results indicate improved cognitive performance in these patients [320], while other results do not confirm this effect [321].
BiguanidesMetformin decreases α-synuclein expression in animal models of PD, and in AD animal models, it has a beneficial effect on cognitive function [322]. This effect is due to the promotion of neurogenesis and prevention of amyloid deposition and tau phosphorylation [320,321]. Performed observations revealed that metformin may have a negative effect in patients. This drug may stimulate the production of Aβ, and in diabetic patients treated with metformin, a significantly higher incidence of PD was observed in comparison with non-metformin diabetic patients [323]. A meta-analysis that studied the effects of metformin on neurodegeneration revealed that this drug has no effect on NDs [323].
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szablewski, L. Human Glucose Transporters in Health and Selected Neurodegenerative Diseases. Int. J. Mol. Sci. 2025, 26, 7392. https://doi.org/10.3390/ijms26157392

AMA Style

Szablewski L. Human Glucose Transporters in Health and Selected Neurodegenerative Diseases. International Journal of Molecular Sciences. 2025; 26(15):7392. https://doi.org/10.3390/ijms26157392

Chicago/Turabian Style

Szablewski, Leszek. 2025. "Human Glucose Transporters in Health and Selected Neurodegenerative Diseases" International Journal of Molecular Sciences 26, no. 15: 7392. https://doi.org/10.3390/ijms26157392

APA Style

Szablewski, L. (2025). Human Glucose Transporters in Health and Selected Neurodegenerative Diseases. International Journal of Molecular Sciences, 26(15), 7392. https://doi.org/10.3390/ijms26157392

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop