Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Biochem Parasitol ; 258: 111616, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38401850

ABSTRACT

Trypanosoma cruzi is a protozoan parasite and the etiological agent of Chagas disease, a debilitating and sometimes fatal disease that continues to spread to new areas. Yet, Chagas disease is still only treated with two related nitro compounds that are insufficiently effective and cause severe side effects. Nucleotide metabolism is one of the known vulnerabilities of T. cruzi, as they are auxotrophic for purines, and nucleoside analogues have been shown to have genuine promise against this parasite in vitro and in vivo. Since purine antimetabolites require efficient uptake through transporters, we here report a detailed characterisation of the T. cruzi NB1 nucleobase transporter with the aim of elucidating the interactions between TcrNB1 and its substrates and finding the positions that can be altered in the design of novel antimetabolites without losing transportability. Systematically determining the inhibition constants (Ki) of purine analogues for TcrNB1 yielded their Gibbs free energy of interaction, ΔG0. Pairwise comparisons of substrate (hypoxanthine, guanine, adenine) and analogues allowed us to determine that optimal binding affinity by TcrNB1 requires interactions with all four nitrogen residues of the purine ring, with N1 and N9, in protonation state, functioning as presumed hydrogen bond donors and unprotonated N3 and N7 as hydrogen bond acceptors. This is the same interaction pattern as we previously described for the main nucleobase transporters of Trypanosoma brucei spp. and Leishmania major and makes it the first of the ENT-family genes that is functionally as well as genetically conserved between the three main kinetoplast pathogens.


Subject(s)
Guanine , Hypoxanthine , Trypanosoma cruzi , Trypanosoma cruzi/metabolism , Trypanosoma cruzi/genetics , Trypanosoma cruzi/chemistry , Guanine/metabolism , Hypoxanthine/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/chemistry , Nucleobase Transport Proteins/metabolism , Nucleobase Transport Proteins/genetics , Nucleobase Transport Proteins/chemistry , Biological Transport , Substrate Specificity , Protein Binding , Nucleosides/metabolism
2.
J Mol Biol ; 434(2): 167393, 2022 01 30.
Article in English | MEDLINE | ID: mdl-34896363

ABSTRACT

SLC23 family members are transporters of either nucleobases or ascorbate. While the mammalian SLC23 ascorbate transporters are sodium-coupled, the non-mammalian nucleobase transporters have been proposed, but not formally shown, to be proton-coupled symporters. This assignment is exclusively based on in vivo transport assays using protonophores. Here, by establishing the first in vitro transport assay for this protein family, we demonstrate that a representative member of the SLC23 nucleobase transporters operates as a uniporter instead. We explain these conflicting assignments by identifying a critical role of uracil phosphoribosyltransferase, the enzyme converting uracil to UMP, in driving uracil uptake in vivo. Detailed characterization of uracil phosphoribosyltransferase reveals that the sharp reduction of uracil uptake in whole cells in presence of protonophores is caused by acidification-induced enzyme inactivation. The SLC23 family therefore consists of both uniporters and symporters in line with the structurally related SLC4 and SLC26 families that have previously been demonstrated to accommodate both transport modes as well.


Subject(s)
Biological Transport/physiology , Ion Transport , Membrane Transport Proteins/chemistry , Protons , Animals , Ascorbic Acid/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Catalytic Domain , Escherichia coli , Humans , Membrane Transport Proteins/metabolism , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/metabolism , Pentosyltransferases/chemistry , Pentosyltransferases/metabolism , Sodium/metabolism , Symporters , Uracil/metabolism
3.
J Mol Biol ; 433(24): 167329, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34710398

ABSTRACT

The relation of sequence with specificity in membrane transporters is challenging to explore. Most relevant studies until now rely on comparisons of present-day homologs. In this work, we study a set of closely related transporters by employing an evolutionary, ancestral-reconstruction approach and reveal unexpected new specificity determinants. We analyze a monophyletic group represented by the xanthine-specific XanQ of Escherichia coli in the Nucleobase-Ascorbate Transporter/Nucleobase-Cation Symporter-2 (NAT/NCS2) family. We reconstructed AncXanQ, the putative common ancestor of this clade, expressed it in E. coli K-12, and found that, in contrast to XanQ, it encodes a high-affinity permease for both xanthine and guanine, which also recognizes adenine, hypoxanthine, and a range of analogs. AncXanQ conserves all binding-site residues of XanQ and differs substantially in only five intramembrane residues outside the binding site. We subjected both homologs to rationally designed mutagenesis and present evidence that these five residues are linked with the specificity change. In particular, we reveal Ser377 of XanQ (Gly in AncXanQ) as a major determinant. Replacement of this Ser with Gly enlarges the specificity of XanQ towards an AncXanQ-phenotype. The ortholog from Neisseria meningitidis retaining Gly at this position is also a xanthine/guanine transporter with extended substrate profile like AncXanQ. Molecular Dynamics shows that the S377G replacement tilts transmembrane helix 12 resulting in rearrangement of Phe376 relative to Phe94 in the XanQ binding pocket. This effect may rationalize the enlarged specificity. On the other hand, the specificity effect of S377G can be masked by G27S or other mutations through epistatic interactions.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli/enzymology , Guanine/metabolism , Neisseria meningitidis/enzymology , Nucleobase Transport Proteins/chemistry , Xanthine/metabolism , Bacterial Proteins/classification , Bacterial Proteins/genetics , Molecular Dynamics Simulation , Mutagenesis , Nucleobase Transport Proteins/classification , Nucleobase Transport Proteins/genetics , Phylogeny , Protein Structure, Secondary , Substrate Specificity/genetics
4.
J Mol Biol ; 433(16): 166814, 2021 08 06.
Article in English | MEDLINE | ID: mdl-33497644

ABSTRACT

Members of the ubiquitous Nucleobase Ascorbate Transporter (NAT) family are H+ or Na+ symporters specific for the cellular uptake of either purines and pyrimidines or L-ascorbic acid. Despite the fact that several bacterial and fungal members have been extensively characterised at a genetic, biochemical or cellular level, and crystal structures of NAT members from Escherichia coli and Aspergillus nidulans have been determined pointing to a mechanism of transport, we have little insight on how substrate selectivity is determined. Here, we present systematic mutational analyses, rational combination of mutations, and novel genetic screens that reveal cryptic context-dependent roles of partially conserved residues in the so-called NAT signature motif in determining the specificity of the UapA transporter of A. nidulans. We show that specific NAT signature motif substitutions, alone and in combinations with each other or with distant mutations in residues known to affect substrate selectivity, lead to novel UapA versions possessing variable transport capacities and specificities for nucleobases. In particular, we show that a UapA version including the quadruple mutation T405S/F406Y/A407S/Q408E in the NAT signature motif (UapA-SYSE) becomes incapable of purine transport, but gains a novel pyrimidine-related profile, which can be further altered to a more promiscuous purine/pyrimidine profile when combined with replacements at distantly located residues, especially at F528. Our results reveal that UapA specificity is genetically highly modifiable and allow us to speculate on how the elevator-type mechanism of transport might account for this flexibility.


Subject(s)
Nucleobase Transport Proteins/metabolism , Purines/metabolism , Amino Acid Motifs , Binding Sites , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mutation , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Protein Binding , Purines/chemistry , Structure-Activity Relationship , Substrate Specificity
5.
Molecules ; 24(12)2019 Jun 12.
Article in English | MEDLINE | ID: mdl-31212849

ABSTRACT

In earlier studies, we generated concentration-response (E/c) curves with CPA (N6-cyclopentyladenosine; a selective A1 adenosine receptor agonist) or adenosine, in the presence or absence of S-(2-hydroxy-5-nitrobenzyl)-6-thioinosine (NBTI, a selective nucleoside transport inhibitor), and with or without a pretreatment with 8-cyclopentyl-N3-[3-(4-(fluorosulfonyl)-benzoyloxy)propyl]-N1-propylxanthine (FSCPX, a chemical known as a selective, irreversible A1 adenosine receptor antagonist), in isolated, paced guinea pig left atria. Meanwhile, we observed a paradoxical phenomenon, i.e. the co-treatment with FSCPX and NBTI appeared to enhance the direct negative inotropic response to adenosine. In the present in silico study, we aimed to reproduce eight of these E/c curves. Four models (and two additional variants of the last model) were constructed, each one representing a set of assumptions, in order to find the model exhibiting the best fit to the ex vivo data, and to gain insight into the paradoxical phenomenon in question. We have obtained in silico evidence for an interference between effects of FSCPX and NBTI upon our ex vivo experimental setting. Regarding the mechanism of this interference, in silico evidence has been gained for the assumption that FSCPX inhibits the effect of NBTI on the level of endogenous (but not exogenous) adenosine. As an explanation, it may be hypothesized that FSCPX inhibits an enzyme participating in the interstitial adenosine formation. In addition, our results suggest that NBTI does not stop the inward adenosine flux in the guinea pig atrium completely.


Subject(s)
Adenosine A1 Receptor Antagonists/chemistry , Nucleobase Transport Proteins/chemistry , Receptor, Adenosine A1/chemistry , Xanthines/chemistry , Adenosine/chemistry , Adenosine/pharmacology , Adenosine A1 Receptor Antagonists/pharmacology , Animals , Dose-Response Relationship, Drug , Guinea Pigs , Nucleobase Transport Proteins/antagonists & inhibitors , Xanthines/pharmacology
6.
Mol Microbiol ; 103(2): 319-332, 2017 01.
Article in English | MEDLINE | ID: mdl-27741561

ABSTRACT

NCS1 proteins are H+ or Na+ symporters responsible for the uptake of purines, pyrimidines or related metabolites in bacteria, fungi and some plants. Fungal NCS1 are classified into two evolutionary and structurally distinct subfamilies, known as Fur- and Fcy-like transporters. These subfamilies have expanded and functionally diversified by gene duplications. The Fur subfamily of the model fungus Aspergillus nidulans includes both major and cryptic transporters specific for uracil, 5-fluorouracil, allantoin or/and uric acid. Here we functionally analyse all four A. nidulans Fcy transporters (FcyA, FcyC, FcyD and FcyE) with previously unknown function. Our analysis shows that FcyD is moderate-affinity, low-capacity, highly specific adenine transporter, whereas FcyE contributes to 8-azaguanine uptake. Mutational analysis of FcyD, supported by homology modelling and substrate docking, shows that two variably conserved residues (Leu356 and Ser359) in transmembrane segment 8 (TMS8) are critical for transport kinetics and specificity differences among Fcy transporters, while two conserved residues (Phe167 and Ser171) in TMS3 are also important for function. Importantly, mutation S359N converts FcyD to a promiscuous nucleobase transporter capable of recognizing adenine, xanthine and several nucleobase analogues. Our results reveal the importance of specific residues in the functional evolution of NCS1 transporters.


Subject(s)
Aspergillus nidulans/genetics , Nucleobase Transport Proteins/genetics , Purines/metabolism , Amino Acid Sequence , Aspergillus nidulans/metabolism , Biological Evolution , Biological Transport , Conserved Sequence , Fungal Proteins/metabolism , Gene Duplication , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/metabolism , Phylogeny , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity
7.
Mol Microbiol ; 98(3): 502-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26192456

ABSTRACT

The xanthine permease XanQ of Escherichia coli is a paradigm for transporters of the evolutionarily broad family nucleobase-cation symporter-2 (NCS2) that transport key metabolites or anti-metabolite analogs. Most functionally known members are xanthine/uric acid transporters related to XanQ and belong to a distinct phylogenetic cluster of the family. Here, we present a comprehensive mutagenesis of XanQ based on the identification and Cys-scanning analysis of conserved sequence motifs in this cluster. Results are interpreted in relation to homology modeling on the structurally known template of UraA and previous data on critical binding-site residues in transmembrane segments (TMs) 3, 8 and 10. The current analysis, of motifs distant to the binding site, revealed a set of functionally important residues in TMs 2, 5, 12 and 13, including seven irreplaceable ones, of which six are Gly residues in the gate domain (159, 369, 370, 383, 409) and in TM2 (Gly-71), and one is polar (Gln-75). Gln-75 (TM2) is probably crucial in a network of hydrogen-bonding interactions in the middle of the core domain involving another essential residue, Asp-304 (TM9). Although the two residues are irreplaceable individually, combinatorial replacement of Gln-75 with Asn and of Asp-304 with Glu rescues significant transport activity.


Subject(s)
Escherichia coli/enzymology , Nucleobase Transport Proteins/genetics , Nucleobase Transport Proteins/metabolism , Xanthine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Biological Transport, Active , Conserved Sequence , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed/methods , Mutation , Nucleobase Transport Proteins/chemistry , Phylogeny , Protein Structure, Secondary , Structure-Activity Relationship , Uric Acid/metabolism , Xanthine/chemistry
8.
Fungal Genet Biol ; 75: 56-63, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25639910

ABSTRACT

The Nucleobase-Ascorbate Transporter (NAT) family includes members in nearly all domains of life. Functionally characterized NAT transporters from bacteria, fungi, plants and mammals are ion-coupled symporters specific for the uptake of purines, pyrimidines and related analogues. The characterized mammalian NATs are specific for the uptake of L-ascorbic acid. In this work we identify in silico a group of fungal putative transporters, named UapD-like proteins, which represent a novel NAT subfamily. To understand the function and specificity of UapD proteins, we cloned and functionally characterized the two Aspergillus brasiliensis NAT members (named AbUapC and AbUapD) by heterologous expression in Aspergillus nidulans. AbUapC represents canonical NATs (UapC or UapA), while AbUapD represents the new subfamily. AbUapC is a high-affinity, high-capacity, H(+)/xanthine-uric acid transporter, which can also recognize other purines with very low affinity. No apparent transport function could be detected for AbUapD. GFP-tagging showed that, unlike AbUapC which is localized in the plasma membrane, AbUapD is ER-retained and degraded in the vacuoles, a characteristic of misfolded proteins. Chimeric UapA/AbUapD molecules are also turned-over in the vacuole, suggesting that UapD includes intrinsic peptidic sequences leading to misfolding. The possible evolutionary implication of such conserved, but inactive proteins is discussed.


Subject(s)
Aspergillus/genetics , Nucleobase Transport Proteins/metabolism , Uric Acid/metabolism , Xanthines/metabolism , Amino Acid Sequence , Aspergillus nidulans/genetics , Aspergillus nidulans/metabolism , Biological Transport , Computer Simulation , Fungal Proteins/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Molecular Sequence Data , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Peptides/chemistry , Phylogeny , Protein Folding , Recombinant Fusion Proteins/chemical synthesis , Recombinant Fusion Proteins/metabolism , Sequence Alignment
9.
Mol Microbiol ; 93(1): 129-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24818808

ABSTRACT

The AzgA purine/H(+) symporter of Aspergillus nidulans is the founding member of a functionally and phylogenetically distinct transporter family present in fungi, bacteria and plants. Here a valid AzgA topological model is built based on the crystal structure of the Escherichia coli uracil transporter UraA, a member of the nucleobase-ascorbate transporter (NAT/NCS2) family. The model consists of 14 transmembrane, mostly α-helical, segments (TMSs) and cytoplasmic N- and C-tails. A distinct compact core of 8 TMSs, made of two intertwined inverted repeats (TMSs 1-4 and 8-11), is topologically distinct from a flexible domain (TMSs 5-7 and 12-14). A putative substrate binding cavity is visible between the core and the gate domains. Substrate docking, molecular dynamics and mutational analysis identified several residues critical for purine binding and/or transport in TMS3, TMS8 and TMS10. Among these, Asn131 (TMS3), Asp339 (TMS8) and Glu394 (TMS10) are proposed to directly interact with substrates, while Asp342 (TMS8) might be involved in subsequent substrate translocation, through H(+) binding and symport. Thus, AzgA and other NAT transporters use topologically similar TMSs and amino acid residues for substrate binding and transport, which in turn implies that AzgA-like proteins constitute a distant subgroup of the ubiquitous NAT family.


Subject(s)
Amino Acids/metabolism , Aspergillus nidulans/physiology , Fungal Proteins/chemistry , Nucleobase Transport Proteins/chemistry , Purines/metabolism , Aspergillus nidulans/chemistry , Binding Sites , DNA Mutational Analysis , Fungal Proteins/genetics , Hydrogen/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Nucleobase Transport Proteins/genetics , Phylogeny , Protein Conformation , Protein Structure, Tertiary , Substrate Specificity
10.
PLoS One ; 9(3): e91343, 2014.
Article in English | MEDLINE | ID: mdl-24621654

ABSTRACT

Plastidic uracil salvage is essential for plant growth and development. So far, PLUTO, the plastidic nucleobase transporter from Arabidopsis thaliana is the only known uracil importer at the inner plastidic membrane which represents the permeability barrier of this organelle. We present the first homology model of PLUTO, the sole plant NCS1 member from Arabidopsis based on the crystal structure of the benzyl hydantoin transporter MHP1 from Microbacterium liquefaciens and validated by molecular dynamics simulations. Polar side chains of residues Glu-227 and backbones of Val-145, Gly-147 and Thr-425 are proposed to form the binding site for the three PLUTO substrates uracil, adenine and guanine. Mutational analysis and competition studies identified Glu-227 as an important residue for uracil and to a lesser extent for guanine transport. A differential response in substrate transport was apparent with PLUTO double mutants E227Q G147Q and E227Q T425A, both of which most strongly affected adenine transport, and in V145A G147Q, which markedly affected guanine transport. These differences could be explained by docking studies, showing that uracil and guanine exhibit a similar binding mode whereas adenine binds deep into the catalytic pocket of PLUTO. Furthermore, competition studies confirmed these results. The present study defines the molecular determinants for PLUTO substrate binding and demonstrates key differences in structure-function relations between PLUTO and other NCS1 family members.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/metabolism , Sequence Homology, Amino Acid , Amino Acid Sequence , Arabidopsis Proteins/genetics , Binding, Competitive , Molecular Sequence Data , Mutation , Nucleobase Transport Proteins/genetics , Structure-Activity Relationship , Substrate Specificity
11.
Plant Cell ; 24(4): 1549-59, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22474184

ABSTRACT

Nucleotide de novo synthesis is highly conserved among organisms and represents an essential biochemical pathway. In plants, the two initial enzymatic reactions of de novo pyrimidine synthesis occur in the plastids. By use of green fluorescent protein fusions, clear support is provided for a localization of the remaining reactions in the cytosol and mitochondria. This implies that carbamoyl aspartate, an intermediate of this pathway, must be exported and precursors of pyrimidine salvage (i.e., nucleobases or nucleosides) are imported into plastids. A corresponding uracil transport activity could be measured in intact plastids isolated from cauliflower (Brassica oleracea) buds. PLUTO (for plastidic nucleobase transporter) was identified as a member of the Nucleobase:Cation-Symporter1 protein family from Arabidopsis thaliana, capable of transporting purine and pyrimidine nucleobases. A PLUTO green fluorescent protein fusion was shown to reside in the plastid envelope after expression in Arabidopsis protoplasts. Heterologous expression of PLUTO in an Escherichia coli mutant lacking the bacterial uracil permease uraA allowed a detailed biochemical characterization. PLUTO transports uracil, adenine, and guanine with apparent affinities of 16.4, 0.4, and 6.3 µM, respectively. Transport was markedly inhibited by low concentrations of a proton uncoupler, indicating that PLUTO functions as a proton-substrate symporter. Thus, a protein for the absolutely required import of pyrimidine nucleobases into plastids was identified.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Biosynthetic Pathways , Nucleobase Transport Proteins/metabolism , Nucleotides/biosynthesis , Plastids/metabolism , Pyrimidines/biosynthesis , Symporters/metabolism , Adenine/metabolism , Amino Acid Sequence , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Biosynthetic Pathways/drug effects , Brassica/metabolism , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Gene Expression Regulation, Plant/drug effects , Green Fluorescent Proteins/metabolism , Guanine/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Models, Biological , Molecular Sequence Data , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Plastids/drug effects , Protein Transport/drug effects , Protoplasts/drug effects , Protoplasts/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sodium Chloride/pharmacology , Substrate Specificity/drug effects , Symporters/chemistry , Symporters/genetics , Time Factors , Uracil/metabolism
12.
J Biol Chem ; 287(19): 15684-95, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22437829

ABSTRACT

The ubiquitous nucleobase-ascorbate transporter (NAT/NCS2) family includes more than 2,000 members, but only 15 have been characterized experimentally. Escherichia coli has 10 members, of which the uracil permease UraA and the xanthine permeases XanQ and XanP are functionally known. Of the remaining members, YgfU is closely related in sequence and genomic locus with XanQ. We analyzed YgfU and showed that it is a proton-gradient dependent, low-affinity (K(m) 0.5 mM), and high-capacity transporter for uric acid. It also shows a low capacity for transport of xanthine at 37 °C but not at 25 °C. Based on the set of positions delineated as important from our previous Cys-scanning analysis of permease XanQ, we subjected YgfU to rationally designed site-directed mutagenesis. The results show that the conserved His-37 (TM1), Glu-270 (TM8), Asp-298 (TM9), and Gln-318 and Asn-319 (TM10) are functionally irreplaceable, and Thr-100 (TM3) is essential for the uric acid selectivity because its replacement with Ala allows efficient uptake of xanthine. The key role of these residues is corroborated by the conservation pattern and homology modeling on the recently described x-ray structure of permease UraA. In addition, site-specific replacements at TM8 (S271A, M274D, V282S) impair expression in the membrane, and V320N (TM10) inactivates the permease, whereas R327G (TM10) or S426N (TM14) reduces the affinity for uric acid (4-fold increased K(m)). Our study shows that comprehensive analysis of structure-function relationships in a newly characterized transporter can be accomplished with relatively few site-directed replacements, based on the knowledge available from Cys-scanning mutagenesis of a prototypic homolog.


Subject(s)
Escherichia coli Proteins/metabolism , Nucleobase Transport Proteins/metabolism , Uric Acid/metabolism , Xanthine/metabolism , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Biological Transport , Blotting, Western , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Protein Structure, Secondary , Sequence Homology, Amino Acid
13.
J Biol Chem ; 285(25): 19422-33, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20406814

ABSTRACT

The nucleobase-ascorbate transporter (NAT) signature motif is a conserved 11-amino acid sequence of the ubiquitous NAT/NCS2 family, essential for function and selectivity of both a bacterial (YgfO) and a fungal (UapA) purine-transporting homolog. We examined the role of NAT motif in more detail, using Cys-scanning and site-directed alkylation analysis of the YgfO xanthine permease of Escherichia coli. Analysis of single-Cys mutants in the sequence 315-339 for sensitivity to inactivation by 2-sulfonatoethyl methanethiosulfonate (MTSES(-)) and N-ethylmaleimide (NEM) showed a similar pattern: highly sensitive mutants clustering at the motif sequence (323-329) and a short alpha-helical face downstream (332, 333, 336). In the presence of substrate, N325C is protected from alkylation with either MTSES(-) or NEM, whereas sensitivity of A323C to inactivation by NEM is enhanced, shifting IC(50) from 34 to 14 microM. Alkylation or sensitivity of the other mutants is unaffected by substrate; the lack of an effect on Q324C is attributed to gross inability of this mutant for high affinity binding. Site-directed mutants G333R and S336N at the alpha-helical face downstream the motif display specific changes in ligand recognition relative to wild type; G333R allows binding of 7-methyl and 8-methylxanthine, whereas S336N disrupts affinity for 6-thioxanthine. Finally, all assayable motif-mutants are highly accessible to MTSES(-) from the periplasmic side. The data suggest that the NAT motif region lines the solvent- and substrate-accessible inner cavity, Asn-325 is at the binding site, Ala-323 responds to binding with a specific conformational shift, and Gly-333 and Ser-336 form part of the purine permeation pathway.


Subject(s)
Alanine/chemistry , Ascorbic Acid/chemistry , Asparagine/chemistry , Escherichia coli Proteins/physiology , Nucleobase Transport Proteins/physiology , Amino Acid Motifs , Amino Acid Sequence , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Ethylmaleimide/chemistry , Inhibitory Concentration 50 , Kinetics , Mesylates/chemistry , Molecular Sequence Data , Mutation , Nucleic Acids/chemistry , Nucleobase Transport Proteins/chemistry , Protein Structure, Secondary , Purines/chemistry , Sequence Homology, Amino Acid
15.
Science ; 322(5902): 709-13, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18927357

ABSTRACT

The nucleobase-cation-symport-1 (NCS1) transporters are essential components of salvage pathways for nucleobases and related metabolites. Here, we report the 2.85-angstrom resolution structure of the NCS1 benzyl-hydantoin transporter, Mhp1, from Microbacterium liquefaciens. Mhp1 contains 12 transmembrane helices, 10 of which are arranged in two inverted repeats of five helices. The structures of the outward-facing open and substrate-bound occluded conformations were solved, showing how the outward-facing cavity closes upon binding of substrate. Comparisons with the leucine transporter LeuT(Aa) and the galactose transporter vSGLT reveal that the outward- and inward-facing cavities are symmetrically arranged on opposite sides of the membrane. The reciprocal opening and closing of these cavities is synchronized by the inverted repeat helices 3 and 8, providing the structural basis of the alternating access model for membrane transport.


Subject(s)
Actinomycetales/chemistry , Bacterial Proteins/chemistry , Nucleobase Transport Proteins/chemistry , Symporters/chemistry , Actinomycetales/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Binding Sites , Cations/chemistry , Cations/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Hydantoins/chemistry , Hydantoins/metabolism , Ion Transport , Models, Molecular , Molecular Sequence Data , Nucleobase Transport Proteins/metabolism , Protein Conformation , Protein Structure, Secondary , Sodium/metabolism , Symporters/metabolism
16.
J Mol Biol ; 382(5): 1121-35, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18718842

ABSTRACT

UapA, a uric acid-xanthine permease of Aspergillus nidulans, has been used as a prototype to study structure-function relationships in the ubiquitous nucleobase-ascorbate transporter (NAT) family. Using novel genetic screens, rational mutational design, chimeric NAT molecules, and extensive transport kinetic analyses, we show that dynamic synergy between three distinct domains, transmembrane segment (TMS)1, the TMS8-9 loop, and TMS12, defines the function and specificity of UapA. The TMS8-9 loop includes four residues absolutely essential for substrate binding and transport (Glu356, Asp388, Gln408, and Asn409), whereas TMS1 and TMS12 seem to control, through steric hindrance or electrostatic repulsion, the differential access of purines to the TMS8-9 domain. Thus, UapA specificity is determined directly by the specific interactions of a given substrate with the TMS8-9 loop and indirectly by interactions of this loop with TMS1 and TMS12. We finally show that intramolecular synergy among UapA domains is highly specific and propose that it forms the basis for the evolution of the unique specificity of UapA for uric acid, a property not present in other NAT members.


Subject(s)
Fungal Proteins/chemistry , Fungal Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Uric Acid/metabolism , Amino Acid Sequence , Amino Acid Substitution , Aspergillus nidulans/genetics , Aspergillus nidulans/growth & development , Aspergillus nidulans/metabolism , Binding Sites/genetics , Fungal Proteins/genetics , Genes, Fungal , Kinetics , Membrane Transport Proteins/genetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Nucleobase Transport Proteins/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid
17.
J Biol Chem ; 283(20): 13666-78, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18359771

ABSTRACT

Transmembrane helix XII of UapA, the major fungal homolog of the nucleobase-ascorbate transporter (NAT/NCS2) family, has been proposed to contain an aromatic residue acting as a purine-selectivity filter, distinct from the binding site. To analyze the role of helix XII more systematically, we employed Cys-scanning mutagenesis of the Escherichia coli xanthine-specific homolog YgfO. Using a functional mutant devoid of Cys residues (C-less), each amino acid residue in sequence 419ILPASIYVLVENPICAGGLTAILLNIILPGGY450 (the putative helix XII is underlined) was replaced individually with Cys. Of the 32 single-Cys mutants, 25 accumulate xanthine to 80-130% of the steady state observed with C-less YgfO, six (P421C, S423C, I424C, Y425C, L427C, G436C) accumulate to low levels (15-40%), and I432C is inactive. Immunoblot analysis shows that P421C and I432C display low expression in the membrane. Extensive mutagenesis reveals that replacement of Ile-432 with equally or more bulky side chains abolishes active transport without affecting expression, whereas replacement with smaller side chains allows activity but impairs affinity for the analogues 1-methyl and 6-thioxanthine. Only three of the single-Cys mutants of helix XII (V426C, N430C, and N443C) are sensitive to inactivation by N-ethylmaleimide. N430C is highly sensitive, with an IC50 of 10 microm, and is completely protected against inactivation in the presence of 2-thioxanthine, a high affinity substrate analogue. Other xanthine analogues are poorly bound by N430C, whereas replacement of Asn-430 with Thr inactivates the permease. The findings suggest that Ile-432 and Asn-430 of helix XII are crucial for purine uptake and affinity, and Asn-430 is probably at the vicinity of the binding site.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Nucleobase Transport Proteins/chemistry , Amino Acid Sequence , Asparagine/chemistry , Binding Sites , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Ethylmaleimide/pharmacology , Gene Expression Regulation, Bacterial , Inhibitory Concentration 50 , Isoleucine/chemistry , Kinetics , Models, Biological , Molecular Sequence Data , Nucleobase Transport Proteins/genetics , Protein Conformation , Protein Structure, Secondary , Sequence Homology, Amino Acid
18.
Int J Parasitol ; 38(6): 641-53, 2008 May.
Article in English | MEDLINE | ID: mdl-18036529

ABSTRACT

Parasitic protozoa are unable to synthesise purines de novo and thus depend on the uptake of nucleosides and nucleobases across their plasma membrane through specific transporters. A number of nucleoside and nucleobase transporters from Trypanosoma brucei brucei and Leishmania major have recently been characterised and shown to belong to the equilibrative nucleoside transporter (ENT) family. A number of studies have demonstrated the functional importance of particular transmembrane segments (TMS) in nucleoside-specific ENT proteins. TbNBT1, one of only three bona fide nucleobase-selective members of the ENT family, has previously been shown to be a high-affinity transporter for purine nucleobases and guanosine. In this study, we use the Saccharomyces cerevisiae expression system to build a biochemical model of how TbNBT1 recognises nucleobases. We next performed random in vitro and site-directed mutagenesis to identify residues critical for TbNBT1 function. The identification of residues likely to contribute to permeant binding, when combined with a structural model of TbNBT1 obtained by homology threading, yield a tentative three-dimensional model of the transporter binding site that is consistent with the binding model emerging from the biochemical data. The model strongly suggests the involvement of TMS5, TMS7 and TMS8 in TbNBT1 function. This situation is very similar to that concerning transporters of the major facilitator superfamily (MFS), one of which was used as a template for the threading. This point raises the possibility that ENT and MFS carriers, despite being considered evolutionarily distinct, might in fact share similar topologies and substrate translocations pathways.


Subject(s)
Nucleobase Transport Proteins , Protozoan Proteins , Saccharomyces cerevisiae/genetics , Trypanosoma brucei brucei/genetics , Animals , Biological Transport/genetics , Gene Expression , Kinetics , Leishmania major/genetics , Leishmania major/metabolism , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Nucleobase Transport Proteins/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Trypanosoma brucei brucei/metabolism
19.
J Biol Chem ; 281(52): 39881-90, 2006 Dec 29.
Article in English | MEDLINE | ID: mdl-17077086

ABSTRACT

The nucleobase-ascorbate transporter (NAT) signature motif is a conserved sequence motif of the ubiquitous NAT/NCS2 family implicated in defining the function and selectivity of purine translocation pathway in the major fungal homolog UapA. To analyze the role of NAT motif more systematically, we employed Cys-scanning mutagenesis of the Escherichia coli xanthine-specific homolog YgfO. Using a functional mutant devoid of Cys residues (C-less), each amino acid residue in sequence (315)GSIPITTFAQNNGVIQMTGVASRYVG(340) (motif underlined) was replaced individually with Cys. Of the 26 single-Cys mutants, 16 accumulate xanthine to > or =50% of the steady state observed with C-less YgfO, 4 accumulate to low levels (10-25% of C-less), F322C, N325C, and N326C accumulate marginally (5-8% of C-less), and P318C, Q324C, and G340C are inactive. When transferred to wild type, F322C(wt) and N326C(wt) are highly active, but P318G(wt), Q324C(wt), N325C(wt), and G340C(wt) are inactive, and G340A(wt) displays low activity. Immunoblot analysis shows that replacements at Pro-318 or Gly-340 are associated with low or negligible expression in the membrane. More extensive mutagenesis reveals that Gln-324 is critical for high affinity uptake and ligand recognition, and Asn-325 is irreplaceable for active xanthine transport, whereas Thr-332 and Gly-333 are important determinants of ligand specificity. All single-Cys mutants react with N-ethylmaleimide, but regarding sensitivity to inactivation, they fall to three regions; positions 315-322 are insensitive to N-ethylmaleimide, with IC(50) values > or =0.4 mM, positions 323-329 are highly sensitive, with IC(50) values of 15-80 microM, and sensitivity of positions 330-340 follows a periodicity, with mutants sensitive to inactivation clustering on one face of an alpha-helix.


Subject(s)
Amino Acid Substitution/genetics , Cysteine/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Nucleobase Transport Proteins/chemistry , Nucleobase Transport Proteins/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence , Asparagine/genetics , Biological Transport, Active/genetics , Cysteine/chemistry , Escherichia coli Proteins/metabolism , Glutamine/genetics , Isoleucine/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleobase Transport Proteins/metabolism , Protein Structure, Secondary/genetics , Valine/genetics , Xanthine/metabolism
20.
Mol Membr Biol ; 22(3): 251-61, 2005.
Article in English | MEDLINE | ID: mdl-16096267

ABSTRACT

The coding potential of the genome of E. coli K-12 includes YgfO and YicE, two members of the evolutionarily conserved NAT/NCS2 transporter family that are highly homologous to each other (45% residue identity) and closely related to UapA of Aspergillus nidulans, a most extensively studied microbial member of this family. YgfO and yicE were cloned from the genome, over-expressed extrachromosomally and assayed for uptake of [(3)H]xanthine and other nucleobases, in E. coli K-12, under conditions of negligible activity of the corresponding endogenous systems. Alternative, essentially equivalent functional versions of YgfO and YicE were engineered by C-terminal tagging with an epitope from the E. coli lactose permease and a biotin-acceptor domain from Klebsiella pneumoniae. Both YgfO and YicE were shown to be present in the plasma membrane of E. coli and function as specific, high-affinity transporters for xanthine (K(m) 4.2-4.6 microM for YgfO, or 2.9-3.8 microM for YicE), in a proton motive force-dependent manner; they display no detectable transport of uracil, hypoxanthine, or uric acid at external concentrations of up to 0.1 mM. Both YgfO and YicE are inefficient in recognizing uric acid or xanthine analogues modified at position 8 of the purine ring (8-methylxanthine, 8-azaxanthine, oxypurinol, allopurinol), which distinguishes them from their fungal homologues UapA and Xut1.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Nucleobase Transport Proteins/chemistry , Animals , Base Sequence , Biological Transport , Cloning, Molecular , Epitopes , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Nucleobase Transport Proteins/genetics , Nucleobase Transport Proteins/metabolism , Plasmids , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship , Substrate Specificity , Uracil/metabolism , Uric Acid/metabolism , Xanthine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...