Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biochem J ; 475(20): 3293-3309, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30254099

ABSTRACT

Human equilibrative nucleoside transporter 1 (hENT1), the first identified member of the ENT family of integral membrane proteins, is the primary mechanism for cellular uptake of physiologic nucleosides and many antineoplastic and antiviral nucleoside drugs. hENT1, which is potently inhibited by nitrobenzylthioinosine (NBMPR), possesses 11 transmembrane helical domains with an intracellular N-terminus and an extracellular C-terminus. As a protein with 10 endogenous cysteine residues, it is sensitive to inhibition by the membrane permeable sulfhydryl-reactive reagent N-ethylmaleimide (NEM) but is unaffected by the membrane impermeable sulfhydryl-reactive reagent p-chloromercuriphenyl sulfonate. To identify the residue(s) involved in NEM inhibition, we created a cysteine-less version of hENT1 (hENT1C-), with all 10 endogenous cysteine residues mutated to serine, and showed that it displays wild-type uridine transport and NBMPR-binding characteristics when produced in the Xenopus oocyte heterologous expression system, indicating that endogenous cysteine residues are not essential for hENT1 function. We then tested NEM sensitivity of recombinant wild-type hENT1, hENT1 mutants C1S to C10S (single cysteine residues replaced by serine), hENT1C- (all cysteine residues replaced by serine), and hENT1C- mutants S1C to S10C (single serine residues converted back to cysteine). Mutants C9S (C416S/hENT1) and S9C (S416C/hENT1C-) were insensitive and sensitive, respectively, to inhibition by NEM, identifying Cys416 as the endofacial cysteine residue in hENT1 responsible for NEM inhibition. Kinetic experiments suggested that NEM modification of Cys416, which is located at the inner extremity of TM10, results in the inhibition of hENT1 uridine transport and NBMPR binding by constraining the protein in its inward-facing conformation.


Subject(s)
Cysteine/metabolism , Equilibrative Nucleoside Transporter 1/metabolism , Ethylmaleimide/metabolism , Animals , Dose-Response Relationship, Drug , Equilibrative Nucleoside Transporter 1/antagonists & inhibitors , Equilibrative Nucleoside Transporter 1/genetics , Female , Humans , Protein Binding/physiology , Thioinosine/analogs & derivatives , Thioinosine/metabolism , Thioinosine/pharmacology , Uridine/metabolism , Uridine/pharmacology , Xenopus laevis
2.
J Biol Chem ; 292(23): 9505-9522, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28385889

ABSTRACT

The human SLC28 family of concentrative nucleoside transporter (CNT) proteins has three members: hCNT1, hCNT2, and hCNT3. Na+-coupled hCNT1 and hCNT2 transport pyrimidine and purine nucleosides, respectively, whereas hCNT3 transports both pyrimidine and purine nucleosides utilizing Na+ and/or H+ electrochemical gradients. Escherichia coli CNT family member NupC resembles hCNT1 in permeant selectivity but is H+-coupled. Using heterologous expression in Xenopus oocytes and the engineered cysteine-less hCNT3 protein hCNT3(C-), substituted cysteine accessibility method analysis with the membrane-impermeant thiol reactive reagent p-chloromercuribenzene sulfonate was performed on the transport domain (interfacial helix 2, hairpin 1, putative transmembrane domain (TM) 7, and TM8), as well as TM9 of the scaffold domain of the protein. This systematic scan of the entire C-terminal half of hCNT3(C-) together with parallel studies of the transport domain of wild-type hCNT1 and the corresponding TMs of cysteine-less NupC(C-) yielded results that validate the newly developed structural homology model of CNT membrane architecture for human CNTs, revealed extended conformationally mobile regions within transport-domain TMs, identified pore-lining residues of functional importance, and provided evidence of an emerging novel elevator-type mechanism of transporter function.


Subject(s)
Membrane Transport Proteins/chemistry , Amino Acid Substitution , Animals , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation, Missense , Protein Domains , Protein Structure, Secondary , Structural Homology, Protein , Structure-Activity Relationship , Xenopus laevis
3.
Am J Physiol Renal Physiol ; 303(4): F527-39, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22647630

ABSTRACT

Human SLC2A9 (GLUT9) is a novel high-capacity urate transporter belonging to the facilitated glucose transporter family. In the present study, heterologous expression in Xenopus oocytes has allowed us to undertake an in-depth radiotracer flux and electrophysiological study of urate transport mediated by both isoforms of SLC2A9 (a and b). Addition of urate to SLC2A9-producing oocytes generated outward currents, indicating electrogenic transport. Urate transport by SLC2A9 was voltage dependent and independent of the Na(+) transmembrane gradient. Urate-induced outward currents were affected by the extracellular concentration of Cl(-), but there was no evidence for exchange of the two anions. [(14)C]urate flux studies under non-voltage-clamped conditions demonstrated symmetry of influx and efflux, suggesting that SLC2A9 functions in urate efflux driven primarily by the electrochemical gradient of the cell. Urate uptake in the presence of intracellular hexoses showed marked differences between the two isoforms, suggesting functional differences between the two splice variants. Finally, the permeant selectivity of SLC2A9 was examined by testing the ability to transport a panel of radiolabeled purine and pyrimidine nucleobases. SLC2A9 mediated the uptake of adenine in addition to urate, but did not function as a generalized nucleobase transporter. The differential expression pattern of the two isoforms of SLC2A9 in the human kidney's proximal convoluted tubule and its electrogenic transport of urate suggest that these transporters play key roles in the regulation of plasma urate levels and are therefore potentially important participants in hyperuricemia and hypouricemia.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Hexoses/metabolism , Uric Acid/metabolism , Animals , Biological Transport , Electrophysiological Phenomena , Gene Expression Regulation/physiology , Glucose Transport Proteins, Facilitative/genetics , Humans , Ion Channel Gating , Oocytes , Protein Isoforms/genetics , Protein Isoforms/metabolism , Xenopus laevis
4.
J Biol Chem ; 286(37): 32552-62, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21795683

ABSTRACT

The human equilibrative nucleoside transporters hENT1 and hENT2 (each with 456 residues) are 40% identical in amino acid sequence and contain 11 putative transmembrane helices. Both transport purine and pyrimidine nucleosides and are distinguished functionally by a difference in sensitivity to inhibition by nanomolar concentrations of nitrobenzylmercaptopurine ribonucleoside (NBMPR), hENT1 being NBMPR-sensitive. Previously, we used heterologous expression in Xenopus oocytes to demonstrate that recombinant hENT2 and its rat ortholog rENT2 also transport purine and pyrimidine bases, h/rENT2 representing the first identified mammalian nucleobase transporter proteins (Yao, S. Y., Ng, A. M., Vickers, M. F., Sundaram, M., Cass, C. E., Baldwin, S. A., and Young, J. D. (2002) J. Biol. Chem. 277, 24938-24948). The same study also revealed lower, but significant, transport of hypoxanthine by h/rENT1. In the present investigation, we have used the enhanced Xenopus oocyte expression vector pGEMHE to demonstrate that hENT1 additionally transports thymine and adenine and, to a lesser extent, uracil and guanine. Fluxes of hypoxanthine, thymine, and adenine by hENT1 were saturable and inhibited by NBMPR. Ratios of V(max) (pmol/oocyte · min(-1)):K(m) (mm), a measure of transport efficiency, were 86, 177, and 120 for hypoxantine, thymine, and adenine, respectively, compared with 265 for uridine. Hypoxanthine influx was competitively inhibited by uridine, indicating common or overlapping nucleobase and nucleoside permeant binding pockets, and the anticancer nucleobase drugs 5-fluorouracil and 6-mercaptopurine were also transported. Nucleobase transport activity was absent from an engineered cysteine-less version hENT1 (hENT1C-) in which all 10 endogenous cysteine residues were mutated to serine. Site-directed mutagenesis identified Cys-414 in transmembrane helix 10 of hENT1 as the residue conferring nucleobase transport activity to the wild-type transporter.


Subject(s)
Equilibrative Nucleoside Transporter 1/metabolism , Nucleosides/metabolism , Amino Acid Substitution , Animals , Binding Sites , Biological Transport, Active/physiology , Equilibrative Nucleoside Transporter 1/chemistry , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative-Nucleoside Transporter 2/chemistry , Equilibrative-Nucleoside Transporter 2/genetics , Equilibrative-Nucleoside Transporter 2/metabolism , Humans , Mutagenesis, Site-Directed , Mutation, Missense , Nucleosides/genetics , Oocytes/cytology , Oocytes/metabolism , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xenopus laevis
5.
J Biol Chem ; 284(25): 17281-17292, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19380585

ABSTRACT

The human SLC28 family of integral membrane CNT (concentrative nucleoside transporter) proteins has three members, hCNT1, hCNT2, and hCNT3. Na(+)-coupled hCNT1 and hCNT2 transport pyrimidine and purine nucleosides, respectively, whereas hCNT3 mediates transport of both pyrimidine and purine nucleosides utilizing Na(+) and/or H(+) electrochemical gradients. These and other eukaryote CNTs are currently defined by a putative 13-transmembrane helix (TM) topology model with an intracellular N terminus and a glycosylated extracellular C terminus. Recent mutagenesis studies, however, have provided evidence supporting an alternative 15-TM membrane architecture. In the absence of CNT crystal structures, valuable information can be gained about residue localization and function using substituted cysteine accessibility method analysis with thiol-reactive reagents, such as p-chloromercuribenzene sulfonate. Using heterologous expression in Xenopus oocytes and the cysteineless hCNT3 protein hCNT3C-, substituted cysteine accessibility method analysis with p-chloromercuribenzene sulfonate was performed on the TM 11-13 region, including bridging extramembranous loops. The results identified residues of functional importance and, consistent with a new revised 15-TM CNT membrane architecture, suggest a novel membrane-associated topology for a region of the protein (TM 11A) that includes the highly conserved CNT family motif (G/A)XKX(3)NEFVA(Y/M/F).


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , 4-Chloromercuribenzenesulfonate/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites/genetics , Cysteine/chemistry , Female , Humans , In Vitro Techniques , Membrane Transport Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Nucleosides/metabolism , Oocytes/drug effects , Oocytes/metabolism , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uridine/metabolism , Uridine/pharmacology , Xenopus laevis
6.
J Biol Chem ; 284(25): 17266-17280, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19380587

ABSTRACT

Human concentrative nucleoside transporter 3 (hCNT3) utilizes electrochemical gradients of both Na(+) and H(+) to accumulate pyrimidine and purine nucleosides within cells. We have employed radioisotope flux and electrophysiological techniques in combination with site-directed mutagenesis and heterologous expression in Xenopus oocytes to identify two conserved pore-lining glutamate residues (Glu-343 and Glu-519) with essential roles in hCNT3 Na(+)/nucleoside and H(+)/nucleoside cotransport. Mutation of Glu-343 and Glu-519 to aspartate, glutamine, and cysteine severely compromised hCNT3 transport function, and changes included altered nucleoside and cation activation kinetics (all mutants), loss or impairment of H(+) dependence (all mutants), shift in Na(+):nucleoside stoichiometry from 2:1 to 1:1 (E519C), complete loss of catalytic activity (E519Q) and, similar to the corresponding mutant in Na(+)-specific hCNT1, uncoupled Na(+) currents (E343Q). Consistent with close-proximity integration of cation/solute-binding sites within a common cation/permeant translocation pore, mutation of Glu-343 and Glu-519 also altered hCNT3 nucleoside transport selectivity. Both residues were accessible to the external medium and inhibited by p-chloromercuribenzene sulfonate when converted to cysteine.


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , 4-Chloromercuribenzenesulfonate/pharmacology , Animals , Binding, Competitive , Cell Membrane/metabolism , Conserved Sequence , Female , Glutamic Acid/chemistry , Guanosine/metabolism , Humans , In Vitro Techniques , Ion Transport , Kinetics , Membrane Transport Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Nucleosides/metabolism , Oocytes/drug effects , Oocytes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium/metabolism , Sodium/pharmacology , Uridine/metabolism , Xenopus
7.
Mol Pharmacol ; 74(5): 1372-80, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669604

ABSTRACT

3'-Deoxy-3'-fluorothymidine (FLT) is a positron emission tomography (PET) tracer used to identify proliferating tumor cells. The purpose of this study was to characterize FLT transport by human nucleoside transporters (hNTs) and to determine the role of hNTs for FLT uptake in various human cancer cell lines. FLT binding to hNTs was monitored by the inhibitory effects of FLT on [(3)H]uridine uptake in yeast cells producing recombinant hNT proteins. hCNT1 displayed the lowest FLT K(i) value for inhibition of [(3)H]uridine uptake, followed by hCNT3, hENT2, hENT1, and hCNT2. [(3)H]FLT was efficiently transported in Xenopus laevis oocytes individually producing hENT1, hENT2, hCNT1, or hCNT3. [(3)H]FLT uptake in MCF-7, A549, U251, A498, MIA PaCa-2, and Capan-2 cells was inhibited at least 50% by the hENT1 inhibitor nitrobenzylmercaptopurine ribonucleoside (NBMPR). According to results of real-time polymerase chain reactions, hENT1 and hENT2 had the most abundant hNT transcripts in all cell lines. Cell lines also underwent 1) [(3)H]NBMPR equilibrium binding assays with or without 5-S-{2-(1-[(fluorescein-5-yl)thioureido]hexanamido)ethyl}-6-N-(4-nitrobenzyl)-5-thioadenosine, a membrane-impermeable NBMPR analog, to determine plasma membrane hENT1 levels, and 2) dose-response NBMPR inhibition of [(3)H]FLT uptake. MCF-7, A549, and Capan-2 cells displayed NBMPR IC(50) values that were smaller or equal to NBMPR K(d) values, suggesting that 50% inhibition of hENT1 reduced [(3)H]FLT uptake by at least 50%. A strong correlation between extracellular NBMPR binding sites/cell and [(3)H]FLT uptake was observed for all cell lines except MIA PaCa-2. These data suggest that plasma membrane hNTs (especially hENT1) are important determinants of cellular FLT uptake.


Subject(s)
Carrier Proteins/physiology , Dideoxynucleosides/metabolism , Nucleosides/metabolism , Animals , Humans , Positron-Emission Tomography , Radioligand Assay , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
8.
J Biol Chem ; 283(36): 24922-34, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18621735

ABSTRACT

In humans, the SLC28 concentrative nucleoside transporter (CNT) protein family is represented by three Na+-coupled members; human CNT1 (hCNT1) and hCNT2 are pyrimidine and purine nucleoside-selective, respectively, whereas hCNT3 transports both purine and pyrimidine nucleosides and nucleoside drugs. Belonging to a phylogenetic CNT subfamily distinct from hCNT1/2, hCNT3 also mediates H+/nucleoside cotransport. Using heterologous expression in Xenopus oocytes, we have characterized a cysteineless version of hCNT3 (hCNT3C-). Processed normally to the cell surface, hCNT3C- exhibited hCNT3-like transport properties, but displayed a decrease in apparent affinity specific for Na+ and not H+. Site-directed mutagenesis experiments in wild-type and hCNT3C- backgrounds identified intramembranous Cys-561 as the residue responsible for this altered Na+-binding phenotype. Alanine at this position restored Na+ binding affinity, whereas substitution with larger neutral amino acids (threonine, valine, and isoleucine) abolished hCNT3 H+-dependent nucleoside transport activity. Independent of these findings, we have established that Cys-561 is located in a mobile region of the hCNT3 translocation pore adjacent to the nucleoside binding pocket and that access of p-chloromercuribenzene sulfonate to this residue reports a specific H+-induced conformational state of the protein ( Slugoski, M. D., Ng, A. M. L., Yao, S. Y. M., Smith, K. M., Lin, C. C., Zhang, J., Karpinski, E., Cass, C. E., Baldwin, S. A., and Young, J. D. (2008) J. Biol. Chem. 283, 8496-8507 ). The present investigation validates hCNT3C- as a template for substituted cysteine accessibility method studies of CNTs and reveals a pivotal functional role for Cys-561 in Na+- as well as H+-coupled modes of hCNT3 nucleoside transport.


Subject(s)
Membrane Transport Proteins/metabolism , Protons , Sodium/metabolism , Amino Acid Substitution , Animals , Binding Sites/physiology , Cysteine/genetics , Cysteine/metabolism , Female , Gene Expression , Humans , Membrane Transport Proteins/genetics , Mutagenesis, Site-Directed , Oocytes/cytology , Point Mutation , Protein Structure, Tertiary/physiology , Xenopus
9.
J Biol Chem ; 283(13): 8496-507, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18199742

ABSTRACT

The concentrative nucleoside transporter (CNT) protein family in humans is represented by three members, hCNT1, hCNT2, and hCNT3. Belonging to a CNT subfamily phylogenetically distinct from hCNT1/2, hCNT3 mediates transport of a broad range of purine and pyrimidine nucleosides and nucleoside drugs, whereas hCNT1 and hCNT2 are pyrimidine and purine nucleoside-selective, respectively. All three hCNTs are Na(+)-coupled. Unlike hCNT1/2, however, hCNT3 is also capable of H(+)-mediated nucleoside cotransport. Using site-directed mutagenesis in combination with heterologous expression in Xenopus oocytes, we have identified a C-terminal intramembranous cysteine residue of hCNT3 (Cys-561) that reversibly binds the hydrophilic thiol-reactive reagent p-chloromercuribenzene sulfonate (PCMBS). Access of this membrane-impermeant probe to Cys-561, as determined by inhibition of hCNT3 transport activity, required H(+), but not Na(+), and was blocked by extracellular uridine. Although this cysteine residue is also present in hCNT1 and hCNT2, neither transporter was affected by PCMBS. We conclude that Cys-561 is located in the translocation pore in a mobile region within or closely adjacent to the nucleoside binding pocket and that access of PCMBS to this residue reports a specific H(+)-induced conformational state of the protein.


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Protons , 4-Chloromercuribenzenesulfonate/pharmacology , Amino Acid Sequence , Animals , Cysteine/genetics , Cysteine/metabolism , Electrophysiology , Humans , Membrane Transport Proteins/genetics , Mesylates/pharmacology , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Structure, Tertiary , Sequence Alignment , Uridine/metabolism , Xenopus laevis
10.
J Biol Chem ; 282(42): 30607-17, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17704058

ABSTRACT

Human concentrative nucleoside transporter 1 (hCNT1), the first discovered of three human members of the SLC28 (CNT) protein family, is a Na+/nucleoside cotransporter with 650 amino acids. The potential functional roles of 10 conserved aspartate and glutamate residues in hCNT1 were investigated by site-directed mutagenesis and heterologous expression in Xenopus oocytes. Initially, each of the 10 residues was replaced by the corresponding neutral amino acid (asparagine or glutamine). Five of the resulting mutants showed unchanged Na+-dependent uridine transport activity (D172N, E338Q, E389Q, E413Q, and D565N) and were not investigated further. Three were retained in intracellular membranes (D482N, E498Q, and E532Q) and thus could not be assessed functionally. The remaining two (E308Q and E322Q) were present in normal quantities at cell surfaces but exhibited low intrinsic transport activities. Charge replacement with the alternate acidic amino acid enabled correct processing of D482E and E498D, but not of E532D, to cell surfaces and also yielded partially functional E308D and E322D. Relative to wild-type hCNT1, only D482E exhibited normal transport kinetics, whereas E308D, E308Q, E322D, E322Q, and E498D displayed increased K50(Na+) and/or Km(uridine) values and diminished Vmax(Na+) and Vmax(uridine) values. E322Q additionally exhibited uridine-gated uncoupled Na+ transport. Together, these findings demonstrate roles for Glu-308, Glu-322, and Glu-498 in Na+/nucleoside cotransport and suggest locations within a common cation/nucleoside translocation pore. Glu-322, the residue having the greatest influence on hCNT1 transport function, exhibited uridine-protected inhibition by p-chloromercuriphenyl sulfonate and 2-aminoethyl methanethiosulfonate when converted to cysteine.


Subject(s)
Amino Acids/metabolism , Cell Membrane/metabolism , Ion Channel Gating/physiology , Membrane Transport Proteins/metabolism , Uridine/metabolism , 4-Chloromercuribenzenesulfonate/pharmacology , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/genetics , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Cell Membrane/chemistry , Cell Membrane/genetics , Enzyme Inhibitors/pharmacology , Ethyl Methanesulfonate/analogs & derivatives , Ethyl Methanesulfonate/pharmacology , Humans , Indicators and Reagents/pharmacology , Ion Channel Gating/drug effects , Kinetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Mutagenesis, Site-Directed , Mutation, Missense , Oocytes/cytology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sodium/chemistry , Sodium/metabolism , Uridine/chemistry , Uridine/genetics , Xenopus laevis
11.
Biochemistry ; 46(6): 1684-93, 2007 Feb 13.
Article in English | MEDLINE | ID: mdl-17279631

ABSTRACT

The Na+/nucleoside cotransporters hCNT1 (650 residues) and hCNT2 (658 residues) are 72% identical in amino acid sequence and contain 13 putative transmembrane helices (TMs). Both transport uridine and adenosine but are otherwise selective for pyrimidine (system cit) and purine (system cif) nucleosides, respectively. Previously, we used site-directed mutagenesis and functional expression in Xenopus oocytes to identify two pairs of adjacent residues in TMs 7 and 8 of hCNT1 (Ser319-Gln320 and Ser353-Leu354) that, when converted to the corresponding residues in hCNT2 (Gly-Met and Thr-Val, respectively), changed the permeant selectivity of the transporter from cit to cif. We now report an investigation of the effects of corresponding mutations in TM 8 alone and demonstrate unique S353T- and L354V-induced changes in nucleoside specificity and cation coupling, respectively. hCNT1 mutation S353T produced a profound decrease in cytidine transport efficiency (Vmax/Km ratio) and, in combination with L354V (S353T/L354V), resulted in a novel uridine-preferring transport phenotype. In addition, the L354V mutation markedly increased the apparent affinity of hCNT1 for Na+ and Li+. Both hCNT1 TM 8 residues exhibited uridine-protectable inhibition by p-chloromercuribenzene sulfonate when converted to Cys, suggesting that they occupy positions within or closely adjacent to a common cation/nucleoside translocation pore.


Subject(s)
Membrane Transport Proteins/genetics , Membrane Transport Proteins/physiology , 4-Chloromercuribenzenesulfonate/pharmacology , Amino Acid Sequence , Animals , Cations, Monovalent/metabolism , Cytidine/metabolism , Humans , Kinetics , Leucine/physiology , Lithium/metabolism , Membrane Transport Proteins/chemistry , Protein Structure, Secondary , Serine/physiology , Sodium/metabolism , Substrate Specificity , Thymidine/metabolism , Uridine/metabolism , Xenopus laevis
12.
Mol Pharmacol ; 69(1): 346-53, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16234483

ABSTRACT

2-Chloro-9-(2'-deoxy-2'-fluoro-beta-d-arabinofuranosyl)adenine (Cl-F-ara-A, clofarabine), a purine nucleoside analog with structural similarity to 2-chloro-2'-deoxyadenosine (Cl-dAdo, cladribine) and 9-beta-d-arabinofuranosyl-2-fluoroadenine (F-ara-A, fludarabine), has activity in adult and pediatric leukemias. Mediated transport of the purine nucleoside analogs is believed to occur through the action of two structurally unrelated protein families, the equilibrative nucleoside transporters (ENTs) and the concentrative nucleoside transporters (CNTs). The current work assessed the transportability of Cl-F-ara-A, Cl-dAdo, and F-ara-A in cultured human leukemic CEM cells that were either nucleoside transport-defective or possessed individual human nucleoside transporter types and in Xenopus laevis oocytes and Saccharomyces cerevisiae yeast that produced individual recombinant human nucleoside transporter types. Cells producing hENT1 or hCNT3 exhibited the highest uptake of Cl-F-ara-A, whereas nucleoside transport-deficient cells and cells producing hCNT1 lacked uptake altogether. When Cl-F-ara-A transport rates by hENT1 were compared with those of Cl-dAdo and F-ara-A, Cl-dAdo had the highest efficiency of transport, although Cl-F-ara-A showed the greatest accumulation during 5-min exposures. In cytotoxicity studies with the CEM lines, Cl-F-ara-A was more cytotoxic to cells producing hENT1 than to the nucleoside transport-deficient cells. The efficiency of Cl-F-ara-A transport by oocytes with recombinant transporters was hCNT3 > hENT2 > hENT1 > hCNT2; no transport was observed with hCNT1. Affinity studies with recombinant transporters produced in yeast showed that hENT1, hENT2, and hCNT3 all had higher affinities for Cl-F-ara-A than for either Cl-dAdo or F-ara-A. These results suggest that the nature and activity of the plasma membrane proteins capable of inward transport of nucleosides are important determinants of Cl-F-ara-A activity in human cells.


Subject(s)
Antineoplastic Agents/metabolism , Arabinonucleosides/metabolism , Cladribine/metabolism , Membrane Transport Proteins/metabolism , Vidarabine/analogs & derivatives , Adenine Nucleotides , Animals , Antineoplastic Agents/pharmacology , Arabinonucleosides/pharmacology , Biological Transport , Cell Line , Cladribine/pharmacology , Clofarabine , Humans , Recombinant Proteins/metabolism , Vidarabine/metabolism , Vidarabine/pharmacology , Xenopus laevis
13.
J Biol Chem ; 280(27): 25436-49, 2005 Jul 08.
Article in English | MEDLINE | ID: mdl-15870078

ABSTRACT

The concentrative nucleoside transporter (CNT) protein family in humans is represented by three members, hCNT1, hCNT2, and hCNT3. hCNT3, a Na+/nucleoside symporter, transports a broad range of physiological purine and pyrimidine nucleosides as well as anticancer and antiviral nucleoside drugs, and belongs to a different CNT subfamily than hCNT1/2. H+-dependent Escherichia coli NupC and Candida albicans CaCNT are also CNT family members. The present study utilized heterologous expression in Xenopus oocytes to investigate the specificity, mechanism, energetics, and structural basis of hCNT3 cation coupling. hCNT3 exhibited uniquely broad cation interactions with Na+, H+, and Li+ not shared by Na+-coupled hCNT1/2 or H+-coupled NupC/CaCNT. Na+ and H+ activated hCNT3 through mechanisms to increase nucleoside apparent binding affinity. Direct and indirect methods demonstrated cation/nucleoside coupling stoichiometries of 2:1 in the presence of Na+ and both Na+ plus H+, but only 1:1 in the presence of H+ alone, suggesting that hCNT3 possesses two Na+-binding sites, only one of which is shared by H+. The H+-coupled hCNT3 did not transport guanosine or 3'-azido-3'-deoxythymidine and 2',3'-dideoxycytidine, demonstrating that Na+- and H+-bound versions of hCNT3 have significantly different conformations of the nucleoside binding pocket and/or translocation channel. Chimeric studies between hCNT1 and hCNT3 located hCNT3-specific cation interactions to the C-terminal half of hCNT3, setting the stage for site-directed mutagenesis experiments to identify the residues involved.


Subject(s)
Hydrogen/metabolism , Membrane Transport Proteins/metabolism , Nucleosides/metabolism , Sodium/metabolism , Animals , Binding Sites , Humans , Hydrogen-Ion Concentration , Kinetics , Lithium/metabolism , Membrane Potentials/physiology , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Oocytes/physiology , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Uridine/metabolism , Xenopus
14.
J Biol Chem ; 280(16): 15880-7, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15701636

ABSTRACT

The first mammalian examples of the equilibrative nucleoside transporter family to be characterized, hENT1 and hENT2, were passive transporters located predominantly in the plasma membranes of human cells. We now report the functional characterization of members of a third subgroup of the family, from human and mouse, which differ profoundly in their properties from previously characterized mammalian nucleoside transporters. The 475-residue human and mouse proteins, designated hENT3 and mENT3, respectively, are 73% identical in amino acid sequence and possess long N-terminal hydrophilic domains that bear typical (DE)XXXL(LI) endosomal/lysosomal targeting motifs. ENT3 transcripts and proteins are widely distributed in human and rodent tissues, with a particular abundance in placenta. However, in contrast to ENT1 and ENT2, the endogenous and green fluorescent protein-tagged forms of the full-length hENT3 protein were found to be predominantly intracellular proteins that co-localized, in part, with lysosomal markers in cultured human cells. Truncation of the hydrophilic N-terminal region or mutation of its dileucine motif to alanine caused the protein to be relocated to the cell surface both in human cells and in Xenopus oocytes, allowing characterization of its transport activity in the latter. The protein proved to be a broad selectivity, low affinity nucleoside transporter that could also transport adenine. Transport activity was relatively insensitive to the classical nucleoside transport inhibitors nitrobenzylthioinosine, dipyridamole, and dilazep and was sodium ion-independent. However, it was strongly dependent upon pH, and the optimum pH value of 5.5 probably reflected the location of the transporter in acidic, intracellular compartments.


Subject(s)
Intracellular Membranes/metabolism , Nucleoside Transport Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , HeLa Cells , Humans , Mice , Molecular Sequence Data , Multigene Family , Nucleoside Transport Proteins/genetics , Oocytes/metabolism , Xenopus
15.
Mol Pharmacol ; 67(1): 273-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15486050

ABSTRACT

Benzamide riboside (BR) and tiazofurin (TR) are converted to analogs of NAD that inhibit IMP dehydrogenase (IMPDH), resulting in cellular depletion of GTP and dGTP and inhibition of proliferation. The current work was undertaken to identify the human nucleoside transporters involved in cellular uptake of BR and TR and to evaluate their role in cytotoxicity. Transportability was examined in Xenopus laevis oocytes and Saccharomyces cerevisiae that produced individual recombinant human concentrative nucleoside transporter (CNT) and equilibrative nucleoside transporter (ENT) types (hENT1, hENT2, hCNT1, hCNT2, or hCNT3). TR was a better permeant than BR with a rank order of transportability in oocytes of hCNT3 >> hENT1 > hENT2 > hCNT2 >> hCNT1. The concentration dependence of inhibition of [(3)H]uridine transport in S. cerevisiae by TR exhibited lower K(i) values than BR: hCNT3 (5.4 versus 226 microM), hENT2 (16 versus 271 microM), hENT1 (57 versus 168 microM), and hCNT1 (221 versus 220 microM). In cytotoxicity experiments, BR was more cytotoxic than TR to cells that were either nucleoside transport-defective or -competent, and transport-competent cells were more sensitive to both drugs. Exposure to nitrobenzylmercaptopurine ribonucleoside conferred resistance to BR and TR cytotoxicity to hENT1-containing CEM cells, thereby demonstrating the importance of transport capacity for manifestation of cytoxicity. A breast cancer cell line with mutant p53 exhibited 9-fold higher sensitivity to BR than the otherwise similar cell line with wild-type p53, suggesting that cells with mutant p53 may be potential targets for IMPDH inhibitors. Further studies are warranted to determine whether this finding can be generalized to other cell types.


Subject(s)
IMP Dehydrogenase/antagonists & inhibitors , Membrane Transport Proteins/metabolism , Nucleosides/pharmacology , Nucleosides/pharmacokinetics , Ribavirin/analogs & derivatives , Ribavirin/pharmacology , Ribavirin/pharmacokinetics , Animals , Biological Transport , Cell Survival/drug effects , Female , Humans , Membrane Transport Proteins/genetics , Oocytes , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Uridine/metabolism , Xenopus laevis
16.
Yeast ; 21(15): 1269-77, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15543539

ABSTRACT

Contigs 19-10196 and 19-20196 of the Stanford Candida albicans genome sequence databank encode two putative allelic isoforms of C. albicans CaCNT, a recently characterized 608 amino acid residue H+-coupled fungal member of the CNT family of concentrative nucleoside transport proteins. The single Ser/Gly difference between CaCNT/19-20196 and CaCNT occurs at position 328 in putative TM 7, and corresponds to a Ser/Gly substitution previously shown to contribute to the contrasting pyrimidine and purine nucleoside selectivities of human (h) and rat (r) Na+-dependent CNT1 and CNT2. CaCNT/19-10196 differs from CaCNT by four amino acids, but has Gly at position 328. These new proteins were recreated by site-directed mutagenesis of CaCNT and characterized functionally by heterologous expression in Xenopus laevis oocytes. In marked contrast to h/rCNT1/2, both CaCNT/19-10196 and CaCNT/19-20196 exhibited permeant selectivities for purine nucleosides (adenosine, guanosine and inosine) and uridine similar to that of CaCNT. However, although H+-coupled, CaCNT/19-20196 exhibited a approximately 10-fold higher apparent Km for uridine than either CaCNT or CaCNT/19-10196. CaCNT/19-20196 also exhibited a low apparent affinity for inosine. We conclude that the three proteins correspond to high-affinity (CaCNT, CaCNT/19-10196) and low-affinity (CaCNT/19-20196) allelic isoforms of the C. albicans CNT nucleoside transporter. This is the first example of a single amino acid residue substitution altering a CNT protein's overall apparent affinity for nucleosides.


Subject(s)
Candida albicans/metabolism , Fungal Proteins/metabolism , Nucleoside Transport Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biological Transport , Candida albicans/genetics , Fungal Proteins/genetics , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleoside Transport Proteins/genetics , Nucleosides/physiology , Oocytes/physiology , Patch-Clamp Techniques , Protein Isoforms , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Xenopus/genetics , Xenopus/physiology
17.
Mol Membr Biol ; 21(5): 323-36, 2004.
Article in English | MEDLINE | ID: mdl-15513740

ABSTRACT

NupG from Escherichia coli is the archetype of a family of nucleoside transporters found in several eubacterial groups and has distant homologues in eukaryotes, including man. To facilitate investigation of its molecular mechanism, we developed methods for expressing an oligohistidine-tagged form of NupG both at high levels (>20% of the inner membrane protein) in E. coli and in Xenopus laevis oocytes. In E. coli recombinant NupG transported purine (adenosine) and pyrimidine (uridine) nucleosides with apparent K(m) values of approximately 20-30 microM and transport was energized primarily by the membrane potential component of the proton motive force. Competition experiments in E. coli and measurements of uptake in oocytes confirmed that NupG was a broad-specificity transporter of purine and pyrimidine nucleosides. Importantly, using high-level expression in E. coli and magic-angle spinning cross-polarization solid-state nuclear magnetic resonance, we have for the first time been able directly to measure the binding of the permeant ([1'-(13)C]uridine) to the protein and to assess its relative mobility within the binding site, under non-energized conditions. Purification of over-expressed NupG to near homogeneity by metal chelate affinity chromatography, with retention of transport function in reconstitution assays, was also achieved. Fourier transform infrared and circular dichroism spectroscopy provided further evidence that the purified protein retained its 3D conformation and was predominantly alpha-helical in nature, consistent with a proposed structure containing 12 transmembrane helices. These findings open the way to elucidating the molecular mechanism of transport in this key family of membrane transporters.


Subject(s)
Escherichia coli Proteins/physiology , Escherichia coli/metabolism , Membrane Transport Proteins/physiology , Nucleoside Transport Proteins/physiology , Adenosine/analysis , Adenosine/metabolism , Amino Acid Sequence , Animals , Biological Transport/physiology , Circular Dichroism , Escherichia coli/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Membrane Transport Proteins/genetics , Membrane Transport Proteins/isolation & purification , Molecular Sequence Data , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/isolation & purification , Oocytes/chemistry , Phylogeny , Protein Structure, Secondary , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sequence Alignment , Spectroscopy, Fourier Transform Infrared , Substrate Specificity , Uridine/analysis , Uridine/metabolism , Xenopus
18.
Mol Membr Biol ; 21(4): 247-59, 2004.
Article in English | MEDLINE | ID: mdl-15371014

ABSTRACT

The genome of Caenorhabditis elegans encodes multiple homologues of the two major families of mammalian equilibrative and concentrative nucleoside transporters. As part of a programme aimed at understanding the biological rationale underlying the multiplicity of eukaryote nucleoside transporters, we have now demonstrated that the nematode genes ZK809.4 (ent-1) and K09A9.3 (ent-2) encode equilibrative transporters, which we designate CeENT1 and CeENT2 respectively. These transporters resemble their human counterparts hENT1 and hENT2 in exhibiting similar broad permeant specificities for nucleosides, while differing in their permeant selectivities for nucleobases. They are insensitive to the classic inhibitors of mammalian nucleoside transport, nitrobenzylthioinosine, dilazep and draflazine, but are inhibited by the vasoactive drug dipyridamole. Use of green fluorescent protein reporter constructs indicated that the transporters are present in a limited number of locations in the adult, including intestine and pharynx. Their potential roles in these tissues were explored by using RNA interference to disrupt gene expression. Although disruption of ent-1 or ent-2 expression alone had no effect, simultaneous disruption of both genes yielded pronounced developmental defects involving the intestine and vulva.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Nucleoside Transport Proteins/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cloning, Molecular , Equilibrative Nucleoside Transporter 1 , Equilibrative-Nucleoside Transporter 2 , Kinetics , Molecular Sequence Data , Nucleoside Transport Proteins/antagonists & inhibitors , Nucleoside Transport Proteins/chemistry , Nucleoside Transport Proteins/genetics , Oocytes/drug effects , Oocytes/metabolism , Phylogeny , RNA Interference , Sequence Alignment , Uridine/pharmacology , Xenopus laevis/genetics , Xenopus laevis/metabolism
19.
J Physiol ; 558(Pt 3): 807-23, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15194733

ABSTRACT

Human concentrative nucleoside transporter 1 (hCNT1) mediates active transport of nucleosides and anticancer and antiviral nucleoside drugs across cell membranes by coupling influx to the movement of Na(+) down its electrochemical gradient. The two-microelectrode voltage-clamp technique was used to measure steady-state and presteady-state currents of recombinant hCNT1 produced in Xenopus oocytes. Transport was electrogenic, phloridzin sensitive and specific for pyrimidine nucleosides and adenosine. Nucleoside analogues that induced inwardly directed Na(+) currents included the anticancer drugs 5-fluorouridine, 5-fluoro-2'-deoxyuridine, cladribine and cytarabine, the antiviral drugs zidovudine and zalcitabine, and the novel thymidine mimics 1-(2-deoxy-beta-d-ribofuranosyl)-2,4-difluoro-5-methylbenzene and 1-(2-deoxy-beta-d-ribofuranosyl)-2,4-difluoro-5-iodobenzene. Apparent K(m) values for 5-fluorouridine, 5-fluoro-2'-deoxyuridine and zidovudine were 18, 15 and 450 microm, respectively. hCNT1 was Na(+) specific, and the kinetics of steady-state uridine-evoked Na(+) currents were consistent with an ordered simultaneous transport model in which Na(+) binds first followed by uridine. Membrane potential influenced both ion binding and carrier translocation. The Na(+)-nucleoside coupling stoichiometry, determined directly by comparing the uridine-induced inward charge movement to [(14)C]uridine uptake was 1: 1. hCNT1 presteady-state currents were used to determine the fraction of the membrane field sensed by Na(+) (61%), the valency of the movable charge (-0.81) and the average number of transporters present in the oocyte plasma membrane (6.8 x 10(10) per cell). The hCNT1 turnover rate at -50 mV was 9.6 molecules of uridine transported per second.


Subject(s)
Membrane Transport Proteins/physiology , Oocytes/metabolism , Recombinant Proteins/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Biological Transport/physiology , Dose-Response Relationship, Drug , Electrophysiology , Female , Humans , Membrane Transport Proteins/metabolism , Nucleosides/chemistry , Nucleosides/metabolism , Nucleosides/pharmacology , Protein Binding/drug effects , Protein Binding/physiology , Xenopus laevis
20.
Yeast ; 20(8): 661-75, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12794928

ABSTRACT

Human and other mammalian concentrative (Na(+)-linked) nucleoside transport proteins belong to a membrane protein family (CNT, TC 2.A.41) that also includes Escherichia coli H(+)-dependent nucleoside transport protein NupC. Here, we report the cDNA cloning and functional characterization of a CNT family member from the pathogenic yeast Candida albicans. This 608 amino acid residue H(+)/nucleoside symporter, designated CaCNT, contains 13 predicted transmembrane domains (TMs), but lacks the exofacial, glycosylated carboxyl-terminus of its mammalian counterparts. When produced in Xenopus oocytes, CaCNT exhibited transport activity for adenosine, uridine, inosine and guanosine but not cytidine, thymidine or the nucleobase hypoxanthine. Apparent K(m) values were in the range 16-64 micro M, with V(max) : K(m) ratios of 0.58-1.31. CaCNT also accepted purine and uridine analogue nucleoside drugs as permeants, including cordycepin (3'-deoxyadenosine), a nucleoside analogue with anti-fungal activity. Electrophysiological measurements under voltage clamp conditions gave a H(+) to [(14)C]uridine coupling ratio of 1 : 1. CaCNT, obtained from logarithmically growing cells, is the first described cation-coupled nucleoside transporter in yeast, and the first member of the CNT family of proteins to be characterized from a unicellular eukaryotic organism.


Subject(s)
Candida albicans/genetics , Fungal Proteins/genetics , Membrane Transport Proteins/genetics , Nucleoside Transport Proteins/genetics , Amino Acid Sequence , Animals , Antifungal Agents/metabolism , Base Sequence , Candida albicans/metabolism , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Deoxyadenosines/metabolism , Fungal Proteins/metabolism , Humans , Kinetics , Membrane Potentials/physiology , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Nucleoside Transport Proteins/metabolism , Nucleosides/metabolism , Patch-Clamp Techniques , Polymerase Chain Reaction , Sequence Homology, Amino Acid , Xenopus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...