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










Publication year range
1.
Viruses ; 13(12)2021 12 04.
Article in English | MEDLINE | ID: mdl-34960704

ABSTRACT

White chick hatchery disease is an emerging disease of broiler chicks with which the virus, chicken astrovirus, has been associated. Adult birds typically show no obvious clinical signs of infection, although some broiler breeder flocks have experienced slight egg drops. Substantial decreases in hatching are experienced over a two-week period, with an increase in mid-to-late embryo deaths, chicks too weak to hatch and pale, runted chicks with high mortality. Chicken astrovirus is an enteric virus, and strains are typically transmitted horizontally within flocks via the faecal-oral route; however, dead-in-shell embryos and weak, pale hatchlings indicate vertical transmission of the strains associated with white chick hatchery disease. Hatch levels are typically restored after two weeks when seroconversion of the hens to chicken astrovirus has occurred. Currently, there are no commercial vaccines available for the virus; therefore, the only means of protection is by good levels of biosecurity. This review aims to outline the current understanding regarding white chick hatchery disease in broiler chick flocks suffering from severe early mortality and increased embryo death in countries worldwide.


Subject(s)
Astroviridae Infections/veterinary , Avastrovirus , Chickens , Communicable Diseases, Emerging/veterinary , Poultry Diseases , Animal Husbandry , Animals , Astroviridae Infections/physiopathology , Astroviridae Infections/prevention & control , Astroviridae Infections/virology , Avastrovirus/isolation & purification , Communicable Diseases, Emerging/physiopathology , Communicable Diseases, Emerging/prevention & control , Communicable Diseases, Emerging/virology , Disease Progression , Poultry Diseases/physiopathology , Poultry Diseases/prevention & control , Poultry Diseases/virology
2.
Malar J ; 20(1): 295, 2021 Jun 30.
Article in English | MEDLINE | ID: mdl-34193175

ABSTRACT

BACKGROUND: Plasmodium species are entirely dependent upon their host as a source of essential iron. Although it is an indispensable micronutrient, oxidation of excess ferrous iron to the ferric state in the cell cytoplasm can produce reactive oxygen species that are cytotoxic. The malaria parasite must therefore carefully regulate the processes involved in iron acquisition and storage. A 273 amino acid membrane transporter that is a member of the vacuolar iron transporter (VIT) family and an orthologue of the yeast Ca2+-sensitive cross complementer (CCC1) protein plays a major role in cytosolic iron detoxification of Plasmodium species and functions in transport of ferrous iron ions into the endoplasmic reticulum for storage. While this transporter, termed PfVIT, is not critical for viability of the parasite evidence from studies of mice infected with VIT-deficient Plasmodium suggests it could still provide an efficient target for chemoprophylactic treatment of malaria. Individual amino acid residues that constitute the Fe2+ binding site of the protein were identified to better understand the structural basis of substrate recognition and binding by PfVIT. METHODS: Using the crystal structure of a recently published plant VIT as a template, a high-quality homology model of PfVIT was constructed to identify the amino acid composition of the transporter's substrate binding site and to act as a guide for subsequent mutagenesis studies. To test the effect of mutation of the substrate binding-site residues on PfVIT function a yeast complementation assay assessed the ability of overexpressed, recombinant wild type and mutant PfVIT to rescue an iron-sensitive deletion strain (ccc1∆) of Saccharomyces cerevisiae yeast from the toxic effects of a high concentration of extracellular iron. RESULTS: The combined in silico and mutagenesis approach identified a methionine residue located within the cytoplasmic metal binding domain of the transporter as essential for PfVIT function and provided insight into the structural basis for the Fe2+-selectivity of the protein. CONCLUSION: The structural model of the metal binding site of PfVIT opens the door for rational design of therapeutics to interfere with iron homeostasis within the malaria parasite.


Subject(s)
Cation Transport Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Binding Sites , Biological Transport , Cation Transport Proteins/metabolism , Iron/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Sequence Alignment , Sequence Analysis, Protein
3.
Chem Sci ; 10(47): 10948-10957, 2019 Dec 21.
Article in English | MEDLINE | ID: mdl-32190252

ABSTRACT

Oligodeoxynucleotides incorporating internucleotide phosphoroselenolate linkages have been prepared under solid-phase synthesis conditions using dimer phosphoramidites. These dimers were constructed following the high yielding Michaelis-Arbuzov (M-A) reaction of nucleoside H-phosphonate derivatives with 5'-deoxythymidine-5'-selenocyanate and subsequent phosphitylation. Efficient coupling of the dimer phosphoramidites to solid-supported substrates was observed under both manual and automated conditions and required only minor modifications to the standard DNA synthesis cycle. In a further demonstration of the utility of M-A chemistry, the support-bound selenonucleoside was reacted with an H-phosphonate and then chain extended using phosphoramidite chemistry. Following initial unmasking of methyl-protected phosphoroselenolate diesters, pure oligodeoxynucleotides were isolated using standard deprotection and purification procedures and subsequently characterised by mass spectrometry and circular dichroism. The CD spectra of both modified and native duplexes derived from self-complementary sequences with A-form, B-form or mixed conformational preferences were essentially superimposable. These sequences were also used to study the effect of the modification upon duplex stability which showed context-dependent destabilisation (-0.4 to -3.1 °C per phosphoroselenolate) when introduced at the 5'-termini of A-form or mixed duplexes or at juxtaposed central loci within a B-form duplex (-1.0 °C per modification). As found with other nucleic acids incorporating selenium, expeditious crystallisation of a modified decanucleotide A-form duplex was observed and the structure solved to a resolution of 1.45 Å. The DNA structure adjacent to the modification was not significantly perturbed. The phosphoroselenolate linkage was found to impart resistance to nuclease activity.

4.
Res Microbiol ; 169(7-8): 461-467, 2018.
Article in English | MEDLINE | ID: mdl-28962921

ABSTRACT

Multidrug resistance is principally a consequence of the active transport of drugs out of the cell by proteins that are integral membrane transporters. In the following review, we present a synthesis of current understanding of the Escherichia coli multidrug resistance transporter, MdtM, a 410 amino acid residue protein that belongs to the large and ubiquitous major facilitator superfamily (MFS).


Subject(s)
Antiporters/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antiporters/chemistry , Antiporters/genetics , Biological Transport , Drug Resistance, Multiple, Bacterial , Escherichia coli/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Multigene Family
6.
Sci Rep ; 7: 42850, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198449

ABSTRACT

Vacuolar iron transporters (VITs) are a poorly understood family of integral membrane proteins that can function in iron homeostasis via sequestration of labile Fe2+ into vacuolar compartments. Here we report on the heterologous overexpression and purification of PfVIT, a vacuolar iron transporter homologue from the human malaria-causing parasite Plasmodium falciparum. Use of synthetic, codon-optimised DNA enabled overexpression of functional PfVIT in the inner membrane of Escherichia coli which, in turn, conferred iron tolerance to the bacterial cells. Cells that expressed PfVIT had decreased levels of total cellular iron compared with cells that did not express the protein. Qualitative transport assays performed on inverted vesicles enriched with PfVIT revealed that the transporter catalysed Fe2+/H+ exchange driven by the proton electrochemical gradient. Furthermore, the PfVIT transport function in this system did not require the presence of any Plasmodium-specific factor such as post-translational phosphorylation. PfVIT purified as a monomer and, as measured by intrinsic protein fluorescence quenching, bound Fe2+ in detergent solution with low micromolar affinity. This study of PfVIT provides material for future detailed biochemical, biophysical and structural studies to advance understanding of the vacuolar iron transporter family of membrane proteins from important human pathogens.


Subject(s)
Cation Transport Proteins/metabolism , Escherichia coli/growth & development , Hydrogen/metabolism , Iron/metabolism , Plasmodium falciparum/metabolism , Cation Transport Proteins/genetics , Cloning, Molecular , Escherichia coli/drug effects , Escherichia coli/genetics , Genes, Synthetic , Humans , Iron/pharmacology , Malaria, Falciparum/parasitology , Microbial Viability , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins/metabolism , Vacuoles/metabolism
7.
Sci Rep ; 6: 22833, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26961153

ABSTRACT

Multidrug resistance arising from the activity of integral membrane transporter proteins presents a global public health threat. In bacteria such as Escherichia coli, transporter proteins belonging to the major facilitator superfamily make a considerable contribution to multidrug resistance by catalysing efflux of myriad structurally and chemically different antimicrobial compounds. Despite their clinical relevance, questions pertaining to mechanistic details of how these promiscuous proteins function remain outstanding, and the role(s) played by individual amino acid residues in recognition, binding and subsequent transport of different antimicrobial substrates by multidrug efflux members of the major facilitator superfamily requires illumination. Using in silico homology modelling, molecular docking and mutagenesis studies in combination with substrate binding and transport assays, we identified several amino acid residues that play important roles in antimicrobial substrate recognition, binding and transport by Escherichia coli MdtM, a representative multidrug efflux protein of the major facilitator superfamily. Furthermore, our studies suggested that 'aromatic clamps' formed by tyrosine and phenylalanine residues located within the substrate binding pocket of MdtM may be important for antimicrobial substrate recognition and transport by the protein. Such 'clamps' may be a structurally and functionally important feature of all major facilitator multidrug efflux proteins.


Subject(s)
Antiporters/metabolism , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Amino Acids/chemistry , Anti-Bacterial Agents/chemistry , Antiporters/chemistry , Antiporters/genetics , Biological Transport , Chloramphenicol/chemistry , Computer Simulation , Escherichia coli/drug effects , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Onium Compounds/chemistry , Organophosphorus Compounds/chemistry , Protein Binding , Protein Conformation
8.
Org Biomol Chem ; 14(4): 1201-5, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26565694

ABSTRACT

Michaelis-Arbuzov reactions of S-aryl disulfide derivatives of 3'-thiothymidine or 5'-thioadenosine with tris(trimethylsilyl) phosphite proceeded in high yields to the corresponding phosphorothiolate monoesters. Subsequent hydrolytic desilylation and phosphate coupling were effected in one-pot using liquid-assisted grinding in a vibration ball mill. Novel 3',5'- and 5',5'-pyrophosphorothiolate-linked dinucleoside cap analogues were thereby prepared.


Subject(s)
Nucleotides/chemical synthesis , Organophosphorus Compounds/chemical synthesis , Sulfhydryl Compounds/chemical synthesis , Molecular Structure , Nucleotides/chemistry , Organophosphorus Compounds/chemistry , Sulfhydryl Compounds/chemistry
9.
Antibiotics (Basel) ; 4(1): 113-35, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-27025617

ABSTRACT

Crystallization of integral membrane proteins is a challenging field and much effort has been invested in optimizing the overexpression and purification steps needed to obtain milligram amounts of pure, stable, monodisperse protein sample for crystallography studies. Our current work involves the structural and functional characterization of the Escherichia coli multidrug resistance transporter MdtM, a member of the major facilitator superfamily (MFS). Here we present a protocol for isolation of MdtM to increase yields of recombinant protein to the milligram quantities necessary for pursuit of structural studies using X-ray crystallography. Purification of MdtM was enhanced by introduction of an elongated His-tag, followed by identification and subsequent removal of chaperonin contamination. For crystallization trials of MdtM, detergent screening using size exclusion chromatography determined that decylmaltoside (DM) was the shortest-chain detergent that maintained the protein in a stable, monodispersed state. Crystallization trials of MdtM performed using the hanging-drop diffusion method with commercially available crystallization screens yielded 3D protein crystals under several different conditions. We contend that the purification protocol described here may be employed for production of high-quality protein of other multidrug efflux members of the MFS, a ubiquitous, physiologically and clinically important class of membrane transporters.

10.
Mol Microbiol ; 92(4): 872-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24684269

ABSTRACT

Resistance to high concentrations of bile salts in the human intestinal tract is vital for the survival of enteric bacteria such as Escherichia coli. Although the tripartite AcrAB-TolC efflux system plays a significant role in this resistance, it is purported that other efflux pumps must also be involved. We provide evidence from a comprehensive suite of experiments performed at two different pH values (7.2 and 6.0) that reflect pH conditions that E. coli may encounter in human gut that MdtM, a single-component multidrug resistance transporter of the major facilitator superfamily, functions in bile salt resistance in E. coli by catalysing secondary active transport of bile salts out of the cell cytoplasm. Furthermore, assays performed on a chromosomal ΔacrB mutant transformed with multicopy plasmid encoding MdtM suggested a functional synergism between the single-component MdtM transporter and the tripartite AcrAB-TolC system that results in a multiplicative effect on resistance. Substrate binding experiments performed on purified MdtM demonstrated that the transporter binds to cholate and deoxycholate with micromolar affinity, and transport assays performed on inverted vesicles confirmed the capacity of MdtM to catalyse electrogenic bile salt/H(+) antiport.


Subject(s)
Antiporters/metabolism , Bile Acids and Salts/metabolism , Bile Acids and Salts/toxicity , Drug Tolerance , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Escherichia coli/metabolism , Bacterial Outer Membrane Proteins/metabolism , Lipoproteins/metabolism , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/metabolism
11.
BMC Microbiol ; 13: 113, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23701827

ABSTRACT

BACKGROUND: In neutralophilic bacteria, monovalent metal cation/H+ antiporters play a key role in pH homeostasis. In Escherichia coli, only four antiporters (NhaA, NhaB, MdfA and ChaA) are identified to function in maintenance of a stable cytoplasmic pH under conditions of alkaline stress. We hypothesised that the multidrug resistance protein MdtM, a recently characterised homologue of MdfA and a member of the major facilitator superfamily, also functions in alkaline pH homeostasis. RESULTS: Assays that compared the growth of an E. coli ΔmdtM deletion mutant transformed with a plasmid encoding wild-type MdtM or the dysfunctional MdtM D22A mutant at different external alkaline pH values (ranging from pH 8.5 to 10) revealed a potential contribution by MdtM to alkaline pH tolerance, but only when millimolar concentrations of sodium or potassium was present in the growth medium. Fluorescence-based activity assays using inverted vesicles generated from transformants of antiporter-deficient (ΔnhaA, ΔnhaB, ΔchaA) E. coli TO114 cells defined MdtM as a low-affinity antiporter that catalysed electrogenic exchange of Na+, K+, Rb+ or Li+ for H+. The K+/H+ antiport reaction had a pH optimum at 9.0, whereas the Na+/H+ exchange activity was optimum at pH 9.25. Measurement of internal cellular pH confirmed MdtM as contributing to maintenance of a stable cytoplasmic pH, acid relative to the external pH, under conditions of alkaline stress. CONCLUSIONS: Taken together, the results support a role for MdtM in alkaline pH tolerance. MdtM can therefore be added to the currently limited list of antiporters known to function in pH homeostasis in the model organism E. coli.


Subject(s)
Alkalies/metabolism , Antiporters/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Homeostasis , Antiporters/genetics , Cations/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Gene Deletion , Genetic Complementation Test , Hydrogen-Ion Concentration , Metals/metabolism
12.
J Antimicrob Chemother ; 68(4): 831-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23221628

ABSTRACT

OBJECTIVES: Quaternary ammonium compounds (QACs) are used extensively as biocides and their misuse may be contributing to the development of bacterial resistance. Although the major intrinsic resistance to QACs of Gram-negative bacteria is mediated by the action of tripartite multidrug transporters of the resistance-nodulation-division family, we aimed to test if the promiscuity of the recently characterized major facilitator superfamily multidrug transporter, MdtM, from Escherichia coli enabled it also to function in the efflux of QACs. METHODS: The ability of the major facilitator mdtM gene product, when overexpressed from multicopy plasmid, to protect E. coli cells from the toxic effects of a panel of seven QACs was determined using growth inhibition assays in liquid medium. Interaction between QACs and MdtM was studied by a combination of substrate binding assays using purified protein in detergent solution and transport assays using inverted vesicles. RESULTS: E. coli cells that overproduced MdtM were less susceptible to the cytotoxic effects of each of the QACs tested compared with cells that did not overproduce the transporter. Purified MdtM bound each QAC with micromolar affinity and the protein utilized the electrochemical proton gradient to transport QACs across the cytoplasmic membrane. Furthermore, the results suggested a functional interaction between MdtM and the tripartite resistance-nodulation-division family AcrAB-TolC efflux system. CONCLUSIONS: The results support a hitherto unidentified capacity for a single-component multidrug transporter of the major facilitator superfamily, MdtM, to function in the efflux of a broad range of QACs and thus contribute to the intrinsic resistance of E. coli to these compounds.


Subject(s)
Antiporters/metabolism , Disinfectants/pharmacology , Drug Resistance, Bacterial , Escherichia coli Proteins/metabolism , Escherichia coli/drug effects , Quaternary Ammonium Compounds/pharmacology , Antiporters/genetics , Biological Transport , Disinfectants/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Protein Binding , Quaternary Ammonium Compounds/metabolism
13.
Mol Biochem Parasitol ; 186(1): 69-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22982092

ABSTRACT

Control of fasciolosis is threatened by the development of anthelmintic resistance. Enhanced triclabendazole (TCBZ) efflux by ABC transporters such as P-glycoprotein (Pgp) has been implicated in this process. A putative full length cDNA coding for a Pgp expressed in adult Fasciola hepatica has been constructed and used to design a primer set capable of amplifying a region encoding part of the second nucleotide binding domain of Pgp when genomic DNA was used as a template. Application of this primer set to genomic DNA from TCBZ-resistant and -susceptible field populations has shown a significant difference in the alleles present. Analysis of an allele occurring at a three-fold higher frequency in the "resistant" population revealed that it was characterised by a serine to arginine substitution at residue 1144. Homology modelling studies have been used to locate this site in the Pgp structure and hence assess its potential to modify functional activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amino Acid Substitution , Anthelmintics/pharmacology , Benzimidazoles/pharmacology , Drug Resistance , Fasciola hepatica/drug effects , Fasciola hepatica/genetics , Alleles , Animals , DNA Primers/genetics , Models, Molecular , Polymerase Chain Reaction , Protein Conformation , Triclabendazole
14.
Biochimie ; 94(6): 1334-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22426385

ABSTRACT

Multidrug resistance (MDR) occurs when bacteria simultaneously acquire resistance to a broad spectrum of structurally dissimilar compounds to which they have not previously been exposed. MDR is principally a consequence of the active transport of drugs out of the cell by proteins that are integral membrane transporters. We characterised and purified the putative Escherichia coli MDR transporter, MdtM, a 410 amino acid residue protein that belongs to the large and ubiquitous major facilitator superfamily. Functional characterisation of MdtM using growth inhibition and whole cell transport assays revealed its role in intrinsic resistance of E. coli cells to the antimicrobials ethidium bromide and chloramphenicol. Site-directed mutagenesis studies implied that the MdtM aspartate 22 residue and the highly conserved arginine at position 108 play a role in proton recognition. MdtM was homologously overexpressed and purified to homogeneity in dodecyl-ß-D-maltopyranoside detergent solution and the oligomeric state and stability of the protein in a variety of detergent solutions was investigated using size-exclusion HPLC. Purified MdtM is monomeric and stable in dodecyl-ß-D-maltopyranoside solution and binds chloramphenicol with nanomolar affinity in the same detergent. This work provides a firm foundation for structural studies on this class of multidrug transporter protein.


Subject(s)
Antiporters/chemistry , Escherichia coli Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Antiporters/genetics , Antiporters/metabolism , Drug Resistance, Multiple , Escherichia coli/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Mutagenesis, Site-Directed
15.
Biophys J ; 97(5): 1346-53, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19720022

ABSTRACT

Major facilitators represent the largest superfamily of secondary active transporter proteins and catalyze the transport of an enormous variety of small solute molecules across biological membranes. However, individual superfamily members, although they may be architecturally similar, exhibit strict specificity toward the substrates they transport. The structural basis of this specificity is poorly understood. A member of the major facilitator superfamily is the glycerol-3-phosphate (G3P) transporter (GlpT) from the Escherichia coli inner membrane. GlpT is an antiporter that transports G3P into the cell in exchange for inorganic phosphate (P(i)). By combining large-scale molecular-dynamics simulations, mutagenesis, substrate-binding affinity, and transport activity assays on GlpT, we were able to identify key amino acid residues that confer substrate specificity upon this protein. Our studies suggest that only a few amino acid residues that line the transporter lumen act as specificity determinants. Whereas R45, K80, H165, and, to a lesser extent Y38, Y42, and Y76 contribute to recognition of both free P(i) and the phosphate moiety of G3P, the residues N162, Y266, and Y393 function in recognition of only the glycerol moiety of G3P. It is the latter interactions that give the transporter a higher affinity to G3P over P(i).


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Amino Acid Sequence , Computer Simulation , Escherichia coli , Fosfomycin/metabolism , Membrane Transport Proteins/genetics , Models, Biological , Models, Molecular , Mutation , Protein Binding/genetics , Protein Transport/genetics
16.
Nat Struct Mol Biol ; 16(6): 652-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19430461

ABSTRACT

Sertraline and fluoxetine are selective serotonin re-uptake inhibitors (SSRIs) that are widely prescribed to treat depression. They exert their effects by inhibiting the presynaptic plasma membrane serotonin transporter (SERT). All SSRIs possess halogen atoms at specific positions, which are key determinants for the drugs' specificity for SERT. For the SERT protein, however, the structural basis of its specificity for SSRIs is poorly understood. Here we report the crystal structures of LeuT, a bacterial SERT homolog, in complex with sertraline, R-fluoxetine or S-fluoxetine. The SSRI halogens all bind to exactly the same pocket within LeuT. Mutation at this halogen-binding pocket (HBP) in SERT markedly reduces the transporter's affinity for SSRIs but not for tricyclic antidepressants. Conversely, when the only nonconserved HBP residue in both norepinephrine and dopamine transporters is mutated into that found in SERT, their affinities for all the three SSRIs increase uniformly. Thus, the specificity of SERT for SSRIs is dependent largely on interaction of the drug halogens with the protein's HBP.


Subject(s)
Amino Acid Transport Systems, Neutral/chemistry , Fluoxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/chemistry , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Sertraline/pharmacology , Amino Acid Transport Systems, Neutral/metabolism , Antidepressive Agents, Tricyclic/pharmacology , Binding Sites , Cell Line , Crystallography, X-Ray/methods , Dopamine/metabolism , Drug Evaluation, Preclinical , Humans , Models, Chemical , Mutation , Norepinephrine/metabolism , Protein Binding , Selective Serotonin Reuptake Inhibitors/pharmacology
17.
Annu Rev Microbiol ; 62: 289-305, 2008.
Article in English | MEDLINE | ID: mdl-18537473

ABSTRACT

The major facilitator superfamily (MFS) represents the largest group of secondary active membrane transporters, and its members transport a diverse range of substrates. Recent work shows that MFS antiporters, and perhaps all members of the MFS, share the same three-dimensional structure, consisting of two domains that surround a substrate translocation pore. The advent of crystal structures of three MFS antiporters sheds light on their fundamental mechanism; they operate via a single binding site, alternating-access mechanism that involves a rocker-switch type movement of the two halves of the protein. In the sn-glycerol-3-phosphate transporter (GlpT) from Escherichia coli, the substrate-binding site is formed by several charged residues and a histidine that can be protonated. Salt-bridge formation and breakage are involved in the conformational changes of the protein during transport. In this review, we attempt to give an account of a set of mechanistic principles that characterize all MFS antiporters.


Subject(s)
Antiporters/chemistry , Antiporters/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli Proteins , Kinetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Models, Molecular , Protein Conformation
18.
J Mol Biol ; 378(4): 828-39, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18395745

ABSTRACT

Active transport of substrates across cytoplasmic membranes is of great physiological, medical and pharmaceutical importance. The glycerol-3-phosphate (G3P) transporter (GlpT) of the E. coli inner membrane is a secondary active antiporter from the ubiquitous major facilitator superfamily that couples the import of G3P to the efflux of inorganic phosphate (P(i)) down its concentration gradient. Integrating information from a novel combination of structural, molecular dynamics simulations and biochemical studies, we identify the residues involved directly in binding of substrate to the inward-facing conformation of GlpT, thus defining the structural basis for the substrate-specificity of this transporter. The substrate binding mechanism involves protonation of a histidine residue at the binding site. Furthermore, our data suggest that the formation and breaking of inter- and intradomain salt bridges control the conformational change of the transporter that accompanies substrate translocation across the membrane. The mechanism we propose may be a paradigm for organophosphate:phosphate antiporters.


Subject(s)
Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Sodium Chloride/chemistry , Amino Acids/genetics , Amino Acids/metabolism , Binding Sites , Computer Simulation , Crystallography, X-Ray , Kinetics , Membrane Transport Proteins/genetics , Models, Molecular , Mutation/genetics , Protein Structure, Tertiary , Protons
19.
Biochemistry ; 46(43): 12190-7, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17915951

ABSTRACT

Secondary active transport of substrate across the cell membrane is crucial to many cellular and physiological processes. The crystal structure of one member of the secondary active transporter family, the sn-glycerol-3-phosphate (G3P) transporter (GlpT) of the inner membrane of Escherichia coli, suggests a mechanism for substrate translocation across the membrane that involves a rocker-switch-type movement of the protein. This rocker-switch mechanism makes two specific predictions with respect to kinetic behavior: the transport rate increases with the temperature, whereas the binding affinity of the transporter to a substrate is temperature-independent. In this work, we directly tested these two predictions by transport kinetics and substrate-binding experiments, integrating the data on this single system into a coherent set of observations. The transport kinetics of the physiologically relevant G3P-phosphate antiport reaction were characterized at different temperatures using both E. coli whole cells and GlpT reconstituted into proteoliposomes. Substrate-binding affinity of the transporter was measured using tryptophan fluorescence quenching in detergent solution. Indeed, the substrate transport velocity of GlpT increased dramatically with temperature. In contrast, neither the apparent Michaelis constant (Km) nor the apparent substrate-binding dissociation constant (Kd) showed temperature dependence. Moreover, GlpT-catalyzed G3P translocation exhibited a completely linear Arrhenius function with an activation energy of 35.2 kJ mol-1 for the transporter reconstituted into proteoliposomes, suggesting that the substrate-loaded transporter is delicately poised between the inward- and outward-facing conformations. When these results are taken together, they are in agreement with a rocker-switch mechanism for GlpT.


Subject(s)
Membrane Transport Proteins/metabolism , Kinetics , Membrane Transport Proteins/chemistry , Protein Binding , Protein Conformation , Protein Transport , Proteolipids
20.
Science ; 317(5843): 1390-3, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17690258

ABSTRACT

Tricyclic antidepressants exert their pharmacological effect-inhibiting the reuptake of serotonin, norepinephrine, and dopamine-by directly blocking neurotransmitter transporters (SERT, NET, and DAT, respectively) in the presynaptic membrane. The drug-binding site and the mechanism of this inhibition are poorly understood. We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine binds at the inner end of the extracellular cavity of the transporter and is held in place by a hairpin loop and by a salt bridge. This binding site is separated from the leucine-binding site by the extracellular gate of the transporter. By directly locking the gate, desipramine prevents conformational changes and blocks substrate transport. Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters.


Subject(s)
Antidepressive Agents, Tricyclic/metabolism , Bacterial Proteins/metabolism , Desipramine/metabolism , Neurotransmitter Uptake Inhibitors/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Amino Acid Sequence , Animals , Antidepressive Agents, Tricyclic/chemistry , Bacterial Proteins/chemistry , Binding Sites , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Cell Line , Conserved Sequence , Crystallography, X-Ray , Desipramine/chemistry , Dopamine/chemistry , Dopamine/metabolism , Dopamine Uptake Inhibitors/chemistry , Dopamine Uptake Inhibitors/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Humans , Leucine/chemistry , Leucine/metabolism , Models, Molecular , Molecular Sequence Data , Neurotransmitter Uptake Inhibitors/chemistry , Norepinephrine/chemistry , Norepinephrine/metabolism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Serotonin/chemistry , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/chemistry , Selective Serotonin Reuptake Inhibitors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...