Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Pflugers Arch ; 443(1): 92-101, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11692272

ABSTRACT

Transfer of glutamine between astrocytes and neurons is an essential part of the glutamate-glutamine cycle in the brain. Here we have investigated how the neural glutamine transporter (rATA1/GlnT) works. Rat ATA1 was expressed in Xenopus laevis oocytes and examined using two-electrode voltage-clamp recordings, ion-sensitive microelectrodes and tracer flux experiments. Glutamine transport via rATA1 was electrogenic and caused inward currents that did not reverse at positive holding potentials. Currents were induced by a variety of neutral amino acids in the following relative order Ala>Ser/Gln/Asn/His/Cys/Met >MeAIB/Gly>Thr/Pro/Tyr/Val, where MeAIB is the amino acid analogue N-methylaminoisobutyric acid. The uptake of glutamine and the corresponding currents depended on Na+ and pH. Hill-coefficient and flux studies with 22NaCl indicated a cotransport stoichiometry 1 Na+ per transport cycle. The transporter also showed uncoupled Na+ transport, particularly when alanine was used as the substrate. Although substrate uptake increased strongly with increasing pH, no change of intracellular pH was observed during transport. A decrease of the intracellular pH similarly inhibited glutamine transport via ATA1, suggesting that the pH dependence was an allosteric effect on the transporter.


Subject(s)
Amino Acid Transport System A/metabolism , Brain/metabolism , Glutamine/metabolism , Sodium/metabolism , Amino Acid Transport System A/genetics , Amino Acids/pharmacology , Animals , Electric Conductivity , Female , Gene Expression , Hydrogen-Ion Concentration , Membrane Potentials , Microelectrodes , Oocytes/metabolism , Patch-Clamp Techniques , Rats , Recombinant Proteins/metabolism , Transfection , Xenopus laevis
2.
Biochem J ; 355(Pt 3): 725-31, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11311135

ABSTRACT

Heterodimeric amino acid transporters are comprised of a type-II membrane protein named the heavy chain (4F2hc or rBAT) that may associate with a number of different polytopic membrane proteins, called light chains. It is thought that the heavy chain is mainly involved in the trafficking of the complex to the plasma membrane, whereas the transport process itself is catalysed by the light chain. The 4F2 heavy chain (4F2hc) associates with at least six different light chains to induce distinct amino acid-transport activites. To test if the light chains are specifically recognized and to identify domains involved in the recognition of light chains, C-terminally truncated mutants of 4F2hc were constructed and co-expressed with the light chains LAT1, LAT2 and y(+)LAT2. The truncated isoform T1, comprised of only 133 amino acids that form the cytosolic N-terminus and the transmembrane helix, displayed only a slight reduction in its ability to promote LAT1 expression at the membrane surface compared with the 529 amino acid wild-type 4F2hc protein. Co-expression of increasingly larger 4F2hc mutants caused a delayed translocation of LAT1. In contrast to the weak effects of 4F2hc truncations on LAT1 expression, surface expression of LAT2 and y(+)LAT2 was almost completely lost with all truncated heavy chains. Co-expression of LAT1 together with the other light chains did not result in displacement of LAT2 and y(+)LAT2. The results suggest that extracellular domains of the heavy chain are responsible mainly for recognition of light chains other than LAT1 and that the extracellular domain ensures proper translocation to the plasma membrane.


Subject(s)
Antigens, CD/metabolism , Carrier Proteins/metabolism , Amino Acid Transport Systems , Animals , Antigens, CD/chemistry , Antigens, CD/genetics , Biological Transport/physiology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Fusion Regulatory Protein-1 , Gene Deletion , Humans , Mutation , Oocytes/metabolism , Protein Structure, Tertiary , Transfection , Xenopus laevis
3.
Neurochem Res ; 25(11): 1485-91, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11071367

ABSTRACT

Of the three isozymes of glycogen phosphorylase (GP) known, the brain (B) and muscle (M) isoforms have been reported to occur in brain. We investigated the regional and cellular occurrence of the three isozymes in various parts of the rat nervous system, fetal brain and astroglia-rich primary cultures by means of electrophoresis of native proteins with subsequent activity stain and by reverse transcriptase polymerase chain reaction. In the cortex, cerebellum, olfactory bulb, brainstem, spinal cord and dorsal root ganglia, both mRNA and enzyme protein were found for the B and M isozymes. In addition, the liver (L) isoform mRNA was detected in fetal brain and cultured astrocytes. Our studies indicate that there is no regional difference in distribution pattern between brain regions, spinal cord and dorsal root ganglia. In immature brain and cultured glial cells, the additional presence of the L isozyme is possible. These results support the idea that astrocytes express two or even three GP isozymes simultaneously.


Subject(s)
Astrocytes/enzymology , Central Nervous System/enzymology , Isoenzymes/metabolism , Phosphorylases/metabolism , Animals , Astrocytes/cytology , Base Sequence , Cells, Cultured , Central Nervous System/cytology , DNA, Complementary , Electrophoresis, Polyacrylamide Gel , Female , Male , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
4.
Am J Physiol Renal Physiol ; 279(3): F584-91, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10966938

ABSTRACT

L-Carnitine is essential for the translocation of acyl-carnitine into the mitochondria for beta-oxidation of long-chain fatty acids. It is taken up into the cells by the recently cloned Na(+)-driven carnitine organic cation transporter OCTN2. Here we expressed hOCTN2 in Xenopus laevis oocytes and investigated with two-electrode voltage- clamp and flux measurements its functional and pharmacological properties as a Na(+)-carnitine cotransporter. L-carnitine transport was electrogenic. The L-carnitine-induced currents were voltage and Na(+) dependent, with half-maximal currents at 0.3 +/- 0.1 mM Na(+) at -60 mV. Furthermore, L-carnitine-induced currents were pH dependent, decreasing with acidification. In contrast to other members of the organic cation transporter family, hOCTN2 functions as a Na(+)-coupled carnitine transporter. Carnitine transport was stereoselective, with an apparent Michaelis-Menten constant (K(m)) of 4.8 +/- 0.3 microM for L-carnitine and 98.3 +/- 38.0 microM for D-carnitine. The substrate specificity of hOCTN2 differs from rOCT-1 and hOCT-2 as hOCTN2 showed only small currents with classic OCT substrates such as choline or tetraethylammonium; by contrast hOCTN2 mediated transport of betaine. hOCTN2 was inhibited by several drugs known to induce secondary carnitine deficiency. Most potent blockers were the antibiotic emetine and the ion channel blockers quinidine and verapamil. The apparent IC(50) for emetine was 4.2 +/- 1.2 microM. The anticonvulsant valproic acid did not induce a significant inhibition of carnitine transport, pointing to a different mode of action. In summary, hOCTN2 mediates electrogenic Na(+)-dependent stereoselective high-affinity transport of L-carnitine and Na(+). hOCTN2 displays transport properties distinct from other members of the OCT family and is directly inhibited by several substances known to induce systemic carnitine deficiency.


Subject(s)
Carnitine/pharmacokinetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organic Cation Transport Proteins , Sodium/metabolism , Amino Acids/chemistry , Amino Acids/pharmacology , Animals , Betaine/chemistry , Betaine/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Carnitine/chemistry , Carnitine/deficiency , Carrier Proteins/chemistry , Emetine/chemistry , Emetine/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Lipotropic Agents/chemistry , Lipotropic Agents/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/chemistry , Microinjections , Oocytes/physiology , Patch-Clamp Techniques , Pentanoic Acids/chemistry , Pentanoic Acids/pharmacology , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , RNA, Complementary/pharmacology , Solute Carrier Family 22 Member 5 , Tetraethylammonium/chemistry , Tetraethylammonium/pharmacology , Tritium , Valproic Acid/chemistry , Valproic Acid/pharmacology , Xenopus laevis
5.
Biochem J ; 349 Pt 3: 787-95, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10903140

ABSTRACT

The cationic amino acid arginine, due to its positive charge, is usually accumulated in the cytosol. Nevertheless, arginine has to be released by a number of cell types, e.g. kidney cells, which supply other organs with this amino acid, or the endothelial cells of the blood-brain barrier which release arginine into the brain. Arginine release in mammalian cells can be mediated by two different transporters, y(+)LAT1 and y(+)LAT2. For insertion into the plasma membrane, these transporters have to be associated with the type-II membrane glycoprotein 4F2hc [Torrents, Estevez, Pineda, Fernandez, Lloberas, Shi, Zorzano and Palacin (1998) J. Biol. Chem. 273, 32437-32445]. The present study elucidates the function and distribution of y(+)LAT2. In contrast to y(+)LAT1, which is expressed mainly in kidney epithelial cells, lung and leucocytes, y(+)LAT2 has a wider tissue distribution, including brain, heart, testis, kidney, small intestine and parotis. When co-expressed with 4F2hc in Xenopus laevis oocytes, y(+)LAT2 mediated uptake of arginine, leucine and glutamine. Arginine uptake was inhibited strongly by lysine, glutamate, leucine, glutamine, methionine and histidine. Mutual inhibition was observed when leucine or glutamine was used as substrate. Inhibition of arginine uptake by neutral amino acids depended on the presence of Na(+), which is a hallmark of y(+)LAT-type transporters. Although arginine transport was inhibited strongly by glutamate, this anionic amino acid was only weakly transported by 4F2hc/y(+)LAT2. Amino acid transport via 4F2hc/y(+)LAT2 followed an antiport mechanism similar to the other members of this new family. Only preloaded arginine could be released in exchange for extracellular amino acids, whereas marginal release of glutamine or leucine was observed under identical conditions. These results indicated that arginine has the highest affinity for the intracellular binding site and that arginine release may be the main physiological function of this transporter.


Subject(s)
Antigens, CD/metabolism , Arginine/metabolism , Carrier Proteins/metabolism , Glutamine/metabolism , Animals , Antigens, CD/genetics , Base Sequence , Biological Transport , Carrier Proteins/genetics , Cells, Cultured , DNA, Complementary , Fusion Regulatory Protein-1 , Humans , Molecular Sequence Data , Rats , Xenopus laevis
6.
J Physiol ; 526 Pt 1: 35-46, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10878097

ABSTRACT

System L is the major Na(+)-independent amino acid transporter of mammalian cells. It is constituted of the type II membrane protein 4F2hc (CD98) which is covalently linked to the polytopic membrane protein LAT1 via a disulfide bridge. The transporter is known to be regulated by the mineral corticoid aldosterone in Xenopus A6 cells. To understand the regulation of the transporter, the 4F2hc/LAT1 heterodimer was functionally expressed in Xenopus laevis oocytes and its transport properties were analysed using flux measurements and the two-electrode voltage-clamp technique. Expression of 4F2hc/LAT1 resulted in a rapid increase in a Na(+)-independent neutral amino acid antiport activity and simultaneously gave rise to a cation conductance. The cation channel was non-rectifying and non-selective, conducting Li(+) > Cs(+) = Na(+) > K(+). After replacement of Na(+) by NMDG, however, the currents were suppressed almost completely. The cation channel was not inhibited by amiloride, Ba2(+), TEA, Hoe293B, flufenamic acid or substrates of the system L amino acid transporter. Significant inhibition, however, was observed in the presence of La3(+), Gd3(+) and quinidine. Channel activity was upregulated by coexpression of 4F2hc/LAT1 with the aldosterone-regulated protein kinase sgk-1. The cation conductance was sensitive to changes in the redox potential, being inhibited following incubation of the oocytes with DTE for 30 min. Mutation of either of the disulfide bridge-constituting cysteines to serine resulted in a loss of ion channel activity whereas amino acid transport was unaffected. It is concluded that the 4F2hc/LAT1 heterodimer regulates a closely associated cation channel or even constitutes a cation channel itself.


Subject(s)
Antigens, CD/metabolism , Carrier Proteins/metabolism , Cations/metabolism , Nuclear Proteins , Oocytes/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Transport Systems , Amino Acids/metabolism , Amino Acids/pharmacology , Animals , Antigens, CD/drug effects , Antigens, CD/genetics , Carrier Proteins/drug effects , Carrier Proteins/genetics , Cells, Cultured , Choline/pharmacology , Dimerization , Disulfides/metabolism , Fusion Regulatory Protein-1 , Humans , Immediate-Early Proteins , Ion Channels/drug effects , Ion Channels/metabolism , Ion Transport/drug effects , Meglumine/pharmacology , Microinjections , Mutagenesis, Site-Directed , Oocytes/cytology , Oocytes/drug effects , Patch-Clamp Techniques , Protein Serine-Threonine Kinases/genetics , RNA, Complementary/administration & dosage , RNA, Complementary/metabolism , Rats , Sodium/metabolism , Transfection , Xenopus
7.
Biochem J ; 346 Pt 3: 705-10, 2000 Mar 15.
Article in English | MEDLINE | ID: mdl-10698697

ABSTRACT

The neutral amino acid transporter ASCT2 mediates electroneutral obligatory antiport but at the same time requires Na(+) for its function. To elucidate the mechanism, ASCT2 was expressed in Xenopus laevis oocytes and transport was analysed by flux studies and two-electrode voltage clamp recordings. Flux studies with (22)NaCl indicated that the uptake of one molecule of glutamine or alanine is accompanied by the uptake of four to seven Na(+) ions. Similarly to the transport of amino acids, the Na(+) uptake was mediated by an obligatory Na(+) exchange mechanism that depended on the presence of amino acids but was not stoichiometrically coupled to the amino acid transport. Other cations could not replace Na(+) in this transport mechanism. When NaCl was replaced by NaSCN in the transport buffer, the superfusion of oocytes with amino acid substrates resulted in large inward currents, indicating the presence of a substrate-gated anion channel in the ASCT2 transporter. The K(m) for glutamine derived from these experiments is in good agreement with the K(m) derived from flux studies; it varied between 40 and 90 microM at holding potentials of -60 and -20 mV respectively. The permeability of the substrate-gated anion conductance decreased in the order SCN(-)>>NO(3)(-)>I(-)>Cl(-) and also required the presence of Na(+).


Subject(s)
Carrier Proteins/metabolism , Ion Channel Gating , Sodium/metabolism , Amino Acid Transport Systems , Animals , Anions , Female , Ion Transport , Oocytes/metabolism , Xenopus laevis
8.
J Neurochem ; 73(5): 2184-94, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10537079

ABSTRACT

Glutamine release from astrocytes is an essential part of the glutamate-glutamine cycle in the brain. Uptake of glutamine into cultured rat astrocytes occurs by at least four different routes. In agreement with earlier studies, a significant contribution of amino acid transport systems ASC, A, L, and N was detected. It has not been determined whether these systems are also involved in glutamine efflux or whether specific efflux transporters exist. We show here that ASCT2, a variant of transport system ASC, is strongly expressed in rat astroglia-rich primary cultures but not in neuron-rich primary cultures. The amino acid sequence of rat astroglial ASCT2 is 83% identical to that of mouse ASCT2. In Xenopus laevis oocytes expressing rat ASCT2, we observed high-affinity uptake of [U-14C]glutamine (Km = 70 microM) that was Na(+)-dependent, concentrative, and unaffected by membrane depolarization. When oocytes were preloaded with [U-14C]glutamine, no glutamine efflux was detected in the absence of extracellular amino acids. Neither lowering intracellular pH nor raising the temperature elicited efflux. However, addition of 0.1 mM unlabeled alanine, serine, cysteine, threonine, glutamine, or leucine to the extracellular solution resulted in a rapid release of glutamine from the ASCT2-expressing oocytes. Amino acids that are not recognized as substrates by ASCT2 were ineffective in this role. Extracellular glutamate stimulated glutamine release weakly at pH 7.5 but was more effective on lowering pH to 5.5, consistent with the pH dependence of ASCT2 affinity for glutamate. Our findings suggest a significant role of ASCT2 in glutamine efflux from astrocytes by obligatory exchange with extracellular amino acids. However, the relative contribution of this pathway to glutamine release from cells in vivo or in vitro remains to be determined.


Subject(s)
Astrocytes/metabolism , Carrier Proteins/metabolism , Glutamine/metabolism , Amino Acid Sequence , Amino Acid Transport Systems , Animals , Animals, Newborn , Base Sequence , Brain/cytology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cells, Cultured , DNA, Complementary/chemistry , Gene Expression , Glutamic Acid/pharmacology , Hydrogen-Ion Concentration , Mice , Molecular Sequence Data , Oocytes/metabolism , Rats , Sequence Homology , Transfection , Xenopus laevis
9.
Biochemistry ; 38(35): 11577-84, 1999 Aug 31.
Article in English | MEDLINE | ID: mdl-10471310

ABSTRACT

Transport of lactate, pyruvate, and the ketone bodies, acetoacetate and beta-hydroxybutyrate, is mediated in many mammalian cells by the monocarboxylate transporter MCT1. To be accepted as a substrate, a carboxyl group and an unpolar side chain are necessary. Site-directed mutagenesis of the rat MCT1 was used to identify residues which are involved in substrate recognition. Helices 8 and 10 but not helix 9 were found to contain critical residues for substrate recognition. Mutation of arginine 306 to threonine in helix 8 resulted in strongly reduced transport activity. Concomitantly, saturable transport was transformed into a nonsaturable dependence of transport activity on lactate concentration, suggesting that binding of the substrate was strongly impaired. Furthermore, proton translocation in the mutant was partially uncoupled from monocarboxylate transport. Mutation of phenylalanine 360 to cysteine in helix 10 resulted in an altered substrate side chain recognition. In contrast to the wild-type transporter, monocarboxylates with more bulky and polar side chains were recognized by the mutated MCT1. Mutation of selected residues in helix 9 and helix 11 (C336A, H337Q, and E391Q) did not cause alterations of the transport properties of MCT1. It is suggested that substrate binding occurs in the carboxy-terminal half of MCT1 and that helices 8 and 10 are involved in the recognition of different parts of the substrate.


Subject(s)
Carboxylic Acids/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Aspartic Acid/genetics , Biological Transport/genetics , Carrier Proteins/genetics , Female , Kinetics , Lactates/metabolism , Molecular Sequence Data , Monocarboxylic Acid Transporters , Mutagenesis, Site-Directed , Oocytes/metabolism , Protein Structure, Secondary , Rats , Threonine/genetics , Xenopus laevis
10.
Biochem J ; 341 ( Pt 3): 529-35, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10417314

ABSTRACT

Observations on lactate transport in brain cells and cardiac myocytes indicate the presence of a high-affinity monocarboxylate transporter. The rat monocarboxylate transporter isoform MCT2 was analysed by expression in Xenopus laevis oocytes and the results were compared with the known characteristics of lactate transport in heart and brain. Monocarboxylate transport via MCT2 was driven by the H(+) gradient over the plasma membrane. Uptake of lactate strongly increased with decreasing pH, showing half-maximal stimulation at pH 7.2. A wide variety of monocarboxylates and ketone bodies, including lactate, pyruvate, beta-hydroxybutyrate, acetoacetate, 2-oxoisovalerate and 2-oxoisohexanoate, were substrates of MCT2. All substrates had a high affinity for MCT2. For lactate a K(m) value of 0.74+/-0.07 mM was determined at pH 7.0. For the other substrates, K(i) values between 100 microM and 1 mM were measured for inhibition of lactate transport, which is about one-tenth of the corresponding values for the ubiquitously expressed monocarboxylate transporter isoform MCT1. Monocarboxylate transport via MCT2 could be inhibited by alpha-cyano-4-hydroxycinnamate, anion-channel inhibitors and flavonoids. It is suggested that cells which express MCT2 preferentially use lactate and ketone bodies as energy sources.


Subject(s)
Carrier Proteins/metabolism , Monocarboxylic Acid Transporters , Animals , Base Sequence , Carrier Proteins/genetics , Cloning, Molecular , DNA Primers , Female , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Oocytes/metabolism , Rats , Substrate Specificity , Xenopus laevis
11.
Biochim Biophys Acta ; 1448(1): 27-36, 1998 Nov 19.
Article in English | MEDLINE | ID: mdl-9824661

ABSTRACT

Expression of aquaporins (AQP) and water permeability were studied in Xenopus laevis oocytes and immobilized glial cells by a pulsed-field gradient spin echo NMR technique and a photometric swelling assay. Oocytes injected with poly(A) RNA from C6-BU-1 cells showed increased swelling behavior under hypoosmotic stress due to expressed water channels as compared to control oocytes. The swelling could be reversibly inhibited by HgCl2. Furthermore, the intracellular relaxation time and the apparent intracellular diffusion coefficient of water in oocytes were determined by diffusion-weighted 1H NMR experiments to be T2=36 ms and Dapp, intra=0.18x10-3 mm2/s. In immobilized C6 and F98 cells the mean exchange time of intracellular water was found to be 51 ms which increased to 75 ms upon chronic treatment (4 days) in hypertonic medium. Additional hybrid depletion experiments with antisense oligonucleotides directed against AQP1 were performed on oocytes and C6 cells. Moreover, different water channel subtypes of glial cells were assessed by a reverse transcriptase polymerase chain reaction assay. With this, the mRNA encoding AQP1 could be detected in primary cultures and glial cell lines, whereas AQP4 mRNA was found in astroglia-rich primary cultures, but not in F98 and C6 cells. Our results show that water permeability in glial cells is mainly mediated by water channels which play an important role in the regulation of water flow in brain under normal and pathological conditions.


Subject(s)
Aquaporins/metabolism , Cell Membrane Permeability , Water/metabolism , Animals , Aquaporin 1 , Aquaporin 4 , Aquaporins/genetics , Aquaporins/isolation & purification , Biological Transport , Cell Size , Cells, Cultured , Diffusion , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oligonucleotides, Antisense , Oocytes/cytology , Oocytes/metabolism , Photometry/methods , Polymerase Chain Reaction , Rats , Xenopus laevis
12.
Biochem J ; 333 ( Pt 3): 549-54, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9677312

ABSTRACT

Expression of the type II membrane proteins of the rbAT/4F2hc family in Xenopus laevis oocytes results in the induction of amino acid transport activity. To elucidate the mechanism of action, amino acid transport was investigated in oocytes expressing the surface antigen 4F2hc. Leucine transport was mediated by a Na+-independent and a Na+-dependent transport mechanism. Both systems could be further discriminated by their stereochemical constraints. Isoleucine, with a branch at the beta-position, shared only the Na+-independent transport system with leucine. Both transport systems were sensitive to inhibition by arginine, but only the Na+-independent system was sensitive to inhibition by 2-aminobicyclo[2,2,1]heptane-2-carboxylic acid. When compared with known transport systems the two transport activities could be described as similar to, but not identical with, mammalian systems b0,+ and y+L. The Na+-independent b0,+-like transport system was found both in rbAT and 4F2hc expressing oocytes, indicating that both proteins act in a similar way.


Subject(s)
Amino Acids/pharmacokinetics , Antigens, CD/metabolism , Carrier Proteins/metabolism , Oocytes/metabolism , Amino Acid Transport Systems , Animals , Antigens, CD/biosynthesis , Binding, Competitive , Biological Transport/physiology , Carrier Proteins/biosynthesis , Female , Fusion Regulatory Protein-1 , Isoleucine/pharmacokinetics , Kinetics , Leucine/pharmacokinetics , RNA, Complementary/metabolism , Rats , Xenopus laevis/metabolism
13.
Biochem J ; 333 ( Pt 1): 167-74, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9639576

ABSTRACT

Several laboratories have investigated monocarboxylate transport in a variety of cell types. The characterization of the cloned transporter isoforms in a suitable expression system is nevertheless still lacking. H+/monocarboxylate co-transport was therefore investigated in monocarboxylate transporter 1 (MCT1)-expressing Xenopus laevis oocytes by using pH-sensitive microelectrodes and [14C]lactate. Superfusion with lactate resulted in intracellular acidification of MCT1-expressing oocytes, but not in non-injected control oocytes. The basic kinetic properties of lactate transport in MCT1-expressing oocytes were determined by analysing the rates of intracellular pH changes under different conditions. The results were in agreement with the known properties of the transporter, with respect to both the dependence on the lactate concentration and the external pH value. Besides lactate, MCT1 mediated the reversible transport of a wide variety of monocarboxylic acids including pyruvate, D,L-3-hydroxybutyrate, acetoacetate, alpha-oxoisohexanoate and alpha-oxoisovalerate, but not of dicarboxylic and tricarboxylic acids. The inhibitor alpha-cyano-4-hydroxycinnamate bound strongly to the transporter without being translocated, but could be displaced by the addition of lactate. In addition to changes in the intracellular pH, lactate transport also induced deviations from the resting membrane potential.


Subject(s)
Carrier Proteins/metabolism , Cytosol/metabolism , Animals , Biological Transport , Carrier Proteins/biosynthesis , Female , Hydrogen-Ion Concentration , Kinetics , Lactic Acid/metabolism , Membrane Potentials , Microelectrodes , Monocarboxylic Acid Transporters , Oocytes/metabolism , Oocytes/ultrastructure , Patch-Clamp Techniques , Xenopus laevis
14.
J Membr Biol ; 164(1): 71-7, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9636245

ABSTRACT

Expression of the protein NaPi-1 in Xenopus oocytes has previously been shown to induce an outwardly rectifying Cl- conductance (GCl), organic anion transport and Na+-dependent Pi-uptake. In the present study we investigated the relation between the NaPi-1 induced GCl and Pi-induced currents and transport. NaPi-1 expression induced Pi-transport, which was not different at 1-20 ng/oocyte NaPi-1 cRNA injection and was already maximal at 1-2 days after cRNA injection. In contrast, GCl was augmented at increased amounts of cRNA injection (1-20 ng/oocyte) and over a five day expression period. Subsequently all experiments were performed on oocytes injected with 20 ng/oocytes cRNA. Pi-induced currents (Ip) could be observed in NaPi-1 expressing oocytes at high concentrations of Pi (>/= 1 mm Pi). The amplitudes of Ip correlated well with GCl. Ip was blocked by the Cl- channel blocker NPPB, partially Na+-dependent and completely abolished in Cl- free solution. In contrast, Pi-transport in NaPi-1 expressing oocytes was not NPPB sensitive, stronger depending on extracellular Na+ and weakly affected by Cl- substitution. Endogenous Pi-uptake in water-injected oocytes amounted in all experiments to 30-50% of the Na+-dependent Pi-transport observed in NaPi-1 expressing oocytes. The properties of the endogenous Pi-uptake system (Km for Pi > 1 mM; partial Na+- and Cl--dependence; lack of NPPB block) were similar to the NaPi-1 induced Pi-uptake, but no Ip could be recorded at Pi-concentrations

Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Chloride Channels/metabolism , Ion Channel Gating/physiology , Oocytes/metabolism , Phosphates/metabolism , Symporters , Animals , Biological Transport/genetics , Carrier Proteins/genetics , Chlorides/physiology , Extracellular Space/chemistry , Gene Expression/drug effects , Microinjections , Nitrobenzoates/pharmacology , RNA, Complementary/pharmacology , Sodium/physiology , Sodium-Phosphate Cotransporter Proteins , Xenopus
15.
Biochem J ; 324 ( Pt 2): 535-41, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9182715

ABSTRACT

Mammalian cells possess a variety of amino acid-transport systems with overlapping substrate specificity. System L is one of the major amino acid-transport systems of non-epithelial cells. By expression cloning we have recently demonstrated that the surface antigen 4F2hc (CD98) is a necessary component for expression of system-L-like amino acid-transport activity in C6-BU-1 rat glioma cells [Bröer, Bröer and Hamprecht (1995) Biochem. J. 312, 863-870]. 4F2hc mRNA was detected in CHO cells, COS cells, activated lymphocytes isolated from mouse spleen and primary cultures of astrocytes. In all these cell types, Na+-independent isoleucine transport was mediated by system L. No contribution of system y+L to isoleucine or arginine transport was detected in C6-BU-1 cells. In lymphocytes, both system-L-like amino acid-transport activity and 4F2hc mRNA levels increased after treatment with phorbol ester plus ionomycin. Antisense oligonucleotides caused modest inhibition of Na+-independent isoleucine transport in C6-BU-1 cells and primary cultures of astroglial cells, whereas arginine transport was unaffected. Overexpression of 4F2hc cDNA in CHO cells resulted in an increase in Na+-independent isoleucine transport.


Subject(s)
Amino Acids/metabolism , Antigens, CD/metabolism , Carrier Proteins/metabolism , Mammals/metabolism , Amino Acid Sequence , Animals , Antigens, CD/genetics , Arginine/metabolism , Astrocytes/metabolism , Biological Transport , Brain/cytology , CHO Cells , COS Cells , Carrier Proteins/genetics , Cells, Cultured , Cricetinae , Cricetulus , DNA, Complementary/genetics , Fusion Regulatory Protein-1 , Glioma/pathology , Isoleucine/metabolism , Lymphocytes/metabolism , Mice , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Recombinant Fusion Proteins/biosynthesis , Sequence Alignment , Sequence Homology , Transfection , Tumor Cells, Cultured
16.
Biochem J ; 312 ( Pt 3): 863-70, 1995 Dec 15.
Article in English | MEDLINE | ID: mdl-8554532

ABSTRACT

Mammalian cells possess a variety of amino acid-transport systems with overlapping substrate specificity. System L is one of the major amino acid-transport systems in all non-epithelial cells. Its molecular structure is not known. To clone the neutral amino acid-transporter system L, we followed an expression cloning strategy using Xenopus laevis oocytes. A cDNA library derived from C6-BU-1 rat glioma cells was used as a source, because high expression of system L activity could be demonstrated with polyadenylated RNA isolated from these cells, when injected into Xenopus laevis oocytes [Bröer, Bröer and Hamprecht (1994) Biochim. Biophys. Acta 1192, 95-100]. A single clone (ILAT) was identified, the sense cRNA of which, on injection into Xenopus laevis oocytes, stimulated sodium-independent isoleucine transport by about 100-fold. Further characterization revealed that transport of cationic amino acids was also stimulated. Sequencing of the cDNA showed that the identified clone is the heavy chain of the rat 4F2 surface antigen, a marker of tumour cells and activated lymphocytes. Uptake of neutral and cationic amino acids was not stimulated by the presence of Na+ ions. Antisense cRNA transcribed from this clone or antisense oligonucleotides, when co-injected with polyadenylated RNA from C6-BU-1 rat glioma cells, completely suppressed system L-like isoleucine-transport activity. We conclude that ILAT is necessary for expression of system L-like amino acid-transport activity by polyadenylated RNA from C6-BU-1 rat glioma cells.


Subject(s)
Amino Acids/metabolism , Antigens, CD/physiology , Antigens, Surface/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Gene Expression , Amino Acid Sequence , Amino Acid Transport Systems , Animals , Antigens, CD/genetics , Base Sequence , Binding, Competitive , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/metabolism , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Female , Fusion Regulatory Protein 1, Heavy Chain , Fusion Regulatory Protein-1 , Gene Transfer Techniques , Isoleucine/metabolism , Kinetics , Molecular Sequence Data , Oocytes/metabolism , Rats , Tumor Cells, Cultured , Xenopus laevis
17.
Biochim Biophys Acta ; 1192(1): 95-100, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-8204656

ABSTRACT

Poly(A)+ RNA from C6-BU-1 rat glioma cells and rat astroglial cells induced isoleucine transport activity when injected into Xenopus laevis oocytes. The Na+-independent component of isoleucine transport was inhibited by leucine, phenylalanine and 2-aminobicyclo[2,2,1]heptane-2-carboxylic acid (BCH) but neither by methylaminoisobutyric acid (MeAIB) nor lysine. A Km value of approx. 100 microM was determined for the Na+-independent transport of isoleucine. These data are in accordance with expression of a system L like transporter. By injection of size fractionated poly(A)+ RNA a length of approx. 1.9 kb was determined for the pertinent mRNA.


Subject(s)
Brain/metabolism , Isoleucine/metabolism , Oocytes/metabolism , Amino Acid Transport Systems , Animals , Biological Transport , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression , Kinetics , Microinjections , Poly A/genetics , RNA, Messenger/isolation & purification , RNA, Messenger/pharmacology , Rats , Rats, Wistar , Sodium/pharmacology , Tumor Cells, Cultured , Xenopus laevis
18.
J Bacteriol ; 175(11): 3480-5, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8501052

ABSTRACT

The arsenic resistance operon of Staphylococcus aureus plasmid pI258 determined lowered net cellular uptake of 73As by an active efflux mechanism. Arsenite was exported from the cells; intracellular arsenate was first reduced to arsenite and then transported out of the cells. Resistant cells showed lower accumulation of 73As originating from both arsenate and arsenite. Active efflux from cells loaded with arsenite required the presence of the plasmid-determined arsB gene. Efflux of arsenic originating as arsenate required the presence of the arsC gene and occurred more rapidly with the addition of arsB. Inhibitor studies with S. aureus loaded with arsenite showed that arsenite efflux was energy dependent and appeared to be driven by the membrane potential. With cells loaded with 73AsO4(3-), a requirement for ATP for energy was observed, leading to the conclusion that ATP was required for arsenate reduction. When the staphylococcal arsenic resistance determinant was cloned into Escherichia coli, lowered accumulation of arsenate and arsenite and 73As efflux from cells loaded with arsenate were also found. Cloning of the E. coli plasmid R773 arsA gene (the determinant of the arsenite-dependent ATPase) in trans to the S. aureus gene arsB resulted in increased resistance to arsenite.


Subject(s)
Arsenic/metabolism , Arsenites , Genes, Bacterial/genetics , Ion Pumps , Multienzyme Complexes , Plasmids/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Adenosine Triphosphatases/genetics , Arsenates/metabolism , Arsenates/pharmacology , Arsenic/pharmacology , Arsenite Transporting ATPases , Biological Transport , Cloning, Molecular , Drug Resistance, Microbial , Energy Metabolism , Escherichia coli/genetics , Operon/genetics , Staphylococcus aureus/drug effects
19.
Eur J Biochem ; 210(1): 269-77, 1992 Nov 15.
Article in English | MEDLINE | ID: mdl-1359967

ABSTRACT

Treatment of the reconstituted aspartate/glutamate carrier from mitochondria with 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole (Nbd-Cl) led to complete inactivation of carrier function. Inhibition could be attributed to chemical modification of one single cysteine in the active site. This residue was specifically protected in the presence of aspartate or glutamate, 50% substrate protection being observed at half-saturation of the external binding site. The bifunctional reagent 4,4'-diisothiocyanostilbene-2,2'-disulfonate (DIDS) also modified the same cysteine and, in addition, an active-site lysine identified previously [Dierks, T., Stappen, R., Salentin, A. & Krämer, R. (1992) Biochim. Biophys. Acta 1103, 13-24]. The proximity of the cysteine [Cys(a)] and the lysine residue was confirmed by a mutual exclusion of the respective reagents when added consecutively. By using a variety of reagents a further cysteine [Cys(b)] and probably a histidine residue could be discriminated from Cys(a) and the lysine. The applied reagents were classified according to functional and structural criteria. Class A reagents, like Nbd-Cl, modified the active-site Cys(a) thereby inhibiting the antiport function. Class B reagents, like HgCl2, reacted with both Cys(a) and Cys(b) leading to a conversion of the carrier from antiport to uniport function [Dierks, T., Salentin, A., Heberger, C. & Krämer, R. (1990) Biochim. Biophys. Acta 1028, 268-280]. DIDS at relatively high concentration (60 microM) also acted as a uniport inducer. Class C reagents finally, like pyridoxal phosphate or diethyl pyrocarbonate, modified the active-site lysine or histidine, respectively, and blocked antiport and uniport activity. By testing the accessibility of the mentioned residues to the various reagents, when applied in different order, topological relationships could be elaborated indicating the location of these amino acids with respect to the exofacial active site of the carrier protein.


Subject(s)
Aspartic Acid/metabolism , Glutamates/metabolism , Mitochondria, Heart/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/analogs & derivatives , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , 4-Chloro-7-nitrobenzofurazan/pharmacology , Animals , Binding Sites , Cattle , Electrophoresis, Polyacrylamide Gel , Glutamic Acid , Mitochondria, Heart/drug effects , Proteolipids , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...