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










Publication year range
1.
Microbiology (Reading) ; 170(3)2024 03.
Article in English | MEDLINE | ID: mdl-38488830

ABSTRACT

Sialic acid (Sia) transporters are critical to the capacity of host-associated bacteria to utilise Sia for growth and/or cell surface modification. While N-acetyl-neuraminic acid (Neu5Ac)-specific transporters have been studied extensively, little is known on transporters dedicated to anhydro-Sia forms such as 2,7-anhydro-Neu5Ac (2,7-AN) or 2,3-dehydro-2-deoxy-Neu5Ac (Neu5Ac2en). Here, we used a Sia-transport-null strain of Escherichia coli to investigate the function of members of anhydro-Sia transporter families previously identified by computational studies. First, we showed that the transporter NanG, from the Glycoside-Pentoside-Hexuronide:cation symporter family, is a specific 2,7-AN transporter, and identified by mutagenesis a crucial functional residue within the putative substrate-binding site. We then demonstrated that NanX transporters, of the Major Facilitator Superfamily, also only transport 2,7-AN and not Neu5Ac2en nor Neu5Ac. Finally, we provided evidence that SiaX transporters, of the Sodium-Solute Symporter superfamily, are promiscuous Neu5Ac/Neu5Ac2en transporters able to acquire either substrate equally well. The characterisation of anhydro-Sia transporters expands our current understanding of prokaryotic Sia metabolism within host-associated microbial communities.


Subject(s)
N-Acetylneuraminic Acid , N-Acetylneuraminic Acid/analogs & derivatives , Organic Anion Transporters , Symporters , N-Acetylneuraminic Acid/chemistry , Symporters/genetics , Symporters/metabolism , Bacteria/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
2.
Microbiology (Reading) ; 170(2)2024 02.
Article in English | MEDLINE | ID: mdl-38363712

ABSTRACT

The twin-arginine protein transport (Tat) system exports folded proteins across the cytoplasmic membranes of prokaryotes and the energy transducing-membranes of plant thylakoids and mitochondria. Proteins are targeted to the Tat machinery by N-terminal signal peptides with a conserved twin-arginine motif, and some substrates are exported as heterodimers where the signal peptide is present on one of the partner proteins. A subset of Tat substrates is found in the membrane. Tat-dependent membrane proteins usually have large globular domains and a single transmembrane helix present at the N- or C-terminus. Five Tat substrates that have C-terminal transmembrane helices have previously been characterized in the model bacterium Escherichia coli. Each of these is an iron-sulfur cluster-containing protein involved in electron transfer from hydrogen or formate. Here we have undertaken a bioinformatic search to identify further tail-anchored Tat substrates encoded in bacterial genomes. Our analysis has revealed additional tail-anchored iron-sulfur proteins associated in modules with either a b-type cytochrome or a quinol oxidase. We also identified further candidate tail-anchored Tat substrates, particularly among members of the actinobacterial phylum, that are not predicted to contain cofactors. Using reporter assays, we show experimentally that six of these have both N-terminal Tat signal peptides and C-terminal transmembrane helices. The newly identified proteins include a carboxypeptidase and a predicted protease, and four sortase substrates for which membrane integration is a prerequisite for covalent attachment to the cell wall.


Subject(s)
Escherichia coli Proteins , Membrane Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Protein Transport , Arginine/metabolism , Carrier Proteins/metabolism , Protein Sorting Signals , Escherichia coli/genetics , Escherichia coli/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
4.
J Biol Chem ; 299(3): 102989, 2023 03.
Article in English | MEDLINE | ID: mdl-36758803

ABSTRACT

The human gastrointestinal (GI) tract harbors diverse microbial communities collectively known as the gut microbiota that exert a profound impact on human health and disease. The repartition and availability of sialic acid derivatives in the gut have a significant impact on the modulation of gut microbes and host susceptibility to infection and inflammation. Although N-acetylneuraminic acid (Neu5Ac) is the main form of sialic acids in humans, the sialic acid family regroups more than 50 structurally and chemically distinct modified derivatives. In the GI tract, sialic acids are found in the terminal location of mucin glycan chains constituting the mucus layer and also come from human milk oligosaccharides in the infant gut or from meat-based foods in adults. The repartition of sialic acid in the GI tract influences the gut microbiota composition and pathogen colonization. In this review, we provide an update on the mechanisms underpinning sialic acid utilization by gut microbes, focusing on sialidases, transporters, and metabolic enzymes.


Subject(s)
Gastrointestinal Microbiome , N-Acetylneuraminic Acid , Infant , Humans , N-Acetylneuraminic Acid/metabolism , Sialic Acids/metabolism , Mucins/metabolism , Polysaccharides/metabolism
5.
Microbiology (Reading) ; 169(2)2023 02.
Article in English | MEDLINE | ID: mdl-36790402

ABSTRACT

The twin arginine transport (Tat) pathway exports folded proteins across the cytoplasmic membranes of prokaryotes and the thylakoid membranes of chloroplasts. In Escherichia coli and other Gram-negative bacteria, the Tat machinery comprises TatA, TatB and TatC components. A Tat receptor complex, formed from all three proteins, binds Tat substrates, which triggers receptor organization and recruitment of further TatA molecules to form the active Tat translocon. The polytopic membrane protein TatC forms the core of the Tat receptor and harbours two binding sites for the sequence-related TatA and TatB proteins. A 'polar' cluster binding site, formed by TatC transmembrane helices (TMH) 5 and 6 is occupied by TatB in the resting receptor and exchanges for TatA during receptor activation. The second binding site, lying further along TMH6, is occupied by TatA in the resting state, but its functional relevance is unclear. Here we have probed the role of this second binding site through a programme of random and targeted mutagenesis. Characterization of three stably produced TatC variants, P221R, M222R and L225P, each of which is inactive for protein transport, demonstrated that the substitutions did not affect assembly of the Tat receptor. Moreover, the substitutions that we analysed did not abolish TatA or TatB binding to either binding site. Using targeted mutagenesis we introduced bulky substitutions into the TatA binding site. Molecular dynamics simulations and crosslinking analysis indicated that TatA binding at this site was substantially reduced by these amino acid changes, but TatC retained function. While it is not clear whether TatA binding at the TMH6 site is essential for Tat activity, the isolation of inactivating substitutions indicates that this region of the protein has a critical function.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Membrane Transport Proteins/metabolism , Arginine/metabolism , Carrier Proteins/metabolism , Binding Sites , Protein Transport/physiology
6.
Nat Commun ; 13(1): 4471, 2022 08 04.
Article in English | MEDLINE | ID: mdl-35927235

ABSTRACT

Tripartite ATP-independent periplasmic (TRAP) transporters are found widely in bacteria and archaea and consist of three structural domains, a soluble substrate-binding protein (P-domain), and two transmembrane domains (Q- and M-domains). HiSiaPQM and its homologs are TRAP transporters for sialic acid and are essential for host colonization by pathogenic bacteria. Here, we reconstitute HiSiaQM into lipid nanodiscs and use cryo-EM to reveal the structure of a TRAP transporter. It is composed of 16 transmembrane helices that are unexpectedly structurally related to multimeric elevator-type transporters. The idiosyncratic Q-domain of TRAP transporters enables the formation of a monomeric elevator architecture. A model of the tripartite PQM complex is experimentally validated and reveals the coupling of the substrate-binding protein to the transporter domains. We use single-molecule total internal reflection fluorescence (TIRF) microscopy in solid-supported lipid bilayers and surface plasmon resonance to study the formation of the tripartite complex and to investigate the impact of interface mutants. Furthermore, we characterize high-affinity single variable domains on heavy chain (VHH) antibodies that bind to the periplasmic side of HiSiaQM and inhibit sialic acid uptake, providing insight into how TRAP transporter function might be inhibited in vivo.


Subject(s)
Bacterial Proteins , N-Acetylneuraminic Acid , Adenosine Triphosphate/metabolism , Archaea/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Membrane Transport Proteins/metabolism , N-Acetylneuraminic Acid/metabolism
7.
Microb Genom ; 7(6)2021 06.
Article in English | MEDLINE | ID: mdl-34184979

ABSTRACT

Located at the tip of cell surface glycoconjugates, sialic acids are at the forefront of host-microbe interactions and, being easily liberated by sialidase enzymes, are used as metabolites by numerous bacteria, particularly by pathogens and commensals living on or near diverse mucosal surfaces. These bacteria rely on specific transporters for the acquisition of host-derived sialic acids. Here, we present the first comprehensive genomic and phylogenetic analysis of bacterial sialic acid transporters, leading to the identification of multiple new families and subfamilies. Our phylogenetic analysis suggests that sialic acid-specific transport has evolved independently at least eight times during the evolution of bacteria, from within four of the major families/superfamilies of bacterial transporters, and we propose a robust classification scheme to bring together a myriad of different nomenclatures that exist to date. The new transporters discovered occur in diverse bacteria, including Spirochaetes, Bacteroidetes, Planctomycetes and Verrucomicrobia, many of which are species that have not been previously recognized to have sialometabolic capacities. Two subfamilies of transporters stand out in being fused to the sialic acid mutarotase enzyme, NanM, and these transporter fusions are enriched in bacteria present in gut microbial communities. Our analysis supports the increasing experimental evidence that competition for host-derived sialic acid is a key phenotype for successful colonization of complex mucosal microbiomes, such that a strong evolutionary selection has occurred for the emergence of sialic acid specificity within existing transporter architectures.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Evolution, Molecular , Organic Anion Transporters/genetics , Symbiosis , Symporters/genetics , Animals , Bacteria/classification , Bacterial Proteins/genetics , Carbohydrate Epimerases/genetics , Humans , Membrane Transport Proteins/genetics , N-Acetylneuraminic Acid/metabolism , Phylogeny
8.
FEMS Microbiol Lett ; 368(10)2021 06 11.
Article in English | MEDLINE | ID: mdl-34057181

ABSTRACT

Their biochemical versatility and biotechnological importance make actinomycete bacteria attractive targets for ambitious genetic engineering using the toolkit of synthetic biology. But their complex biology also poses unique challenges. This mini review discusses some of the recent advances in synthetic biology approaches from an actinomycete perspective and presents examples of their application to the rational improvement of industrially relevant strains.


Subject(s)
Actinobacteria/genetics , Synthetic Biology/methods , Actinobacteria/metabolism , Industrial Microbiology/methods , Industrial Microbiology/trends , Metabolic Engineering , Synthetic Biology/trends
9.
J Biol Chem ; 295(40): 13724-13736, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32669363

ABSTRACT

The human gut symbiont Ruminococcus gnavus scavenges host-derived N-acetylneuraminic acid (Neu5Ac) from mucins by converting it to 2,7-anhydro-Neu5Ac. We previously showed that 2,7-anhydro-Neu5Ac is transported into R. gnavus ATCC 29149 before being converted back to Neu5Ac for further metabolic processing. However, the molecular mechanism leading to the conversion of 2,7-anhydro-Neu5Ac to Neu5Ac remained elusive. Using 1D and 2D NMR, we elucidated the multistep enzymatic mechanism of the oxidoreductase (RgNanOx) that leads to the reversible conversion of 2,7-anhydro-Neu5Ac to Neu5Ac through formation of a 4-keto-2-deoxy-2,3-dehydro-N-acetylneuraminic acid intermediate and NAD+ regeneration. The crystal structure of RgNanOx in complex with the NAD+ cofactor showed a protein dimer with a Rossman fold. Guided by the RgNanOx structure, we identified catalytic residues by site-directed mutagenesis. Bioinformatics analyses revealed the presence of RgNanOx homologues across Gram-negative and Gram-positive bacterial species and co-occurrence with sialic acid transporters. We showed by electrospray ionization spray MS that the Escherichia coli homologue YjhC displayed activity against 2,7-anhydro-Neu5Ac and that E. coli could catabolize 2,7-anhydro-Neu5Ac. Differential scanning fluorimetry analyses confirmed the binding of YjhC to the substrates 2,7-anhydro-Neu5Ac and Neu5Ac, as well as to co-factors NAD and NADH. Finally, using E. coli mutants and complementation growth assays, we demonstrated that 2,7-anhydro-Neu5Ac catabolism in E. coli depended on YjhC and on the predicted sialic acid transporter YjhB. These results revealed the molecular mechanisms of 2,7-anhydro-Neu5Ac catabolism across bacterial species and a novel sialic acid transport and catabolism pathway in E. coli.


Subject(s)
Bacterial Proteins/chemistry , Clostridiales/enzymology , N-Acetylneuraminic Acid/chemistry , Oxidoreductases/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Clostridiales/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Complementation Test , Humans , Mucins/chemistry , Mucins/metabolism , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism
10.
Microbiology (Reading) ; 165(8): 805-818, 2019 08.
Article in English | MEDLINE | ID: mdl-30964430

ABSTRACT

In the fight against antimicrobial resistance (AMR), antibiotic biosynthetic gene clusters are constantly being discovered. These clusters often include genes for membrane transporters that are involved in the export of the produced natural product during biosynthesis and/or subsequent resistance through active efflux. Despite transporter genes being integral parts of these clusters, study of the function of antibiotic export in natural producers such as Streptomyces spp. remains underexplored, in many cases lagging far behind our understanding of the biosynthetic enzymes. More efficient release of antibiotics by producing cells has potential benefits to industrial biotechnology and understanding the relationships between exporters in natural producers and resistance-associated efflux pumps in pathogens can inform our efforts to understand how AMR spreads. Herein we compile and critically assess the literature on the identification and characterization of antibiotic exporters and their contribution to production in natural antibiotic producers. We evaluate examples of how this knowledge could be used in biotechnology to increase yields of the final product or modulate its chemical nature. Finally, we consider the evidence that natural exporters form a reservoir of protein functions that could be hijacked by pathogens as efflux pumps and emphasize the need for much greater understanding of these exporters to fully exploit their potential for applications around human health.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacteria/metabolism , Biological Products/metabolism , Membrane Transport Proteins , Bacteria/genetics , Biotechnology , Drug Resistance, Multiple/genetics , Drug Resistance, Multiple/physiology , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multigene Family , Secondary Metabolism/genetics , Streptomyces/genetics , Streptomyces/metabolism
11.
J Biol Chem ; 290(45): 27113-27123, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26342690

ABSTRACT

Tripartite ATP-independent periplasmic (TRAP) transporters are secondary transporters that have evolved an obligate dependence on a substrate-binding protein (SBP) to confer unidirectional transport. Different members of the DctP family of TRAP SBPs have binding sites that recognize a diverse range of organic acid ligands but appear to only share a common electrostatic interaction between a conserved arginine and a carboxylate group in the ligand. We investigated the significance of this interaction using the sialic acid-specific SBP, SiaP, from the Haemophilus influenzae virulence-related SiaPQM TRAP transporter. Using in vitro, in vivo, and structural methods applied to SiaP, we demonstrate that the coordination of the acidic ligand moiety of sialic acid by the conserved arginine (Arg-147) is essential for the function of the transporter as a high affinity scavenging system. However, at high substrate concentrations, the transporter can function in the absence of Arg-147 suggesting that this bi-molecular interaction is not involved in further stages of the transport cycle. As well as being required for high affinity binding, we also demonstrate that the Arg-147 is a strong selectivity filter for carboxylate-containing substrates in TRAP transporters by engineering the SBP to recognize a non-carboxylate-containing substrate, sialylamide, through water-mediated interactions. Together, these data provide biochemical and structural support that TRAP transporters function predominantly as high affinity transporters for carboxylate-containing substrates.


Subject(s)
Arginine/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Arginine/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites/genetics , Crystallography, X-Ray , Genes, Bacterial , Haemophilus influenzae/genetics , Haemophilus influenzae/metabolism , Kinetics , Ligands , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
FEMS Microbiol Lett ; 304(1): 47-54, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20100283

ABSTRACT

The function of sialic acids in the biology of bacterial pathogens is reflected by the diverse range of solute transporters that can recognize these sugar acids. Here, we use an Escherichia coliDeltananT strain to characterize the function of known and proposed bacterial sialic acid transporters. We discover that the STM1128 gene from Salmonella enterica serovar Typhimurium, which encodes a member of the sodium solute symporter family, is able to restore growth on sialic acid to the DeltananT strain and is able to transport [(14)C]-sialic acid. Using the DeltananT genetic background, we performed a direct in vivo comparison of the transport properties of the STM1128 protein with those of sialic acid transporters of the major facilitator superfamily and tripartite ATP-independent periplasmic families, E. coli NanT and Haemophilus influenzae SiaPQM, respectively. This revealed that both STM1128 and SiaPQM are sodium-dependent and, unlike SiaPQM, both STM1128 and NanT are reversible secondary carriers, demonstrating qualitative functional differences in the properties of sialic acid transporters used by bacteria that colonize humans.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Organic Anion Transporters/metabolism , Salmonella typhimurium/metabolism , Symporters/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Haemophilus influenzae , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Organic Anion Transporters/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Symporters/genetics
13.
Proc Natl Acad Sci U S A ; 106(6): 1778-83, 2009 Feb 10.
Article in English | MEDLINE | ID: mdl-19179287

ABSTRACT

Substrate-binding protein-dependent secondary transporters are widespread in prokaryotes and are represented most frequently by members of the tripartite ATP-independent periplasmic (TRAP) transporter family. Here, we report the membrane reconstitution of a TRAP transporter, the sialic acid-specific SiaPQM system from Haemophilus influenzae, and elucidate its mechanism of energy coupling. Uptake of sialic acid via membrane-reconstituted SiaQM depends on the presence of the sialic acid-binding protein, SiaP, and is driven by the electrochemical sodium gradient. The interaction between SiaP and SiaQM is specific as transport is not reconstituted using the orthologous sialic acid-binding protein VC1779. Importantly, the binding protein also confers directionality on the transporter, and reversal of sialic acid transport from import to export is only possible in the presence of an excess of unliganded SiaP.


Subject(s)
Haemophilus influenzae/chemistry , Membrane Transport Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , Organic Anion Transporters/metabolism , Symporters/metabolism , Biological Transport , Energy Metabolism , Protein Binding , Substrate Specificity , Viral Proteins/metabolism
14.
J Biol Chem ; 283(8): 4841-9, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18063573

ABSTRACT

The acquisition of host-derived sialic acid is an important virulence factor for some bacterial pathogens, but in vivo this sugar acid is sequestered in sialoconjugates as the alpha-anomer. In solution, however, sialic acid is present mainly as the beta-anomer, formed by a slow spontaneous mutarotation. We studied the Escherichia coli protein YjhT as a member of a family of uncharacterized proteins present in many sialic acid-utilizing pathogens. This protein is able to accelerate the equilibration of the alpha- and beta-anomers of the sialic acid N-acetylneuraminic acid, thus describing a novel sialic acid mutarotase activity. The structure of this periplasmic protein, solved to 1.5A resolution, reveals a dimeric 6-bladed unclosed beta-propeller, the first of a bacterial Kelch domain protein. Mutagenesis of conserved residues in YjhT demonstrated an important role for Glu-209 and Arg-215 in mutarotase activity. We also present data suggesting that the ability to utilize alpha-N-acetylneuraminic acid released from complex sialoconjugates in vivo provides a physiological advantage to bacteria containing YjhT.


Subject(s)
Carbohydrate Epimerases/chemistry , Escherichia coli K12/enzymology , Escherichia coli Proteins/chemistry , Virulence Factors/chemistry , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Catalysis , Crystallography, X-Ray , Escherichia coli K12/genetics , Escherichia coli K12/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/genetics , N-Acetylneuraminic Acid/metabolism , Protein Structure, Tertiary , Virulence Factors/genetics , Virulence Factors/metabolism
15.
Microbiology (Reading) ; 153(Pt 9): 2817-2822, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17768226

ABSTRACT

Sialic acid occupies the terminal position within glycan molecules on the surfaces of many vertebrate cells, where it functions in diverse cellular processes such as intercellular adhesion and cell signalling. Pathogenic bacteria have evolved to use this molecule beneficially in at least two different ways: they can coat themselves in sialic acid, providing resistance to components of the host's innate immune response, or they can use it as a nutrient. Sialic acid itself is either synthesized de novo by these bacteria or scavenged directly from the host. In this mini-review we will summarize recent findings relating to sialic acid transport, modification of sialic acid by O-acetylation, and the mechanisms of sialic acid-mediated complement resistance.


Subject(s)
Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacterial Infections/microbiology , N-Acetylneuraminic Acid/metabolism , Streptococcal Infections/microbiology , Streptococcus agalactiae/pathogenicity , Animals , Gram-Negative Bacteria/metabolism , Humans , N-Acetylneuraminic Acid/biosynthesis , Streptococcus agalactiae/metabolism
16.
Mol Membr Biol ; 24(2): 161-71, 2007.
Article in English | MEDLINE | ID: mdl-17453422

ABSTRACT

The ammonium transport (Amt) proteins are a highly conserved family of integral membrane proteins found in eubacteria, archaea, fungi and plants. Genetic, biochemical and bioinformatic analyses suggest that they have a common tertiary structure comprising eleven trans-membrane helices with an N-out, C-in topology. The cytoplasmic C-terminus is variable in length but includes a core region of some 22 residues with considerable sequence conservation. Previous studies have indicated that this C-terminus is not absolutely required for Amt activity but that mutations that alter C-terminal residues can have very marked effects. Using the Escherichia coli AmtB protein as a model system for Amt proteins, we have carried out an extensive site-directed mutagenesis study to investigate the possible role of this region of the protein. Our data indicate that nearly all mutations fall into two phenotypic classes that are best explained in terms of two distinct effects of the C-terminal region on AmtB activity. Residues within the C-terminus play a significant role in normal AmtB function and the C-terminal region might also mediate co-operativity between the three subunits of AmtB.


Subject(s)
Cation Transport Proteins/chemistry , Cation Transport Proteins/physiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/physiology , Quaternary Ammonium Compounds/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Conserved Sequence , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Models, Molecular , Mutation/physiology , Nucleotidyltransferases/metabolism , PII Nitrogen Regulatory Proteins/metabolism , Protein Binding , Protein Denaturation/genetics , Protein Structure, Tertiary/physiology
17.
J Biol Chem ; 281(31): 22212-22222, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16702222

ABSTRACT

Extracytoplasmic solute receptors (ESRs) are important components of solute uptake systems in bacteria, having been studied extensively as parts of ATP binding cassette transporters. Herein we report the first crystal structure of an ESR protein from a functionally characterized electrochemical ion gradient dependent secondary transporter. This protein, SiaP, forms part of a tripartite ATP-independent periplasmic transporter specific for sialic acid in Haemophilus influenzae. Surprisingly, the structure reveals an overall topology similar to ATP binding cassette ESR proteins, which is not apparent from the sequence, demonstrating that primary and secondary transporters can share a common structural component. The structure of SiaP in the presence of the sialic acid analogue 2,3-didehydro-2-deoxy-N-acetylneuraminic acid reveals the ligand bound in a deep cavity with its carboxylate group forming a salt bridge with a highly conserved Arg residue. Sialic acid binding, which obeys simple bimolecular association kinetics as determined by stopped-flow fluorescence spectroscopy, is accompanied by domain closure about a hinge region and the kinking of an alpha-helix hinge component. The structure provides insight into the evolution, mechanism, and substrate specificity of ESR-dependent secondary transporters that are widespread in prokaryotes.


Subject(s)
Haemophilus influenzae/chemistry , Membrane Transport Proteins/chemistry , N-Acetylneuraminic Acid/chemistry , Organic Anion Transporters/chemistry , Symporters/chemistry , Virulence Factors/chemistry , ATP-Binding Cassette Transporters/chemistry , Amino Acid Sequence , Binding Sites , Crystallization , Crystallography, X-Ray , Kinetics , N-Acetylneuraminic Acid/analogs & derivatives , Protein Conformation , Sequence Alignment , Substrate Specificity
18.
Mol Microbiol ; 58(4): 1173-85, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16262798

ABSTRACT

Sialylation of the lipopolysaccharide (LPS) is an important mechanism used by the human pathogen Haemophilus influenzae to evade the innate immune response of the host. We have demonstrated that N-acetylneuraminic acid (Neu5Ac or sialic acid) uptake in H. influenzae is essential for the subsequent modification of the LPS and that this uptake is mediated through a single transport system which is a member of the tripartite ATP-independent periplasmic (TRAP) transporter family. Disruption of either the siaP (HI0146) or siaQM (HI0147) genes, that encode the two subunits of this transporter, results in a complete loss of uptake of [14C]-Neu5Ac. Mutant strains lack sialylated glycoforms in their LPS and are more sensitive to killing by human serum than the parent strain. The SiaP protein has been purified and demonstrated to bind a stoichiometric amount of Neu5Ac by electrospray mass spectrometry. This binding was of high affinity with a Kd of approximately 0.1 microM as determined by protein fluorescence. The inactivation of the SiaPQM TRAP transporter also results in decreased growth of H. influenzae in a chemically defined medium containing Neu5Ac, supporting an additional nutritional role of sialic acid in H. influenzae physiology.


Subject(s)
Haemophilus influenzae/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/metabolism , N-Acetylneuraminic Acid/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Biological Transport , Blood Bactericidal Activity , Gene Deletion , Gene Order , Genes, Bacterial , Haemophilus influenzae/genetics , Haemophilus influenzae/physiology , Lipopolysaccharides/chemistry , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/isolation & purification , Mutagenesis, Insertional , Protein Binding , Protein Subunits/genetics , Spectrometry, Mass, Electrospray Ionization , Synteny
19.
Plant Physiol ; 136(1): 2875-86, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15347797

ABSTRACT

Recent experiments indicate that nitric oxide (NO) plays a pivotal role in disease resistance and several other physiological processes in plants. However, most of the current information about the function of NO in plants is based on pharmacological studies, and additional approaches are therefore required to ascertain the role of NO as an important signaling molecule in plants. We have expressed a bacterial nitric oxide dioxygenase (NOD) in Arabidopsis plants and/or avirulent Pseudomonas syringae pv tomato to study incompatible plant-pathogen interactions impaired in NO signaling. NOD expression in transgenic Arabidopsis resulted in decreased NO levels in planta and attenuated a pathogen-induced NO burst. Moreover, NOD expression in plant cells had very similar effects on plant defenses compared to NOD expression in avirulent Pseudomonas. The defense responses most affected by NO reduction during the incompatible interaction were decreased H(2)O(2) levels during the oxidative burst and a blockage of Phe ammonia lyase expression, the key enzyme in the general phenylpropanoid pathway. Expression of the NOD furthermore blocked UV light-induced Phe ammonia lyase and chalcone synthase gene expression, indicating a general signaling function of NO in the activation of the phenylpropanoid pathway. NO possibly functions in incompatible plant-pathogen interactions by inhibiting the plant antioxidative machinery, and thereby ensuring locally prolonged H(2)O(2) levels. Additionally, albeit to a lesser extent, we observed decreases in salicylic acid production, a diminished development of hypersensitive cell death, and a delay in pathogenesis-related protein 1 expression during these NO-deficient plant-pathogen interactions. Therefore, this genetic approach confirms that NO is an important regulatory component in the signaling network of plant defense responses.


Subject(s)
Arabidopsis/genetics , Arabidopsis/metabolism , Nitric Oxide/metabolism , Arabidopsis/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cell Death , DNA, Bacterial/genetics , DNA, Recombinant/genetics , Dickeya chrysanthemi/enzymology , Dickeya chrysanthemi/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Expression , Genes, Bacterial , Genes, Plant , Hemeproteins/genetics , Hemeproteins/metabolism , Hydrogen Peroxide/metabolism , Molecular Sequence Data , Oxygenases/genetics , Oxygenases/metabolism , Plants, Genetically Modified , Pseudomonas syringae/pathogenicity , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Respiratory Burst , Salicylic Acid/metabolism , Signal Transduction/genetics
20.
J Biol Chem ; 279(10): 8530-8, 2004 Mar 05.
Article in English | MEDLINE | ID: mdl-14668330

ABSTRACT

The Amt proteins are high affinity ammonium transporters that are conserved in all domains of life. In bacteria and archaea the Amt structural genes (amtB) are invariably linked to glnK, which encodes a member of the P(II) signal transduction protein family, proteins that regulate many facets of nitrogen metabolism. We have now shown that Escherichia coli AmtB is inactivated by formation of a membrane-bound complex with GlnK. Complex formation is reversible and occurs within seconds in response to micromolar changes in the extracellular ammonium concentration. Regulation is mediated by the uridylylation/deuridylylation of GlnK in direct response to fluctuations in the intracellular glutamine pool. Furthermore under physiological conditions AmtB activity is required for GlnK deuridylylation. Hence the transporter is an integral part of the signal transduction cascade, and AmtB can be formally considered to act as an ammonium sensor. This system provides an exquisitely sensitive mechanism to control ammonium flux into the cell, and the conservation of glnK linkage to amtB suggests that this regulatory mechanism may occur throughout prokaryotes.


Subject(s)
Cation Transport Proteins/metabolism , Escherichia coli Proteins/metabolism , Protein Kinases/metabolism , Quaternary Ammonium Compounds/metabolism , Transcription Factors/metabolism , Biological Transport , Escherichia coli/metabolism , Models, Biological , Nucleotidyltransferases , PII Nitrogen Regulatory Proteins , Protein Binding , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...