Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters











Publication year range
1.
Microbiology (Reading) ; 170(1)2024 01.
Article in English | MEDLINE | ID: mdl-38189440

ABSTRACT

One of the mechanisms employed by the opportunistic pathogen Burkholderia cenocepacia to acquire the essential element iron is the production and release of two ferric iron chelating compounds (siderophores), ornibactin and pyochelin. Here we show that B. cenocepacia is also able to take advantage of a range of siderophores produced by other bacteria and fungi ('xenosiderophores') that chelate iron exclusively by means of hydroxamate groups. These include the tris-hydroxamate siderophores ferrioxamine B, ferrichrome, ferricrocin and triacetylfusarinine C, the bis-hydroxamates alcaligin and rhodotorulic acid, and the monohydroxamate siderophore cepabactin. We also show that of the 24 TonB-dependent transporters encoded by the B. cenocepacia genome, two (FhuA and FeuA) are involved in the uptake of hydroxamate xenosiderophores, with FhuA serving as the exclusive transporter of iron-loaded ferrioxamine B, triacetylfusarinine C, alcaligin and rhodotorulic acid, while both FhuA and FeuA are able to translocate ferrichrome-type siderophores across the outer membrane. Finally, we identified FhuB, a putative cytoplasmic membrane-anchored ferric-siderophore reductase, as being obligatory for utilization of all of the tested bis- and tris-hydroxamate xenosiderophores apart from alcaligin.


Subject(s)
Burkholderia cenocepacia , Ferrichrome , Burkholderia cenocepacia/genetics , Siderophores , Iron
2.
Nucleic Acids Res ; 50(7): 3709-3726, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35234897

ABSTRACT

Burkholderia cenocepacia is an opportunistic pathogen that causes severe infections of the cystic fibrosis (CF) lung. To acquire iron, B. cenocepacia secretes the Fe(III)-binding compound, ornibactin. Genes for synthesis and utilisation of ornibactin are served by the iron starvation (IS) extracytoplasmic function (ECF) σ factor, OrbS. Transcription of orbS is regulated in response to the prevailing iron concentration by the ferric uptake regulator (Fur), such that orbS expression is repressed under iron-sufficient conditions. Here we show that, in addition to Fur-mediated regulation of orbS, the OrbS protein itself responds to intracellular iron availability. Substitution of cysteine residues in the C-terminal region of OrbS diminished the ability to respond to Fe(II) in vivo. Accordingly, whilst Fe(II) impaired transcription from and recognition of OrbS-dependent promoters in vitro by inhibiting the binding of OrbS to core RNA polymerase (RNAP), the cysteine-substituted OrbS variant was less responsive to Fe(II). Thus, the cysteine residues within the C-terminal region of OrbS contribute to an iron-sensing motif that serves as an on-board 'anti-σ factor' in the presence of Fe(II). A model to account for the presence two regulators (Fur and OrbS) that respond to the same intracellular Fe(II) signal to control ornibactin synthesis and utilisation is discussed.


Subject(s)
Bacterial Proteins , Burkholderia cenocepacia , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia Infections/microbiology , Burkholderia cenocepacia/genetics , Cystic Fibrosis/complications , Ferrous Compounds/metabolism , Gene Expression Regulation, Bacterial , Humans , Iron/metabolism
3.
Med Microbiol Immunol ; 209(5): 643-652, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32495068

ABSTRACT

In the original article, incorrect  figures were published with incorrect captions. The correct figures and captions are given below.

4.
Med Microbiol Immunol ; 209(4): 473-487, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32253503

ABSTRACT

Burkholderia pseudomallei is the causative agent of melioidosis, a disease with high morbidity that is endemic in South East Asia and northern Australia. An unusual feature of the bacterium is its ability to induce multinucleated giant cell formation (MNGC), which appears to be related to bacterial pathogenicity. The mechanism of MNGC formation is not fully understood, but host cell factors as well as known bacterial virulence determinants are likely to contribute. Since members of the tetraspanin family of membrane proteins are involved in various types of cell:cell fusion, their role in MNGC formation induced by Burkholderia thailandensis, a mildly pathogenic species closely related to B. pseudomallei, was investigated. The effect of antibodies to tetraspanins CD9, CD81, and CD63 in MNGC formation induced by B. thailandensis in infected mouse J774.2 and RAW macrophage cell lines was assessed along with that of recombinant proteins corresponding to the large extracellular domain (EC2) of the tetraspanins. B. thailandensis-induced fusion was also examined in macrophages derived from CD9 null and corresponding WT mice, and in J774.2 macrophages over-expressing CD9. Antibodies to CD9 and CD81 promoted MNGC formation induced by B. thailandensis, whereas EC2 proteins of CD9, CD81, and CD63 inhibited MNGC formation. Enhanced MNGC formation was observed in CD9 null macrophages, whereas a decrease in MNGC formation was associated with overexpression of CD9. Overall our findings show that tetraspanins are involved in MNGC formation induced by B. thailandensis and by implication, B. pseudomallei, with CD9 and CD81 acting as negative regulators of this process.


Subject(s)
Burkholderia , Cell Fusion , Giant Cells/metabolism , Macrophages/microbiology , Tetraspanins/metabolism , Animals , Burkholderia pseudomallei , Cell Line , Giant Cells/microbiology , Melioidosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/metabolism , Tetraspanin 28/metabolism , Tetraspanin 29/metabolism , Tetraspanin 30/metabolism
5.
Microbiologyopen ; 8(7): e00774, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30628184

ABSTRACT

Burkholderia cenocepacia is an opportunistic bacterial pathogen that poses a significant threat to individuals with cystic fibrosis by provoking a strong inflammatory response within the lung. It possesses a type VI secretion system (T6SS), a secretory apparatus that can perforate the cellular membrane of other bacterial species and/or eukaryotic targets, to deliver an arsenal of effector proteins. The B. cenocepacia T6SS (T6SS-1) has been shown to be implicated in virulence in rats and contributes toward actin rearrangements and inflammasome activation in B. cenocepacia-infected macrophages. Here, we present bioinformatics evidence to suggest that T6SS-1 is the archetype T6SS in the Burkholderia genus. We show that B. cenocepacia T6SS-1 is active under normal laboratory growth conditions and displays antibacterial activity against other Gram-negative bacterial species. Moreover, B. cenocepacia T6SS-1 is not required for virulence in three eukaryotic infection models. Bioinformatics analysis identified several candidate T6SS-dependent effectors that may play a role in the antibacterial activity of B. cenocepacia T6SS-1. We conclude that B. cenocepacia T6SS-1 plays an important role in bacterial competition for this organism, and probably in all Burkholderia species that possess this system, thereby broadening the range of species that utilize the T6SS for this purpose.

6.
J Bacteriol ; 201(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30455278

ABSTRACT

OrbS and PvdS are extracytoplasmic function (ECF) σ factors that regulate transcription of operons required for the biosynthesis of the siderophores ornibactin and pyoverdine in the Burkholderia cepacia complex and Pseudomonas spp., respectively. Here we show that promoter recognition by OrbS requires specific tetrameric -35 and -10 element sequences that are strikingly similar to those of the consensus PvdS-dependent promoter. However, whereas Pseudomonas aeruginosa PvdS can serve OrbS-dependent promoters, OrbS cannot utilize PvdS-dependent promoters. To identify features present at OrbS-dependent promoters that facilitate recognition by OrbS, we carried out a detailed analysis of the nucleotide sequence requirements for promoter recognition by both OrbS and PvdS. This revealed that DNA sequence features located outside the sigma binding elements are required for efficient promoter utilization by OrbS. In particular, the presence of an A-tract extending downstream from the -35 element at OrbS-dependent promoters was shown to be an important contributor to OrbS specificity. Our observations demonstrate that the nature of the spacer sequence can have a major impact on promoter recognition by some ECF σ factors through modulation of the local DNA architecture.IMPORTANCE ECF σ factors regulate subsets of bacterial genes in response to environmental stress signals by directing RNA polymerase to promoter sequences known as the -35 and -10 elements. In this work, we identify the -10 and -35 elements that are recognized by the ECF σ factor OrbS. Furthermore, we demonstrate that efficient promoter utilization by this σ factor also requires a polyadenine tract located downstream of the -35 region. We propose that the unique architecture of A-tract DNA imposes conformational features on the -35 element that facilitates efficient recognition by OrbS. Our results show that sequences located between the core promoter elements can make major contributions to promoter recognition by some ECF σ factors.


Subject(s)
Burkholderia cenocepacia/metabolism , DNA, Bacterial/metabolism , Promoter Regions, Genetic , Pseudomonas aeruginosa/metabolism , Sigma Factor/metabolism , Substrate Specificity , Burkholderia cenocepacia/genetics , DNA Mutational Analysis , DNA, Bacterial/genetics , Iron/metabolism , Protein Binding , Pseudomonas aeruginosa/genetics , Trace Elements/metabolism
7.
Nat Commun ; 9(1): 4765, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420757

ABSTRACT

The type VI secretion system (T6SS) is a multi-protein complex that injects bacterial effector proteins into target cells. It is composed of a cell membrane complex anchored to a contractile bacteriophage tail-like apparatus consisting of a sharpened tube that is ejected by the contraction of a sheath against a baseplate. We present structural and biochemical studies on TssA subunits from two different T6SSs that reveal radically different quaternary structures in comparison to the dodecameric E. coli TssA that arise from differences in their C-terminal sequences. Despite this, the different TssAs retain equivalent interactions with other components of the complex and position their highly conserved N-terminal ImpA_N domain at the same radius from the centre of the sheath as a result of their distinct domain architectures, which includes additional spacer domains and highly mobile interdomain linkers. Together, these variations allow these distinct TssAs to perform a similar function in the complex.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Secretion Systems , Protein Subunits/chemistry , Protein Subunits/metabolism , Amino Acid Sequence , Bacterial Proteins/ultrastructure , Computational Biology , Phylogeny , Protein Domains , Proteolysis , Structure-Activity Relationship
8.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 9): 578-582, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30198891

ABSTRACT

TssA is a core subunit of the type VI secretion system, which is a major player in interspecies competition in Gram-negative bacteria. Previous studies on enteroaggregative Escherichia coli TssA suggested that it is comprised of three putative domains: a conserved N-terminal domain, a middle domain and a ring-forming C-terminal domain. X-ray studies of the latter two domains have identified their respective structures. Here, the results of the expression and purification of full-length and domain constructs of TssA from Aeromonas hydrophila are reported, resulting in diffraction-quality crystals for the middle domain (Nt2) and a construct including the middle and C-terminal domains (Nt2-CTD).


Subject(s)
Aeromonas hydrophila/chemistry , Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Type VI Secretion Systems/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Domains , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
9.
Article in English | MEDLINE | ID: mdl-30181989

ABSTRACT

[This corrects the article DOI: 10.3389/fcimb.2017.00460.].

10.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 9): 536-542, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30198885

ABSTRACT

TssA is a core component of the type VI secretion system, and phylogenetic analysis of TssA subunits from different species has suggested that these proteins fall into three distinct clades. Whilst representatives of two clades, TssA1 and TssA2, have been the subjects of investigation, no members of the third clade (TssA3) have been studied. Constructs of TssA from Burkholderia cenocepacia, a representative of clade 3, were expressed, purified and subjected to crystallization trials. Data were collected from crystals of constructs of the N-terminal and C-terminal domains. Analysis of the data from the crystals of these constructs and preliminary structure determination indicates that the C-terminal domain forms an assembly of 32 subunits in D16 symmetry, whereas the N-terminal domain is not involved in subunit assocation.


Subject(s)
Bacterial Proteins/chemistry , Burkholderia cenocepacia/chemistry , Electrons , Membrane Proteins/chemistry , Protein Subunits/chemistry , Type VI Secretion Systems/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Burkholderia cenocepacia/classification , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phylogeny , Protein Conformation, alpha-Helical , Protein Domains , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Article in English | MEDLINE | ID: mdl-29164069

ABSTRACT

Burkholderia is a genus within the ß-Proteobacteriaceae that contains at least 90 validly named species which can be found in a diverse range of environments. A number of pathogenic species occur within the genus. These include Burkholderia cenocepacia and Burkholderia multivorans, opportunistic pathogens that can infect the lungs of patients with cystic fibrosis, and are members of the Burkholderia cepacia complex (Bcc). Burkholderia pseudomallei is also an opportunistic pathogen, but in contrast to Bcc species it causes the tropical human disease melioidosis, while its close relative Burkholderia mallei is the causative agent of glanders in horses. For these pathogens to survive within a host and cause disease they must be able to acquire iron. This chemical element is essential for nearly all living organisms due to its important role in many enzymes and metabolic processes. In the mammalian host, the amount of accessible free iron is negligible due to the low solubility of the metal ion in its higher oxidation state and the tight binding of this element by host proteins such as ferritin and lactoferrin. As with other pathogenic bacteria, Burkholderia species have evolved an array of iron acquisition mechanisms with which to capture iron from the host environment. These mechanisms include the production and utilization of siderophores and the possession of a haem uptake system. Here, we summarize the known mechanisms of iron acquisition in pathogenic Burkholderia species and discuss the evidence for their importance in the context of virulence and the establishment of infection in the host. We have also carried out an extensive bioinformatic analysis to identify which siderophores are produced by each Burkholderia species that is pathogenic to humans.


Subject(s)
Burkholderia Infections/metabolism , Burkholderia Infections/microbiology , Burkholderia/metabolism , Burkholderia/pathogenicity , Iron/metabolism , Virulence , Animals , Burkholderia/classification , Burkholderia/genetics , Burkholderia gladioli/metabolism , Burkholderia gladioli/pathogenicity , Burkholderia mallei/metabolism , Burkholderia mallei/pathogenicity , Burkholderia pseudomallei/metabolism , Burkholderia pseudomallei/pathogenicity , Computational Biology , Cystic Fibrosis/microbiology , Ferritins/metabolism , Glanders , Heme/metabolism , Horses , Humans , Lactoferrin/metabolism , Lung/microbiology , Melioidosis/microbiology , Siderophores/metabolism
12.
Plasmid ; 89: 49-56, 2017 01.
Article in English | MEDLINE | ID: mdl-27825973

ABSTRACT

To elucidate the function of a gene in bacteria it is vital that targeted gene inactivation (allelic replacement) can be achieved. Allelic replacement is often carried out by disruption of the gene of interest by insertion of an antibiotic-resistance marker followed by subsequent transfer of the mutant allele to the genome of the host organism in place of the wild-type gene. However, due to their intrinsic resistance to many antibiotics only selected antibiotic-resistance markers can be used in members of the genus Burkholderia, including the Burkholderia cepacia complex (Bcc). Here we describe the construction of improved antibiotic-resistance cassettes that specify resistance to kanamycin, chloramphenicol or trimethoprim effectively in the Bcc and related species. These were then used in combination with and/or to construct a series enhanced suicide vectors, pSHAFT2, pSHAFT3 and pSHAFT-GFP to facilitate effective allelic replacement in the Bcc. Validation of these improved suicide vectors was demonstrated by the genetic inactivation of selected genes in the Bcc species Burkholderia cenocepacia and B. lata, and in the non-Bcc species, B. thailandensis.


Subject(s)
Burkholderia/genetics , DNA, Bacterial , Mutation , Plasmids/genetics , Anti-Bacterial Agents/pharmacology , Burkholderia/drug effects , Drug Resistance, Bacterial , Gene Order , Humans
13.
Microbiology (Reading) ; 160(Pt 2): 261-269, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24217175

ABSTRACT

Pyocins are toxic proteins produced by some strains of Pseudomonas aeruginosa that are lethal for related strains of the same species. Some soluble pyocins (S2, S3 and S4) were previously shown to use the pyoverdine siderophore receptors to enter the cell. The P. aeruginosa PAO1 pore-forming pyocin S5 encoding gene (PAO985) was cloned into the expression vector pET15b, and the affinity-purified protein product tested for its killing activity against different P. aeruginosa strains. The results, however, did not show any correlation with a specific ferripyoverdine receptor. To further identify the S5 receptor, transposon mutants were generated. Pooled mutants were exposed to pyocin S5 and the resistant colonies growing in the killing zone were selected. The majority of S5-resistant mutants had an insertion in the fptA gene encoding the receptor for the siderophore pyochelin. Complementation of an fptA transposon mutant with the P. aeruginosa fptA gene in trans restored the sensitivity to S5. In order to define the receptor-binding domain of pyocin S5, two hybrid pyocins were constructed containing different regions from pyocin S5 fused to the C-terminal translocation and DNase killing domains of pyocin S2. Only the protein containing amino acid residues 151 to 300 from S5 showed toxicity, indicating that the pyocin S5 receptor-binding domain is not at the N-terminus of the protein as in other S-type pyocins. Pyocin S5 was, however, unable to kill Burkholderia cenocepacia strains producing a ferripyochelin FptA receptor, nor was the B. cenocepacia fptA gene able to restore the sensitivity of the resistant fptA mutant P. aeruginosa strain.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Microbial Viability/drug effects , Pseudomonas aeruginosa/drug effects , Pyocins/metabolism , Pyocins/toxicity , Receptors, Cell Surface/metabolism , Burkholderia cenocepacia/drug effects , Burkholderia cenocepacia/genetics , DNA Mutational Analysis , DNA Transposable Elements , Gene Knockout Techniques , Genetic Complementation Test , Mutagenesis, Insertional , Protein Interaction Mapping , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/physiology , Pyocins/isolation & purification
15.
Protein Sci ; 20(6): 986-95, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21416542

ABSTRACT

The biochemical characterization of the bacterial transcription cycle has been greatly facilitated by the production and characterization of targeted RNA polymerase (RNAP) mutants. Traditionally, RNAP preparations containing mutant subunits have been produced by reconstitution of denatured RNAP subunits, a process that is undesirable for biophysical and structural studies. Although schemes that afford the production of in vivo-assembled, recombinant RNAP containing amino acid substitutions, insertions, or deletions in either the monomeric ß or ß' subunits have been developed, there is no such system for the production of in vivo-assembled, recombinant RNAP with mutations in the homodimeric α-subunits. Here, we demonstrate a strategy to generate in vivo-assembled, recombinant RNAP preparations free of the α C-terminal domain. Furthermore, we describe a modification of this approach that would permit the purification of in vivo-assembled, recombinant RNAP containing any α-subunit variant, including those variants that are lethal. Finally, we propose that these related approaches can be extended to generate in vivo-assembled, recombinant variants of other protein complexes containing homomultimers for biochemical, biophysical, and structural analyses.


Subject(s)
DNA-Directed RNA Polymerases/chemistry , DNA-Directed RNA Polymerases/genetics , Escherichia coli/enzymology , Protein Engineering/methods , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , DNA-Directed RNA Polymerases/isolation & purification , Escherichia coli/genetics , Molecular Sequence Data , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/isolation & purification , Recombinant Proteins/isolation & purification , Sequence Deletion , Up-Regulation
16.
J Bacteriol ; 193(8): 1843-53, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21317335

ABSTRACT

The genome of Burkholderia cenocepacia contains two genes encoding closely related LysR-type transcriptional regulators, CysB and SsuR, involved in control of sulfur assimilation processes. In this study we show that the function of SsuR is essential for the utilization of a number of organic sulfur sources of either environmental or human origin. Among the genes upregulated by SsuR identified here are the tauABC operon encoding a predicted taurine transporter, three tauD-type genes encoding putative taurine dioxygenases, and atsA encoding a putative arylsulfatase. The role of SsuR in expression of these genes/operons was characterized through (i) construction of transcriptional reporter fusions to candidate promoter regions and analysis of their expression in the presence/absence of SsuR and (ii) testing the ability of SsuR to bind SsuR-responsive promoter regions. We also demonstrate that expression of SsuR-activated genes is not repressed in the presence of inorganic sulfate. A more detailed analysis of four SsuR-responsive promoter regions indicated that ~44 bp of the DNA sequence preceding and/or overlapping the predicted -35 element of such promoters is sufficient for SsuR binding. The DNA sequence homology among SsuR "recognition motifs" at different responsive promoters appears to be limited.


Subject(s)
Burkholderia cenocepacia/genetics , Burkholderia cenocepacia/metabolism , Gene Expression Regulation, Bacterial , Metabolic Networks and Pathways/genetics , Sulfur/metabolism , Transcription Factors/metabolism , Artificial Gene Fusion , Base Sequence , DNA Footprinting , Electrophoretic Mobility Shift Assay , Gene Expression Profiling , Genes, Reporter , Humans , Molecular Sequence Data , Operon , Promoter Regions, Genetic , Protein Binding
17.
Microbiology (Reading) ; 157(Pt 2): 349-361, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20966087

ABSTRACT

The opportunistic pathogen Burkholderia cenocepacia produces the siderophores ornibactin and pyochelin under iron-restricted conditions. Biosynthesis of both siderophores requires the involvement of non-ribosomal peptide synthetases (NRPSs). Using a transposon containing the lacZ reporter gene, two B. cenocepacia mutants were isolated which were deficient in siderophore production. Mutant IW10 was shown to produce normal amounts of ornibactin but only trace amounts of pyochelin, whereas synthesis of both siderophores was abolished in AHA27. Growth of AHA27, but not IW10, was inhibited under iron-restricted conditions. In both mutants, the transposon had integrated into the pobA gene, which encodes a polypeptide exhibiting similarity to the Sfp-type phosphopantetheinyltransferases (PPTases). These enzymes are responsible for activation of NRPSs by the covalent attachment of the 4'-phosphopantetheine (P-pant) moiety of coenzyme A. Previously characterized PPTase genes from other bacteria were shown to efficiently complement both mutants for siderophore production when provided in trans. The B. cenocepacia pobA gene was also able to efficiently complement an Escherichia coli entD mutant for production of the siderophore enterobactin. Using mutant IW10, in which the lacZ gene carried by the transposon is inserted in the same orientation as pobA, it was shown that pobA is not appreciably iron-regulated. Finally, we confirmed that Sfp-type bacterial PPTases can be subdivided into two distinct groups, and we present the amino acid signature sequences which characterize each of these groups.


Subject(s)
Bacterial Proteins/metabolism , Burkholderia cenocepacia/genetics , Siderophores/biosynthesis , Transferases (Other Substituted Phosphate Groups)/metabolism , Bacterial Proteins/genetics , Burkholderia cenocepacia/metabolism , DNA Transposable Elements , Escherichia coli/genetics , Genes, Bacterial , Genetic Complementation Test , Iron/metabolism , Mutagenesis, Insertional , Mutation , Phenols/metabolism , Thiazoles/metabolism , Transferases (Other Substituted Phosphate Groups)/genetics
18.
Virulence ; 1(6): 535-40, 2010.
Article in English | MEDLINE | ID: mdl-21178498

ABSTRACT

The recently identified bacterial type VI secretion system (T6SS) has rapidly become one of the most interesting areas of research in microbiology. In a relatively short period of time the relationship between the T6SS and the bacteriophage T4 tail and baseplate has been established. However, a number of questions concerning the T6SS remain the focus of a large number of researchers worldwide. Key questions that need to be addressed include how this system assembles in the cell envelope and the mechanism by which it translocates effector proteins across two membranes, the identification of such effectors and their function, how this secretion system contributes to virulence, interbacterial interactions and/or adaptation to the environment, and the evolutionary relationship between T6SS machine and bacteriophage T4. Focused on how the proteins constituting the secretion system interact, we recently identified a sub-complex of the T6SS comprised of four cell envelope proteins: the inner membrane-anchored TssL, TssM and TagL proteins and the outer membrane-associated TssJ lipoprotein. We further demonstrated that the TagL subunit carries a specific domain allowing anchorage of the secretion system to the peptidoglycan (PG) layer. Herein, we discuss these results, examine whether PG-binding motifs are found within other T6SS subunits and express hypotheses regarding the role of PG-binding motifs in type VI secretion.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems/physiology , Cell Membrane/metabolism , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/pathogenicity , Peptidoglycan/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/chemistry , Bacteriophage T4/metabolism , Cell Wall/metabolism , Gram-Negative Bacteria/ultrastructure , Protein Binding , Protein Structure, Tertiary , Virulence Factors
19.
J Bacteriol ; 191(19): 6094-104, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19633076

ABSTRACT

The Escherichia coli guaB promoter (P(guaB)) is responsible for directing transcription of the guaB and guaA genes, which specify the biosynthesis of the nucleotide GMP. P(guaB) is subject to growth rate-dependent control (GRDC) and possesses an UP element that is required for this regulation. In addition, P(guaB) contains a discriminator, three binding sites for the nucleoid-associated protein FIS, and putative binding sites for the regulatory proteins DnaA, PurR, and cyclic AMP receptor protein (CRP). Here we show that the CRP-cyclic AMP (cAMP) complex binds to a site located over 100 bp upstream of the guaB transcription start site, where it serves to downregulate P(guaB). The CRP-mediated repression of P(guaB) activity increases in media that support lower growth rates. Inactivation of the crp or cyaA gene or ablation/translocation of the CRP site relieves repression by CRP and results in a loss of GRDC of P(guaB). Thus, GRDC of P(guaB) involves a progressive increase in CRP-mediated repression of the promoter as the growth rate decreases. Our results also suggest that the CRP-cAMP complex does not direct GRDC at P(guaB) and that at least one other regulatory factor is required for conferring GRDC on this promoter. However, PurR and DnaA are not required for this regulatory mechanism.


Subject(s)
Cyclic AMP Receptor Protein/physiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Promoter Regions, Genetic/genetics , Base Sequence , Blotting, Western , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Cyclic AMP Receptor Protein/genetics , Cyclic AMP Receptor Protein/metabolism , DNA Footprinting , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Electrophoretic Mobility Shift Assay , Escherichia coli/growth & development , Escherichia coli Proteins/physiology , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/physiology , Molecular Sequence Data , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/physiology
20.
Microbiology (Reading) ; 154(Pt 6): 1729-1738, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18524927

ABSTRACT

The Escherichia coli guaB promoter (P(guaB)) regulates transcription of two genes, guaB and guaA, that are required for the synthesis of guanosine 5'-monophosphate (GMP), a precursor for the synthesis of guanine nucleoside triphosphates. Transcription from P(guaB) increases as a function of increasing cellular growth rate, and this is referred to as growth rate-dependent control (GRDC). Here we investigated the role of the factor for inversion stimulation (FIS) in the regulation of this promoter. The results showed that there are three binding sites for FIS centred near positions -11, +8 and +29 relative to the guaB transcription start site. Binding of FIS to these sites results in repression of P(guaB) in vitro but not in vivo. Deletion of the fis gene results in increased P(guaB) activity in vivo, but GRDC of P(guaB) is maintained.


Subject(s)
Down-Regulation , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Factor For Inversion Stimulation Protein/metabolism , Gene Expression Regulation, Bacterial , IMP Dehydrogenase/metabolism , Promoter Regions, Genetic/genetics , Binding Sites/genetics , DNA Footprinting , Deoxyribonuclease I/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Genes, Bacterial/genetics , IMP Dehydrogenase/genetics , Physical Chromosome Mapping
SELECTION OF CITATIONS
SEARCH DETAIL