Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29866904

ABSTRACT

Salmonella enterica serovars are associated with diarrhea and gastroenteritis and are a helpful model for understanding host-pathogen mechanisms. Salmonella enterica serovar Typhimurium regulates the distribution of O antigen (OAg) and presents a trimodal distribution based on Wzy polymerase and the WzzST (long-chain-length OAg [L-OAg]) and WzzfepE (very-long-chain-length OAg [VL-OAg]) copolymerases; however, several mechanisms regulating this process remain unclear. Here, we report that LPS modifications modulate the infectious process and that OAg chain length determination plays an essential role during infection. An increase in VL-OAg is dependent on Wzy polymerase, which is promoted by a growth condition resembling the environment of Salmonella-containing vacuoles (SCVs). The virulence- and stress-related periplasmic protein (VisP) participates in OAg synthesis, as a ΔvisP mutant presents a semirough OAg phenotype. The ΔvisP mutant has greatly decreased motility and J774 macrophage survival in a colitis model of infection. Interestingly, the phenotype is restored after mutation of the wzzST or wzzfepE gene in a ΔvisP background. Loss of both the visP and wzzST genes promotes an imbalance in flagellin secretion. L-OAg may function as a shield against host immune systems in the beginning of an infectious process, and VL-OAg protects bacteria during SCV maturation and facilitates intramacrophage replication. Taken together, these data highlight the roles of OAg length in generating phenotypes during S Typhimurium pathogenesis and show the periplasmic protein VisP as a novel protein in the OAg biosynthesis pathway.


Subject(s)
Bacterial Proteins/metabolism , O Antigens/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/metabolism , Animals , Bacterial Load , Cell Line , Colitis/microbiology , Colitis/pathology , Disease Models, Animal , Feces/microbiology , Female , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Microbial Viability , Phagocytosis
2.
Nature ; 492(7427): 113-7, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23160491

ABSTRACT

The mammalian gastrointestinal tract provides a complex and competitive environment for the microbiota. Successful colonization by pathogens requires scavenging nutrients, sensing chemical signals, competing with the resident bacteria and precisely regulating the expression of virulence genes. The gastrointestinal pathogen enterohaemorrhagic Escherichia coli (EHEC) relies on inter-kingdom chemical sensing systems to regulate virulence gene expression. Here we show that these systems control the expression of a novel two-component signal transduction system, named FusKR, where FusK is the histidine sensor kinase and FusR the response regulator. FusK senses fucose and controls expression of virulence and metabolic genes. This fucose-sensing system is required for robust EHEC colonization of the mammalian intestine. Fucose is highly abundant in the intestine. Bacteroides thetaiotaomicron produces multiple fucosidases that cleave fucose from host glycans, resulting in high fucose availability in the gut lumen. During growth in mucin, B. thetaiotaomicron contributes to EHEC virulence by cleaving fucose from mucin, thereby activating the FusKR signalling cascade, modulating the virulence gene expression of EHEC. Our findings suggest that EHEC uses fucose, a host-derived signal made available by the microbiota, to modulate EHEC pathogenicity and metabolism.


Subject(s)
Bacterial Proteins/metabolism , Bacteroides/metabolism , Enterohemorrhagic Escherichia coli/growth & development , Fucose/metabolism , Gastrointestinal Tract/microbiology , Animals , Bacteroides/enzymology , Bacteroides/growth & development , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Bacterial , Mucins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Rabbits , Receptors, Adrenergic/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence/genetics , Virulence Factors/genetics , alpha-L-Fucosidase/metabolism
3.
Infect Immun ; 85(12)2017 12.
Article in English | MEDLINE | ID: mdl-28947641

ABSTRACT

Intestinal bacteria employ microbial metabolites from the microbiota and chemical signaling during cell-to-cell communication to regulate several cellular functions. Pathogenic bacteria are extremely efficient in orchestrating their response to these signals through complex signaling transduction systems. Precise coordination and interpretation of these multiple chemical cues is important within the gastrointestinal (GI) tract. Enteric foodborne pathogens, such as enterohemorrhagic Escherichia coli (EHEC) and Salmonella enterica serovar Typhimurium, or the surrogate murine infection model for EHEC, Citrobacter rodentium, are all examples of microorganisms that modulate the expression of their virulence repertoire in response to signals from the microbiota or the host, such as autoinducer-3 (AI-3), epinephrine (Epi), and norepinephrine (NE). The QseBC and QseEF two-component systems, shared by these pathogens, are involved in sensing these signals. We review how these signaling systems sense and relay these signals to drive bacterial gene expression; specifically, to modulate virulence. We also review how bacteria chat via chemical signals integrated with metabolite recognition and utilization to promote successful associations among enteric pathogens, the microbiota, and the host.


Subject(s)
Citrobacter rodentium/drug effects , Escherichia coli/drug effects , Gastrointestinal Microbiome , Host-Pathogen Interactions , Salmonella typhimurium/drug effects , Signal Transduction , Virulence Factors/biosynthesis , Animals , Mice
4.
Adv Exp Med Biol ; 874: 247-61, 2016.
Article in English | MEDLINE | ID: mdl-26589223

ABSTRACT

Epinephrine/norepinephrine/AI-3 signaling is used as an interkingdom chemical signaling system between microbes and their hosts. This system is also exploited by pathogens to regulate virulence traits. In enterohemorrhagic E. coli (EHEC) O157:H7, it is essential for pathogenesis and flagella motility. These three signals activate expression of a pathogenicity island named locus of enterocyte effacement (LEE), Shiga toxin, and the flagella regulon. These signals are sensed by the two-component system QseBC, whereas the bacterial membrane receptor QseC autophosphorylates and phosphorylates the QseB response regulator initiating a complex phosphorelay signaling cascade that activates the expression of a second two-component system, QseEF. The QseEF two-component system is also involved in the expression of the virulence genes, and it senses epinephrine, phosphate, and sulfate. This complex signaling cascade still needs to be completely elucidated.


Subject(s)
Epinephrine/physiology , Escherichia coli O157/pathogenicity , Norepinephrine/physiology , Signal Transduction/physiology , Animals , Escherichia coli Proteins/physiology , Humans , Lactones , Phosphoproteins/physiology , Quorum Sensing , Receptors, Adrenergic/physiology , Virulence
5.
Proc Natl Acad Sci U S A ; 110(4): 1470-5, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23302685

ABSTRACT

Gram-negative bacteria have an outer membrane containing LPS. LPS is constituted of an oligosaccharide portion and a lipid-A moiety that embeds this molecule within the outer membrane. LPS is a pathogen-associated molecular pattern, and several pathogens modify their lipid-A as a stealth strategy to avoid recognition by the innate immune system and gain resistance to host factors that disrupt the bacterial cell envelope. An essential feature of Salmonella enterica Typhimurium pathogenesis is its ability to replicate within vacuoles in professional macrophages. S. Typhimurium modifies its lipid-A by hydroxylation by the Fe2+/α-ketoglutarate-dependent dioxygenase enzyme (LpxO). Here, we show that a periplasmic protein of the bacterial oligonucleotide/oligosaccharide-binding fold family, herein named virulence and stress-related periplasmic protein (VisP), on binding to the sugar moiety of peptidoglycan interacts with LpxO. This interaction inhibits LpxO function, leading to decreased LpxO-dependent lipid-A modifications and increasing resistance to stressors within the vacuole environment during intramacrophage replication promoting systemic disease. Consequently, ΔvisP is avirulent in systemic murine infections, where VisP acts through LpxO. Several Gram-negative pathogens harbor both VisP and LpxO, suggesting that this VisP-LpxO mechanism of lipid-A modifications has broader implications in bacterial pathogenesis. Bacterial species devoid of LpxO (e.g., Escherichia coli) have no lipid-A phenotypes associated with the lack of VisP; however, VisP also controls LpxO-independent phenotypes. VisP and LpxO act independently in the S. Typhimurium murine colitis model, with both mutants being attenuated for diverging reasons; ΔvisP is less resistant to cationic antimicrobial peptides, whereas ΔlpxO is deficient for epithelial cell invasion. VisP converges bacterial cell wall homeostasis, stress responses, and pathogenicity.


Subject(s)
Bacterial Proteins/physiology , Host-Pathogen Interactions/physiology , Periplasmic Proteins/physiology , Salmonella typhimurium/pathogenicity , Virulence Factors/physiology , Amino Acid Sequence , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Line , Female , Genes, Bacterial , HeLa Cells , Host-Pathogen Interactions/genetics , Humans , Lipid A/chemistry , Lipid A/metabolism , Macrophages/microbiology , Macrophages/physiology , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Periplasmic Proteins/chemistry , Periplasmic Proteins/genetics , Regulon , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/physiopathology , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Sequence Homology, Amino Acid , Virulence/genetics , Virulence/physiology , Virulence Factors/chemistry , Virulence Factors/genetics
6.
Int Braz J Urol ; 41(1): 67-77, 2015.
Article in English | MEDLINE | ID: mdl-25928511

ABSTRACT

PURPOSE: The treatment of urinary tract infections (UTI) with antibiotics is commonly used, but recurrence and antibiotic resistance have been growing and concerning clinicians. We studied whether the rapid onset of a protective biofilm may be responsible for the lack of effectiveness of antibiotics against selected bacteria. MATERIALS AND METHODS: Two established uropathogenic Escherichia coli strains, UTI89 and CFT073, and two Pseudomonas aeruginosa strains, PA01 and Boston-41501, were studied to establish a reliable biofilm formation process. Bacterial growth (BG) was determined by optical density at 600 nm (OD 600) using a spectrophotometer, while biofilm formation (BF) using crystal violet staining was measured at OD 550. Next, these bacterial strains were treated with clinically relevant antibiotics, ciprofloxacin HCl (200 ng/mL and 2 µg/mL), nitrofurantoin (20 µg/mL and 40 µg/mL) and ampicillin (50 µg/mL) at time points of 0 (T0) or after 6 hours of culture (T6). All measurements, including controls (bacteria -1% DMSO), were done in triplicates and repeated three times for consistency. RESULTS: The tested antibiotics effectively inhibited both BG and BF when administered at T0 for UPEC strains, but not when the antibiotic administration started 6 hours later. For Pseudomonas strains, only Ciprofloxacin was able to significantly inhibit bacterial growth at T0 but only at the higher concentration of 2 µg/mL for T6. CONCLUSION: When established UPEC and Pseudomonas bacteria were allowed to culture for 6 hours before initialization of treatment, the therapeutic effect of selected antibiotics was greatly suppressed when compared to immediate treatment, probably as a result of the protective nature of the biofilm.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biofilms/drug effects , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Ampicillin/administration & dosage , Anti-Infective Agents, Urinary , Biofilms/growth & development , Ciprofloxacin/administration & dosage , Drug Resistance, Bacterial , Escherichia coli/physiology , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Nitrofurantoin/administration & dosage , Pseudomonas aeruginosa/physiology , Reference Values , Reproducibility of Results , Time Factors , Urinary Tract Infections/drug therapy
7.
Infect Immun ; 80(12): 4344-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23027532

ABSTRACT

The bacterial adrenergic sensor kinases QseC and QseE respond to epinephrine and/or norepinephrine to initiate a complex phosphorelay regulatory cascade that modulates virulence gene expression in several pathogens. We have previously shown that QseC activates virulence gene expression in Salmonella enterica serovar Typhimurium. Here we report the role of QseE in S. Typhimurium pathogenesis as well as the interplay between these two histidine sensor kinases in gene regulation. An S. Typhimurium qseE mutant is hampered in the invasion of epithelial cells and intramacrophage replication. The ΔqseC strain is highly attenuated for intramacrophage survival but has only a minor defect in invasion. However, the ΔqseEC strain has only a slight attenuation in invasion, mirroring the ΔqseC strain, and has an intermediary intramacrophage replication defect in comparison to the ΔqseE and ΔqseC strains. The expressions of the sipA and sopB genes, involved in the invasion of epithelial cells, are activated by epinephrine via QseE. The expression levels of these genes are still decreased in the ΔqseEC double mutant, albeit to a lesser extent, congruent with the invasion phenotype of this mutant. The expression level of the sifA gene, important for intramacrophage replication, is decreased in the qseE mutant and the ΔqseEC double mutant grown in vitro. However, as previously reported by us, the epinephrine-dependent activation of this gene occurs via QseC. In the systemic model of S. Typhimurium infection of BALB/c mice, the qseC and qseE mutants are highly attenuated, while the double mutant has an intermediary phenotype. Altogether, these data suggest that both adrenergic sensors play an important role in modulating several aspects of S. Typhimurium pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Protein Kinases/metabolism , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/genetics , Cell Line , Epinephrine/pharmacology , Epithelial Cells/microbiology , Female , HeLa Cells , Histidine Kinase , Humans , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Mutation , Protein Kinases/genetics , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/mortality , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics , Virulence
8.
Braz J Microbiol ; 53(2): 557-564, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35303296

ABSTRACT

Salmonella Typhimurium is a pathogen of clinical relevance and a model of study in host-pathogen interactions. The virulence and stress-related periplasmic protein VisP is important during S. Typhimurium pathogenesis. It supports bacteria invading host cells, surviving inside macrophages, swimming, and succeeding in murine colitis model, O-antigen assembly, and responding to cationic antimicrobial peptides. This study aimed to investigate the role of the O-antigen molecular ruler WzzST and the periplasmic protein VisP in swarming motility and osmotic stress response. Lambda red mutagenesis was performed to generate single and double mutants, followed by swarming motility, qRT-PCR, Western blot, and growth curves. Here we demonstrate that the deletion of visP affects swarming under osmotic stress and changes the expression levels of genes responsible for chemotaxis, flagella assembly, and general stress response. The deletion of the gene encoding for the O-antigen co-polymerase wzzST increases swarming motility but not under osmotic stress. A second mutation in O-antigen co-polymerase wzzST in a ΔvisP background affected gene expression levels. The ΔvisP growth was affected by sodium and magnesium levels on N-minimum media. These data indicate that WzzST has a role in swarming the motility of S. Typhimurium, as the VisP is involved in chemotaxis and osmotic stress, specifically in response to MgCl2 and NaCl.


Subject(s)
O Antigens , Salmonella typhimurium , Animals , Bacterial Proteins/metabolism , Chemotaxis/genetics , Flagella/physiology , Mice , O Antigens/genetics , O Antigens/metabolism , Osmoregulation
9.
Braz J Microbiol ; 52(3): 1067-1075, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34105110

ABSTRACT

Urinary tract infections (UTIs) are a major public health concern in both community and hospital settings worldwide. Uropathogenic Escherichia coli (UPEC) is the main causative agent of UTI and increasingly associated with antibiotic resistance. Herein, we report the draft genome sequence of 9 fluoroquinolone-resistant UPEC isolates from Brazil and examine selected major phenotypic features, such as antimicrobial resistance profile, phylogroup, serotype, sequence type (ST), virulence genes, and resistance marks. Besides the quinolone resistance, beta-lactams, ESBL production, aminoglycosides, and tetracycline resistance were observed. High prevalence of 20 virulence genes was detected in all isolates, such as those encoding type 1 fimbriae, acid tolerance system, and hemolysin E, particularly within E. coli B2 phylogroup, as ST131 and ST1193 strains, among other genomic analyses as genomic islands, resistance plasmids, and integron identification.


Subject(s)
Escherichia coli Infections , Genome, Bacterial , Urinary Tract Infections , Uropathogenic Escherichia coli , Brazil , Drug Resistance, Bacterial , Escherichia coli Infections/microbiology , Fluoroquinolones/pharmacology , Humans , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/drug effects , Uropathogenic Escherichia coli/genetics , Virulence Factors/genetics
10.
Infect Immun ; 78(3): 914-26, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028809

ABSTRACT

The autoinducer-3 (AI-3)/epinephrine (Epi)/norepinephrine (NE) interkingdom signaling system mediates chemical communication between bacteria and their mammalian hosts. The three signals are sensed by the QseC histidine kinase (HK) sensor. Salmonella enterica serovar Typhimurium is a pathogen that uses HKs to sense its environment and regulate virulence. Salmonella serovar Typhimurium invades epithelial cells and survives within macrophages. Invasion of epithelial cells is mediated by the type III secretion system (T3SS) encoded in Salmonella pathogenicity island 1 (SPI-1), while macrophage survival and systemic disease are mediated by the T3SS encoded in SPI-2. Here we show that QseC plays an important role in Salmonella serovar Typhimurium pathogenicity. A qseC mutant was impaired in flagellar motility, in invasion of epithelial cells, and in survival within macrophages and was attenuated for systemic infection in 129x1/SvJ mice. QseC acts globally, regulating expression of genes within SPI-1 and SPI-2 in vitro and in vivo (during infection of mice). Additionally, dopamine beta-hydroxylase knockout (Dbh(-)(/)(-)) mice that do not produce Epi or NE showed different susceptibility to Salmonella serovar Typhimurium infection than wild-type mice. These data suggest that the AI-3/Epi/NE signaling system is a key factor during Salmonella serovar Typhimurium pathogenesis in vitro and in vivo. Elucidation of the role of this interkingdom signaling system in Salmonella serovar Typhimurium should contribute to a better understanding of the complex interplay between the pathogen and the host during infection.


Subject(s)
Gene Expression Regulation, Bacterial , Protein Kinases/physiology , Salmonella typhimurium/pathogenicity , Trans-Activators/physiology , Virulence Factors/biosynthesis , Animals , Cell Line , Colony Count, Microbial , Epithelial Cells/microbiology , Gene Expression Profiling , Gene Knockout Techniques , Genomic Islands , Histidine Kinase , Humans , Liver/microbiology , Locomotion , Macrophages/microbiology , Mice , Mice, Knockout , Protein Kinases/genetics , Salmonella Infections, Animal/microbiology , Spleen/microbiology , Survival Analysis , Trans-Activators/genetics , Virulence
11.
J Inorg Biochem ; 212: 111247, 2020 11.
Article in English | MEDLINE | ID: mdl-32920435

ABSTRACT

The aims of this work were to evaluate the antibacterial and antiproliferative potential in vitro of the metal complex with 4-aminobenzoic acid (Ag-pABA) and a drug delivery system based on bacterial cellulose (BC-Ag-pABA). The Ag-pABA complex was characterized by elemental analysis, high resolution mass spectrometry and single-crystal X-ray diffraction techniques, which indicated a 1:2 metal/pABA composition plus a nitrate ion coordinated to silver by the oxygen atom, with the coordination formula [Ag (C7H7NO2)2(NO3)]. The coordination of pABA to the silver ion occurred by the nitrogen atom. The in vitro antibacterial activity of the complex evaluated by minimum inhibitory concentration assays demonstrated the effective growth inhibitory activity against Gram-positive, Gram-negative biofilm producers and acid-alcohol resistant Bacillus. The antiproliferative activities against a panel of eight tumor cells demonstrated the activity of the complex with a significant selectivity index (SI). The DNA interaction capacity and the Ames Test indicated the absence of mutagenicity. The BC-Ag-pABA composite showed an effective capacity of sustained release of Ag-pABA. The observed results validate further studies on its mechanisms of action and the conditions that mediate the in vivo biological effects using animal models to confirm its safety and effectiveness for treatment of skin and soft tissues infected by bacterial pathogens, urinary tract infections and cancer.


Subject(s)
4-Aminobenzoic Acid/chemistry , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Proliferation/drug effects , Cellulose/chemistry , Silver/pharmacology , Anti-Bacterial Agents/chemistry , Delayed-Action Preparations , Microbial Sensitivity Tests , Silver/chemistry
12.
FEMS Immunol Med Microbiol ; 51(2): 319-26, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17714491

ABSTRACT

Proteus mirabilis is an important cause of urinary tract infection (UTI) in patients with complicated urinary tracts. Thirty-five strains of P. mirabilis isolated from UTI were examined for the adherence capacity to epithelial cells. All isolates displayed the aggregative adherence (AA) to HEp-2 cells, a phenotype similarly presented in LLC-MK(2) cells. Biofilm formation on polystyrene was also observed in all strains. The mannose-resistant Proteus-like fimbriae (MR/P), Type I fimbriae and AAF/I, II and III fimbriae of enteroaggregative Escherichia coli were searched by the presence of their respective adhesin-encoding genes. Only the MR/P fimbrial subunits encoding genes mrpA and mrpH were detected in all isolates, as well as MR/P expression. A mutation in mrpA demonstrated that MR/P is involved in aggregative adherence to HEp-2 cells, as well as in biofilm formation. However, these phenotypes are multifactorial, because the mrpA mutation reduced but did not abolish both phenotypes. The present results reinforce the importance of MR/P as a virulence factor in P. mirabilis due to its association with AA and biofilm formation, which is an important step for the establishment of UTI in catheterized patients.


Subject(s)
Bacterial Adhesion/physiology , Epithelial Cells/microbiology , Proteus mirabilis/physiology , Urinary Tract Infections/microbiology , Adhesins, Bacterial/genetics , Adhesins, Escherichia coli/genetics , Adolescent , Adult , Aged , Animals , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Biofilms/growth & development , Cell Line , Child , Child, Preschool , Female , Fimbriae Proteins/genetics , Gene Deletion , Humans , Infant , Macaca mulatta , Male , Middle Aged , Polystyrenes , Proteus mirabilis/genetics , Proteus mirabilis/isolation & purification
13.
mBio ; 7(3)2016 06 07.
Article in English | MEDLINE | ID: mdl-27273829

ABSTRACT

UNLABELLED: Enteric pathogens such as enterohemorrhagic Escherichia coli (EHEC) and Citrobacter rodentium, which is largely used as a surrogate EHEC model for murine infections, are exposed to several host neurotransmitters in the gut. An important chemical exchange within the gut involves the neurotransmitters epinephrine and/or norepinephrine, extensively reported to increase virulence gene expression in EHEC, acting through two bacterial adrenergic sensors: QseC and QseE. However, EHEC is unable to establish itself and cause its hallmark lesions, attaching and effacing (AE) lesions, on murine enterocytes. To address the role of these neurotransmitters during enteric infection, we employed C. rodentium Both EHEC and C. rodentium harbor the locus of enterocyte effacement (LEE) that is necessary for AE lesion formation. Here we show that expression of the LEE, as well as that of other virulence genes in C. rodentium, is also activated by epinephrine and/or norepinephrine. Both QseC and QseE are required for LEE gene activation in C. rodentium, and the qseC and qseE mutants are attenuated for murine infection. C. rodentium has a decreased ability to colonize dopamine ß-hydroxylase knockout (Dbh(-/-)) mice, which do not produce epinephrine and norepinephrine. Both adrenergic sensors are required for C. rodentium to sense these neurotransmitters and activate the LEE genes during infection. These data indicate that epinephrine and norepinephrine are sensed by bacterial adrenergic receptors during enteric infection to promote activation of their virulence repertoire. This is the first report of the role of these neurotransmitters during mammalian gastrointestinal (GI) infection by a noninvasive pathogen. IMPORTANCE: The epinephrine and norepinephrine neurotransmitters play important roles in gut physiology and motility. Of note, epinephrine and norepinephrine play a central role in stress responses in mammals, and stress has profound effects on GI function. Bacterial enteric pathogens exploit these neurotransmitters as signals to coordinate the regulation of their virulence genes. The bacterial QseC and QseE adrenergic sensors are at the center of this regulatory cascade. C. rodentium is a noninvasive murine pathogen with a colonization mechanism similar to that of EHEC, enabling the investigation of host signals in mice. The presence of these neurotransmitters in the gut is necessary for C. rodentium to fully activate its virulence program, in a QseC/QseE-dependent manner, to successfully colonize its murine host. Our study data provide the first example of epinephrine and norepinephrine signaling within the gut to stimulate infection by a bacterial pathogen in a natural animal infection.


Subject(s)
Citrobacter rodentium/pathogenicity , Enterobacteriaceae Infections/microbiology , Enterohemorrhagic Escherichia coli/pathogenicity , Gastrointestinal Tract/microbiology , Gene Expression Regulation, Bacterial , Phosphoproteins/genetics , Receptors, Adrenergic/genetics , Animals , Citrobacter rodentium/genetics , Dopamine beta-Hydroxylase/genetics , Enterocytes/microbiology , Enterohemorrhagic Escherichia coli/genetics , Epinephrine/genetics , Epinephrine/metabolism , Escherichia coli Infections , Escherichia coli Proteins/genetics , Genes, Bacterial , Host-Pathogen Interactions , Mice , Mice, Knockout , Norepinephrine/genetics , Norepinephrine/metabolism , Vasoconstrictor Agents , Virulence/genetics
14.
FEMS Microbiol Lett ; 226(1): 79-85, 2003 Sep 12.
Article in English | MEDLINE | ID: mdl-13129611

ABSTRACT

Enteroaggregative Escherichia coli (EAEC) is distinguished by its characteristic aggregative adherence (AA) pattern to cultured epithelial cells. In this study we investigated the role of type I fimbriae (TIF) in the AA pattern to HEp-2 cells and in biofilm formation. Accentuation of this pattern was observed when the adherence assay was performed in the absence of mannose. This effect was observed in the prototype EAEC strain 042 (O44:H18), O128:H35 strains and for other EAEC serotypes. Antiserum against TIF decreased AA by 70% and 90% for strains 042 and 18 (O128:H35 prototype strain), respectively. A non-polar knockout of fimD, the TIF usher, in strains 042 and 18 resulted in inhibition of the accentuated AA pattern of approximately 80% and 70% respectively, and biofilm formation diminution of 49% for 042::fimD and 76% for 18::fimD. Our data evidence a role for TIF in the AA pattern and in EAEC biofilm formation, demonstrating that these phenotypes are multifactorial.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Proteins , Biofilms/growth & development , Escherichia coli Proteins , Escherichia coli/physiology , Fimbriae, Bacterial/physiology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/physiology , Antibodies, Bacterial , Antigens, Bacterial , Bacterial Adhesion/genetics , Cell Line, Tumor , Colony Count, Microbial , Escherichia coli/pathogenicity , Fimbriae Proteins/genetics , Fimbriae Proteins/physiology , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/immunology , Humans , Mannose/metabolism , Microscopy , Microscopy, Immunoelectron , Mutagenesis, Insertional
15.
Urol Sci ; 25(1): 1-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-27499825

ABSTRACT

Recurrent urinary tract infections (RUTI) are prevalent and pose significant clinical challenges. Although the term RUTI has long been vaguely defined, a consensus definition has emerged in recent years. The exact etiology behind RUTI remains under debate, with valid arguments for both ascending reinfections as well as persistent infection inside the bladder. These persistent infections exist in the form of quiescent intracellular reservoirs in the mouse model and may represent a novel concept to explain UTI recurrence in humans. Manageable risk factors such as behavioral patterns alongside nonmanageable risk factors including genetic susceptibility are growing fields of investigation. Acute UTI have been studied through two model bacterial strains: Escherichia coli UTI89 and CFT073. However, the clinical relevance to RUTI of these two strains has not been firmly established. Current treatment strategies for RUTI are limited and remain dominated by antibiotic usage despite variable efficacy. The majority of studies in humans have focused on younger groups of women with little information available about the postmenopausal population despite a heightened risk of RUTI in this age group.

16.
mBio ; 5(6): e02165, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25389178

ABSTRACT

UNLABELLED: Invasive pathogens interface with the host and its resident microbiota through interkingdom signaling. The bacterial receptor QseC, which is a membrane-bound histidine sensor kinase, responds to the host stress hormones epinephrine and norepinephrine and the bacterial signal AI-3, integrating interkingdom signaling at the biochemical level. Importantly, the QseC signaling cascade is exploited by many bacterial pathogens to promote virulence. Here, we translated this basic science information into development of a potent small molecule inhibitor of QseC, LED209. Extensive structure activity relationship (SAR) studies revealed that LED209 is a potent prodrug that is highly selective for QseC. Its warhead allosterically modifies lysines in QseC, impairing its function and preventing the activation of the virulence program of several Gram-negative pathogens both in vitro and during murine infection. LED209 does not interfere with pathogen growth, possibly leading to a milder evolutionary pressure toward drug resistance. LED209 has desirable pharmacokinetics and does not present toxicity in vitro and in rodents. This is a unique antivirulence approach, with a proven broad-spectrum activity against multiple Gram-negative pathogens that cause mammalian infections. IMPORTANCE: There is an imminent need for development of novel treatments for infectious diseases, given that one of the biggest challenges to medicine in the foreseeable future is the emergence of microbial antibiotic resistance. Here, we devised a broad-spectrum antivirulence approach targeting a conserved histidine kinase, QseC, in several Gram-negative pathogens that promotes their virulence expression. The LED209 QseC inhibitor has a unique mode of action by acting as a prodrug scaffold to deliver a warhead that allosterically modifies QseC, impeding virulence in several Gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents/metabolism , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/physiology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Quorum Sensing/drug effects , Sulfonamides/pharmacology , Animals , Histidine Kinase , Mice , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/isolation & purification , Structure-Activity Relationship , Sulfonamides/chemistry , Virulence/drug effects
17.
mBio ; 3(5): e00280-12, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23073764

ABSTRACT

Gastrointestinal (GI) bacteria sense diverse environmental signals as cues for differential gene regulation and niche adaptation. Pathogens such as enterohemorrhagic Escherichia coli (EHEC), which causes bloody diarrhea, use these signals for the temporal and energy-efficient regulation of their virulence factors. One of the main virulence strategies employed by EHEC is the formation of attaching and effacing (AE) lesions on enterocytes. Most of the genes necessary for the formation of these lesions are grouped within a pathogenicity island, the locus of enterocyte effacement (LEE), whose expression requires the LEE-encoded regulator Ler. Here we show that growth of EHEC in glycolytic environments inhibits the expression of ler and consequently all other LEE genes. Conversely, growth within a gluconeogenic environment activates expression of these genes. This sugar-dependent regulation is achieved through two transcription factors: KdpE and Cra. Both Cra and KdpE directly bind to the ler promoter, and Cra's affinity to this promoter is catabolite dependent. Moreover, we show that the Cra and KdpE proteins interact in vitro and that KdpE's ability to bind DNA is enhanced by the presence of Cra. Cra is important for AE lesion formation, and KdpE contributes to this Cra-dependent regulation. The deletion of cra and kdpE resulted in the ablation of AE lesions. One of the many challenges that bacteria face within the GI tract is to successfully compete for carbon sources. Linking carbon metabolism to the precise coordination of virulence expression is a key step in the adaptation of pathogens to the GI environment. IMPORTANCE An appropriate and prompt response to environmental cues is crucial for bacterial survival. Cra and KdpE are two proteins found in both nonpathogenic and pathogenic bacteria that regulate genes in response to differences in metabolite concentration. In this work, we show that, in the deadly pathogen enterohemorrhagic Escherichia coli (EHEC) O157:H7, which causes bloody diarrhea, these two proteins influence important virulence traits. We also propose that their control of one or more of these virulence traits is due to the direct interaction of the Cra and KdpE proteins with each other, as well as with their DNA targets. This work shows how EHEC coopts established mechanisms for sensing the metabolites and stress cues in the environment, to induce virulence factors in a temporal and energy-efficient manner, culminating in disease. Understanding how pathogens commandeer nonpathogenic systems can help us develop measures to control them.


Subject(s)
Bacterial Proteins/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Repressor Proteins/metabolism , Trans-Activators/metabolism , Bacterial Proteins/genetics , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/physiology , Escherichia coli Proteins/genetics , Gene Deletion , Gluconeogenesis , Glucose/metabolism , Glycolysis , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Repressor Proteins/genetics , Trans-Activators/genetics , Virulence
18.
Int. braz. j. urol ; 41(1): 67-77, jan-feb/2015. tab, graf
Article in English | LILACS | ID: lil-742860

ABSTRACT

Purpose The treatment of urinary tract infections (UTI) with antibiotics is commonly used, but recurrence and antibiotic resistance have been growing and concerning clinicians. We studied whether the rapid onset of a protective biofilm may be responsible for the lack of effectiveness of antibiotics against selected bacteria. Materials and Methods Two established uropathogenic Escherichia coli strains, UTI89 and CFT073, and two Pseudomonas aeruginosa strains, PA01 and Boston-41501, were studied to establish a reliable biofilm formation process. Bacterial growth (BG) was determined by optical density at 600 nm (OD 600) using a spectrophotometer, while biofilm formation (BF) using crystal violet staining was measured at OD 550. Next, these bacterial strains were treated with clinically relevant antibiotics, ciprofloxacin HCl (200 ng/mL and 2 μg/mL), nitrofurantoin (20 μg/mL and 40 μg/mL) and ampicillin (50 μg/mL) at time points of 0 (T0) or after 6 hours of culture (T6). All measurements, including controls (bacteria -1% DMSO), were done in triplicates and repeated three times for consistency. Results The tested antibiotics effectively inhibited both BG and BF when administered at T0 for UPEC strains, but not when the antibiotic administration started 6 hours later. For Pseudomonas strains, only Ciprofloxacin was able to significantly inhibit bacterial growth at T0 but only at the higher concentration of 2 μg/mL for T6. Conclusion When established UPEC and Pseudomonas bacteria were allowed to culture for 6 hours before initialization of treatment, the therapeutic effect of selected antibiotics was greatly suppressed when compared to immediate treatment, probably as a result of the protective nature of the biofilm. .


Subject(s)
Anti-Bacterial Agents/administration & dosage , Biofilms/drug effects , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Anti-Infective Agents, Urinary , Ampicillin/administration & dosage , Biofilms/growth & development , Ciprofloxacin/administration & dosage , Drug Resistance, Bacterial , Escherichia coli/physiology , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Nitrofurantoin/administration & dosage , Pseudomonas aeruginosa/physiology , Reference Values , Reproducibility of Results , Time Factors , Urinary Tract Infections/drug therapy
19.
Science ; 321(5892): 1078-80, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18719281

ABSTRACT

Many bacterial pathogens rely on a conserved membrane histidine sensor kinase, QseC, to respond to host adrenergic signaling molecules and bacterial signals in order to promote the expression of virulence factors. Using a high-throughput screen, we identified a small molecule, LED209, that inhibits the binding of signals to QseC, preventing its autophosphorylation and consequently inhibiting QseC-mediated activation of virulence gene expression. LED209 is not toxic and does not inhibit pathogen growth; however, this compound markedly inhibits the virulence of several pathogens in vitro and in vivo in animals. Inhibition of signaling offers a strategy for the development of broad-spectrum antimicrobial drugs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterohemorrhagic Escherichia coli/pathogenicity , Escherichia coli Proteins/metabolism , Francisella tularensis/pathogenicity , Gram-Negative Bacterial Infections/drug therapy , Protein Kinases/metabolism , Salmonella typhimurium/pathogenicity , Sulfonamides/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/genetics , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Infections/drug therapy , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/genetics , Francisella tularensis/drug effects , Francisella tularensis/genetics , Francisella tularensis/metabolism , Gene Expression Regulation, Bacterial/drug effects , Histidine Kinase , Mice , Norepinephrine/metabolism , Phosphorylation , Protein Kinases/genetics , Rabbits , Salmonella Infections, Animal/drug therapy , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Signal Transduction/drug effects , Small Molecule Libraries , Sulfonamides/administration & dosage , Sulfonamides/chemistry , Sulfonamides/therapeutic use , Tularemia/drug therapy , Virulence Factors/genetics
20.
J Bacteriol ; 188(11): 3952-61, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16707687

ABSTRACT

Microcolony formation is one of the initial steps in biofilm development, and in enteropathogenic Escherichia coli (EPEC) it is mediated by several adhesins, including the bundle-forming pilus (BFP) and the EspA filament. Here we report that EPEC forms biofilms on plastic under static conditions and a flowthrough continuous culture system. The abilities of several EPEC isogenic mutants to form biofilms were assessed. Adhesins such as BFP and EspA, important in microcolony formation on epithelial cells, are also involved in bacterial aggregation during biofilm formation on abiotic surfaces. Mutants that do not express BFP or EspA form more-diffuse biofilms than does the wild type. We also determined, using gfp transcriptional fusions, that, consistent with the role of these adhesins in biofilms, the genes encoding BFP and EspA are expressed during biofilm formation. Finally, expression of espA is controlled by a quorum-sensing (QS) regulatory mechanism, and the EPEC qseA QS mutant also forms altered biofilms, suggesting that this signaling mechanism plays an important role in EPEC biofilm development. Taken together, these studies allowed us to propose a model of EPEC biofilm formation.


Subject(s)
Biofilms/growth & development , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Escherichia coli/pathogenicity , Fimbriae, Bacterial/physiology , Genotype , Kinetics , Mutagenesis , Recombinant Proteins/metabolism , Signal Transduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL