Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38081970

ABSTRACT

StayGold is an exceptionally bright and stable fluorescent protein that is highly resistant to photobleaching. Despite favorable fluorescence properties, use of StayGold as a fluorescent tag is limited because it forms a natural dimer. Here we report the 1.6 Å structure of StayGold and generate a derivative, mStayGold, that retains the brightness and photostability of the original protein while being fully monomeric.

2.
Proc Natl Acad Sci U S A ; 120(24): e2302580120, 2023 06 13.
Article in English | MEDLINE | ID: mdl-37276423

ABSTRACT

AmiA and AmiB are peptidoglycan-hydrolyzing enzymes from Escherichia coli that are required to break the peptidoglycan layer during bacterial cell division and maintain integrity of the cell envelope. In vivo, the activity of AmiA and AmiB is tightly controlled through their interactions with the membrane-bound FtsEX-EnvC complex. Activation of AmiA and AmiB requires access to a groove in the amidase-activating LytM domain of EnvC which is gated by ATP-driven conformational changes in FtsEX-EnvC complex. Here, we present a high-resolution structure of the isolated AmiA protein, confirming that it is autoinhibited in the same manner as AmiB and AmiC, and a complex of the AmiB enzymatic domain bound to the activating EnvC LytM domain. In isolation, the active site of AmiA is blocked by an autoinhibitory helix that binds directly to the catalytic zinc and fills the volume expected to accommodate peptidoglycan binding. In the complex, binding of the EnvC LytM domain induces a conformational change that displaces the amidase autoinhibitory helix and reorganizes the active site for activity. Our structures, together with complementary mutagenesis work, defines the conformational changes required to activate AmiA and/or AmiB through their interaction with their cognate activator EnvC.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/metabolism , Peptidoglycan/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Escherichia coli/metabolism , Amidohydrolases/metabolism , Bacterial Proteins/metabolism
3.
Proc Natl Acad Sci U S A ; 120(8): e2215237120, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36787358

ABSTRACT

Acinetobacter baumannii is a gram-negative bacterial pathogen that causes challenging nosocomial infections. ß-lactam targeting of penicillin-binding protein (PBP)-mediated cell wall peptidoglycan (PG) formation is a well-established antimicrobial strategy. Exposure to carbapenems or zinc (Zn)-deprived growth conditions leads to a rod-to-sphere morphological transition in A. baumannii, an effect resembling that caused by deficiency in the RodA-PBP2 PG synthesis complex required for cell wall elongation. While it is recognized that carbapenems preferentially acylate PBP2 in A. baumannii and therefore block the transpeptidase function of the RodA-PBP2 system, the molecular details underpinning cell wall elongation inhibition upon Zn starvation remain undefined. Here, we report the X-ray crystal structure of A. baumannii PBP2, revealing an unexpected Zn coordination site in the transpeptidase domain required for protein stability. Mutations in the Zn-binding site of PBP2 cause a loss of bacterial rod shape and increase susceptibility to ß-lactams, therefore providing a direct rationale for cell wall shape maintenance and Zn homeostasis in A. baumannii. Furthermore, the Zn-coordinating residues are conserved in various ß- and γ-proteobacterial PBP2 orthologs, consistent with a widespread Zn-binding requirement for function that has been previously unknown. Due to the emergence of resistance to virtually all marketed antibiotic classes, alternative or complementary antimicrobial strategies need to be explored. These findings offer a perspective for dual inhibition of Zn-dependent PG synthases and metallo-ß-lactamases by metal chelating agents, considered the most sought-after adjuvants to restore ß-lactam potency against gram-negative bacteria.


Subject(s)
Acinetobacter baumannii , Peptidyl Transferases , Acinetobacter baumannii/metabolism , Peptidyl Transferases/metabolism , Zinc/metabolism , Cell Shape , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Penicillin-Binding Proteins/metabolism , beta-Lactams/pharmacology , Carbapenems/pharmacology , Chelating Agents/pharmacology , Binding Sites , Bacterial Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 117(45): 28355-28365, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33097670

ABSTRACT

FtsEX is a bacterial ABC transporter that regulates the activity of periplasmic peptidoglycan amidases via its interaction with the murein hydrolase activator, EnvC. In Escherichia coli, FtsEX is required to separate daughter cells after cell division and for viability in low-osmolarity media. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for amidase activation, but the process itself is poorly understood. Here we present the 2.1 Å structure of the FtsX periplasmic domain in complex with its periplasmic partner, EnvC. The EnvC-FtsX periplasmic domain complex has a 1-to-2 stoichiometry with two distinct FtsX-binding sites located within an antiparallel coiled coil domain of EnvC. Residues involved in amidase activation map to a previously identified groove in the EnvC LytM domain that is here found to be occluded by a "restraining arm" suggesting a self-inhibition mechanism. Mutational analysis, combined with bacterial two-hybrid screens and in vivo functional assays, verifies the FtsEX residues required for EnvC binding and experimentally test a proposed mechanism for amidase activation. We also define a predicted link between FtsEX and integrity of the outer membrane. Both the ATPase activity of FtsEX and its periplasmic interaction with EnvC are required for resistance to membrane-attacking antibiotics and detergents to which E. coli would usually be considered intrinsically resistant. These structural and functional data provide compelling mechanistic insight into FtsEX-mediated regulation of EnvC and its downstream control of periplasmic peptidoglycan amidases.


Subject(s)
Bacteria/metabolism , Bacterial Proteins/chemistry , Cell Cycle Proteins/chemistry , Cell Division/physiology , Endopeptidases/chemistry , Periplasm/metabolism , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Crystallography, X-Ray , DNA Mutational Analysis , Endopeptidases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins , Models, Molecular , Mutation , N-Acetylmuramoyl-L-alanine Amidase/genetics , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Periplasm/chemistry , Protein Binding , Protein Conformation , Protein Domains , Protein Interaction Domains and Motifs
5.
Front Microbiol ; 9: 2318, 2018.
Article in English | MEDLINE | ID: mdl-30310383

ABSTRACT

[This corrects the article DOI: 10.3389/fmicb.2018.00950.].

6.
Proc Natl Acad Sci U S A ; 115(31): E7389-E7397, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30012603

ABSTRACT

In Gram-negative bacteria, outer-membrane lipoproteins are essential for maintaining cellular integrity, transporting nutrients, establishing infections, and promoting the formation of biofilms. The LolCDE ABC transporter, LolA chaperone, and LolB outer-membrane receptor form an essential system for transporting newly matured lipoproteins from the outer leaflet of the cytoplasmic membrane to the innermost leaflet of the outer membrane. Here, we present a crystal structure of LolA in complex with the periplasmic domain of LolC. The structure reveals how a solvent-exposed ß-hairpin loop (termed the "Hook") and trio of surface residues (the "Pad") of LolC are essential for recruiting LolA from the periplasm and priming it to receive lipoproteins. Experiments with purified LolCDE complex demonstrate that association with LolA is independent of nucleotide binding and hydrolysis, and homology models based on the MacB ABC transporter predict that LolA recruitment takes place at a periplasmic site located at least 50 Å from the inner membrane. Implications for the mechanism of lipoprotein extraction and transfer are discussed. The LolA-LolC structure provides atomic details on a key protein interaction within the Lol pathway and constitutes a vital step toward the complete molecular understanding of this important system.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Escherichia coli Proteins/metabolism , Periplasm/metabolism , Periplasmic Binding Proteins/metabolism , ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/chemistry , Escherichia coli Proteins/chemistry , Hydrolysis , Models, Molecular , Periplasmic Binding Proteins/chemistry , Protein Interaction Mapping , Protein Transport
7.
Front Microbiol ; 9: 950, 2018.
Article in English | MEDLINE | ID: mdl-29892271

ABSTRACT

The MacB ABC transporter forms a tripartite efflux pump with the MacA adaptor protein and TolC outer membrane exit duct to expel antibiotics and export virulence factors from Gram-negative bacteria. Here, we review recent structural and functional data on MacB and its homologs. MacB has a fold that is distinct from other structurally characterized ABC transporters and uses a unique molecular mechanism termed mechanotransmission. Unlike other bacterial ABC transporters, MacB does not transport substrates across the inner membrane in which it is based, but instead couples cytoplasmic ATP hydrolysis with transmembrane conformational changes that are used to perform work in the extra-cytoplasmic space. In the MacAB-TolC tripartite pump, mechanotransmission drives efflux of antibiotics and export of a protein toxin from the periplasmic space via the TolC exit duct. Homologous tripartite systems from pathogenic bacteria similarly export protein-like signaling molecules, virulence factors and siderophores. In addition, many MacB-like ABC transporters do not form tripartite pumps, but instead operate in diverse cellular processes including antibiotic sensing, cell division and lipoprotein trafficking.

8.
Proc Natl Acad Sci U S A ; 114(47): 12572-12577, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109272

ABSTRACT

MacB is an ABC transporter that collaborates with the MacA adaptor protein and TolC exit duct to drive efflux of antibiotics and enterotoxin STII out of the bacterial cell. Here we present the structure of ATP-bound MacB and reveal precise molecular details of its mechanism. The MacB transmembrane domain lacks a central cavity through which substrates could be passed, but instead conveys conformational changes from one side of the membrane to the other, a process we term mechanotransmission. Comparison of ATP-bound and nucleotide-free states reveals how reversible dimerization of the nucleotide binding domains drives opening and closing of the MacB periplasmic domains via concerted movements of the second transmembrane segment and major coupling helix. We propose that the assembled tripartite pump acts as a molecular bellows to propel substrates through the TolC exit duct, driven by MacB mechanotransmission. Homologs of MacB that do not form tripartite pumps, but share structural features underpinning mechanotransmission, include the LolCDE lipoprotein trafficking complex and FtsEX cell division signaling protein. The MacB architecture serves as the blueprint for understanding the structure and mechanism of an entire ABC transporter superfamily and the many diverse functions it supports.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Adenosine Triphosphate/chemistry , Aggregatibacter actinomycetemcomitans/metabolism , Bacterial Outer Membrane Proteins/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Aggregatibacter actinomycetemcomitans/genetics , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Biological Transport , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
9.
Biochemistry ; 55(23): 3198-203, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27185533

ABSTRACT

Membrane-intrinsic nitric oxide reductases (NORs) are key components of bacterial denitrification pathways with a close evolutionary relationship to the cytochrome oxidase (COX) complex found in aerobic respiratory chains. A key distinction between COX and NOR is the identity of the metal directly opposite heme b3 within the active site. In NOR, this metal is iron (FeB), whereas in COX, it is copper (CuB). The purified NOR of Roseobacter denitrificans contains copper and has modest oxidase activity, raising the possibility that a COX-like active site might have independently arisen within the context of a NOR-like protein scaffold. Here we present the crystal structure of the Roseobacter denitrificans NorBC complex and anomalous scattering experiments probing the identity of each metal center. Our results refute the hypothesis that copper occupies the active site and instead reveal a new metal center in the small subunit not seen in any other NOR or COX.


Subject(s)
Cell Membrane/metabolism , Copper/metabolism , Heme/metabolism , Iron/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Roseobacter/enzymology , Catalytic Domain , Crystallization , Crystallography, X-Ray , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/metabolism , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 112(23): E3058-66, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26016525

ABSTRACT

Secreted pore-forming toxins of pathogenic Gram-negative bacteria such as Escherichia coli hemolysin (HlyA) insert into host-cell membranes to subvert signal transduction and induce apoptosis and cell lysis. Unusually, these toxins are synthesized in an inactive form that requires posttranslational activation in the bacterial cytosol. We have previously shown that the activation mechanism is an acylation event directed by a specialized acyl-transferase that uses acyl carrier protein (ACP) to covalently link fatty acids, via an amide bond, to specific internal lysine residues of the protoxin. We now reveal the 2.15-Å resolution X-ray structure of the 172-aa ApxC, a toxin-activating acyl-transferase (TAAT) from pathogenic Actinobacillus pleuropneumoniae. This determination shows that bacterial TAATs are a structurally homologous family that, despite indiscernible sequence similarity, form a distinct branch of the Gcn5-like N-acetyl transferase (GNAT) superfamily of enzymes that typically use acyl-CoA to modify diverse bacterial, archaeal, and eukaryotic substrates. A combination of structural analysis, small angle X-ray scattering, mutagenesis, and cross-linking defined the solution state of TAATs, with intermonomer interactions mediated by an N-terminal α-helix. Superposition of ApxC with substrate-bound GNATs, and assay of toxin activation and binding of acyl-ACP and protoxin peptide substrates by mutated ApxC variants, indicates the enzyme active site to be a deep surface groove.


Subject(s)
Actinobacillus pleuropneumoniae/enzymology , Acyltransferases/chemistry , Acyltransferases/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Processing, Post-Translational , Sequence Homology, Amino Acid
11.
FEBS Lett ; 588(17): 3147-53, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-24996185

ABSTRACT

Periplasmic adaptor proteins are key components of bacterial tripartite efflux pumps. The 2.85 Å resolution structure of an MFS (major facilitator superfamily) pump adaptor, Aquifex aeolicus EmrA, shows linearly arranged α-helical coiled-coil, lipoyl, and ß-barrel domains, but lacks the fourth membrane-proximal domain shown in other pumps to interact with the inner membrane transporter. The adaptor α-hairpin, which binds outer membrane TolC, is exceptionally long at 127 Å, and the ß-barrel contains a conserved disordered loop. The structure extends the view of adaptors as flexible, modular components that mediate diverse pump assembly, and suggests that in MFS tripartite pumps a hexamer of adaptors could provide a periplasmic seal.


Subject(s)
Bacterial Proteins/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Aquifoliaceae , Bacterial Proteins/metabolism , Conserved Sequence , Crystallography, X-Ray , Ligands , Membrane Transport Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary
12.
Proteins ; 82(3): 517-27, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123467

ABSTRACT

Streptococcus pyogenes and other Gram-positive bacterial pathogens present long macromolecular filaments known as pili on their surface that mediate adhesion and colonization. These pili are covalent polymers, assembled by sortases. Typically, they comprise a putative adhesin at their tip, a backbone subunit present in multiple copies and a basal subunit that is covalently anchored to the peptidoglycan layer of the cell surface. The crystal structures of pilin subunits revealed the presence of unusual covalent linkages in these proteins, including intramolecular isopeptide and internal thioester bonds. The intramolecular isopeptide bonds in backbone pilins are important for protein stability. Here, using both the wild-type protein and a set of mutants, we assessed the proteolytic and thermal stability of the S. pyogenes pilus tip adhesin Spy0125, in the presence and absence of its intramolecular isopeptide and internal thioester bonds. We also determined a crystal structure of the internal thioester bond variant Spy0125(Cys426Ala). We find that mutations in the intramolecular isopeptide bonds compromise the stability of Spy0125. Using limited proteolysis and thermal denaturation assays, we could separate the contribution of each intramolecular isopeptide bond to Spy0125 stability. In contrast, mutation in the internal thioester bond had a lesser effect on protein stability and the crystal structure is essentially identical to wild type. This work suggests that the internal thioester in Spy0125, although having a minor contributory role, is not required for protein stability and must have a different primary function, most likely mediating a covalent interaction with host cell ligands.


Subject(s)
Adhesins, Bacterial/chemistry , Fimbriae, Bacterial/chemistry , Streptococcus pyogenes/genetics , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Circular Dichroism , Crystallography, X-Ray , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Models, Molecular , Protein Stability , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/enzymology
13.
FEBS Lett ; 587(18): 2984-8, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23851070

ABSTRACT

Periplasmic adaptor proteins are essential components of bacterial tripartite multidrug efflux pumps. Here we report the 2.35 Å resolution crystal structure of the BesA adaptor from the spirochete Borrelia burgdorferi solved using selenomethionine derivatized protein. BesA shows the archetypal linear, flexible, multi-domain architecture evident among proteobacteria and retains the lipoyl, ß-barrel and membrane-proximal domains that interact with the periplasmic domains of the inner membrane transporter. However, it lacks the α-hairpin domain shown to establish extensive coiled-coil interactions with the periplasmic entrance helices of the outer membrane-anchored TolC exit duct. This has implications for the modelling of assembled tripartite efflux pumps.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Borrelia burgdorferi/chemistry , Multidrug Resistance-Associated Proteins/chemistry , Periplasm/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Borrelia burgdorferi/genetics , Borrelia burgdorferi/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Gene Expression , Molecular Dynamics Simulation , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Selenomethionine/chemistry , Selenomethionine/metabolism
14.
Proc Natl Acad Sci U S A ; 109(27): E1830-8, 2012 Jul 03.
Article in English | MEDLINE | ID: mdl-22691497

ABSTRACT

The cycle inhibiting factors (Cifs) are a family of translocated effector proteins, found in diverse pathogenic bacteria, that interfere with the host cell cycle by catalyzing the deamidation of a specific glutamine residue (Gln40) in NEDD8 and the related protein ubiquitin. This modification prevents recycling of neddylated cullin-RING ligases, leading to stabilization of various cullin-RING ligase targets, and also prevents polyubiquitin chain formation. Here, we report the crystal structures of two Cif/NEDD8 complexes, revealing a conserved molecular interface that defines enzyme/substrate recognition. Mutation of residues forming the interface suggests that shape complementarity, rather than specific individual interactions, is a critical feature for complex formation. We show that Cifs from diverse bacteria bind NEDD8 in vitro and conclude that they will all interact with their substrates in the same way. The "occluding loop" in Cif gates access to Gln40 by forcing a conformational change in the C terminus of NEDD8. We used native PAGE to follow the activity of Cif from the human pathogen Yersinia pseudotuberculosis and selected variants, and the position of Gln40 in the active site has allowed us to propose a catalytic mechanism for these enzymes.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Photorhabdus/enzymology , Ubiquitins/chemistry , Ubiquitins/metabolism , Yersinia pseudotuberculosis/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Crystallization , Glutamine/genetics , HeLa Cells , Host-Parasite Interactions/physiology , Humans , Molecular Sequence Data , Mutagenesis/physiology , NEDD8 Protein , Oncogene Protein p21(ras)/metabolism , Photorhabdus/genetics , Polyubiquitin/metabolism , Protein Binding/physiology , Protein Structure, Tertiary , Ubiquitins/genetics , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism , Yersinia pseudotuberculosis/genetics , Yersinia pseudotuberculosis Infections/metabolism , Yersinia pseudotuberculosis Infections/microbiology
15.
J Biol Chem ; 285(44): 33858-66, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20729215

ABSTRACT

Many bacterial pathogens present adhesins at the tips of long macromolecular filaments known as pili that are often important virulence determinants. Very little is known about how pili presented by Gram-positive pathogens mediate host cell binding. The crystal structure of a pilus adhesin from the important human pathogen Streptococcus pyogenes reveals an internal thioester bond formed between the side chains of a cysteine and a glutamine residue. The presence of the thioester was verified using UV-visible spectroscopy and mass spectrometry. This unusual bond has only previously been observed in thioester domains of complement and complement-like proteins where it is used to form covalent attachment to target molecules. The structure also reveals two intramolecular isopeptide bonds, one of these formed through a Lys/Asp residue pair, which are strategically positioned to confer protein stability. Removal of the internal thioester by allele-replacement mutagenesis in S. pyogenes severely compromises bacterial adhesion to model host cells. Although current paradigms of bacterial/host cell interaction envisage strong non-covalent interactions, the present study suggests cell adhesion could also involve covalent bonds.


Subject(s)
Adhesins, Bacterial/chemistry , Esters/chemistry , Fimbriae, Bacterial/metabolism , Streptococcus pyogenes/metabolism , Alleles , Aspartic Acid/chemistry , Cell Adhesion , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Lysine/chemistry , Mutagenesis , Peptides/chemistry , Protein Binding , Spectrophotometry, Ultraviolet/methods , Streptococcus pneumoniae/metabolism
16.
Biochim Biophys Acta ; 1800(8): 732-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20388533

ABSTRACT

BACKGROUND: To satisfy their requirement for iron while at the same time countering the toxicity of this highly reactive metal ion, prokaryotes have evolved proteins belonging to two distinct sub-families of the ferritin family: the bacterioferritins (BFRs) and the bacterial ferritins (Ftns). Recently, Ftn homologues have also been identified and characterised in archaeon species. All of these prokaryotic ferritins function by solubilising and storing large amounts of iron in the form of a safe but bio-available mineral. SCOPE OF REVIEW: The mechanism(s) by which the iron mineral is formed by these proteins is the subject of much current interest. Here we review the available information on these proteins, with particular emphasis on significant advances resulting from recent structural, spectroscopic and kinetic studies. MAJOR CONCLUSIONS: Current understanding indicates that at least two distinct mechanisms are in operation in prokaryotic ferritins. In one, the ferroxidase centre acts as a true catalytic centre in driving Fe(2+) oxidation in the cavity; in the other, the centre acts as a gated iron pore by oxidising Fe(2+) and transferring the resulting Fe(3+) into the central cavity. GENERAL SIGNIFICANCE: The prokaryotic ferritins exhibit a wide variation in mechanisms of iron core mineralisation. The basis of these differences lies, at least in part, in structural differences at and around the catalytic centre. However, it appears that more subtle differences must also be important in controlling the iron chemistry of these remarkable proteins.


Subject(s)
Ferritins/chemistry , Ferritins/metabolism , Iron/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Ferric Compounds/chemistry , Ferric Compounds/metabolism , Iron/chemistry , Minerals/chemistry , Minerals/metabolism , Models, Biological , Models, Molecular , Molecular Sequence Data , Prokaryotic Cells/metabolism , Sequence Homology, Amino Acid
17.
Biochemistry ; 48(38): 9031-9, 2009 Sep 29.
Article in English | MEDLINE | ID: mdl-19705876

ABSTRACT

Ferritins solubilize and detoxify the essential metal iron through formation of a ferric mineral within the protein's central cavity. Key to this activity is an intrasubunit catalytic dinuclear iron center called the ferroxidase center. Here we show that the fluorescence intensity of Escherichia coli bacterioferritin (BFR), due to the presence of two tryptophan residues (Trp35 and Trp133) in each of the 24 subunits, is highly sensitive to the iron status of the ferroxidase center and is quenched to different extents by Fe2+ and Fe3+. Recovery of the quench following oxidation of Fe2+ to Fe3+ at the ferroxidase center was not observed, indicating that the di-Fe3+ form of the center is stable. Studies of the single-tryptophan variants W35F and W133F showed that Trp133, which lies approximately 10 A from the ferroxidase center, is primarily responsible for the observed fluorescence sensitivity to iron, while studies of a stable E. coli BFR subunit dimer demonstrated that the observed quench properties are principally derived from the interaction of iron with tryptophan residues within the subunit dimer. A double-tryptophan variant (W35F/W133F) was found to exhibit fluorescence from the seven tyrosine residues present in each subunit, which was also sensitive to the iron status of the ferroxidase center. Finally, we demonstrate using Zn2+, a potent competitive inhibitor of Fe2+ binding and oxidation, that the fluorescence response can be used to monitor the loss of iron from the ferroxidase center.


Subject(s)
Bacterial Proteins/chemistry , Cytochrome b Group/chemistry , Escherichia coli Proteins/chemistry , Ferritins/chemistry , Amino Acid Substitution , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , DNA Primers/genetics , Dimerization , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ferritins/genetics , Ferritins/metabolism , Iron/chemistry , Iron/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Tryptophan/chemistry
18.
J Biol Chem ; 284(35): 23719-33, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19535335

ABSTRACT

BdbD is a thiol:disulfide oxidoreductase (TDOR) from Bacillus subtilis that functions to introduce disulfide bonds in substrate proteins/peptides on the outside of the cytoplasmic membrane and, as such, plays a key role in disulfide bond management. Here we demonstrate that the protein is membrane-associated in B. subtilis and present the crystal structure of the soluble part of the protein lacking its membrane anchor. This reveals that BdbD is similar in structure to Escherichia coli DsbA, with a thioredoxin-like domain with an inserted helical domain. A major difference, however, is the presence in BdbD of a metal site, fully occupied by Ca(2+), at an inter-domain position some 14 A away from the CXXC active site. The midpoint reduction potential of soluble BdbD was determined as -75 mV versus normal hydrogen electrode, and the active site N-terminal cysteine thiol was shown to have a low pK(a), consistent with BdbD being an oxidizing TDOR. Equilibrium unfolding studies revealed that the oxidizing power of the protein is based on the instability introduced by the disulfide bond in the oxidized form. The crystal structure of Ca(2+)-depleted BdbD showed that the protein remained folded, with only minor conformational changes. However, the reduced form of Ca(2+)-depleted BdbD was significantly less stable than reduced Ca(2+)-containing protein, and the midpoint reduction potential was shifted by approximately -20 mV, suggesting that Ca(2+) functions to boost the oxidizing power of the protein. Finally, we demonstrate that electron exchange does not occur between BdbD and B. subtilis ResA, a low potential extra-cytoplasmic TDOR.


Subject(s)
Bacterial Proteins/chemistry , Calcium/chemistry , Protein Disulfide Reductase (Glutathione)/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Calcium/metabolism , Crystallization , Molecular Conformation , Molecular Sequence Data , Oxidation-Reduction , Protein Binding , Protein Disulfide Reductase (Glutathione)/genetics , Protein Disulfide Reductase (Glutathione)/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid
19.
PLoS One ; 4(5): e5582, 2009.
Article in English | MEDLINE | ID: mdl-19440549

ABSTRACT

A pre-requisite for bacterial pathogenesis is the successful interaction of a pathogen with a host. One mechanism used by a broad range of Gram negative bacterial pathogens is to deliver effector proteins directly into host cells through a dedicated type III secretion system where they modulate host cell function. The cycle inhibiting factor (Cif) family of effector proteins, identified in a growing number of pathogens that harbour functional type III secretion systems and have a wide host range, arrest the eukaryotic cell cycle. Here, the crystal structures of Cifs from the insect pathogen/nematode symbiont Photorhabdus luminescens (a gamma-proteobacterium) and human pathogen Burkholderia pseudomallei (a beta-proteobacterium) are presented. Both of these proteins adopt an overall fold similar to the papain sub-family of cysteine proteases, as originally identified in the structure of a truncated form of Cif from Enteropathogenic E. coli (EPEC), despite sharing only limited sequence identity. The structure of an N-terminal region, referred to here as the 'tail-domain' (absent in the EPEC Cif structure), suggests a surface likely to be involved in host-cell substrate recognition. The conformation of the Cys-His-Gln catalytic triad is retained, and the essential cysteine is exposed to solvent and addressable by small molecule reagents. These structures and biochemical work contribute to the rapidly expanding literature on Cifs, and direct further studies to better understand the molecular details of the activity of these proteins.


Subject(s)
Bacterial Proteins/chemistry , Burkholderia pseudomallei/chemistry , Photorhabdus/chemistry , Amino Acid Sequence , Burkholderia pseudomallei/metabolism , Catalytic Domain , Chromatography, Gel , Crystallography, X-Ray , Molecular Sequence Data , Photorhabdus/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary
20.
J Am Chem Soc ; 131(19): 6808-13, 2009 May 20.
Article in English | MEDLINE | ID: mdl-19391621

ABSTRACT

Ferritin proteins function to detoxify, solubilize and store cellular iron by directing the synthesis of a ferric oxyhydroxide mineral solubilized within the protein's central cavity. Here, through the application of X-ray crystallographic and kinetic methods, we report significant new insight into the mechanism of mineralization in a bacterioferritin (BFR). The structures of nonheme iron-free and di-Fe(2+) forms of BFR showed that the intrasubunit catalytic center, known as the ferroxidase center, is preformed, ready to accept Fe(2+) ions with little or no reorganization. Oxidation of the di-Fe(2+) center resulted in a di-Fe(3+) center, with bridging electron density consistent with a mu-oxo or hydro bridged species. The mu-oxo bridged di-Fe(3+) center appears to be stable, and there is no evidence that Fe(3+)species are transferred into the core from the ferroxidase center. Most significantly, the data also revealed a novel Fe(2+) binding site on the inner surface of the protein, lying approximately 10 A directly below the ferroxidase center, coordinated by only two residues, His46 and Asp50. Kinetic studies of variants containing substitutions of these residues showed that the site is functionally important. In combination, the data support a model in which the ferroxidase center functions as a true catalytic cofactor, rather than as a pore for the transfer of iron into the central cavity, as found for eukaryotic ferritins. The inner surface iron site appears to be important for the transfer of electrons, derived from Fe(2+) oxidation in the cavity, to the ferroxidase center. Bacterioferritin may represent an evolutionary link between ferritins and class II di-iron proteins not involved in iron metabolism.


Subject(s)
Bacterial Proteins/chemistry , Ceruloplasmin/chemistry , Cytochrome b Group/chemistry , Ferritins/chemistry , Iron/chemistry , Minerals/chemistry , Crystallography, X-Ray , Evolution, Molecular , Oxidation-Reduction , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...