Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 1429, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082341

ABSTRACT

The passive transport of glucose and related hexoses in human cells is facilitated by members of the glucose transporter family (GLUT, SLC2 gene family). GLUT3 is a high-affinity glucose transporter primarily responsible for glucose entry in neurons. Changes in its expression have been implicated in neurodegenerative diseases and cancer. GLUT3 inhibitors can provide new ways to probe the pathophysiological role of GLUT3 and tackle GLUT3-dependent cancers. Through in silico screening of an ~ 8 million compounds library against the inward- and outward-facing models of GLUT3, we selected ~ 200 ligand candidates. These were tested for in vivo inhibition of GLUT3 expressed in hexose transporter-deficient yeast cells, resulting in six new GLUT3 inhibitors. Examining their specificity for GLUT1-5 revealed that the most potent GLUT3 inhibitor (G3iA, IC50 ~ 7 µM) was most selective for GLUT3, inhibiting less strongly only GLUT2 (IC50 ~ 29 µM). None of the GLUT3 inhibitors affected GLUT5, three inhibited GLUT1 with equal or twofold lower potency, and four showed comparable or two- to fivefold better inhibition of GLUT4. G3iD was a pan-Class 1 GLUT inhibitor with the highest preference for GLUT4 (IC50 ~ 3.9 µM). Given the prevalence of GLUT1 and GLUT3 overexpression in many cancers and multiple myeloma's reliance on GLUT4, these GLUT3 inhibitors may discriminately hinder glucose entry into various cancer cells, promising novel therapeutic avenues in oncology.


Subject(s)
Drug Discovery , Glucose Transporter Type 3/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/drug effects , Small Molecule Libraries/pharmacology , Binding Sites , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Glucose Transporter Type 3/antagonists & inhibitors , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 5/antagonists & inhibitors , Glucose Transporter Type 5/chemistry , Glucose Transporter Type 5/genetics , Glucose Transporter Type 5/metabolism , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Neoplasms/drug therapy , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemistry
2.
Life Sci Alliance ; 4(4)2021 04.
Article in English | MEDLINE | ID: mdl-33536238

ABSTRACT

The human glucose transporters GLUT1 and GLUT3 have a central role in glucose uptake as canonical members of the Sugar Porter (SP) family. GLUT1 and GLUT3 share a fully conserved substrate-binding site with identical substrate coordination, but differ significantly in transport affinity in line with their physiological function. Here, we present a 2.4 Å crystal structure of GLUT1 in an inward open conformation and compare it with GLUT3 using both structural and functional data. Our work shows that interactions between a cytosolic "SP motif" and a conserved "A motif" stabilize the outward conformational state and increases substrate apparent affinity. Furthermore, we identify a previously undescribed Cl- ion site in GLUT1 and an endofacial lipid/glucose binding site which modulate GLUT kinetics. The results provide a possible explanation for the difference between GLUT1 and GLUT3 glucose affinity, imply a general model for the kinetic regulation in GLUTs and suggest a physiological function for the defining SP sequence motif in the SP family.


Subject(s)
Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Models, Molecular , Protein Conformation , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Biological Transport , Glucose/chemistry , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/genetics , Humans , Ligands , Oocytes , Protein Binding , Protein Interaction Domains and Motifs , Protein Isoforms , Structure-Activity Relationship , Sugars , Xenopus
3.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33402433

ABSTRACT

Artemisinin-resistant malaria parasites have emerged and have been spreading, posing a significant public health challenge. Antimalarial drugs with novel mechanisms of action are therefore urgently needed. In this report, we exploit a "selective starvation" strategy by inhibiting Plasmodium falciparum hexose transporter 1 (PfHT1), the sole hexose transporter in P. falciparum, over human glucose transporter 1 (hGLUT1), providing an alternative approach to fight against multidrug-resistant malaria parasites. The crystal structure of hGLUT3, which shares 80% sequence similarity with hGLUT1, was resolved in complex with C3361, a moderate PfHT1-specific inhibitor, at 2.3-Å resolution. Structural comparison between the present hGLUT3-C3361 and our previously reported PfHT1-C3361 confirmed the unique inhibitor binding-induced pocket in PfHT1. We then designed small molecules to simultaneously block the orthosteric and allosteric pockets of PfHT1. Through extensive structure-activity relationship studies, the TH-PF series was identified to selectively inhibit PfHT1 over hGLUT1 and potent against multiple strains of the blood-stage P. falciparum Our findings shed light on the next-generation chemotherapeutics with a paradigm-shifting structure-based design strategy to simultaneously target the orthosteric and allosteric sites of a transporter.


Subject(s)
Antimalarials/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 3/ultrastructure , Malaria, Falciparum/drug therapy , Monosaccharide Transport Proteins/ultrastructure , Protozoan Proteins/ultrastructure , Allosteric Site , Amino Acid Sequence/genetics , Animals , Crystallography, X-Ray , Glucose/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 3/chemistry , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Monosaccharide Transport Proteins/antagonists & inhibitors , Monosaccharide Transport Proteins/genetics , Plasmodium falciparum/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Conformation/drug effects , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Structure-Activity Relationship
4.
Food Chem ; 317: 126398, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32086122

ABSTRACT

Malvidin-3-O-glucoside, malvidin-3,5-O-diglucoside, malvidin-3-O-(6-O-coumaroyl)-glucoside-5-O-glucoside from Chinese Vitis davidii red wine were used to investigate the role of glucoside, diglucoside and coumaroylated glucoside moieties on their transport efficiency through MKN-28 gastric and Caco-2 intestinal cells. Due to the already described conversion of 3-O-glucosylated anthocyanins into 3-O-glucuronidated, the 3-O-glucuronidated metabolite of malvidin-3-O-glucoside was also tested. The antiproliferative activity was higher for the glucuronidated metabolite in both cell lines. All anthocyanins were transported through MKN-28 gastric cells and Caco-2 intestinal cells with transport efficiencies ranging from 4% to 9% in MKN-28 and from 3% to 5% in Caco-2. No significant differences on transport efficiencies were observed at 180 min among the different anthocyanins in MKN-28. The transport efficiency of malvidin-3-O-glucuronide at 180 min was about 3-4% in Caco-2 and MKN-28 cells. Computational studies were performed to evaluate the interaction between anthocyanins and glucose gastric transporters GLUT1 and GLUT3, which supported the experimental findings.


Subject(s)
Anthocyanins/pharmacokinetics , Vitis/chemistry , Wine , Anthocyanins/analysis , Anthocyanins/chemistry , Caco-2 Cells , Cell Survival/drug effects , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Glucosides/analysis , Glucosides/chemistry , Glucosides/pharmacokinetics , Humans , Inactivation, Metabolic , Intestinal Absorption , Molecular Docking Simulation
5.
Chembiochem ; 21(1-2): 45-52, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31553512

ABSTRACT

Glucose addiction is observed in cancer and other diseases that are associated with hyperproliferation. The development of compounds that restrict glucose supply and decrease glycolysis has great potential for the development of new therapeutic approaches. Addressing facilitative glucose transporters (GLUTs), which are often upregulated in glucose-dependent cells, is therefore of particular interest. This article reviews a selection of potent, isoform-selective GLUT inhibitors and their biological characterization. Potential therapeutic applications of GLUT inhibitors in oncology and other diseases that are linked to glucose addiction are discussed.


Subject(s)
Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 2/antagonists & inhibitors , Glucose Transporter Type 3/antagonists & inhibitors , Glucose Transporter Type 4/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Glucose/antagonists & inhibitors , Glucose/metabolism , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 2/chemistry , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 4/chemistry , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry
6.
ACS Chem Neurosci ; 10(11): 4444-4448, 2019 11 20.
Article in English | MEDLINE | ID: mdl-31617996

ABSTRACT

Glucose transporters (GLUTs) are an essential kind of protein that exists in the neuron and are responsible for glucose transport. In the present study, we performed molecular dynamic simulations to deeply understand the glucose uptake mechanism. According to our results, we reconstruct the glucose uptake model of the GLUT3, which is similar to the working model of GLUTs raised by Yan et al., and find a new intermediate state ( Yan, N., et al. ( 2015 ) Molecular basis of ligand recognition and transport by glucose transporters , Nature 526 , 391 - 396 ). In addition, we discover the bottleneck residues for the protein conformational switch. Water molecules are also important for the conformational switch by influencing the hydrogen bond networks of the glucose-protein complex, which can cause the obvious rearrangement of corresponding transmembrane segments. Our findings may shed light on the glucose uptake process of this key neuronic transmembrane protein and the functional relationships between the multiple intermediate states.


Subject(s)
Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Molecular Dynamics Simulation , Neurons/metabolism , Humans , Protein Conformation , Protein Structure, Secondary
7.
Methods Mol Biol ; 1713: 15-29, 2018.
Article in English | MEDLINE | ID: mdl-29218514

ABSTRACT

Overexpression, purification, and crystallization of eukaryotic membrane proteins represent a major challenge for structural biology. In recent years, we have solved the crystal structures of the human glucose transporters GLUT1 in the inward-open conformation at 3.17 Å resolution and GLUT3 in the outward-open and occluded conformations at 2.4 and 1.5 Å resolutions, respectively. Structural elucidation of these transporters in three distinct functional states reveal the molecular basis for the alternating access transport cycle of this prototypal solute carrier family. It established the molecular foundation for future dynamic and kinetic investigations of these GLUTs, and will likely facilitate structure-based ligand development. In this chapter, we present the detailed protocols of recombinant protein expression, purification, and crystallization of GLUT1 and GLUT3, which may help the pursuit of structural elucidation of other eukaryotic membrane proteins.


Subject(s)
Crystallization , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 3/chemistry , Protein Conformation , Gene Expression , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Humans , Plasmids/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
8.
J Biol Chem ; 291(52): 26762-26772, 2016 Dec 23.
Article in English | MEDLINE | ID: mdl-27836974

ABSTRACT

WZB117 (2-fluoro-6-(m-hydroxybenzoyloxy) phenyl m-hydroxybenzoate) inhibits passive sugar transport in human erythrocytes and cancer cell lines and, by limiting glycolysis, inhibits tumor growth in mice. This study explores how WZB117 inhibits the erythrocyte sugar transporter glucose transport protein 1 (GLUT1) and examines the transporter isoform specificity of inhibition. WZB117 reversibly and competitively inhibits erythrocyte 3-O-methylglucose (3MG) uptake with Ki(app) = 6 µm but is a noncompetitive inhibitor of sugar exit. Cytochalasin B (CB) is a reversible, noncompetitive inhibitor of 3MG uptake with Ki(app) = 0.3 µm but is a competitive inhibitor of sugar exit indicating that WZB117 and CB bind at exofacial and endofacial sugar binding sites, respectively. WZB117 inhibition of GLUTs expressed in HEK293 cells follows the order of potency: insulin-regulated GLUT4 ≫ GLUT1 ≈ neuronal GLUT3. This may explain WZB117-induced murine lipodystrophy. Molecular docking suggests the following. 1) The WZB117 binding envelopes of exofacial GLUT1 and GLUT4 conformers differ significantly. 2) GLUT1 and GLUT4 exofacial conformers present multiple, adjacent glucose binding sites that overlap with WZB117 binding envelopes. 3) The GLUT1 exofacial conformer lacks a CB binding site. 4) The inward GLUT1 conformer presents overlapping endofacial WZB117, d-glucose, and CB binding envelopes. Interrogating the GLUT1 mechanism using WZB117 reveals that subsaturating WZB117 and CB stimulate erythrocyte 3MG uptake. Extracellular WZB117 does not affect CB binding to GLUT1, but intracellular WZB117 inhibits CB binding. These findings are incompatible with the alternating conformer carrier for glucose transport but are consistent with either a multisubunit, allosteric transporter, or a transporter in which each subunit presents multiple, interacting ligand binding sites.


Subject(s)
3-O-Methylglucose/metabolism , Erythrocytes/metabolism , Glucose Transporter Type 1/metabolism , Glucose/metabolism , Hydroxybenzoates/pharmacology , Animals , Binding Sites , Biological Transport , Crystallography, X-Ray , Cytochalasin B/metabolism , Erythrocytes/drug effects , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , HEK293 Cells , Humans , Kinetics , Mice , Molecular Docking Simulation , Protein Conformation
9.
J Biol Chem ; 291(33): 17271-82, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27302065

ABSTRACT

The regulated movement of glucose across mammalian cell membranes is mediated by facilitative glucose transporters (GLUTs) embedded in lipid bilayers. Despite the known importance of phospholipids in regulating protein structure and activity, the lipid-induced effects on the GLUTs remain poorly understood. We systematically examined the effects of physiologically relevant phospholipids on glucose transport in liposomes containing purified GLUT4 and GLUT3. The anionic phospholipids, phosphatidic acid, phosphatidylserine, phosphatidylglycerol, and phosphatidylinositol, were found to be essential for transporter function by activating it and stabilizing its structure. Conical lipids, phosphatidylethanolamine and diacylglycerol, enhanced transporter activity up to 3-fold in the presence of anionic phospholipids but did not stabilize protein structure. Kinetic analyses revealed that both lipids increase the kcat of transport without changing the Km values. These results allowed us to elucidate the activation of GLUT by plasma membrane phospholipids and to extend the field of membrane protein-lipid interactions to the family of structurally and functionally related human solute carriers.


Subject(s)
Glucose Transporter Type 3 , Glucose Transporter Type 4 , Phospholipids , Biological Transport, Active/physiology , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Glucose Transporter Type 4/chemistry , Glucose Transporter Type 4/metabolism , HEK293 Cells , Humans , Liposomes/chemistry , Phospholipids/chemistry , Phospholipids/metabolism
10.
Nature ; 526(7573): 391-6, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26176916

ABSTRACT

The major facilitator superfamily glucose transporters, exemplified by human GLUT1-4, have been central to the study of solute transport. Using lipidic cubic phase crystallization and microfocus X-ray diffraction, we determined the structure of human GLUT3 in complex with D-glucose at 1.5 Å resolution in an outward-occluded conformation. The high-resolution structure allows discrimination of both α- and ß-anomers of D-glucose. Two additional structures of GLUT3 bound to the exofacial inhibitor maltose were obtained at 2.6 Å in the outward-open and 2.4 Å in the outward-occluded states. In all three structures, the ligands are predominantly coordinated by polar residues from the carboxy terminal domain. Conformational transition from outward-open to outward-occluded entails a prominent local rearrangement of the extracellular part of transmembrane segment TM7. Comparison of the outward-facing GLUT3 structures with the inward-open GLUT1 provides insights into the alternating access cycle for GLUTs, whereby the C-terminal domain provides the primary substrate-binding site and the amino-terminal domain undergoes rigid-body rotation with respect to the C-terminal domain. Our studies provide an important framework for the mechanistic and kinetic understanding of GLUTs and shed light on structure-guided ligand design.


Subject(s)
Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Glucose/chemistry , Glucose/metabolism , Binding Sites , Biological Transport , Cell Membrane/metabolism , Crystallography, X-Ray , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/metabolism , Humans , Kinetics , Ligands , Maltose/chemistry , Maltose/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Rotation , Structure-Activity Relationship
11.
J Agric Food Chem ; 63(35): 7685-92, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-25858301

ABSTRACT

The gastric absorption of red wine anthocyanins was evaluated using a gastric MKN-28 cell barrier model. Anthocyanin transport was not affected by the presence of 4% ethanol and decreased with the increase of pH. Gastric cells pretreated with anthocyanins were found to increase anthocyanin transport. The presence of d-(+)-glucose was found to decrease anthocyanin uptake, suggesting the involvement of glucose transporters. RT-PCR assays revealed that GLUT1, GLUT3, and MCT1 transporters were expressed in MKN-28 cells. Computational studies were performed to provide a structural characterization of the binding site of hGLUT1 to glucose or different anthocyanins under different forms. Docking results demonstrated that anthocyanins can bind to glucose transporters from both intracellular and extracellular sides. Anthocyanins seem to enter into the transporter by two main conformations: B ring or glucose. From MD simulations, hGLUT1 was found to form complexes with all anthocyanins tested in the different protonation states.


Subject(s)
Anthocyanins/metabolism , Gastric Mucosa/metabolism , Wine/analysis , Anthocyanins/chemistry , Biological Transport , Cell Line , Glucose/metabolism , Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , Humans , Models, Biological , Stomach/chemistry
12.
J Biol Chem ; 288(28): 20734-44, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720776

ABSTRACT

The human blood-brain barrier glucose transport protein (GLUT1) forms homodimers and homotetramers in detergent micelles and in cell membranes, where the GLUT1 oligomeric state determines GLUT1 transport behavior. GLUT1 and the neuronal glucose transporter GLUT3 do not form heterocomplexes in human embryonic kidney 293 (HEK293) cells as judged by co-immunoprecipitation assays. Using homology-scanning mutagenesis in which GLUT1 domains are substituted with equivalent GLUT3 domains and vice versa, we show that GLUT1 transmembrane helix 9 (TM9) is necessary for optimal association of GLUT1-GLUT3 chimeras with parental GLUT1 in HEK cells. GLUT1 TMs 2, 5, 8, and 11 also contribute to a less abundant heterocomplex. Cell surface GLUT1 and GLUT3 containing GLUT1 TM9 are 4-fold more catalytically active than GLUT3 and GLUT1 containing GLUT3 TM9. GLUT1 and GLUT3 display allosteric transport behavior. Size exclusion chromatography of detergent solubilized, purified GLUT1 resolves GLUT1/lipid/detergent micelles as 6- and 10-nm Stokes radius particles, which correspond to GLUT1 dimers and tetramers, respectively. Studies with GLUTs expressed in and solubilized from HEK cells show that HEK cell GLUT1 resolves as 6- and 10-nm Stokes radius particles, whereas GLUT3 resolves as a 6-nm particle. Substitution of GLUT3 TM9 with GLUT1 TM9 causes chimeric GLUT3 to resolve as 6- and 10-nm Stokes radius particles. Substitution of GLUT1 TM9 with GLUT3 TM9 causes chimeric GLUT1 to resolve as a mixture of 6- and 4-nm particles. We discuss these findings in the context of determinants of GLUT oligomeric structure and transport function.


Subject(s)
Glucose Transporter Type 1/chemistry , Glucose Transporter Type 3/chemistry , Protein Multimerization , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Deoxyglucose/metabolism , Deoxyglucose/pharmacokinetics , Detergents/chemistry , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/genetics , Glucose Transporter Type 3/metabolism , HEK293 Cells , Humans , Kinetics , Micelles , Molecular Sequence Data , Mutation , Protein Engineering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Genetics ; 193(4): 1135-47, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23288934

ABSTRACT

The bithorax complex in Drosophila melanogaster includes three homeobox-containing genes--Ultrabithorax (Ubx), abdominal--A (abd-A), and Abdominal-B (Abd-B)-which are required for the proper differentiation of the posterior 10 segments of the body. Each of these genes has multiple distinct regulatory regions; there is one for each segmental unit of the body plan where the genes are expressed. One additional protein- coding gene in the bithorax complex, Glut3, a sugar-transporter homolog, can be deleted without phenotype. We focus here on the upstream regulatory region for Ubx, the bithoraxoid (bxd) domain, and its border with the adjacent infraabdominal-2 (iab-2) domain, which controls abdA. These two domains can be defined by the phenotypes of rearrangement breakpoints, and by the expression patterns of enhancer traps. In D. virilis, the homeotic cluster is split between Ubx and abd-A, and so the border can also be located by a sequence comparison between species. When the border region is deleted in melanogaster, the flies show a dominant phenotype called Front-ultraabdominal (Fub); the first abdominal segment is transformed into a copy of the second abdominal segment. Thus, the border blocks the spread of activation from the bxd domain into the iab-2 domain.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Breakpoints , Chromosomes, Insect/genetics , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/genetics , Molecular Sequence Data , Phenotype , Sequence Deletion , Sequence Homology
14.
Am J Physiol Endocrinol Metab ; 295(2): E242-53, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18577699

ABSTRACT

Glucose metabolism is vital to most mammalian cells, and the passage of glucose across cell membranes is facilitated by a family of integral membrane transporter proteins, the GLUTs. There are currently 14 members of the SLC2 family of GLUTs, several of which have been the focus of this series of reviews. The subject of the present review is GLUT3, which, as implied by its name, was the third glucose transporter to be cloned (Kayano T, Fukumoto H, Eddy RL, Fan YS, Byers MG, Shows TB, Bell GI. J Biol Chem 263: 15245-15248, 1988) and was originally designated as the neuronal GLUT. The overriding question that drove the early work on GLUT3 was why would neurons need a separate glucose transporter isoform? What is it about GLUT3 that specifically suits the needs of the highly metabolic and oxidative neuron with its high glucose demand? More recently, GLUT3 has been studied in other cell types with quite specific requirements for glucose, including sperm, preimplantation embryos, circulating white blood cells, and an array of carcinoma cell lines. The last are sufficiently varied and numerous to warrant a review of their own and will not be discussed here. However, for each of these cases, the same questions apply. Thus, the objective of this review is to discuss the properties and tissue and cellular localization of GLUT3 as well as the features of expression, function, and regulation that distinguish it from the rest of its family and make it uniquely suited as the mediator of glucose delivery to these specific cells.


Subject(s)
Glucose Transporter Type 3/metabolism , Animals , Glucose/metabolism , Glucose Transporter Type 3/chemistry , Humans , Models, Molecular , Neurons/metabolism
15.
FEBS J ; 274(11): 2843-53, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17459098

ABSTRACT

The hexose transporter family, which mediates facilitated uptake in mammalian cells, consists of more than 10 members containing 12 membrane-spanning segments with a single N-glycosylation site. We previously demonstrated that glucose transporter 1 is organized into a raft-like detergent-resistant membrane domain but that glucose transporter 3 distributes to fluid membrane domains in nonpolarized mammalian cells. In this study, we further examined the structural basis responsible for the distribution by using a series of chimeric constructs. Glucose transporter 1 and glucose transporter 3 with a FLAG-tagged N-terminus were expressed in detergent-resistant membranes and non-detergent-resistant membranes of CHO-K1 cells, respectively. Replacement of either the C-terminal or N-terminal cytosolic portion of FLAG-tagged glucose transporter 1 and glucose transporter 3 did not affect the membrane distribution. However, a critical sorting signal may exist within the N-terminal half of the isoforms without affecting transport activity and its inhibition by cytochalasin B. Further shortening of these regions altered the critical distribution, suggesting that a large proportion or several parts of the intrinsic structure, including the N-terminus of each isoform, are involved in the regulation.


Subject(s)
Glucose Transporter Type 1/chemistry , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/chemistry , Glucose Transporter Type 3/metabolism , Membrane Microdomains/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Glucose/metabolism , Green Fluorescent Proteins/chemistry , Oligopeptides , Peptides/chemistry , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...