Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 16(12): 1794-801, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24212673

ABSTRACT

Prior to the advent of synthetic nematocides, natural products such as seaweed were used to control nematode infestations. The nematocidal agent in seaweed is betaine, an amino acid that functions as an osmolyte and methyl donor. However, the molecular mechanisms of betaine toxicity are unknown. We identified the betaine transporter SNF-3 and the betaine receptor ACR-23 in the nematode C. elegans. Mutating snf-3 in a sensitized background caused the worms to be hypercontracted and paralyzed, presumably as a result of excess extracellular betaine. These behavioral defects were suppressed by mutations in acr-23, which encodes a ligand-gated cation channel of the cys-loop family. ACR-23 was activated by betaine and functioned in the mechanosensory neurons to maintain basal levels of locomotion. However, overactivation of the receptor by excess betaine or by the allosteric modulator monepantel resulted in hypercontraction and death of the nematode. Thus, monepantel targets a betaine signaling pathway in nematodes.


Subject(s)
Antinematodal Agents/pharmacology , Betaine/metabolism , Betaine/pharmacology , Ion Channel Gating/drug effects , Nervous System/metabolism , Animals , Animals, Genetically Modified , Antinematodal Agents/metabolism , Body Size/genetics , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Ion Channel Gating/genetics , Larva , Ligand-Gated Ion Channels/genetics , Mechanoreceptors/drug effects , Mechanoreceptors/metabolism , Membrane Potentials/drug effects , Mutation/genetics , Neurotransmitter Agents/pharmacology , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism
2.
Life Sci ; 82(19-20): 1004-11, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18407297

ABSTRACT

Taurine and its transporter (TauT) are expressed in preimplantation embryos, but their role in embryogenesis is not known. To investigate the role of TauT during embryonic development, we cloned and functionally characterized the zebrafish TauT. The zebrafish TauT cDNA codes for a protein of 625 amino acids which is highly homologous to mammalian TauT. When expressed in mammalian cells, zebrafish TauT mediates taurine uptake in a Na(+)/Cl(-)-dependent manner with a Na(+):Cl(-):taurine stoichiometry of 2:1:1. In the zebrafish embryo, taurine and TauT mRNA are present during early cleavage stages, indicating that both the transporter and its substrate are maternally derived. During embryogenesis, zygotic expression of TauT mRNA is evident in the retina, brain, heart, kidney, and blood vessels. Knockdown of TauT by antisense morpholino oligonucleotides leads to cell death in the central nervous system and increased mortality. These findings suggest a specific role for TauT during development in vertebrates.


Subject(s)
Embryonic Development , Membrane Glycoproteins/biosynthesis , Membrane Transport Proteins/biosynthesis , Zebrafish/embryology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Cloning, Molecular , Embryonic Development/drug effects , Epithelial Cells/metabolism , Humans , In Situ Hybridization , Membrane Glycoproteins/genetics , Membrane Transport Proteins/genetics , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Sequence Homology, Amino Acid , Substrate Specificity , Zebrafish/metabolism
3.
Pharm Res ; 23(6): 1209-16, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16729224

ABSTRACT

PURPOSE: Sodium-coupled monocarboxylate transporter 1 (SMCT1) is a Na+-coupled transporter for monocarboxylates. Many nonsteroidal anti-inflammatory drugs (NSAIDs) are monocarboxylates. Therefore, we investigated the interaction of these drugs with human SMCT1 (hSMCT1). METHODS: We expressed hSMCT1 in a mammalian cell line and in Xenopus laevis oocytes and used the uptake of nicotinate and propionate-induced currents to monitor its transport function, respectively. We also used [14C]-nicotinate and [3H]-ibuprofen for direct measurements of uptake in oocytes. RESULTS: In mammalian cells, hSMCT1-mediated nicotinate uptake was inhibited by ibuprofen and other structurally related NSAIDs. The inhibition was Na+ dependent. With ibuprofen, the concentration necessary for 50% inhibition was 64 +/- 16 microM. In oocytes, the transport function of hSMCT1 was associated with inward currents in the presence of propionate. Under identical conditions, ibuprofen and other structurally related NSAIDs failed to induce inward currents. However, these compounds blocked propionate-induced currents. With ibuprofen, the blockade was dose dependent, Na+ dependent, and competitive. However, there was no uptake of [3H]-ibuprofen into oocytes expressing hSMCT1, although the uptake of [14C]-nicotinate was demonstrable under identical conditions. CONCLUSIONS: Ibuprofen and other structurally related NSAIDs interact with hSMCT1 as blockers of its transport function rather than as its transportable substrates.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Fenoprofen/pharmacology , Ibuprofen/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Dose-Response Relationship, Drug , Fenoprofen/chemistry , Humans , Ibuprofen/chemistry , Membrane Potentials , Molecular Structure , Monocarboxylic Acid Transporters , Niacin/metabolism , Oocytes/metabolism , Propionates , Sodium/metabolism , Structure-Activity Relationship , Transfection , Xenopus laevis
4.
Methods Mol Med ; 122: 285-300, 2006.
Article in English | MEDLINE | ID: mdl-16511988

ABSTRACT

We use two different heterologous expression systems to characterize the functional features of plasma membrane transporters cloned from placenta. The first is the vaccinia virus expression system that utilizes a recombinant vaccinia virus carrying a transgene for T7 RNA polymerase. Mammalian cells, when infected with this virus, are able to produce T7 RNA polymerase. If these cells are then transfected with a transporter cDNA which is under the control of T7 promoter in the plasmid, the T7 RNA polymerase will transcribe the cDNA and the cellular machinery will synthesize the transporter protein from the resultant mRNA and target it to the plasma membrane. The transport function of the heterologously expressed transporter can then be monitored by uptake of suitable substrates in these cells. The second is the Xenopus laevis oocyte expression system in which any transporter can be expressed heterologously by injection of the oocytes with corresponding cRNA. The functional features of the expressed transporter can then be monitored either by uptake of suitable substrates or by electrophysiological means if the transporter function is electrogenic. Both methods are complementary to each other and together provide an effective means to delineate the functional features of various transporters, irrespective of their independent transport mechanisms.


Subject(s)
Membrane Transport Proteins/metabolism , Placenta/metabolism , Transfection/methods , Animals , Cell Line , DNA, Complementary , Electrophysiology , Gene Expression , Humans , Microinjections , Oocytes , Plasmids , Radioisotopes , Vaccinia virus , Xenopus
5.
Mol Pharmacol ; 69(5): 1542-53, 2006 May.
Article in English | MEDLINE | ID: mdl-16446384

ABSTRACT

We characterized the electrophysiology, kinetics, and quantitative structure-activity relationship (QSAR) of the human concentrative nucleoside transporter 3 (hCNT3) expressed in Xenopus laevis oocytes by measuring substrate-induced inward currents using a two-microelectrode voltage-clamp system. At membrane potentials between -30 and -150 mV, sodium activation of gemcitabine transport was sigmoidal, with a K0.5 of 8.5+/-0.3 mM for Na+ and a Hill coefficient of 2.2+/-0.25 independent of membrane potential. We measured the Imax and K0.5 for substrate at -50 mV for the nucleoside analog drugs gemcitabine (638+/-58 nA, 59.7+/-17.5 microM), ribavirin (546+/-37 nA, 61.0+/-13.2 microM), AZT (420+/-4 nA, 310+/-9 microM), and 3-deazauridine (506+/-30 nA, 50.8+/-9.90 microM). K0.5 and Imax for substrate were dependent on membrane potential (both increasing as the membrane became more hyperpolarized) for all four drugs. hCNT3 also exhibited pre-steady-state currents. The quantitative structure-activity relationship (QSAR) was examined using comparative molecular field analysis and comparative molecular similarity indices analysis of the inward currents induced by 27 nucleoside analogs with substitutions at both the ribose and the nucleobase. Two statistically significant QSAR models identified electrostatic interaction as the major force in hCNT3 transport and attributed a critical role to the 3'-hydroxyl position of hCNT3 substrates. Steric hindrance at the 3-position and positive charge at the 5-position of the pyrimidine ring were favorable for transport. Two hCNT3 pharmacophore models revealed the minimal features required for hCNT3 transport as two hydrogen bond acceptors at 3'-OH and 5'-O and the hydrophobic center occupied by the base ring.


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/physiology , Algorithms , Animals , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacokinetics , Electrophysiology/methods , Female , Humans , Kinetics , Membrane Potentials/physiology , Nucleosides/chemistry , Nucleosides/metabolism , Oocytes/physiology , Purines/metabolism , Pyrimidines/metabolism , Quantitative Structure-Activity Relationship , Transfection , Xenopus laevis , Gemcitabine
6.
Biochem J ; 392(Pt 3): 655-64, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16104846

ABSTRACT

We report in the present paper, on the isolation and functional characterization of slc5a12, the twelfth member of the SLC5 gene family, from mouse kidney. The slc5a12 cDNA codes for a protein of 619 amino acids. Heterologous expression of slc5a12 cDNA in mammalian cells induces Na+-dependent transport of lactate and nicotinate. Several other short-chain monocarboxylates compete with nicotinate for the cDNA-induced transport process. Expression of slc5a12 in Xenopus oocytes induces electrogenic and Na+-dependent transport of lactate, nicotinate, propionate and butyrate. The substrate specificity of slc5a12 is similar to that of slc5a8, an Na+-coupled transporter for monocarboxylates. However, the substrate affinities of slc5a12 were much lower than those of slc5a8. slc5a12 mRNA is expressed in kidney, small intestine and skeletal muscle. In situ hybridization with sagittal sections of mouse kidney showed predominant expression of slc5a12 in the outer cortex. This is in contrast with slc5a8, which is expressed in the cortex as well as in the medulla. The physiological function of slc5a12 in the kidney is likely to mediate the reabsorption of lactate. In the intestinal tract, slc5a12 is expressed in the proximal parts, whereas slc5a8 is expressed in the distal parts. The expression of slc5a12 in the proximal parts of the intestinal tract, where there is minimal bacterial colonization, suggests that the physiological function of slc5a12 is not to mediate the absorption of short-chain monocarboxylates derived from bacterial fermentation but rather to mediate the absorption of diet-derived short-chain monocarboxylates. Based on the functional and structural similarities between slc5a8 and slc5a12, we suggest that the two transporters be designated as SMCT1 (sodium-coupled monocarboxylate transporter 1) and SMCT2 respectively.


Subject(s)
Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Sodium-Glucose Transport Proteins/genetics , Sodium-Glucose Transport Proteins/metabolism , Sodium/metabolism , Amino Acid Sequence , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Cloning, Molecular , Humans , Intestine, Small/metabolism , Kidney/metabolism , Mice , Molecular Sequence Data , Muscle, Skeletal/metabolism , Oocytes , Organ Specificity , Phylogeny , Sequence Alignment , Sequence Homology, Amino Acid , Xenopus laevis
7.
Biochem J ; 388(Pt 1): 309-16, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15651982

ABSTRACT

SMCT (sodium-coupled monocarboxylate transporter; slc5a8) is a Na+-coupled transporter for lactate, pyruvate and short-chain fatty acids. Similar to these already known substrates of SMCT, the water-soluble B-complex vitamin nicotinic acid also exists as a monocarboxylate anion (nicotinate) under physiological conditions. Therefore we evaluated the ability of SMCT to mediate the uptake of nicotinate. In mammalian cells, the cloned mouse SMCT (slc5a8) induced the uptake of nicotinate. The SMCT-induced uptake was Na+-dependent. The Michaelis constant for the uptake process was 296+/-88 microM. The Na+-activation kinetics indicated that at least two Na+ ions are involved in the process. Among the various structural analogues tested, nicotinate was the most effective substrate. Nicotinamide and methylnicotinate were not recognized by the transporter. 2-pyrazine carboxylate and isonicotinate interacted with the transporter to a moderate extent. SMCT-mediated uptake of nicotinate was inhibited by lactate and pyruvate. In the Xenopus laevis oocyte expression system, SMCT-mediated nicotinate transport was electrogenic, as evident from the nicotinate-induced inward currents under voltage-clamp conditions. Substrate-induced currents in this expression system corroborated the substrate specificity determined in the mammalian cell expression system. The kinetic parameters with regard to the affinity of the transporter for nicotinate and the Hill coefficient for Na+ activation, determined by using the oocyte expression system, were also similar to those obtained from the mammalian cell expression system. We conclude that SMCT functions not only as a Na+-coupled transporter for short-chain fatty acids and lactate but also as a Na+-coupled transporter for the water-soluble vitamin nicotinic acid.


Subject(s)
Biological Transport, Active , Cation Transport Proteins/metabolism , Electrophysiology , Niacin/metabolism , Sodium/physiology , Animals , Cells, Cultured , Epithelial Cells/metabolism , Gene Expression , Humans , Molecular Structure , Monocarboxylic Acid Transporters , Niacin/chemistry , Oocytes , Retinal Pigment Epithelium/cytology , Substrate Specificity , Xenopus laevis
8.
J Biol Chem ; 280(3): 2065-77, 2005 Jan 21.
Article in English | MEDLINE | ID: mdl-15542610

ABSTRACT

GABA functions as an inhibitory neurotransmitter in body muscles and as an excitatory neurotransmitter in enteric muscles in Caenorhabditis elegans. Whereas many of the components of the GABA-ergic neurotransmission in this organism have been identified at the molecular and functional levels, no transporter specific for this neurotransmitter has been identified to date. Here we report on the cloning and functional characterization of a GABA transporter from C. elegans (ceGAT-1) and on the functional relevance of the transporter to the biology of body muscles and enteric muscles. ceGAT-1 is coded by snf-11 gene, a member of the sodium-dependent neurotransmitter symporter gene family in C. elegans. The cloned ceGAT-1 functions as a Na(+)/Cl(-)-coupled high-affinity transporter selective for GABA with a K(t) of approximately 15 microm. The Na(+):Cl(-):GABA stoichiometry for ceGAT-1-mediated transport process is 2:1:1. The transport process is electrogenic as evidenced from GABA-induced inward currents in Xenopus laevis oocytes that express ceGAT-1 heterologously. The transporter is expressed exclusively in GABA-ergic neurons and in two other additional neurons. We also investigated the functional relevance of ceGAT-1 to the biology of body muscles and enteric muscles by ceGAT-1-specific RNA interference (RNAi) in rrf-3 mutant, a strain of C. elegans in which neurons are not refractory to RNAi as in the wild type strain. The down-regulation of ceGAT-1 by RNAi leads to an interesting phenotype associated with altered function of body muscles (as evident from changes in thrashing frequency) and enteric muscles (as evident from the rates of defecation failure) and also with altered sensitivity to aldicarb-induced paralysis. These findings provide unequivocal evidence for a modulatory role of GABA and ceGAT-1 in the biology of cholinergic neurons and in the function of body muscles and enteric muscles in this organism.


Subject(s)
Caenorhabditis elegans/metabolism , Membrane Transport Proteins/metabolism , Muscles/physiology , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans Proteins , Cloning, Molecular , DNA Primers , GABA Plasma Membrane Transport Proteins , Kinetics , Membrane Transport Proteins/genetics , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Vesicular Inhibitory Amino Acid Transport Proteins , Xenopus laevis
9.
J Biol Chem ; 279(43): 44522-32, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15322102

ABSTRACT

We report here on the expression of slc5a8 in kidney and its relevance to Na(+)-coupled reabsorption of lactate. slc5a8 is the murine ortholog of SLC5A8, a candidate tumor suppressor gene, which we recently cloned from human intestine and demonstrated its functional identity as a Na(+)-coupled transporter for short-chain fatty acids and lactate. The slc5a8 cDNA, cloned from mouse kidney, codes for a protein consisting of 611 amino acids. When expressed heterologously in mammalian cells or Xenopus oocytes, slc5a8 mediates Na(+)-coupled electrogenic transport of lactate/pyruvate as well as short-chain fatty acids (e.g. acetate, propionate, and butyrate). The Na+/fatty acid stoichiometry varies depending on the fatty acid substrate (2:1 for lactate and 4:1 for propionate). This phenomenon of variable Na+/substrate stoichiometry depending on the fatty acid substrate is also demonstrable with human SLC5A8. In situ hybridization with sagittal sections of mouse kidney demonstrates abundant expression of the transcripts in the cortex as well as the medulla. Brush border membrane vesicles prepared from rabbit kidney are able to transport lactate in a Na(+)-coupled manner. The transport process exhibits the overshoot phenomenon, indicating uphill lactate transport in response to the transmembrane Na+ gradient. The Na(+)-coupled lactate transport in these membrane vesicles is inhibitable by short-chain fatty acids. We conclude that slc5a8 is expressed abundantly in the kidney and that it plays a role in the active reabsorption of lactate. slc5a8 is the first transporter known to be expressed in mammalian kidney that has the ability to mediate the Na(+)-coupled reabsorption of lactate.


Subject(s)
Cation Transport Proteins/biosynthesis , Kidney/metabolism , Lactates/metabolism , Sodium/metabolism , Animals , Biological Transport , Blotting, Northern , Cloning, Molecular , Coumaric Acids/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Fatty Acids/metabolism , Humans , Hydrogen-Ion Concentration , In Situ Hybridization , Intestinal Mucosa/metabolism , Kinetics , Mice , Microvilli/metabolism , Models, Biological , Molecular Sequence Data , Monocarboxylic Acid Transporters , Oocytes/metabolism , Propionates/metabolism , Pyruvates/metabolism , RNA, Complementary/metabolism , Rabbits , Substrate Specificity , Time Factors , Tissue Distribution , Transfection , Xenopus , Xenopus laevis
10.
J Biol Chem ; 279(14): 13293-6, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14966140

ABSTRACT

SLC5A8, a tumor suppressor gene down-regulated in human colon cancer, codes for a transporter in the Na(+)/glucose cotransporter gene family, but the definitive functional identity of the transporter protein is not known. Since this gene is expressed abundantly in the colon where short-chain fatty acids are generated by bacterial fermentation, we tested the hypothesis that it codes for a Na(+)-coupled transporter for these fatty acids. The coding region of SLC5A8 mRNA was amplified from human intestine and expressed heterologously in Xenopus laevis oocytes. Transport function was monitored by uptake of radiolabeled substrates and by substrate-induced currents under voltage-clamp conditions. Uptake of short-chain fatty acids (lactate, pyruvate, acetate, propionate, and butyrate) in oocytes expressing SLC5A8 was severalfold higher than in uninjected oocytes. Exposure of SLC5A8-expressing oocytes to these fatty acids induced inward currents under voltage-clamp conditions in a Na(+)-dependent manner. These currents were saturable and the substrate concentrations needed for half-maximal induction of the current were in the range of 0.08-2.5 mm. The substrate-induced currents decreased as the carbon chain length of the substrates increased. The Na(+)-activation kinetics indicated involvement of more than one Na(+) ion in the activation process. Direct measurements of substrate (propionate) and charge transfer showed that three positive charges are transferred into oocytes per substrate molecule. These studies establish the functional identity of SLC5A8 as a Na(+)-coupled transporter for short-chain fatty acids.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/physiopathology , Fatty Acids, Volatile/metabolism , Animals , Binding, Competitive , Cation Transport Proteins/chemistry , Down-Regulation , Electrochemistry , Gene Expression Regulation, Neoplastic , Humans , Iodides/metabolism , Membrane Potentials/physiology , Monocarboxylic Acid Transporters , Oocytes/physiology , Patch-Clamp Techniques , RNA, Messenger/analysis , Sodium/metabolism , Xenopus
11.
Biochem J ; 378(Pt 3): 949-57, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-14656221

ABSTRACT

In the present study, we report on the molecular cloning and functional characterization of mouse NaCT (Na+-coupled citrate transporter), the mouse orthologue of Drosophila Indy. Mouse NaCT consists of 572 amino acids and is highly similar to rat and human NaCTs in primary sequence. The mouse nact gene coding for the transporter is approx. 23 kb long and consists of 12 exons. When expressed in mammalian cells, the cloned transporter mediates the Na+-coupled transport of citrate and succinate. Competition experiments reveal that mouse NaCT also recognizes other tricarboxylic acid cycle intermediates such as malate, fumarate and 2-oxo-glutarate as excellent substrates. The Michaelis-Menten constant for the transport process is 38+/-5 mM for citrate and 37+/-6 mM for succinate at pH 7.5. The transport process is electrogenic and exhibits an obligatory requirement for Na+. Na+-activation kinetics indicates that multiple Na+ ions are involved in the activation process. Extracellular pH has a differential effect on the transport function of mouse NaCT depending on whether the transported substrate is citrate or succinate. The Michaelis-Menten constants for these substrates are also influenced markedly by pH. When examined in the Xenopus laevis oocyte expression system with the two-microelectrode voltage-clamp technique, the transport process mediated by mouse NaCT is electrogenic. The charge-to-substrate ratio is 1 for citrate and 2 for succinate. The most probable transport mechanism predicted by these studies involves the transport of citrate as a tervalent anion and succinate as a bivalent anion with a fixed Na+/substrate stoichiometry of 4:1. The present study provides the first unequivocal evidence for the electrogenic nature of mammalian NaCT.


Subject(s)
Citric Acid Cycle , Symporters/genetics , Symporters/metabolism , Amino Acid Sequence , Animals , Biological Transport/drug effects , Cells, Cultured , Citric Acid/metabolism , Cloning, Molecular , Dicarboxylic Acid Transporters , Exons , Genome , Humans , Introns , Mice , Molecular Sequence Data , Oocytes/metabolism , Sodium/pharmacology , Substrate Specificity , Succinic Acid/metabolism , Symporters/chemistry , Xenopus laevis
12.
Biochem J ; 379(Pt 1): 191-8, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14678010

ABSTRACT

We have cloned and functionally characterized an Na+-coupled citrate transporter from Caenorhabditis elegans (ceNAC-2). This transporter shows significant sequence homology to Drosophila Indy and the mammalian Na+-coupled citrate transporter NaCT (now known as NaC2). When heterologously expressed in a mammalian cell line or in Xenopus oocytes, the cloned ceNAC-2 mediates the Na+-coupled transport of various intermediates of the citric acid cycle. However, it transports the tricarboxylate citrate more efficiently than dicarboxylates such as succinate, a feature different from that of ceNAC-1 (formerly known as ceNaDC1) and ceNAC-3 (formerly known as ceNaDC2). The transport process is electrogenic, as evidenced from the substrate-induced inward currents in oocytes expressing the transporter under voltage-clamp conditions. Expression studies using a reporter-gene fusion method in transgenic C. elegans show that the gene is expressed in the intestinal tract, the organ responsible for not only the digestion and absorption of nutrients but also for the storage of energy in this organism. Functional knockdown of the transporter by RNAi (RNA interference) not only leads to a significant increase in life span, but also causes a significant decrease in body size and fat content. The substrates of ceNAC-2 play a critical role in metabolic energy production and in the biosynthesis of cholesterol and fatty acids. The present studies suggest that the knockdown of these metabolic functions by RNAi is linked to an extension of life span and a decrease in fat content and body size.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Citric Acid/metabolism , Organic Anion Transporters/physiology , Sodium/metabolism , Adipose Tissue/anatomy & histology , Animals , Animals, Genetically Modified , Biological Transport , Body Composition/genetics , Body Constitution/genetics , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/isolation & purification , Cell Line , Citric Acid Cycle , Cloning, Molecular , Genes, Helminth , Genes, Reporter , Humans , Intestinal Mucosa/metabolism , Longevity/genetics , Molecular Sequence Data , Oocytes , Organ Specificity , Organic Anion Transporters/genetics , Organic Anion Transporters/isolation & purification , Patch-Clamp Techniques , Pigment Epithelium of Eye/cytology , RNA Interference , Recombinant Fusion Proteins/metabolism , Xenopus laevis
13.
J Pharmacol Exp Ther ; 308(3): 1138-47, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14617696

ABSTRACT

We evaluated the potential of the Na(+)- and Cl(-)-coupled amino acid transporter ATB(0,+) as a delivery system for amino acid-based prodrugs. Immunofluorescence analysis indicated that ATB(0,+) is expressed abundantly on the luminal surface of cells lining the lumen of the large intestine and the airways of the lung and in various ocular tissues, including the conjunctival epithelium, the tissues easily amenable for drug delivery. We screened a variety of beta-carboxyl derivatives of aspartate and gamma-carboxyl derivatives of glutamate as potential substrates for this transporter using heterologous expression systems. In mammalian cells expressing the cloned ATB(0,+), several of the aspartate and glutamate derivatives inhibited glycine transport via ATB(0,+). Direct evidence for ATB(0,+)-mediated transport of these derivatives was obtained in Xenopus laevis oocytes using electrophysiological methods. Exposure of oocytes, which express ATB(0,+) heterologously, to aspartate beta-benzyl ester as a model derivative induced inward currents in a Na(+)- and Cl(-)-dependent manner with a Na(+)/Cl(-)/aspartate beta-benzyl ester stoichiometry of 2:1:1. ATB(0,+) transported not only the beta-carboxyl derivatives of aspartate and the gamma-carboxyl derivatives of glutamate but also valacyclovir, which is an alpha-carboxyl ester of acyclovir with valine. The transport of valacyclovir via ATB(0,+) was demonstrable in both heterologous expression systems. This process was dependent on Na(+) and Cl(-). The ability of ATB(0,+) to transport valacyclovir was comparable with that of the peptide transporter PEPT1. These findings suggest that ATB(0,+) has significant potential as a delivery system for amino acid-based drugs and prodrugs.


Subject(s)
Acyclovir/analogs & derivatives , Amino Acid Transport System ASC/metabolism , Amino Acids/metabolism , Chlorides/metabolism , Prodrugs/metabolism , Sodium/metabolism , Valine/analogs & derivatives , Acyclovir/chemistry , Acyclovir/pharmacokinetics , Amino Acid Transport Systems/metabolism , Animals , Biological Transport , Drug Delivery Systems , Humans , Mice , Minor Histocompatibility Antigens , Oocytes/metabolism , Valacyclovir , Valine/pharmacokinetics , Xenopus laevis
14.
Exp Eye Res ; 77(6): 687-97, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14609557

ABSTRACT

Reduced-folate transporter-1 (RFT-1) transports reduced-folates, such as N5-methyltetrahydrofolate (MTF), the predominant circulating form of folate. In RPE, RFT-1 is localized to the apical membrane and is thought to transport folate from RPE to photoreceptor cells. Folate is required for DNA, RNA, protein synthesis and the conversion of homocysteine (Hcy) to methionine. Decreased folate levels are associated with increased Hcy levels. In the present study, we asked whether RFT-1 activity in RPE is altered under high Hcy conditions and examined the transport mechanism for Hcy in RPE. Treatment of ARPE-19 cells, a human RPE cell line, with Hcy at concentrations higher than 50 microM led to a significant decrease in RFT-1 activity. This effect increased as the treatment time increased. The inhibitory effect of Hcy on RFT-1 activity was not non-specific, as the activities of several other nutrient transporters were not affected under identical conditions. The effect of Hcy on RFT-1 was associated primarily with a decrease in the maximal velocity with no detectable change in substrate affinity. The decrease in RFT-1 activity was accompanied by parallel changes in RFT-1 mRNA and protein. Uptake of Hcy in ARPE-19 cells occurred via several transport systems, including Na+-independent systems L and b(0,+) and the Na+-dependent systems B0, ATB(0,+) and A. Studies of the interaction of Hcy with one of the cloned transporters (ATB(0,+)) provided direct evidence for the translocation of Hcy across the membrane via the transporter. We conclude that several transport systems operate in ARPE-19 cells for the entry of Hcy and that high levels of Hcy have deleterious effects on the expression and activity of RFT-1 in these cells.


Subject(s)
Homocysteine/pharmacology , Pigment Epithelium of Eye/drug effects , Biological Transport , Blotting, Western/methods , Carrier Proteins/metabolism , Cell Line , Culture Media , Dose-Response Relationship, Drug , Eye Proteins/analysis , Folic Acid/metabolism , Gene Expression , Humans , Membrane Proteins/metabolism , Membrane Transport Proteins , Pigment Epithelium of Eye/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Sodium/metabolism , Time Factors
15.
J Physiol ; 546(Pt 2): 349-61, 2003 Jan 15.
Article in English | MEDLINE | ID: mdl-12527723

ABSTRACT

The human orthologue of the H(+)-coupled amino acid transporter (hPAT1) was cloned from the human intestinal cell line Caco-2 and its functional characteristics evaluated in a mammalian cell heterologous expression system. The cloned hPAT1 consists of 476 amino acids and exhibits 85 % identity with rat PAT1. Among the various human tissues examined by Northern blot, PAT1 mRNA was expressed most predominantly in the intestinal tract. When expressed heterologously in mammalian cells, hPAT1 mediated the transport of alpha-(methylamino)isobutyric acid (MeAIB). The cDNA-induced transport was Na(+)-independent, but was energized by an inwardly directed H(+) gradient. hPAT1 interacted with glycine, L-alanine, L-proline, alpha-aminoisobutyrate (AIB) and gamma-aminobutyrate (GABA), as evidenced from direct transport measurements and from competition experiments with MeAIB as a transport substrate. hPAT1 also recognized the D-isomers of alanine and proline. With serine and cysteine, though the L-isomers did not interact with hPAT1 to any significant extent, the corresponding D-isomers were recognized as substrates. With proline and alanine, the affinity was similar for L- and D-isomers. However, with cysteine and serine, the D-isomers showed 6- to 8-fold higher affinity for hPAT1 than the corresponding L-isomers. These functional characteristics of hPAT1 closely resemble those that have been described previously for the H(+)-coupled amino acid transport system in Caco-2 cells. Furthermore, there was a high degree of correlation (r(2) = 0.93) between the relative potencies of various amino acids to inhibit the H(+)-coupled MeAIB transport measured with native Caco-2 cells and with hPAT1 in the heterologous expression system. Immunolocalization studies showed that PAT1 was expressed exclusively in the apical membrane of Caco-2 cells. These data suggest that hPAT1 is responsible for the H(+)-coupled amino acid transport expressed in the apical membrane of Caco-2 cells.


Subject(s)
Amino Acid Transport Systems/chemistry , Amino Acid Transport Systems/metabolism , Intestinal Mucosa/metabolism , Amino Acid Sequence/genetics , Amino Acid Transport Systems/genetics , Caco-2 Cells , DNA, Complementary/genetics , Digestive System/metabolism , Exons , Humans , Immunologic Techniques , Intestines/cytology , Introns , Molecular Sequence Data , RNA, Messenger/metabolism , Structure-Activity Relationship , Symporters , Tissue Distribution
16.
J Biol Chem ; 278(8): 6136-44, 2003 Feb 21.
Article in English | MEDLINE | ID: mdl-12480943

ABSTRACT

We have cloned and functionally characterized two Na(+)-coupled dicarboxylate transporters, namely ceNaDC1 and ceNaDC2, from Caenorhabditis elegans. These two transporters show significant sequence homology with the product of the Indy gene identified in Drosophila melanogaster and with the Na(+)-coupled dicarboxylate transporters NaDC1 and NaDC3 identified in mammals. In a mammalian cell heterologous expression system, the cloned ceNaDC1 and ceNaDC2 mediate Na(+)-coupled transport of various dicarboxylates. With succinate as the substrate, ceNaDC1 exhibits much lower affinity compared with ceNaDC2. Thus, ceNaDC1 and ceNaDC2 correspond at the functional level to the mammalian NaDC1 and NaDC3, respectively. The nadc1 and nadc2 genes are not expressed at the embryonic stage, but the expression is detectable all through the early larva stage to the adult stage. Tissue-specific expression pattern studies using a reporter gene fusion approach in transgenic C. elegans show that both genes are coexpressed in the intestinal tract, an organ responsible for not only the digestion and absorption of nutrients but also for the storage of energy in this organism. Independent knockdown of the function of these two transporters in C. elegans using the strategy of RNA interference suggests that NaDC1 is not associated with the regulation of average life span in this organism, whereas the knockdown of NaDC2 function leads to a significant increase in the average life span. Disruption of the function of the high affinity Na(+)-coupled dicarboxylate transporter NaDC2 in C. elegans may lead to decreased availability of dicarboxylates for cellular production of metabolic energy, thus creating a biological state similar to that of caloric restriction, and consequently leading to life span extension.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Carrier Proteins/physiology , Dicarboxylic Acid Transporters/physiology , Gene Expression Regulation, Developmental , Membrane Proteins/physiology , Organic Anion Transporters, Sodium-Dependent/physiology , Symporters/physiology , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cloning, Molecular , Cosmids , DNA Primers , Dicarboxylic Acid Transporters/chemistry , Dicarboxylic Acid Transporters/genetics , Escherichia coli/genetics , Life Expectancy , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Organic Anion Transporters, Sodium-Dependent/chemistry , Organic Anion Transporters, Sodium-Dependent/genetics , Polymerase Chain Reaction/methods , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Symporters/chemistry , Symporters/genetics
17.
Biochem J ; 367(Pt 2): 313-9, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12186628

ABSTRACT

Indy is a gene in Drosophila melanogaster which, when made dysfunctional, leads to an extension of the average adult life span of the organism. The present study was undertaken to clone the Indy gene-product and to establish its functional identity. We isolated a full-length Indy cDNA from a D. melanogaster cDNA library. The cDNA codes for a protein of 572 amino acids [( Drosophila Indy (drIndy)]. In its amino acid sequence, drIndy exhibits comparable similarity to the two known Na(+)-coupled dicarboxylate transporters in mammals; namely, NaDC1 (35% identity) and NaDC3 (34% identity). We elucidated the functional characteristics of drIndy in two different heterologous expression systems by using mammalian cells and Xenopus laevis oocytes. These studies show that drIndy is a cation-independent electroneutral transporter for a variety of tricarboxylic acid-cycle intermediates, with preference for citrate compared with succinate. These characteristics of drIndy differ markedly from those of NaDC1 and NaDC3, indicating that neither of these latter transporters is the mammalian functional counterpart of drIndy. Since drIndy is a transporter for tricarboxylic acid-cycle intermediates, dysfunction of the Indy gene may lead to decreased production of metabolic energy in cells, analogous to caloric restriction. This might provide the molecular basis for the observation that disruption of the Indy gene function in Drosophila leads to extension of the average adult life span of the organism.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Symporters/metabolism , Animals , Cations/metabolism , Cells, Cultured , Citric Acid/metabolism , Citric Acid Cycle/physiology , Cloning, Molecular , Dicarboxylic Acid Transporters/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Molecular Sequence Data , Oocytes/metabolism , Patch-Clamp Techniques , Pyruvic Acid/metabolism , Sodium/metabolism , Substrate Specificity , Succinic Acid/metabolism , Symporters/genetics , Xenopus laevis
18.
Biol Psychiatry ; 51(9): 723-32, 2002 May 01.
Article in English | MEDLINE | ID: mdl-11983186

ABSTRACT

BACKGROUND: Fifty-one patients with major depression were classified for 5-HTT promoter region polymorphism and platelet 5-HTT kinetics before treatment with fluoxetine, and then examined for treatment outcome. METHODS: Dose was stratified from 1.25 mg to 40 mg per day to allow for the possibility that one genotype could express a lower-dose fluoxetine response. A repeated-measures analysis of variance of 24-item Hamilton depression change through baseline, 1-week placebo lead-in, and 6, 12, and 18 weeks treatment was done to test a genotype effect on outcome. RESULTS: Genotype had a significant effect on outcome (F = 4.7, p <.02), with the initial affinity constant (K(m)) (F = 11.9, p =.001), and dose (F = 6.0, p <.02) being significant covariates on outcome as well. The gene effect, however, was complex in that the 5-HTT promoter region insertion showed two effects: both a placebo response effect (F = 4, p <.025), and a drug dose response effect (r =.40, p <.01). The long allele group was more responsive to placebo, as well as more responsive to drug dose than was the short allele group. CONCLUSIONS: This is the first study to examine the antidepressant dose-response relationship to 5-HTT kinetics and genetics. The findings indicate that both the initial affinity and genotype of 5-HTT may contribute in unique ways to the variation in the outcome of depression treatment trials.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Depressive Disorder/drug therapy , Fluoxetine/therapeutic use , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Transport Proteins , Nerve Tissue Proteins , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adolescent , Adult , Aged , Analysis of Variance , Depressive Disorder/psychology , Dose-Response Relationship, Drug , Female , Fluoxetine/metabolism , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics , Psychiatric Status Rating Scales , Serotonin Plasma Membrane Transport Proteins , Selective Serotonin Reuptake Inhibitors/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...