Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
J Gen Physiol ; 130(2): 157-68, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17635959

ABSTRACT

Cytoplasmic ATP inhibits human erythrocyte glucose transport protein (GLUT1)-mediated glucose transport in human red blood cells by reducing net glucose transport but not exchange glucose transport (Cloherty, E.K., D.L. Diamond, K.S. Heard, and A. Carruthers. 1996. Biochemistry. 35:13231-13239). We investigated the mechanism of ATP regulation of GLUT1 by identifying GLUT1 domains that undergo significant conformational change upon GLUT1-ATP interaction. ATP (but not GTP) protects GLUT1 against tryptic digestion. Immunoblot analysis indicates that ATP protection extends across multiple GLUT1 domains. Peptide-directed antibody binding to full-length GLUT1 is reduced by ATP at two specific locations: exofacial loop 7-8 and the cytoplasmic C terminus. C-terminal antibody binding to wild-type GLUT1 expressed in HEK cells is inhibited by ATP but binding of the same antibody to a GLUT1-GLUT4 chimera in which loop 6-7 of GLUT1 is substituted with loop 6-7 of GLUT4 is unaffected. ATP reduces GLUT1 lysine covalent modification by sulfo-NHS-LC-biotin by 40%. AMP is without effect on lysine accessibility but antagonizes ATP inhibition of lysine modification. Tandem electrospray ionization mass spectrometry analysis indicates that ATP reduces covalent modification of lysine residues 245, 255, 256, and 477, whereas labeling at lysine residues 225, 229, and 230 is unchanged. Exogenous, intracellular GLUT1 C-terminal peptide mimics ATP modulation of transport whereas C-terminal peptide-directed IgGs inhibit ATP modulation of glucose transport. These findings suggest that transport regulation involves ATP-dependent conformational changes in (or interactions between) the GLUT1 C terminus and the C-terminal half of GLUT1 cytoplasmic loop 6-7.


Subject(s)
Adenosine Triphosphate/physiology , Glucose Transporter Type 1/metabolism , Amino Acids/metabolism , Biological Transport/physiology , Cytoplasm/metabolism , Erythrocytes/metabolism , Glucose/metabolism , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/chemistry , Humans , Protein Binding/physiology , Protein Conformation
3.
Mov Disord ; 21(2): 241-5, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16149086

ABSTRACT

Glucose transport protein deficiency due to mutation in the GLUT1 gene is characterized by infantile onset and chronic seizure disorder, microcephaly, global developmental delays, and hypoglycorrhachia. We describe a 10-year-old normocephalic male with prominent ataxia, dystonia, choreoathetosis, and GLUT1 deficiency whose motor abnormalities improved with a ketogenic diet. We illustrate the motor abnormalities, at baseline and after ketogenic diet, that characterize this unusual case. This case broadens the phenotype of GLUT1 deficiency and illustrates the importance of cerebrospinal fluid (CSF) evaluation in detecting potentially treatable conditions in children with undiagnosed movement disorders.


Subject(s)
Developmental Disabilities/genetics , Dietary Fats/administration & dosage , Glucose Transporter Type 1/deficiency , Microcephaly/genetics , Movement Disorders/genetics , Seizures/genetics , Athetosis/diagnosis , Athetosis/diet therapy , Athetosis/genetics , Blood Glucose/metabolism , Child , Chorea/diagnosis , Chorea/diet therapy , Chorea/genetics , Developmental Disabilities/diagnosis , Developmental Disabilities/diet therapy , Erythrocyte Membrane/metabolism , Genetic Carrier Screening , Glucose Transporter Type 1/genetics , Humans , Male , Microcephaly/diagnosis , Microcephaly/diet therapy , Movement Disorders/diagnosis , Movement Disorders/diet therapy , Mutagenesis, Insertional , Seizures/diet therapy
4.
Biochemistry ; 44(15): 5606-16, 2005 Apr 19.
Article in English | MEDLINE | ID: mdl-15823019

ABSTRACT

Human erythrocyte hexose transfer is mediated by the glucose transport protein GLUT1 and is characterized by a complexity that is unexplained by available hypotheses for carrier-mediated sugar transport [Cloherty, E. K., Heard, K. S., and Carruthers, A. (1996) Biochemistry 35, 10411-10421]. The study presented here examines the possibility that the operational properties of GLUT1 are determined by host cell environment. A glucose transport-null strain of Saccharomyces cerevisiae (RE700A) was transfected with the p426 GPD yeast expression vector containing DNA encoding the wild-type human glucose transport protein (GLUT1), mutant GLUT1 (GLUT1(338)(-)(A3)), or carboxy-terminal hemagglutinin-polyHis-tagged GLUT1 (GLUT1-HA-H6). GLUT1 and GLUT1-HA-H6 are expressed at the yeast cell membrane and restore 2-deoxy-d-glucose, 3-O-methylglucose, and d-glucose transport capacity to RE700A. GLUT1-HA-H6 confers GLUT1-specific sugar transport characteristics to transfected RE700A, including inhibition by cytochalasin B and high-affinity transport of the nonmetabolized sugar 3-O-methylglucose. GLUT1(338)(-)(A3), a catalytically inactive GLUT1 mutant, is expressed but fails to restore RE700A sugar uptake capacity or growth on glucose. In contrast to transport in human red cells, K(m(app)) for 2-deoxy-d-glucose uptake equals K(i(app)) for 2-deoxy-d-glucose inhibition of 3-O-methylglucose uptake. Unlike transport in human red cells or transport in human embryonic kidney cells transfected with GLUT1-HA-H6, unidirectional sugar uptake in RE700A-GLUT1-HA-H6 is not inhibited by reductant and is not stimulated by intracellular sugar. Net uptake of subsaturating 3-O-methylglucose by RE700A-GLUT1-HA-H6 is a simple, first-order process. These findings support the hypothesis that red cell sugar transport complexity is host cell-specific.


Subject(s)
Erythrocytes/metabolism , Monosaccharide Transport Proteins/blood , Amino Acid Substitution , Biological Transport, Active , Cell Compartmentation , Gene Deletion , Genes, Fungal , Glucose/metabolism , Glucose Transporter Type 1 , Humans , In Vitro Techniques , Kinetics , Models, Biological , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Transfection
5.
Biochemistry ; 41(42): 12629-38, 2002 Oct 22.
Article in English | MEDLINE | ID: mdl-12379105

ABSTRACT

Intracellular ATP inhibits human erythrocyte net sugar transport by binding cooperatively to the glucose transport protein (GluT1). ATP binding produces altered transporter affinity for substrate and promotes substrate occlusion within a post-translocation vestibule formed by GluT1 cytosolic domains. The accompanying paper (Cloherty, E. K., Levine, K. B., Graybill, C., and Carruthers, A. (2002) Biochemistry 41, 12639-12651) demonstrates that reduced intracellular pH promotes high-affinity ATP binding to GluT1 but inhibits ATP-modulation of GluT1-mediated sugar transport. The present study explores the role of GluT1 residues 326-343 (a proposed GluT1 ATP-binding site subdomain) in GluT1 ATP binding by using alanine scanning mutagenesis. Cos-7 and HEK cells were transfected with a cDNA encoding full-length human GluT1 terminating in a carboxyl-terminal hemagglutinin (HA)-His6 epitope. The transporter (GluT1.HA.H6) is expressed at the surface of both cell-types and is catalytically active. In HEK cells, both parental GluT1- and GluT1.HA.H6-mediated sugar transport are acutely sensitive to cellular metabolic inhibition. Isolated, detergent-solubilized GluT1.HA.H6 is photolabeled by [gamma-32P]-azidoATP in an ATP-protectable manner. Alanine substitution of E329 or G332/R333/R334 enhances GluT1.HA.H6 [gamma-32P]azidoATP photoincorporation but blocks acute modulation of net sugar transport by cellular metabolic inhibition. These actions resemble those of reduced pH on ATP binding to and modulation of red cell GluT1. It is proposed that cooperative nucleotide binding to GluT1 and nucleotide modulation of GluT1-mediated sugar transport are regulated by a proton-sensitive saltbridge (Glu329-Arg333/334).


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/chemistry , Deoxyglucose/chemistry , Deoxyglucose/metabolism , Monosaccharide Transport Proteins/chemistry , Monosaccharide Transport Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Azides/metabolism , Biological Transport, Active/genetics , COS Cells/metabolism , Cell Line/metabolism , Glucose Transporter Type 1 , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/biosynthesis , Monosaccharide Transport Proteins/genetics , Mutagenesis, Site-Directed , Photoaffinity Labels/metabolism , Transfection
6.
Biochemistry ; 41(42): 12639-51, 2002 Oct 22.
Article in English | MEDLINE | ID: mdl-12379106

ABSTRACT

The human erythrocyte glucose transport protein (GluT1) is an adenine nucleotide binding protein. When complexed with cytosolic ATP, GluT1 exhibits increased affinity for the sugar export site ligand cytochalasin B, prolonged substrate occlusion, reduced net sugar import capacity, and diminished reactivity with carboxyl terminal peptide-directed antibodies. The present study examines the kinetics of nucleotide interaction with GluT1. When incorporated into resealed human red blood cell ghosts, (2,3)-trinitrophenyl-adenosine-triphosphate (TNP-ATP) mimics the ability of cytosolic ATP to promote high-affinity 3-O-methylglucose uptake. TNP-ATP fluorescence increases upon interaction with purified human red cell GluT1. TNP-ATP binding to GluT1 is rapid (t(1/2) approximately 0.5 s at 50 microM TNP-ATP), cooperative, and pH-sensitive and is stimulated by ATP and by the exit site ligand cytochalasin B. Dithiothreitol inhibits TNP-ATP binding to GluT1. GluT1 preirradiation with saturating, unlabeled azidoATP enhances subsequent GluT1 photoincorporation of [gamma-32P]azidoATP. Reduced pH enhances azidoATP photoincorporation into isolated red cell GluT1 but inhibits ATP modulation of sugar transport in resealed red cell ghosts and in GluT1 proteoliposomes. We propose that cooperative nucleotide binding to reductant-sensitive, oligomeric GluT1 is modulated by a proton-sensitive saltbridge. The effects of ATP on GluT1-mediated sugar transport may be determined by the number of ATP molecules complexed with the transporter.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/blood , Erythrocyte Membrane/metabolism , Monosaccharide Transport Proteins/metabolism , 3-O-Methylglucose/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Azides/metabolism , Binding Sites , Biological Transport, Active , Erythrocyte Membrane/chemistry , Glucose Transporter Type 1 , Humans , Hydrogen-Ion Concentration , Models, Chemical , Monosaccharide Transport Proteins/blood , Monosaccharide Transport Proteins/chemistry , Photoaffinity Labels/metabolism , Proteolipids/chemistry , Proteolipids/metabolism , Spectrometry, Fluorescence
SELECTION OF CITATIONS
SEARCH DETAIL
...