Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
J Membr Biol ; 189(2): 131-41, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12235488

ABSTRACT

Changes in amino-acid sequence of the unique pore-forming protein of H. influenzae (OmpP2; porin) have been associated with increased antimicrobial resistance in H. influenzae strains isolated from cystic fibrosis patients. From patients who were subjected to long-term antimicrobial therapy, H. influenzae strains 67d and 69a (patient 27) and strains 77a and 77f (patient 30) were isolated. Strains 67d and 77a were previously shown to have elevated values for minimal inhibitory concentrations of antibiotics compared to strains 69a and 77f. Porins were extracted from all four H. influenzae strains by detergent treatment and purified to homogeneity by ion exchange chromatography. By reconstitution of the clinical Hi porins into planar lipid bilayers, single-channel conductance, ionic selectivity, and voltage-gating characteristics were assessed. Porins 77a and 77f displayed similar single-channel conductance and ionic selectivity. Current-voltage relationships were determined for the different porins: porin 77f displayed substantial voltage gating at both positive and negative polarity; porin 77a gated at negative polarity only. Porins 67d and 69a showed substantial differences in their pore-forming properties: the single-channel conductance of porin 69a was significantly increased (1.05 nS) relative to porin 67d (0.73 nS). Porin 67d was twice as permeable to cations as porin 69a, and at both positive and negative polarities the extent of voltage gating was greater for porin 67d relative to porin 69a. Expression of the porins in an isogenic, porin-deleted H. influenzae background allowed for assessment of the contribution of each porin to the minimum inhibitory concentrations of various antimicrobial compounds. Porin 67d was found to have a decreased susceptibility to the antimicrobials novobiocin and streptomycin. This decreased susceptibility of porin 67d to novobiocin and streptomycin correlates with its decrease in single-channel conductance.


Subject(s)
Drug Resistance, Bacterial , Haemophilus influenzae/classification , Haemophilus influenzae/genetics , Ion Channels/physiology , Lipid Bilayers/chemistry , Porins/chemistry , Amino Acid Sequence , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Electric Conductivity , Escherichia coli/classification , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Haemophilus influenzae/drug effects , Haemophilus influenzae/metabolism , Humans , Ion Channel Gating/physiology , Lactams/classification , Lactams/pharmacology , Membrane Potentials , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis, Site-Directed , Porins/genetics , Porins/isolation & purification , Porins/metabolism , Proteolipids/chemistry , Reference Values , Sensitivity and Specificity , Species Specificity , Statistics as Topic
2.
Biochemistry ; 40(48): 14621-8, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11724575

ABSTRACT

Porin (341 amino acids; M(r) 37 782) of Haemophilus influenzae type b mediates exchange of solutes between the external environment and the periplasm of this Gram-negative bacterium. Positively charged residues in the extracellular loops have been shown to be involved in the voltage gating of this protein. To further elucidate our observations on the functional properties of this channel, we mutated seven lysines (Lys(48), Lys(161), Lys(165), Lys(170), Lys(248), Lys(250), and Lys(253)) to glutamic acid. The selected residues were previously shown to be accessible to chemical modification, and they map to three locations: loop 4 and loop 6, and within the barrel lumen. The seven mutant proteins were purified, and each was reconstituted into planar lipid bilayers to characterize its channel forming properties. The single substitution mutant porins displayed increased single channel conductances in 1 M KCl ranging between 134 and 178% of the single channel conductance for wild-type Hib porin. Six of the seven mutant porins also displayed altered current-voltage relationships when compared to wild-type Hib porin. Whereas Lys(170)Glu had activity similar to wild-type Hib porin, Lys(48)Glu, Lys(248)Glu, and Lys(253)Glu showed substantial voltage gating at both positive and negative polarities. Lys(161)Glu and Lys(250)Glu gated only at negative potentials, and Lys(165)Glu gated only at positive potentials. Rather than ascribing one specific loop in gating, our analyses of these mutant Hib porins suggest that voltage gating can be attributed to contributions from loops 4 and 6 and a residue within the barrel lumen.


Subject(s)
Haemophilus influenzae type b/genetics , Ion Channel Gating/physiology , Porins/genetics , Amino Acid Sequence , Amino Acid Substitution , Cell Membrane Permeability/physiology , DNA Primers/chemistry , Electric Conductivity , Haemophilus influenzae type b/chemistry , Lipid Bilayers/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Porins/chemistry , Protein Structure, Secondary
3.
Structure ; 9(8): 707-16, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11587645

ABSTRACT

BACKGROUND: FhuA, an integral membrane protein of Escherichia coli, actively transports ferrichrome and the structurally related antibiotic albomycin across the outer membrane. The transport is coupled to the proton motive force, which energizes FhuA through the inner-membrane protein TonB. FhuA also transports the semisynthetic rifamycin derivative CGP 4832, although the chemical structure of this antibiotic differs markedly from that of ferric hydroxamates. RESULTS: X-ray crystallography revealed that rifamycin CGP 4832 occupies the same ligand binding site as ferrichrome and albomycin, thus demonstrating a surprising lack of selectivity. However, the binding of rifamycin CGP 4832 is deviant from the complexes of FhuA with hydroxamate-type ligands in that it does not result in the unwinding of the switch helix but only in its destabilization, as reflected by increased B factors. Unwinding of the switch helix is proposed to be required for efficient binding of TonB to FhuA and for coupling the proton motive force of the cytoplasmic membrane with energy-dependent ligand transport. The transport data from cells expressing mutant FhuA proteins indicated conserved structural and mechanistic requirements for the transport of both types of compounds. CONCLUSIONS: We conclude that the binding of rifamycin CGP 4832 destabilizes the switch helix and promotes the formation of a transport-competent FhuA-TonB complex, albeit with lower efficiency than ferrichrome. Active transport of this rifamycin derivative explains the 200-fold increase in potency as compared to rifamycin, which is not a FhuA-specific ligand and permeates across the cell envelope by passive diffusion only.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins/chemistry , Receptors, Virus/chemistry , Rifamycins/chemistry , Rifamycins/pharmacology , Allosteric Site , Bacterial Proteins/chemistry , Binding Sites , Biological Transport , Biological Transport, Active , Cell Membrane/metabolism , Crystallography, X-Ray , Dose-Response Relationship, Drug , Escherichia coli/chemistry , Ferrichrome/chemistry , Ligands , Membrane Proteins/chemistry , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary , Spectrometry, Fluorescence , Time Factors , Tryptophan/chemistry
4.
Protein Sci ; 9(5): 956-63, 2000 May.
Article in English | MEDLINE | ID: mdl-10850805

ABSTRACT

One alternative method for drug delivery involves the use of siderophore-antibiotic conjugates. These compounds represent a specific means by which potent antimicrobial agents, covalently linked to iron-chelating siderophores, can be actively transported across the outer membrane of gram-negative bacteria. These "Trojan Horse" antibiotics may prove useful as an efficient means to combat multi-drug-resistant bacterial infections. Here we present the crystallographic structures of the natural siderophore-antibiotic conjugate albomycin and the siderophore phenylferricrocin, in complex with the active outer membrane transporter FhuA from Escherichia coli. To our knowledge, this represents the first structure of an antibiotic bound to its cognate transporter. Albomycins are broad-host range antibiotics that consist of a hydroxamate-type iron-chelating siderophore, and an antibiotically active, thioribosyl pyrimidine moiety. As observed with other hydroxamate-type siderophores, the three-dimensional structure of albomycin reveals an identical coordination geometry surrounding the ferric iron atom. Unexpectedly, this antibiotic assumes two conformational isomers in the binding site of FhuA, an extended and a compact form. The structural information derived from this study provides novel insights into the diverse array of antibiotic moieties that can be linked to the distal portion of iron-chelating siderophores and offers a structural platform for the rational design of hydroxamate-type siderophore-antibiotic conjugates.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Escherichia coli Proteins , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Binding Sites , Crystallography, X-Ray , Escherichia coli/chemistry , Ferrichrome/analogs & derivatives , Ferrichrome/chemistry , Ferrichrome/metabolism , Ligands , Models, Chemical , Models, Molecular , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
5.
Structure ; 8(6): 585-92, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10873859

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS), a lipoglycan from the outer membrane of Gram-negative bacteria, is an immunomodulatory molecule that stimulates the innate immune response. High levels of LPS cause excessive release of inflammatory mediators and are responsible for the septic shock syndrome. The interaction of LPS with its cognate binding proteins has not, as yet, been structurally elucidated. RESULTS: The X-ray crystallographic structure of LPS in complex with the integral outer membrane protein FhuA from Escherichia coli K-12 is reported. It is in accord with data obtained using mass spectroscopy and nuclear magnetic resonance. Most of the important hydrogen-bonding or electrostatic interactions with LPS are provided by eight positively charged residues of FhuA. Residues in a similar three-dimensional arrangement were searched for in all structurally known proteins using a fast template-matching algorithm, and a subset of four residues was identified that is common to known LPS-binding proteins. CONCLUSIONS: These four residues, three of which form specific interactions with lipid A, appear to provide the structural basis of pattern recognition in the innate immune response. Their arrangement can serve to identify LPS-binding sites on proteins known to interact with LPS, and could serve as a template for molecular modeling of a LPS scavenger designed to reduce the septic shock syndrome.


Subject(s)
Acute-Phase Proteins , Carrier Proteins/metabolism , Escherichia coli Proteins , Lipopolysaccharides/chemistry , Lipopolysaccharides/metabolism , Membrane Glycoproteins , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Carbohydrate Sequence , Carrier Proteins/chemistry , Crystallography, X-Ray , Escherichia coli/chemistry , Humans , Hydrogen Bonding , Mass Spectrometry , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Static Electricity
6.
J Membr Biol ; 178(3): 185-93, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11140274

ABSTRACT

Porin of Haemophilus influenzae type b (341 amino acids; M(r) 37782) determines the permeability of the outer membrane to low molecular mass compounds. Purified Hib porin was subjected to chemical modification of lysine residues by succinic anhydride. Electrospray ionization mass spectrometry identified up to 12 modifications per porin molecule. Tryptic digestion of modified Hib porin followed by reverse phase chromatography and matrix assisted laser desorption ionization time-of-flight mass spectrometry mapped the succinylation sites. Most modified lysines are positioned in surface-located loops, numbers 1 and 4 to 7. Succinylated porin was reconstituted into planar lipid bilayers, and biophysical properties were analyzed and compared to Hib porin: there was an increased average single channel conductance compared to Hib porin (1.24 +/- 0.41 vs. 0.85 +/- 0.40 nanosiemens). The voltage-gating activity of succinylated porin differed considerably from that of Hib porin. The threshold voltage for gating was decreased from 75 to 40 mV. At 80 mV, steady-state conductance for succinylated porin was 50-55% of the instantaneous conductance. Hib porin at 80 mV showed a decrease to 89-91% of the instantaneous current levels. We propose that surface-located lysine residues are determinants of voltage gating for porin of Haemophilus influenzae type b.


Subject(s)
Haemophilus influenzae/chemistry , Ion Channel Gating/physiology , Porins/chemistry , Amino Acid Sequence , Cell Membrane Permeability/physiology , Electric Conductivity , Lipid Bilayers , Lysine/chemistry , Models, Molecular , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protein Conformation , Protein Structure, Tertiary , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Succinic Acid/chemistry
7.
Gene ; 233(1-2): 49-57, 1999 Jun 11.
Article in English | MEDLINE | ID: mdl-10375620

ABSTRACT

The porA gene encodes the class 1 outer membrane protein (OMP1) in Neisseria meningitidis and is under transcriptional control. Promoter regions of porA from different clinical isolates were sequenced and were found to differ in the number of guanosine residues in a poly(G) track located upstream of the -10 region. Isolates that did not express OMP1 had up to nine G residues in the poly(G) track or an adenosine residue within this poly(G) track. Using beta-galactosidase as a reporter gene, the transcriptional activities of the promoter regions of the porA gene from three strains, two of which do not express OMP1, were assayed in both Escherichia coli and N. meningitidis. Mutations in the poly(G) track were created by site-directed mutagenesis and promoter fusions were further analyzed in E. coli and N. meningitidis. The number of nucleotides in the poly(G) track influenced promoter activity: reduction of a poly(G) track of 12nt by one and by two guanosine residues reduced promoter activity. Within the poly(G) track, replacement of an adenosine residue by a guanosine residue increased the promoter activity; replacement of a guanosine residue by an adenosine residue decreased the activity. The similar transcriptional activities for the mutated promoters in E. coli and N. meningitidis are compatible with similar control mechanisms for transcriptional control in both organisms.


Subject(s)
Neisseria meningitidis/genetics , Porins/genetics , Promoter Regions, Genetic , Base Sequence , DNA Primers , Escherichia coli/genetics , Mutagenesis, Site-Directed , Sequence Homology, Nucleic Acid
8.
Antimicrob Agents Chemother ; 43(2): 226-32, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9925510

ABSTRACT

We investigated the relationship between susceptibility to beta-lactam antibiotics and variation in the major outer membrane protein P2 (OmpP2; also called porin) of persistent nonencapsulated Haemophilus influenzae isolated from cystic fibrosis patients. Nine OmpP2 variants were selected from two distinct H. influenzae strains from two patients extensively treated with beta-lactam antibiotics. The variants differed in their susceptibilities to at least two beta-lactam antibiotics. By detergent extraction and column chromatography, OmpP2 was purified from two variants that were derived from strain 70 and that differed notably in their susceptibilities to beta-lactam antibiotics. The proteins were reconstituted into black lipid membranes for measurement of porin function. OmpP2 from the more resistant isolate (isolate 70b) had a smaller channel conductance than OmpP2 of the more susceptible isolate (isolate 70f). DNA sequencing of ompP2 of these isolates revealed single nonsynonymous base differences; there were changes in the amino acid sequence corresponding to surface-exposed loops 4, 5, 6, and 8. Changes in loops 4, 5, and 6 were previously shown to result in antigenic differences. Beside these mutations, variants of strain 70 showed additional mutations in loop 1 and nonexposed loop 3. Taken together, our results suggest that in variants of strain 70, nonsynonymous point mutations accumulated both in the sequences of ompP2 coding for antigen-variable loops and in other loops, notably, loops 1 and 3. The latter changes are suggested to affect the permeability of the porin channel.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins , Cystic Fibrosis/microbiology , Haemophilus influenzae/physiology , Hexosyltransferases , Peptidyl Transferases , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/analysis , DNA, Bacterial/analysis , Genetic Variation , Haemophilus influenzae/chemistry , Haemophilus influenzae/genetics , Haemophilus influenzae/isolation & purification , Humans , Microbial Sensitivity Tests , Molecular Sequence Data , Muramoylpentapeptide Carboxypeptidase/analysis , Penicillin-Binding Proteins , Sequence Homology, Amino Acid
9.
Science ; 282(5397): 2215-20, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9856937

ABSTRACT

FhuA, the receptor for ferrichrome-iron in Escherichia coli, is a member of a family of integral outer membrane proteins, which, together with the energy-transducing protein TonB, mediate the active transport of ferric siderophores across the outer membrane of Gram-negative bacteria. The three-dimensional structure of FhuA is presented here in two conformations: with and without ferrichrome-iron at resolutions of 2.7 and 2.5 angstroms, respectively. FhuA is a beta barrel composed of 22 antiparallel beta strands. In contrast to the typical trimeric arrangement found in porins, FhuA is monomeric. Located within the beta barrel is a structurally distinct domain, the "cork," which mainly consists of a four-stranded beta sheet and four short alpha helices. A single lipopolysaccharide molecule is noncovalently associated with the membrane-embedded region of the protein. Upon binding of ferrichrome-iron, conformational changes are transduced to the periplasmic pocket of FhuA, signaling the ligand-loaded status of the receptor. Sequence homologies and mutagenesis data are used to propose a structural mechanism for TonB-dependent siderophore-mediated transport across the outer membrane.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Escherichia coli Proteins , Escherichia coli/chemistry , Ferric Compounds/metabolism , Ferrichrome/metabolism , Lipopolysaccharides/metabolism , Protein Conformation , Receptors, Virus/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Biological Transport, Active , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Diffusion , Escherichia coli/metabolism , Hydrogen Bonding , Ligands , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Protein Structure, Secondary , Receptors, Virus/metabolism , Signal Transduction
10.
Protein Sci ; 7(7): 1636-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9684898

ABSTRACT

FhuA (Mr 78,992, 714 amino acids), siderophore receptor for ferrichrome-iron in the outer membrane of Escherichia coli, was affinity tagged, rapidly purified, and crystallized. To obtain FhuA in quantities sufficient for crystallization, a hexahistidine tag was genetically inserted into the fhuA gene after amino acid 405, which resides in a known surface-exposed loop. Recombinant FhuA405.H6 was overexpressed in an E. coli strain that is devoid of several major porins and using metal-chelate chromatography was purified in large amounts to homogeneity. FhuA crystals were grown using the hanging drop vapor diffusion technique and were suitable for X-ray diffraction analysis. On a rotating anode X-ray source, diffraction was observed to 3.0 A resolution. The crystals belong to space group P6(1) or P6(5) with unit cell dimensions of a=b=174 A, c=88 A (alpha=beta=90 degrees, gamma=120 degrees).


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/isolation & purification , Escherichia coli Proteins , Escherichia coli/chemistry , Receptors, Virus/chemistry , Receptors, Virus/isolation & purification , Chelating Agents , Chromatography, Affinity , Crystallization , Crystallography, X-Ray , Ferrichrome , Histidine/chemistry , Iron , Nickel , Protein Conformation , Protein Engineering , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
11.
Mol Microbiol ; 28(4): 675-81, 1998 May.
Article in English | MEDLINE | ID: mdl-9643536

ABSTRACT

Cells growing in aerobic environments have developed intricate strategies to overcome the scarcity of iron, an essential nutrient. In gram-negative bacteria, high-affinity iron acquisition requires outer membrane-localized proteins that bind iron chelates at the cell surface and promote their uptake. Transport of bound chelates across the outer membrane depends upon TonB-ExbB-ExbD, a cytoplasmic membrane-localized complex that transduces energy from the proton motive force to high-affinity receptors in the outer membrane. Upon ligand binding to iron chelate receptors, conformational changes are induced, some of which are detected in the periplasm. These structural alterations signal the ligand-loaded status of the receptor and, therefore, the requirement for TonB-dependent energy transduction. Thus, TonB interacts preferentially and directly with ligand-loaded receptors. Such a mechanism ensures the productive use of cellular energy to drive active transport at the outer membrane.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Membrane Proteins/metabolism , Siderophores/metabolism , Bacterial Outer Membrane Proteins/metabolism , Biological Transport, Active , Ligands , Receptors, Cell Surface/metabolism , Signal Transduction
12.
Can J Microbiol ; 44(1): 56-63, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9522450

ABSTRACT

Subtyping Neisseria meningitidis by methods that rely on monoclonal antibody (mAb) reactivity results in an unusually high number of strains that are not subtypeable. To subtype 48 strains isolated (1993-1994) in the province of Quebec that were not subtypeable by mAb-based techniques, we used DNA sequencing of the variable regions of porA, a gene that encodes the class 1 outer membrane protein. We assigned subtypes to all the previously nonserosubtypeable isolates and identified some novel subtypes. Because our sequencing strategy included the promoter region of porA, different isolates were compared in their sequences of the porA promoter region. A poly(G) stretch lies between the -10 and -35 regions of the promoter; replacement of a G residue by an A residue in this region resulted in loss of expression of porA. No correlation was found between the number of G residues in the poly(G) stretch and the level of expression; a minimum of 10 G residues is required in this stretch for expression of porA. One isolate expressed no class 1 outer membrane protein because of the insertion sequence IS1301 in the coding region of porA. Another isolate did not express the protein owing to a frame-shift mutation within the coding region of porA. Sequencing of porA allowed assignments of subtypes to previously uncharacterized isolates and provided insights about the regulation of expression of this gene in N. meningitidis.


Subject(s)
Meningococcal Infections/microbiology , Neisseria meningitidis/classification , Porins/genetics , Amino Acid Sequence , Antigenic Variation , DNA Transposable Elements , Humans , Molecular Sequence Data , Neisseria meningitidis/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Sequence Analysis, DNA , Sequence Homology, Amino Acid
13.
J Biol Chem ; 272(45): 28391-7, 1997 Nov 07.
Article in English | MEDLINE | ID: mdl-9353297

ABSTRACT

The ferrichrome-iron receptor of Escherichia coli is FhuA, an outer membrane protein that is dependent upon the energy-coupling protein TonB to enable active transport of specific hydroxamate siderophores, infection by certain phages, and cell killing by the protein antibiotics colicin M and microcin 25. In vivo cross-linking studies were performed to establish at the biochemical level the interaction between FhuA and TonB. In an E. coli strain in which both proteins were expressed from the chromosome, a high molecular mass complex was detected when the ferrichrome homologue ferricrocin was added immediately prior to addition of cross-linker. The complex included both proteins; it was absent from strains of E. coli that were devoid of either FhuA or TonB, and it was detected with anti-FhuA and anti-TonB monoclonal antibodies. These results indicate that, in vivo, the binding of ferricrocin to FhuA enhances complex formation between the receptor and TonB. An in vitro system was established with which to examine the FhuA-TonB interaction. Incubation of TonB with histidine-tagged FhuA followed by addition of Ni2+-nitrilotriacetate-agarose led to the specific recovery of both TonB and FhuA. Addition of ferricrocin or colicin M to FhuA in this system greatly increased the coupling between FhuA and TonB. Conversely, a monoclonal antibody that binds near the N terminus of FhuA reduced the retention of TonB by histidine-tagged FhuA. These studies demonstrate the significance of ligand binding at the external surface of the cell to mediate signal transduction across the outer membrane.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Escherichia coli Proteins , Escherichia coli/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface , Receptors, Virus/metabolism , Signal Transduction , Animals , Biological Transport , Carrier Proteins/metabolism , Ferrichrome/analogs & derivatives , Ferrichrome/metabolism , Goats , Ligands , Mice , Protein Binding , Protein Conformation
14.
J Biol Chem ; 272(21): 13614-21, 1997 May 23.
Article in English | MEDLINE | ID: mdl-9153210

ABSTRACT

Porin (341 amino acids; mass of 37,782 Da) in the outer membrane of Haemophilus influenzae type b (Hib) permits diffusion into the periplasm of small solutes up to a molecular mass of 1400 Da. Molecular modeling of Hib porin identified its structural similarities to OmpF of Escherichia coli and disclosed for Hib porin a shorter length of loop 3 and a longer length of loop 4. By site-directed mutagenesis of the porin gene ompP2, mutant porins were constructed to contain 6 or 12 amino acid deletions either in loop 3 or in surface-exposed loop 4. Wild type Hib porin and mutant porins were expressed in a nontypeable H. influenzae strain deleted for the ompP2 gene. The mutant porins were purified and reconstituted into planar bilayers, tested for channel formation and compared with wild type Hib porin. Mutant Haemophilus porin possessing a 6-amino acid deletion in loop 3 displayed a broad distribution of single channel conductance values, while deletion of 12 amino acids from the same loop destabilized the porin channel. By comparison, deletion of 6 or of 12 amino acids from loop 4 of Hib porin resulted in an increased single channel conductance (1.15 and 1.05 nanosiemens, respectively) compared with wild type Hib porin (0. 85 nanosiemens). The C3 epitope of the poliovirus VP1 capsid protein was inserted either into loop 3 or into loop 4 of Hib porin. By flow cytometry, the C3 epitope was detected as surface-exposed in strains expressing C3 insertion in loop 4; in strains expressing C3 insertion in loop 3, the epitope was inaccessible. We propose that loop 4 of Hib porin, although surface-accessible, is oriented toward the central axis of the pore and that deletions in this loop increase the single channel conductance by widening the pore entrance.


Subject(s)
Haemophilus influenzae/genetics , Porins/genetics , Amino Acid Sequence , Base Sequence , Escherichia coli , Flow Cytometry , Haemophilus influenzae/chemistry , Immunosorbent Techniques , Lipid Bilayers/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Porins/chemistry , Protein Folding , Protein Structure, Tertiary , Sequence Alignment
15.
Can J Microbiol ; 43(3): 234-8, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9090112

ABSTRACT

Routine serosubtyping of Neisseria meningitidis relies upon reactivity of whole cells to monoclonal antibodies (mAbs). This procedure is limited in providing maximum serosubtype information because some epitopes in whole cells are masked and because mAbs are currently unavailable for some epitopes. To address masking of epitopes in whole cells, we isolated outer membrane vesicles (OMVs) from nine representative meningococcal strains that were isolated (1991-1993) in the province of Quebec, Canada; the OMVs were used in enzyme-linked immunosorbent assay for reactivity to mAbs, and improved serosubtyping information was obtained. A recent proposal assigns subtypes based on deduced amino acid sequences in the variable regions of the class 1 outer membrane protein. This scheme maintains the subtyping nomenclature that is based on reactivity to mAbs by defining the sequences in the epitopes recognized by the mAbs. We used this technique to assign subtypes to the meningococcal strains isolated in Quebec. For the strains tested, serosubtyping using mAbs and subtyping based on deduced amino acid sequences were in complete agreement. Subtyping using deduced amino acid sequences is superior because it does not depend on the availability of mAbs.


Subject(s)
Amino Acid Sequence/genetics , Neisseria meningitidis/genetics , Neisseria meningitidis/immunology , Serotyping , Antibodies, Monoclonal/immunology , Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/isolation & purification , Canada , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Molecular Sequence Data , Polymerase Chain Reaction
16.
Curr Microbiol ; 34(1): 18-22, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8939796

ABSTRACT

The currently practiced protocol for routine serosubtyping of Neisseria meningitidis relies on reactivity of whole cells to monoclonal antibodies against the class 1 outer membrane protein (OMP) in ELISAs or dot-blots. This procedure, however, failed to yield serosubtyping information in 28% (48/174) of clinical isolates (1993-1994) in the province of Québec, Canada. These 48 strains were characterized by OMP profiles and ELISAs with outer membrane vesicles (OMVs). Forty out of the 48 strains expressed class 1 OMP, indicating that the inability to assign a serosubtype was not owing to the absence of the class 1 OMP. Of these, 15 (38%) were serosubtypable in ELISAs with outer membrane vesicles. Thus, 81% (141/174) of all meningococcal strains were serosubtypable with ELISAs using whole-cells or OMVs. Because the routinely used procedure for serosubtyping of meningococci is limited in providing serosubtype information, alternate procedures are proposed to obtain comprehensive information for epidemiological identification of this bacterium.


Subject(s)
Bacterial Outer Membrane Proteins/analysis , Neisseria meningitidis/classification , Serotyping/methods , Cell Membrane/chemistry , Enzyme-Linked Immunosorbent Assay/methods , Genes, Bacterial/genetics , Porins/genetics , Quebec
17.
Mol Microbiol ; 22(3): 459-71, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8939430

ABSTRACT

Ferrichrome-iron is actively transported across the outer membrane of Escherichia coli by the TonB-dependent receptor FhuA. To obtain FhuA in a form suitable for secondary-structure analyses, a hexahistidine tag was inserted into a surface-located site and the recombinant protein was purified by metal chelate chromatography. Functional studies indicated that the presence of the hexahistidine tag did not interfere with FhuA localization or with ligand-binding activity. Ferrichrome protected lysine 67 but not lysine 5 of purified recombinant FhuA from trypsinolysis. Results from trypsin digestion were interpreted as a conformational change in FhuA which had occurred upon ferrichrome binding, thereby preventing access of trypsin to lysine 67. Circular dichroism and Fourier transform infrared spectroscopy revealed a predominance of beta-sheet structure for the purified protein. In the presence of ferrichrome, FhuA exhibited a secondary structure and a thermostability which were similar to FhuA without ligand. The addition of ferrichrome to purified FhuA reduced the ability of certain anti-FhuA monoclonal antibodies to bind to the receptor. All antibodies which could in this manner discriminate between FhuA and FhuA bound to ferrichrome had their determinants within a loop which is toward the N-terminus and which is exposed to the periplasm. These data indicate that the binding of ferrichrome induces a structural change that is propogated across the outer membrane and results in an altered conformation of a periplasmically exposed loop of FhuA. It is proposed that by such an alteration of FhuA conformation, TonB is triggered to energize the active transport of the bound ligand across the outer membrane.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Escherichia coli Proteins , Escherichia coli/chemistry , Escherichia coli/genetics , Ferrichrome/pharmacokinetics , Iron/pharmacokinetics , Protein Structure, Secondary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Antibodies, Monoclonal/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/physiology , Biological Transport, Active/physiology , Chromatography, Agarose , Circular Dichroism , Enzyme-Linked Immunosorbent Assay , Escherichia coli/physiology , Flow Cytometry , Gene Expression Regulation, Bacterial , Histidine/genetics , Lysine/metabolism , Membrane Proteins/physiology , Receptors, Virus/immunology , Recombination, Genetic , Spectroscopy, Fourier Transform Infrared , Trypsin/metabolism
18.
FEBS Lett ; 392(3): 304-8, 1996 Sep 02.
Article in English | MEDLINE | ID: mdl-8774868

ABSTRACT

The major diffusion channel in the outer membrane of Haemophilus influenzae type b (Hib) is porin (341 amino acids; Mr 37 782). The Hib porin gene was cloned and overexpressed in Bacillus subtilis. Recombinant Hib porin (Bac porin), having aggregated into inclusion bodies, was purified under denaturing conditions and subsequently refolded. To compare Bac porin that is intrinsically devoid of lipooligosaccharides versus native Hib porin, the properties of Bac porin were assessed by the following four criteria: circular dichroism spectroscopy, channel formation in planar bilayers, resistance to trypsin digestion and formation of the conformational epitope recognized by an anti-Hib porin monoclonal antibody. We conclude that in the absence of lipooligosaccharides, Bac porin was refolded into a functional form which closely resembled the structure of Hib porin.


Subject(s)
Bacillus subtilis/genetics , Haemophilus influenzae/chemistry , Porins/chemistry , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Bacillus subtilis/chemistry , Bacillus subtilis/metabolism , Circular Dichroism , Detergents , Haemophilus influenzae/genetics , Lipopolysaccharides/pharmacology , Mice , Porins/immunology , Porins/metabolism , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Trypsin/metabolism , Trypsin/pharmacology
19.
J Bacteriol ; 177(21): 6118-25, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7592376

ABSTRACT

Ferrichrome-iron transport in Escherichia coli is initiated by the outer membrane receptor FhuA. Thirty-five anti-FhuA monoclonal antibodies (MAbs) were isolated to examine the surface accessibility of FhuA sequences and their contribution to ligand binding. The determinants of 32 of the MAbs were mapped to eight distinct regions in the primary sequence of FhuA by immunoblotting against (i) five internal deletion FhuA proteins and (ii) four FhuA peptides generated by cyanogen bromide cleavage. Two groups of MAbs bound to FhuA in outer membrane vesicles but not to intact cells, indicating that their determinants, located between residues 1 and 20 and 21 and 59, are exposed to the periplasm. One of the 28 strongly immunoblot-reactive MAbs bound to FhuA on intact cells in flow cytometry, indicating that its determinant, located between amino acids 321 and 381, is cell surface exposed. This MAb and four others which in flow cytometry bound to cells expressing FhuA were tested for the ability to block ligand binding. While no MAb inhibited growth promotion by ferrichrome or cell killing by microcin 25, some prevented killing by colicin M and were partially able to inhibit the inactivation of T5 phage. These data provide evidence for spatially distinct ligand binding sites on FhuA. The lack of surface reactivity of most of the immunoblot-reactive MAbs suggests that the majority of FhuA sequences which lie external to the outer membrane may adopt a tightly ordered organization with little accessible linear sequence.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Escherichia coli Proteins , Receptors, Virus/chemistry , Receptors, Virus/immunology , Amino Acid Sequence , Antibodies, Bacterial , Antibodies, Monoclonal , Bacillus subtilis/genetics , Base Sequence , Binding, Competitive , Colicins/pharmacology , Epitope Mapping , Escherichia coli , Ferrichrome , Flow Cytometry , Hybridomas , Immunoblotting , Iron , Ligands , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , T-Phages
20.
J Bacteriol ; 176(14): 4250-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7517392

ABSTRACT

The ferrichrome-iron receptor of Escherichia coli K-12 is FhuA (M(r), 78,992), the first component of an energy-dependent, high-affinity iron uptake pathway. FhuA is also the cognate receptor for bacteriophages T5, T1, phi 80, and UC-1, for colicin M and microcin 25, and for albomycin. To probe the topological organization of FhuA which enables recognition of these different ligands, we generated a library of 16 insertion mutations within the fhuA gene. Each insertion spliced a 13-amino-acid antigenic determinant (the C3 epitope of poliovirus) at a different position within FhuA. Immunoblotting of outer membranes with anti-FhuA and anti-C3 antibodies indicated that 15 of 16 FhuA.C3 proteins were present in the outer membrane in amounts similar to that observed for plasmid-encoded wild-type FhuA. One chimeric protein with the C3 epitope inserted after amino acid 440 of FhuA was present in the outer membrane in greatly reduced amounts. Strains overexpressing FhuA.C3 proteins were subjected to flow cytometric analysis using anti-FhuA monoclonal antibodies. Such analysis showed that (i) the chimeric proteins were properly localized and (ii) the wild-type FhuA protein structure had not been grossly altered by insertion of the C3 epitope. Twelve of sixteen strains expressing FhuA.C3 proteins were proficient in ferrichrome transport and remained sensitive to FhuA-specific phages. Three FhuA.C3 proteins, with insertions after amino acid 321, 405, or 417 of FhuA, were detected at the cell surface by flow cytometry using anti-C3 antibodies. These three chimeric proteins were all biologically active. We conclude that amino acids 321, 405, and 417 are surface accessible in wild-type FhuA.


Subject(s)
Bacterial Outer Membrane Proteins/analysis , Escherichia coli Proteins , Escherichia coli/chemistry , Receptors, Virus/analysis , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Base Sequence , Capsid/analysis , Capsid/genetics , Capsid/immunology , Capsid Proteins , Cell Membrane/chemistry , Epitopes/analysis , Epitopes/chemistry , Epitopes/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Genes, Bacterial , Immunoblotting , Molecular Sequence Data , Mutagenesis, Insertional , Phenotype , Poliovirus/immunology , Protein Folding , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...