Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Int Microbiol ; 23(4): 597-605, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32451737

ABSTRACT

Human infection by Legionella pneumophila (Lpn) only occurs via contaminated water from man-made sources, and eradication of these bacteria from man-made water systems is complicated by biofilm colonization. Using a continuously fed biofilm reactor model, we grew a biofilm consortium from potable water that was able to prolong recovery of Lpn CFU from biofilms. This effect was recreated using a subset of those species in a simplified consortium composed of eight bacterial isolates from the first biofilm reactor. In the reactor with the eight-species consortium, Lpn biofilm CFU was relatively stable over a 12-day trial. An isolate of Acidovorax from the consortium was, as a single species biofilm, able to promote Lpn surface attachment. Other isolates from the Pelomonas genus grew as equally robust biofilms alone, but did not promote surface attachment of Lpn. This attachment was disrupted by cationic polysaccharides and loss of the Lpn Lcl collagen-like adhesin protein. This work demonstrates that, while Lpn was fairly incompetent at attachment to surfaces to form a biofilm alone, pre-existing biofilms allowed attachment of Lpn as secondary colonizers. In addition, we demonstrate that initial attachment of Lpn to Acidovorax biofilms is likely via the Lcl-adhesin protein.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Biofilms/growth & development , Comamonadaceae/metabolism , Legionella pneumophila/metabolism , Adhesins, Bacterial/genetics , Drinking Water/microbiology , Humans , Polysaccharides, Bacterial/metabolism , Water Microbiology
2.
Curr Microbiol ; 75(6): 736-744, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29468303

ABSTRACT

Human infection by bacteria of the genus Legionella most often result in the pneumonia known as Legionnaires Disease. Legionella is found as a resident of adherent biofilms in man-made water systems. Disinfection efforts to prevent Legionella infections require a better understanding of the structures that promote Legionella surface attachment and biofilm colonization. Various enzymatic treatments, including multiple carbohydrate-targeting mixtures, failed to disrupt Legionella biofilms, despite the presence of carbohydrates in the biofilms as shown by biochemical methods and concanavalin-A lectin staining. Moreover, Legionella biofilms contained amyloids as detected by three microscopic staining methods (congo red, thioflavin T, and the amyloid-specific antibody WO2). Amyloid structures were seen in biofilms of both L. pneumophila and L. longbeachae, the two Legionella species most associated with human infection. Inhibition of amyloid assembly by congo red and thioflavin T limited both self-aggregation and surface attachment of L. pneumophila, indicating that functional amyloid structures have a key role in initial biofilm formation by these pathogenic bacteria.


Subject(s)
Legionella pneumophila/metabolism , Amyloid/metabolism , Biofilms , Water Microbiology
3.
Biochemistry ; 56(51): 6734-6742, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29190068

ABSTRACT

Campylobacter jejuni is the most common bacterial cause of gastroenteritis and a major contributor to infant mortality in the developing world. The increasing incidence of antibiotic-resistant C. jejuni only adds to the urgency to develop effective therapies. Because of the essential role that polyamines play, particularly in protection from oxidative stress, enzymes involved in the biosynthesis of these metabolites are emerging as promising antibiotic targets. The recent description of an alternative pathway for polyamine synthesis, distinct from that in human cells, in C. jejuni suggests this pathway could be a target for novel therapies. To that end, we determined X-ray crystal structures of C. jejuni agmatine deiminase (CjADI) and demonstrated that loss of CjADI function contributes to antibiotic sensitivity, likely because of polyamine starvation. The structures provide details of key molecular features of the active site of this protein. Comparison of the unliganded structure (2.1 Å resolution) to that of the CjADI-agmatine complex (2.5 Å) reveals significant structural rearrangements that occur upon substrate binding. The shift of two helical regions of the protein and a large conformational change in a loop near the active site generate a narrow binding pocket around the bound substrate. This change optimally positions the substrate for catalysis. In addition, kinetic analysis of this enzyme demonstrates that CjADI is an iminohydrolase that effectively deiminates agmatine. Our data suggest that C. jejuni agmatine deiminase is a potentially important target for combatting antibiotic resistance, and these results provide a valuable framework for guiding future drug development.


Subject(s)
Campylobacter jejuni/enzymology , Drug Resistance, Bacterial/drug effects , Hydrolases/antagonists & inhibitors , Aminoglycosides/pharmacology , Campylobacter jejuni/drug effects , Catalytic Domain , Crystallography, X-Ray , Hydrolases/chemistry , Hydrolases/genetics , Hydrolases/metabolism , Kinetics , Protein Conformation
4.
Infect Immun ; 81(11): 4182-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23980114

ABSTRACT

Iron acquisition is critical to the growth and virulence of Legionella pneumophila. Previously, we found that L. pneumophila uses both a ferrisiderophore pathway and ferrous iron transport to obtain iron. We now report that two molecules secreted by L. pneumophila, homogentisic acid (HGA) and its polymerized variant (HGA-melanin, a pyomelanin), are able to directly mediate the reduction of various ferric iron salts. Furthermore, HGA, synthetic HGA-melanin, and HGA-melanin derived from bacterial supernatants enhanced the ability of L. pneumophila and other species of Legionella to take up radiolabeled iron. Enhanced iron uptake was not observed with a ferrous iron transport mutant. Thus, HGA and HGA-melanin mediate ferric iron reduction, with the resulting ferrous iron being available to the bacterium for uptake. Upon further testing of L. pneumophila culture supernatants, we found that significant amounts of ferric and ferrous iron were associated with secreted HGA-melanin. Importantly, a pyomelanin-containing fraction obtained from a wild-type culture supernatant was able to stimulate the growth of iron-starved legionellae. That the corresponding supernatant fraction obtained from a nonpigmented mutant culture did not stimulate growth demonstrated that HGA-melanin is able to both promote iron uptake and enhance growth under iron-limiting conditions. Indicative of a complementary role in iron acquisition, HGA-melanin levels were inversely related to the levels of siderophore activity. Compatible with a role in the ecology and pathogenesis of L. pneumophila, HGA and HGA-melanin were effective at reducing and releasing iron from both insoluble ferric hydroxide and the mammalian iron chelates ferritin and transferrin.


Subject(s)
Iron/metabolism , Legionella pneumophila/growth & development , Legionella pneumophila/metabolism , Melanins/metabolism , Homogentisic Acid/metabolism , Iron Compounds/metabolism , Iron Radioisotopes/metabolism , Isotope Labeling , Oxidation-Reduction
5.
Methods Mol Biol ; 954: 151-62, 2013.
Article in English | MEDLINE | ID: mdl-23150393

ABSTRACT

This chapter describes methods for culturing Legionella pneumophila in both complex and defined media. The first protocol describes the use of buffered charcoal yeast extract (BCYE) agar, the solid medium that is most commonly used for culturing L. pneumophila. The next procedure details the cultivation of L. pneumophila in buffered yeast extract (BYE) broth, i.e., the liquid medium version of BCYE agar. We describe how culturing in BYE broth can also be used for investigating proteins that are secreted by the type II secretion system of L. pneumophila. The next part of the chapter explains the cultivation of L. pneumophila in a chemically defined liquid media (CDM). CDM contains a mixture of amino acids, metals, α-ketoglutarate, and pyruvate. Because of its defined nature, CDM provides a simple means for controlling the concentration of nutrients and thereby allows for investigations of physiology and metabolism. To illustrate this point, the use of deferrated CDM for the purpose of assessing Legionella siderophore production is outlined. Finally, the chapter ends with a brief discussion of the storage and shipping of L. pneumophila.


Subject(s)
Culture Media , Legionella pneumophila/growth & development , Enzyme Activation , Legionella pneumophila/metabolism , Peptide Hydrolases/metabolism , Preservation, Biological , Siderophores/metabolism , Specimen Handling
6.
PLoS One ; 7(9): e46209, 2012.
Article in English | MEDLINE | ID: mdl-23049981

ABSTRACT

BACKGROUND: Legionella pneumophila is a pathogenic bacterium that can cause Legionnaires' disease and other non-pneumonic infections in humans. This bacterium produces a pyomelanin pigment, a potential virulence factor with ferric reductase activity. In this work, we have investigated the role of phenylalanine hydroxylase from L. pneumophila (lpPAH), the product of the phhA gene, in the synthesis of the pyomelanin pigment and the growth of the bacterium in defined compositions. METHODOLOGY/PRINCIPAL FINDINGS: Comparative studies of wild-type and phhA mutant corroborate that lpPAH provides the excess tyrosine for pigment synthesis. phhA and letA (gacA) appear transcriptionally linked when bacteria were grown in buffered yeast extract medium at 37°C. phhA is expressed in L. pneumophila growing in macrophages. We also cloned and characterized lpPAH, which showed many characteristics of other PAHs studied so far, including Fe(II) requirement for activity. However, it also showed many particular properties such as dimerization, a high conformational thermal stability, with a midpoint denaturation temperature (T(m)) = 79 ± 0.5°C, a high specific activity at 37°C (10.2 ± 0.3 µmol L-Tyr/mg/min) and low affinity for the substrate (K(m) (L-Phe) = 735 ± 50 µM. CONCLUSIONS/SIGNIFICANCE: lpPAH has a major functional role in the synthesis of pyomelanin and promotes growth in low-tyrosine media. The high thermal stability of lpPAH might reflect the adaptation of the enzyme to withstand relatively high survival temperatures.


Subject(s)
Legionella pneumophila/enzymology , Melanins/biosynthesis , Phenylalanine Hydroxylase/metabolism , Bacterial Proteins , Enzyme Stability , Temperature , Tyrosine/metabolism
7.
Microbiology (Reading) ; 158(Pt 3): 721-735, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22160401

ABSTRACT

The Gram-negative bacterium Legionella pneumophila elaborates the siderophore legiobactin. We previously showed that cytoplasmic LbtA helps mediate legiobactin synthesis, inner-membrane LbtB promotes export of legiobactin, and outer-membrane LbtU acts as the ferrisiderophore receptor. RT-PCR analyses now identified lbtC as an iron-repressed gene that is the final gene in an operon containing lbtA and lbtB. In silico analysis predicted that LbtC is an inner-membrane protein that belongs to the major facilitator superfamily (MFS). Although capable of normal growth in standard media, lbtC mutants were defective for growth on iron-depleted agar media. While producing normal levels of legiobactin, lbtC mutants were unable to utilize supplied legiobactin to stimulate growth on iron-depleted media and displayed an impaired ability to take up radiolabelled iron. All lbtC mutant phenotypes were complemented by reintroduction of an intact copy of lbtC. When a cloned copy of both lbtC and lbtU was introduced into a heterologous bacterium (Legionella longbeachae), the organism acquired the ability to utilize legiobactin to grow better on low-iron media. Together, these data indicate that LbtC is involved in the uptake of legiobactin, and based upon its predicted location is most likely the mediator of ferrilegiobactin transport across the inner membrane. The data are also a unique documentation of how an MFS protein can promote bacterial iron-siderophore import, standing in contrast to the vast majority of studies which have defined ABC-type permeases as the mediators of siderophore import across the Gram-negative inner membrane or the Gram-positive cytoplasmic membrane.


Subject(s)
Bacterial Proteins/metabolism , Iron/metabolism , Legionella pneumophila/metabolism , Membrane Proteins/metabolism , Culture Media/chemistry , Gene Deletion , Genetic Complementation Test , Legionella longbeachae/growth & development , Legionella longbeachae/metabolism , Legionella pneumophila/growth & development , Membrane Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
J Bacteriol ; 193(7): 1563-75, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21278293

ABSTRACT

Gram-negative Legionella pneumophila produces a siderophore (legiobactin) that promotes lung infection. We previously determined that lbtA and lbtB are required for the synthesis and secretion of legiobactin. DNA sequence and reverse transcription-PCR (RT-PCR) analyses now reveal the presence of an iron-repressed gene (lbtU) directly upstream of the lbtAB-containing operon. In silico analysis predicted that LbtU is an outer membrane protein consisting of a 16-stranded transmembrane ß-barrel, multiple extracellular domains, and short periplasmic tails. Immunoblot analysis of cell fractions confirmed an outer membrane location for LbtU. Although replicating normally in standard media, lbtU mutants, like lbtA mutants, were impaired for growth on iron-depleted agar media. While producing typical levels of legiobactin, lbtU mutants were unable to use supplied legiobactin to stimulate growth on iron-depleted media and displayed an inability to take up iron. Complemented lbtU mutants behaved as the wild type did. The lbtU mutants were also impaired for infection in a legiobactin-dependent manner. Together, these data indicate that LbtU is involved in the uptake of legiobactin and, based upon its location, is most likely the Legionella siderophore receptor. The sequence and predicted two-dimensional (2D) and 3D structures of LbtU were distinct from those of all known siderophore receptors, which generally contain a 22-stranded ß-barrel and an extended N terminus that binds TonB in order to transduce energy from the inner membrane. This observation coupled with the fact that L. pneumophila does not encode TonB suggests that LbtU is a new type of receptor that participates in a form of iron uptake that is mechanistically distinct from the existing paradigm.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation/physiology , Legionella pneumophila/metabolism , Siderophores/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Iron/metabolism , Legionella pneumophila/genetics , Mutation , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Siderophores/genetics
9.
Infect Immun ; 77(7): 2887-95, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398549

ABSTRACT

When cultured in a low-iron medium, Legionella pneumophila secretes a siderophore (legiobactin) that is both reactive in the chrome azurol S (CAS) assay and capable of stimulating the growth of iron-starved legionellae. Using anion-exchange high-pressure liquid chromatography (HPLC), we purified legiobactin from culture supernatants of a virulent strain of L. pneumophila. In the process, we detected the ferrated form of legiobactin as well as other CAS-reactive substances. Purified legiobactin had a yellow-gold color and absorbed primarily from 220 nm and below. In accordance, nuclear magnetic resonance spectroscopy revealed that legiobactin lacks aromatic carbons, and among the 13 aliphatics present, there were 3 carbonyls. When examined by HPLC, supernatants from L. pneumophila mutants inactivated for lbtA and lbtB completely lacked legiobactin, indicating that the LbtA and LbtB proteins are absolutely required for siderophore activity. Independently derived lbtA mutants, but not a complemented derivative, displayed a reduced ability to infect the lungs of A/J mice after intratracheal inoculation, indicating that legiobactin is required for optimal intrapulmonary survival by L. pneumophila. This defect, however, was not evident when the lbtA mutant and its parental strain were coinoculated into the lung, indicating that legiobactin secreted by the wild type can promote growth of the mutant in trans. Legiobactin mutants grew normally in murine lung macrophages and alveolar epithelial cells, suggesting that legiobactin promotes something other than intracellular infection of resident lung cells. Overall, these data represent the first documentation of a role for siderophore expression in the virulence of L. pneumophila.


Subject(s)
Bacterial Proteins/isolation & purification , Bacterial Proteins/physiology , Legionella pneumophila/pathogenicity , Legionnaires' Disease/microbiology , Virulence Factors/isolation & purification , Virulence Factors/physiology , Animals , Bacterial Proteins/chemistry , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Colony Count, Microbial , Epithelial Cells/microbiology , Female , Gene Deletion , Genes, Bacterial , Genetic Complementation Test , Lung/microbiology , Macrophages, Alveolar/microbiology , Magnetic Resonance Spectroscopy , Mice , Microbial Viability , Spectrophotometry , Virulence
10.
Infect Immun ; 75(8): 4062-70, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17548481

ABSTRACT

The virulence of Legionella pneumophila is dependent upon its capacity to acquire iron. To identify genes involved in expression of its siderophore, we screened a mutagenized population of L. pneumophila for strains that were no longer able to rescue the growth of a ferrous transport mutant. However, an unusual mutant was obtained that displayed a strong inhibitory effect on the feoB mutant. Due to an insertion in hmgA that encodes homogentisate 1,2-dioxygenase, the mutant secreted increased levels of pyomelanin, the L. pneumophila pigment that is derived from secreted homogentisic acid (HGA). Thus, we hypothesized that L. pneumophila-secreted HGA-melanin has intrinsic ferric reductase activity, converting Fe(3+) to Fe(2+), but that hyperpigmentation results in excessive reduction of iron that can, in the case of the feoB mutant, be inhibitory to growth. In support of this hypothesis, we demonstrated, for the first time, that wild-type L. pneumophila secretes ferric reductase activity. Moreover, whereas the hyperpigmented mutant had increased secreted activity, an lly mutant specifically impaired for pigment production lacked the activity. Compatible with the nature of HGA-melanins, the secreted ferric reductase activity was positively influenced by the amount of tyrosine in the growth medium, resistant to protease, acid precipitable, and heterogeneous in size. Together, these data represent the first demonstration of pyomelanin-mediated ferric reduction by a pathogenic bacterium.


Subject(s)
FMN Reductase/metabolism , Legionella pneumophila/enzymology , Melanins/metabolism , Bacterial Proteins/genetics , DNA Transposable Elements , FMN Reductase/chemistry , Genetic Complementation Test , Melanins/chemistry , Molecular Weight , Mutagenesis, Insertional , Peptide Hydrolases/metabolism
11.
Microbiology (Reading) ; 151(Pt 2): 625-631, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15699211

ABSTRACT

Streptococcus mutans is the primary odontopathogen present in supragingival plaque and causes the oral disease known as dental caries. Colonization of the oral cavity by S. mutans requires the bacteria to adhere to the tooth surface and occurs by both sucrose-dependent and -independent mechanisms. Sucrose-independent adhesion of S. mutans in vitro has been shown to involve an ORF (ORF0317) encoding a homologue (39 %) to LytR, a regulator of autolysin activity in Bacillus subtilis. The protein encoded by ORF0317, LytR, belongs to the LytR/CpsA/Psr protein family. This family has a putative role in cell-wall structural maintenance, possibly through autolysin regulation. Autolysins have also been shown to be important in surface adhesion in Lactococcus lactis and in the pathogenic properties of Streptococcus pneumoniae. To investigate the role of autolysins in the adhesion and pathogenesis of S. mutans, a LytR mutant was constructed. The mutant grows in long chains, which may indicate a defect in cell division. Further experiments with the mutant strain show increased autolytic activity, indicating that LytR attenuates S. mutans autolytic activity, possibly through regulation of the expression of autolytic enzymes. No defect in cell-to-surface adherence or biofilm growth was seen in the LytR mutant. However, a connection between cell growth phase and transcription of lytR was found.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Streptococcus mutans/growth & development , Streptococcus mutans/physiology , Transcription Factors/metabolism , Amino Acid Sequence , Bacterial Adhesion , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Division , Humans , Molecular Sequence Data , Mutation , Streptococcus mutans/genetics , Streptococcus mutans/pathogenicity , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...