Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Front Cell Infect Microbiol ; 14: 1414188, 2024.
Article in English | MEDLINE | ID: mdl-38979511

ABSTRACT

In Escherichia coli, the disaccharide trehalose can be metabolized as a carbon source or be accumulated as an osmoprotectant under osmotic stress. In hypertonic environments, E. coli accumulates trehalose in the cell by synthesis from glucose mediated by the cytosolic enzymes OtsA and OtsB. Trehalose in the periplasm can be hydrolyzed into glucose by the periplasmic trehalase TreA. We have previously shown that a treA mutant of extraintestinal E. coli strain BEN2908 displayed increased resistance to osmotic stress by 0.6 M urea, and reduced production of type 1 fimbriae, reduced invasion of avian fibroblasts, and decreased bladder colonization in a murine model of urinary tract infection. Since loss of TreA likely results in higher periplasmic trehalose concentrations, we wondered if deletion of otsA and otsB genes, which would lead to decreased internal trehalose concentrations, would reduce resistance to stress by 0.6 M urea and promote type 1 fimbriae production. The BEN2908ΔotsBA mutant was sensitive to osmotic stress by urea, but displayed an even more pronounced reduction in production of type 1 fimbriae, with the consequent reduction in adhesion/invasion of avian fibroblasts and reduced bladder colonization in the murine urinary tract. The BEN2908ΔtreAotsBA mutant also showed a reduction in production of type 1 fimbriae, but in contrast to the ΔotsBA mutant, resisted better than the wild type in the presence of urea. We hypothesize that, in BEN2908, resistance to stress by urea would depend on the levels of periplasmic trehalose, but type 1 fimbriae production would be influenced by the levels of cytosolic trehalose.


Subject(s)
Fimbriae, Bacterial , Osmoregulation , Trehalose , Urinary Bladder , Urinary Tract Infections , Animals , Trehalose/metabolism , Mice , Urinary Bladder/microbiology , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/genetics , Urinary Tract Infections/microbiology , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Escherichia coli/metabolism , Escherichia coli/genetics , Disease Models, Animal , Female , Osmotic Pressure , Extraintestinal Pathogenic Escherichia coli/metabolism , Extraintestinal Pathogenic Escherichia coli/genetics , Urea/metabolism , Trehalase/metabolism , Trehalase/genetics , Gene Deletion , Glucose/metabolism
2.
Microbiol Res ; 283: 127647, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38452551

ABSTRACT

The Type VI secretion system (T6SS) functions as a protein transport nanoweapon in several stages of bacterial life. Even though bacterial competition is the primary function of T6SS, different bacteria exhibit significant variations. Particularly in Extraintestinal pathogenic Escherichia coli (ExPEC), research into T6SS remains relatively limited. This study identified the uncharacterized gene evfG within the T6SS cluster of ExPEC RS218. Through our experiments, we showed that evfG is involved in T6SS expression in ExPEC RS218. We also found evfG can modulate T6SS activity by competitively binding to c-di-GMP, leading to a reduction in the inhibitory effect. Furthermore, we found that evfG can recruit sodA to alleviate oxidative stress. The research shown evfG controls an array of traits, both directly and indirectly, through transcriptome and additional tests. These traits include cell adhesion, invasion, motility, drug resistance, and pathogenicity of microorganisms. Overall, we contend that evfG serves as a multi-functional regulator for the T6SS and several crucial activities. This forms the basis for the advancement of T6SS function research, as well as new opportunities for vaccine and medication development.


Subject(s)
Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Type VI Secretion Systems , Type VI Secretion Systems/genetics , Type VI Secretion Systems/metabolism , Extraintestinal Pathogenic Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Virulence , Virulence Factors/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
3.
Infect Immun ; 91(11): e0003923, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37815368

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) is responsible for severe bloodstream infections in humans and animals. However, the mechanisms underlying ExPEC's serum resistance remain incompletely understood. Through the transposon-directed insertion-site sequencing approach, our previous study identified nhaA, the gene encoding a Na+/H+ antiporter, as a crucial factor for infection in vivo. In this study, we investigated the role of NhaA in ExPEC virulence utilizing both in vitro models and systemic infection models involving avian and mammalian animals. Genetic mutagenesis analysis revealed that nhaA deletion resulted in filamentous bacterial morphology and rendered the bacteria more susceptible to sodium dodecyl sulfate, suggesting the role of nhaA in maintaining cell envelope integrity. The nhaA mutant also displayed heightened sensitivity to complement-mediated killing compared to the wild-type strain, attributed to augmented deposition of complement components (C3b and C9). Remarkably, NhaA played a more crucial role in virulence compared to several well-known factors, including Iss, Prc, NlpI, and OmpA. Our findings revealed that NhaA significantly enhanced virulence across diverse human ExPEC prototype strains within B2 phylogroups, suggesting widespread involvement in virulence. Given its pivotal role, NhaA could serve as a potential drug target for tackling ExPEC infections.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Animals , Humans , Extraintestinal Pathogenic Escherichia coli/metabolism , Virulence/genetics , Escherichia coli Infections/microbiology , Virulence Factors/genetics , Birds/metabolism , Birds/microbiology , Mammals , Sodium-Hydrogen Exchangers , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Lipoproteins
4.
Curr Microbiol ; 80(1): 50, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-36542185

ABSTRACT

The type II toxin-antitoxin (T-A) HicAB system is abundant in several bacteria and archaea, such as Escherichia coli, Burkholderia Pseudomallei, Yersinia pestis, Pseudomonas aeruginosa, and Streptococcus pneumoniae. This system engages in stress response, virulence, and bacterial persistence. This study showed that the biofilm-forming ability of the hicAB deletion mutant was significantly decreased to moderate ability compared to the extra-intestinal pathogenic Escherichia coli (ExPEC) parent strain and the complemented strain, which are strong biofilm producers. Congo red assay showed that the hicAB mutant maintained the ability to form curli fimbriae. Using RNA-seq and comparative real-time quantitative RT-PCR, we observed the difference in gene expression between the hicAB mutant and the parent strain, which was associated with biofilm formation. Our data indicate that the HicAB type II T-A system has a key role in biofilm formation by ExPEC, which may be associated with outer membrane protein (OMP) gene expression. Collectively, our results indicate that the hicAB type II T-A system is involved in ExPEC biofilm formation.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Toxin-Antitoxin Systems , Humans , Escherichia coli , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Toxin-Antitoxin Systems/genetics , Biofilms , Escherichia coli Infections/microbiology
5.
Front Immunol ; 13: 929740, 2022.
Article in English | MEDLINE | ID: mdl-36059454

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) is a well-known critical pathogenic zoonosis that causes extraintestinal infections in humans and animals by affecting their immune organs. Recently, research on the outer membrane protein of E. coli, tolerant colicin (TolC), a virulent protein in the formation of the ExPEC efflux pump, has been an attractive subject. However, the pathogenic mechanisms remain unclear. This study aimed to explore the role of TolC in the pathogenesis of the ExPEC strain PPECC42; a complementation strain (Cm-TolC) and an isogenic mutant (ΔTolC) were constructed. Loss of TolC drastically impaired the virulence of ExPEC in an experimental mouse model. ΔTolC showed a substantial decrease in the porcine aortic vascular endothelial cell (PAVEC) adherence, invasion, and pro-inflammatory response, in contrast to that of the wild type, with a reduced survival ratio in both the bacterial load and whole blood in mice. ΔTolC also showed decreased expression of necroptosis signals such as receptor-interacting protein kinase 1, phosphorylated mixed-lineage kinase domain-like protein, and mitochondrial proteins such as phosphoglycerate mutase family member 5. Our data suggest that TolC is closely associated with ExPEC pathogenesis. These results provide scientific grounds for exploring the potential of TolC as an effective drug target for controlling ExPEC infection, screening new inhibitors, and developing new drugs. This will allow for further prevention and control of ExPEC infection.


Subject(s)
Colicins , Escherichia coli Infections , Extraintestinal Pathogenic Escherichia coli , Animals , Colicins/metabolism , Escherichia coli/genetics , Escherichia coli Infections/microbiology , Extraintestinal Pathogenic Escherichia coli/metabolism , Humans , Mice , Swine , Virulence
6.
Int J Antimicrob Agents ; 60(4): 106649, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35934230

ABSTRACT

Extended-spectrum ß-lactamase (ESBL)-producing extraintestinal pathogenic Escherichia coli (ExPEC), particularly high-risk lineages, are responsible for severe infections and increased mortality and hospital costs worldwide, with a major burden in low-income countries. Here we determined the antimicrobial susceptibility and performed whole-genome sequencing of E. coli isolates from extraintestinal infections of patients during 2017-2018 at Maputo Central Hospital (Mozambique). Multidrug resistance was displayed by 71% of isolates (17/24). All isolates resistant to cefotaxime and ceftazidime were positive for ESBL genes (16/24; 67%) and were co-resistant to amoxicillin/clavulanate (14/16; 88%), piperacillin/tazobactam (8/16; 50%), gentamicin (12/16; 75%), trimethoprim/sulfamethoxazole (15/16; 94%) and ciprofloxacin (11/16; 69%). Several major high-risk ExPEC lineages were identified, such as H30Rx-ST131, fimH41-ST131, H24Rx-ST410, ST617, ST361 and ST69 harbouring blaCTX-M-15, and H30R-ST131, ST38 and ST457 carrying blaCTX-M-27. Dissemination of CTX-M transposition units (ISEcp1-blaCTX-M-15-orf477 and ISEcp1-blaCTX-M-27-IS903B) among different sequence types could be occurring through the mobility of IncF plasmids. Additionally, all H24Rx-ST410 isolates carried ISEcp1-mediated blaCMY-2 AmpC and specific mutations in PBP3/OmpC proteins, potentially contributing to carbapenem resistance even in the absence of carbapenemase genes. Genome analysis highlighted a high assortment of ExPEC/UPEC virulence-associated genes mainly involved in adhesion, invasion, iron uptake and secretory systems among isolates, and an ExPEC/EAEC hybrid pathotype (fimH27-ST131_O18-ac:H4) showing the highest virulence gene content. cgMLST showed clonality and closely related isolates, particularly among ST131 and ST410, suggesting hospital-acquired infections and long-term ward persistence. Our study provides new insights into ExPEC clones, urging measures to prevent and contain their diffusion in this hospital and Mozambique.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Amoxicillin , Anti-Bacterial Agents/pharmacology , Carbapenems , Cefotaxime , Ceftazidime , Ciprofloxacin , Clavulanic Acid , Escherichia coli , Escherichia coli Infections/epidemiology , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Gentamicins , Hospitals , Humans , Iron , Mozambique/epidemiology , Piperacillin , Tazobactam , Trimethoprim, Sulfamethoxazole Drug Combination , beta-Lactamases/genetics , beta-Lactamases/metabolism
7.
PLoS Pathog ; 18(6): e1010582, 2022 06.
Article in English | MEDLINE | ID: mdl-35700218

ABSTRACT

Extra-intestinal pathogenic Escherichia coli (ExPEC) belong to a critical priority group of antibiotic resistant pathogens. ExPEC establish gut reservoirs that seed infection of the urinary tract and bloodstream, but the mechanisms of gut colonisation remain to be properly understood. Ucl fimbriae are attachment organelles that facilitate ExPEC adherence. Here, we investigated cellular receptors for Ucl fimbriae and Ucl expression to define molecular mechanisms of Ucl-mediated ExPEC colonisation of the gut. We demonstrate differential expression of Ucl fimbriae in ExPEC sequence types associated with disseminated infection. Genome editing of strains from two common sequence types, F11 (ST127) and UTI89 (ST95), identified a single nucleotide polymorphism in the ucl promoter that changes fimbriae expression via activation by the global stress-response regulator OxyR, leading to altered gut colonisation. Structure-function analysis of the Ucl fimbriae tip-adhesin (UclD) identified high-affinity glycan receptor targets, with highest affinity for sialyllacto-N-fucopentose VI, a structure likely to be expressed on the gut epithelium. Comparison of the UclD adhesin to the homologous UcaD tip-adhesin from Proteus mirabilis revealed that although they possess a similar tertiary structure, apart from lacto-N-fucopentose VI that bound to both adhesins at low-micromolar affinity, they recognize different fucose- and glucose-containing oligosaccharides. Competitive surface plasmon resonance analysis together with co-structural investigation of UcaD in complex with monosaccharides revealed a broad-specificity glycan binding pocket shared between UcaD and UclD that could accommodate these interactions. Overall, our study describes a mechanism of adaptation that augments establishment of an ExPEC gut reservoir to seed disseminated infections, providing a pathway for the development of targeted anti-adhesion therapeutics.


Subject(s)
Escherichia coli Infections , Extraintestinal Pathogenic Escherichia coli , Adhesins, Bacterial/metabolism , Adhesins, Escherichia coli/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Humans , Intestinal Diseases , Polysaccharides/metabolism
8.
Appl Environ Microbiol ; 88(13): e0142121, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35758695

ABSTRACT

Fimbrial adhesins promote bacterial adherence and biofilm formation. Sequencing of avian pathogenic Escherichia coli (APEC) strain QT598 identified new fimbriae belonging to the π group, which we named PL (P-like) fimbriae since the genetic organization and sequence are similar to those of P and related fimbriae. Genes encoding PL fimbriae located on IncF plasmids are present in diverse E. coli isolates from poultry, human systemic infections, and other sources. As with P fimbriae, PL fimbriae exhibit divergence in adhesin-encoding genes and could be divided into 5 classes based on sequence differences in the PlfG adhesin. plf genes from two predominant PlfG adhesin classes, PlfG class I (PlfGI) and PlfGII, were cloned. PL fimbriae were visualized by electron microscopy, associated with increased biofilm, demonstrated distinct hemagglutination profiles, and promoted adherence to human bladder and kidney epithelial cells. The genes encoding hybrid fimbriae were comprised of genes from plfQT598, wherein plfG was replaced by papG; the adhesin-encoding genes were also functional and mediated adherence to epithelial cells, demonstrating compatibility between the components of these two types of fimbriae. Deletion of plf genes did not reduce colonization of the mouse urinary tract in a single-strain infection model. In contrast, loss of plf genes significantly reduced competitive colonization in the mouse kidneys. Furthermore, plf gene expression was increased over 40-fold in the bladder compared to during in vitro culture. Overall, PL fimbriae represent a new group of fimbriae demonstrating both functional differences from and similarities to P fimbriae, which mediated adherence to host cells and improved competitive colonization of the mouse kidney. IMPORTANCE Fimbriae are important colonization factors in many bacterial species. The identification of a new type of fimbriae encoded on some IncF plasmids in E. coli was investigated. Genomic sequences demonstrated these fimbrial gene clusters have genetic diversity, particularly in the adhesin-encoding plfG gene. Functional studies demonstrated differences in hemagglutination specificity, although both types of Plf adhesin under study mediated adherence to human urinary epithelial cells. A plf mutant also showed decreased colonization of the kidneys in a mouse competitive infection model. PL fimbriae may represent previously unrecognized adhesins that could contribute to host specificity and tissue tropism of some E. coli strains.


Subject(s)
Escherichia coli Infections , Extraintestinal Pathogenic Escherichia coli , Fimbriae Proteins , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Animals , Bacterial Adhesion/physiology , Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/veterinary , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Fimbriae Proteins/genetics , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Humans , Mice
9.
J Bacteriol ; 204(4): e0061221, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35311558

ABSTRACT

Flagellum-mediated bacterial motility is important for bacteria to take up nutrients, adapt to environmental changes, and establish infection. The twin-arginine translocation system (Tat) is an important protein export system, playing a critical role in bacterial physiology and pathogenesis. It has been observed for a long time that the Tat system is critical for bacterial motility. However, the underlying mechanism remains unrevealed. In this study, a comparative transcriptomics analysis was performed with extraintestinal pathogenic Escherichia coli (ExPEC), which identified a considerable number of genes differentially expressed when the Tat system was disrupted. Among them, a large proportion of flagellar biosynthesis genes showed downregulation, indicating that transcription regulation plays an important role in mediating the motility defects. We further identified three Tat substrate proteins, MdoD, AmiA, and AmiC, that were responsible for the nonmotile phenotype. The Rcs system was deleted in the Δtat, the ΔmdoD, and the ΔamiAΔamiC strains, which restored the motility of ΔmdoD and partially restored the motility of Δtat and ΔamiAΔamiC. The flagella were also observed in all of the ΔtatΔrcsDB, ΔmdoDΔrcsDB, and ΔamiAΔamiCΔrcsDB strains, but not in the Δtat, ΔmdoD, and ΔamiAΔamiC strains, by using transmission electron microscopy. Quantitative reverse transcription-PCR data revealed that the regulons of the Rcs system displayed differential expression in the tat mutant, indicating that the Rcs signaling was activated. Our results suggest that the Rcs system plays an important role in mediating the motility defects of the tat mutant of ExPEC. IMPORTANCE The Tat system is an important protein export system critical for bacterial physiology and pathogenesis. It has been observed for a long time that the Tat system is critical for bacterial motility. However, the underlying mechanism remains unrevealed. In this study, we combine transcriptomics analysis and bacterial genetics, which reveal that transcription regulation plays an important role in mediating the motility defects of the tat mutant of extraintestinal pathogenic Escherichia coli. The Tat substrate proteins responsible for the motility defects are identified. We further show that the Rcs system contributes to the motility suppression. We for the first time reveal the link between the Tat system and bacterial motility, which is important for understanding the physiological functions of the Tat system.


Subject(s)
Escherichia coli Proteins , Extraintestinal Pathogenic Escherichia coli , Twin-Arginine-Translocation System , Arginine/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Flagella/metabolism , Protein Transport , Twin-Arginine-Translocation System/genetics , Twin-Arginine-Translocation System/metabolism
10.
J Microbiol ; 59(9): 854-860, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34382147

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) is an important zoonotic pathogen that places severe burdens on public health and animal husbandry. There are many pathogenic factors in E. coli. The type VI secretion system (T6SS) is a nano-microbial weapon that can assemble quickly and inject toxic effectors into recipient cells when danger is encountered. T6SSs are encoded in the genomes of approximately 25% of sequenced Gram-negative bacteria. When these bacteria come into contact with eukaryotic cells or prokaryotic microbes, the T6SS assembles and secretes associated effectors. In the porcine ExPEC strain PCN033, we identified four classic rearrangement hotspot (Rhs) genes. We determined the functions of the four Rhs proteins through mutant construction and protein expression. Animal infection experiments showed that the Δrhs-1CT, Δrhs-2CT, Δrhs-3CT, and Δrhs-4CT caused a significant decrease in the multiplication ability of PCN033 in vivo. Cell infection experiments showed that the Rhs protein is involved in anti-phagocytosis activities and bacterial adhesion and invasion abilities. The results of this study demonstrated that rhs1, rhs3, and rh4 plays an important role in the interaction between PCN033 and host cell. Rhs2 has contribution to cell and mice infection. This study helps to elucidate the pathogenic mechanism governing PCN033 and may help to establish a foundation for further research seeking to identify potential T6SS effectors.


Subject(s)
Escherichia coli Infections/veterinary , Escherichia coli Proteins/metabolism , Swine Diseases/microbiology , Animals , Bacterial Adhesion , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Female , Intestines/microbiology , Mice , Multigene Family , Swine
11.
Infect Immun ; 89(10): e0035721, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34228495

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) strains are major causes of urinary and bloodstream infections. ExPEC reservoirs are not completely understood. Some mastitis-associated E. coli (MAEC) strains carry genes associated with ExPEC virulence, including metal scavenging, immune avoidance, and host attachment functions. In this study, we investigated the role of the high-affinity zinc uptake (znuABC) system in the MAEC strain M12. Elimination of znuABC moderately decreased fitness during mouse mammary gland infections. The ΔznuABC mutant strain exhibited an unexpected growth delay in the presence of bile salts, which was alleviated by the addition of excess zinc. We isolated suppressor mutants with improved growth in bile salts, several of which no longer produced the K96 capsule made by strain M12. The addition of bile salts also reduced capsule production by strain M12 and ExPEC strain CP9, suggesting that capsule synthesis may be detrimental when bile salts are present. To better understand the role of the capsule, we compared the virulence of mastitis strain M12 with that of its unencapsulated ΔkpsCS mutant in two models of ExPEC disease. The wild-type strain successfully colonized mouse bladders and kidneys and was highly virulent in intraperitoneal infections. Conversely, the ΔkpsCS mutant was unable to colonize kidneys and was unable to cause sepsis. These results demonstrate that some MAEC strains may be capable of causing human ExPEC illness. The virulence of strain M12 in these infections is dependent on its capsule. However, capsule may interfere with zinc homeostasis in the presence of bile salts while in the digestive tract.


Subject(s)
Bacterial Capsules/metabolism , Bile Acids and Salts/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/metabolism , Mastitis/metabolism , Zinc/metabolism , Animals , Escherichia coli Infections/microbiology , Female , Male , Mastitis/microbiology , Mice , Mice, Inbred C57BL , Sepsis/metabolism , Sepsis/microbiology , Virulence/physiology , Virulence Factors/metabolism
12.
Appl Environ Microbiol ; 87(17): e0056721, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34132592

ABSTRACT

Avian pathogenic Escherichia coli (APEC), an extraintestinal pathogenic E. coli (ExPEC), causes colibacillosis in chickens and is reportedly associated with urinary tract infections and meningitis in humans. Development of resistance is a major limitation of current ExPEC antibiotic therapy. New antibacterials that can circumvent resistance problem such as antimicrobial peptides (AMPs) are critically needed. Here, we evaluated the efficacy of Lactobacillus rhamnosus GG (LGG)-derived peptides against APEC and uncovered their potential antibacterial targets. Three peptides (NPSRQERR [P1], PDENK [P2], and VHTAPK [P3]) displayed inhibitory activity against APEC. These peptides were effective against APEC in biofilm and chicken macrophage HD11 cells. Treatment with these peptides reduced the cecum colonization (0.5 to 1.3 log) of APEC in chickens. Microbiota analysis revealed two peptides (P1 and P2) decreased Enterobacteriaceae abundance with minimal impact on overall cecal microbiota of chickens. Bacterial cytological profiling showed peptides disrupt APEC membranes either by causing membrane shedding, rupturing, or flaccidity. Furthermore, gene expression analysis revealed that peptides downregulated the expression of ompC (>13.0-fold), ompF (>11.3-fold), and mlaA (>4.9-fold), genes responsible for the maintenance of outer membrane (OM) lipid asymmetry. Consistently, immunoblot analysis also showed decreased levels of OmpC and MlaA proteins in APEC treated with peptides. Alanine scanning studies revealed residues crucial (P1, N, E, R and P; P2, D and E; P3, T, P, and K) for their activity. Overall, our study identified peptides with a new antibacterial target that can be developed to control APEC infections in chickens, thereby curtailing poultry-originated human ExPEC infections. IMPORTANCE Avian pathogenic Escherichia coli (APEC) is a subgroup of extraintestinal pathogenic E. coli (ExPEC) and considered a foodborne zoonotic pathogen transmitted through consumption of contaminated poultry products. APEC shares genetic similarities with human ExPECs, including uropathogenic E. coli (UPEC) and neonatal meningitis E. coli (NMEC). Our study identified Lactobacillus rhamnosus GG (LGG)-derived peptides (P1 [NPSRQERR], P2 [PDENK], and P3 [VHTAPK]) effective in reducing APEC infection in chickens. Antimicrobial peptides (AMPs) are regarded as ideal candidates for antibacterial development because of their low propensity for resistance development and ability to kill resistant bacteria. Mechanistic studies showed peptides disrupt the APEC membrane by affecting the MlaA-OmpC/F system responsible for the maintenance of outer membrane (OM) lipid asymmetry, a promising new druggable target to overcome resistance problems in Gram-negative bacteria. Altogether, these peptides can provide a valuable approach for development of novel anti-ExPEC therapies, including APEC, human ExPECs, and other related Gram-negative pathogens. Furthermore, effective control of APEC infections in chickens can curb poultry-originated ExPEC infections in humans.


Subject(s)
Escherichia coli Infections/veterinary , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/drug effects , Phospholipid Transfer Proteins/metabolism , Pore Forming Cytotoxic Proteins/pharmacology , Porins/metabolism , Poultry Diseases/microbiology , Animals , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , Biofilms/drug effects , Chickens/microbiology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/growth & development , Extraintestinal Pathogenic Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/drug effects , Phospholipid Transfer Proteins/genetics , Porins/genetics , Poultry Diseases/drug therapy
13.
Vet Microbiol ; 249: 108830, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32920372

ABSTRACT

Porcine extraintestinal pathogenic Escherichia coli (ExPEC) is occurring with increasing frequency in China, and leads to significant economic and welfare costs in the swine industry. The underlying mechanisms of porcine ExPEC in blood colonization during systematic infection is poorly understood. Here we measured the gene expression of porcine ExPEC in infected animal bloodstream in vivo and fresh swine blood in vitro. Using comparisons with P values of ≤ 0.01, we identified 354 and 313 genes as being significantly up- or down-regulated at least 2-fold change during bloodstream infection, respectively. Excepting for an array of iron acquisition systems, numerous genes involved in carbon central metabolism and anaerobic respiratory chains were upregulated here. These genes were categorized into several clusters including the TCA-cycle (frdABCD, citCEFXG), d-ribose transporter (rbsDACB), nickel transporter (nikABCDER), NiFe hydrogenase (hybOABCDEF, hycBCDEFG), Hyp-complex (hypABCDE), DMSO reductase (dmsABC and ynfEFGHI), format dehydrogenase (fdnGHI) and NADH dehydrogenase I (nuoA-N). The mutant with simultaneous inactivation of ribose and citrate imports showed significant reduced fitness in host blood, suggesting these two carbohydrates are utilized by central metabolism network as important carbon-source during bloodstream infection. Similar deficiency was also observed in the mutant double deleted NiFe hydrogenase 2 and 3 anaerobic respiratory chains. Further study found that FNR (a global regulator facilitating bacterial adaptation to anaerobic conditions) is an important regulator in response to bloodstream to activate center metabolism and anaerobic respiratory chains, thus contribute to the full-virulence of porcine ExPEC. These findings provide compelling evidence to support the notion that carbon central metabolism network and anaerobic respiratory chains play key roles for porcine ExPEC fitness within host bloodstream.


Subject(s)
Bacteremia/veterinary , Escherichia coli Infections/veterinary , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Swine Diseases/microbiology , Anaerobiosis/genetics , Animals , Bacteremia/microbiology , Carbon/metabolism , China , Electron Transport/genetics , Escherichia coli Infections/microbiology , Extraintestinal Pathogenic Escherichia coli/metabolism , Swine
14.
BMC Res Notes ; 13(1): 51, 2020 Jan 31.
Article in English | MEDLINE | ID: mdl-32005127

ABSTRACT

OBJECTIVE: Extraintestinal Pathogenic E. coli (ExPEC), are responsible for host diseases such as Neonatal Meningitis Escherichia coli (NMEC), the second-leading cause of neonatal bacterial meningitis, Avian Pathogenic E. coli (APEC), a cause of extraintestinal disease in poultry, and Uropathogenic E. coli (UPEC), the most common cause of urinary tract infections. Virulence factors associated with NMEC include outer membrane protein A (OmpA) and type I fimbriae (FimH), which also occur in APEC and UPEC. OmpA contributes to NMEC's ability to cross the blood-brain barrier, persist in the bloodstream and has been identified as a potential vaccine target for ExPEC, however the protein has amino acid variants, which may influence virulence of strains or alter vaccine efficacy. Although OmpA is present in virtually all E. coli, differences in its amino acid residues have yet to be surveyed in ExPEC. RESULTS: Here the ompA gene (n = 399) from ExPEC collections were sequenced and translated in silico. Twenty-five different OmpA polymorphism patterns were identified. Seven polymorphism patterns were significantly associated with an ExPEC subpathotype, but chromosomal history most likely accounts for most differences found. The differences in OmpA protein sequences suggest that OmpA may influence variation in virulence and host specificity within ExPEC subpathotypes.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/metabolism , Animals , Bacterial Outer Membrane Proteins/chemistry , Birds/microbiology , Humans , Infant, Newborn , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/metabolism
15.
Article in English | MEDLINE | ID: mdl-31964797

ABSTRACT

Two multidrug-resistant and carbapenemase-producing Escherichia coli clones of sequence type 410 were isolated from fecal samples of a dog with skin infection on admission to an animal hospital in Portugal and 1 month after discharge. Whole-genome sequencing revealed a 126,409-bp Col156/IncFIA/IncFII multidrug resistance plasmid and a 51,479-bp IncX3 blaOXA-181-containing plasmid. The chromosome and plasmids carried virulence genes characteristic for uropathogenic E. coli, indicating that dogs may carry multidrug-resistant E. coli isolates related to those causing urinary tract infections in humans.


Subject(s)
Dog Diseases/microbiology , Escherichia coli Infections/veterinary , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/isolation & purification , beta-Lactamases/genetics , Animals , Bacterial Proteins/metabolism , Cat Diseases/microbiology , Cats , Dogs , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/metabolism , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Feces/microbiology , Gastrointestinal Microbiome , Genome, Bacterial , Phylogeny , Plasmids , Portugal , Skin Diseases, Bacterial/microbiology , Skin Diseases, Bacterial/veterinary , Urinary Tract Infections/microbiology , Urinary Tract Infections/veterinary , Virulence Factors/genetics , beta-Lactamases/metabolism
16.
Sci Rep ; 9(1): 2600, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30796316

ABSTRACT

Pathoadaptive mutations linked to c-di-GMP signalling were investigated in neonatal meningitis-causing Escherichia coli (NMEC). The results indicated that NMEC strains deficient in RpoS (the global stress regulator) maintained remarkably low levels of c-di-GMP, a major bacterial sessility-motility switch. Deletion of ycgG2, shown here to encode a YcgG allozyme with c-di-GMP phosphodiesterase activity, and the restoration of RpoS led to a decrease in S-fimbriae, robustly produced in artificial urine, hinting that the urinary tract could serve as a habitat for NMEC. We showed that NMEC were skilled in aerobic citrate utilization in the presence of glucose, a property that normally does not exist in E. coli. Our data suggest that this metabolic novelty is a property of extraintestinal pathogenic E. coli since we reconstituted this ability in E. coli UTI89 (a cystitis isolate) via deactivation rpoS; additionally, a set of pyelonephritis E. coli isolates were shown here to aerobically use citrate in the presence of glucose. We found that the main reason for this metabolic capability is RpoS inactivation leading to the production of the citrate transporter CitT, exploited by NMEC for ferric citrate uptake dependent on YcgG2 (an allozyme with c-di-GMP phosphodiesterase activity).


Subject(s)
Bacterial Proteins/genetics , Cyclic GMP/analogs & derivatives , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Meningitis, Escherichia coli/microbiology , Sigma Factor/genetics , Citric Acid/metabolism , Cyclic GMP/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Glucose/metabolism , Organic Anion Transporters/metabolism
17.
Front Immunol ; 9: 788, 2018.
Article in English | MEDLINE | ID: mdl-29719540

ABSTRACT

The extraintestinal pathogenic Escherichia coli (ExPEC) is a typical facultative intracellular bacterial pathogen. Sensing the environmental stimuli and undertaking adaptive change are crucial for ExPEC to successfully colonize in specific extraintestinal niches. The previous studies show that pathogens exploit two-component systems (TCSs) in response to the host environments during its infection. The PhoP/PhoQ is a typical TCS which is ubiquitous in Gram-negative bacteria. However, there is an incompletely understanding about critical regulatory roles of PhoP/PhoQ in ExPEC pathogenesis. Conjugative ColV-related plasmids are responsible for ExPEC virulence, which is associated with ExPEC zoonotic risk. In this study, the molecular characteristics of HlyF, Mig-14 ortholog (Mig-14p), and OmpT variant (OmpTp) encoded by ColV plasmids were identified. Mig-14p and OmpTp played important roles in conferring ExPEC resistance to cationic antimicrobial peptides (CAMPs) during the infection. Moreover, HlyF and Mig-14p acted as intracellular survival factors to promote ExPEC resistance to macrophages killing. The hlyF and Mig-14p formed an operon in ExPEC ColV plasmid, and PhoP acted as a transcriptional activator of hlyF operon by directly binding to the P hlyF promoter. The acidic pH and CAMPs could additively stimulate ExPEC PhoQ/PhoP activities to upregulate the expression of HlyF and Mig-14p. Our studies revealed that the novel PhoP/PhoQ-HlyF signaling pathway directly upregulates the production of ExPEC outer membrane vesicles. Furthermore, our study first clarified that this PhoP/PhoQ-HlyF pathway was essential for ExPEC intracellular survival in macrophages. It was required to prevent the fusion of ExPEC-containing phagosomes with lysosomes. Moreover, PhoP/PhoQ-HlyF pathway facilitated the inhibition of the phagolysosomal acidification and disruption of the phagolysosomal membranes. In addition, this pathway might promote the formation of ExPEC-containing autophagosome during ExPEC replication in macrophages. Collectively, our studies suggested that PhoP/PhoQ system and CloV plasmids could facilitate ExPEC survival and replication within macrophages.


Subject(s)
Escherichia coli Proteins/metabolism , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Host-Pathogen Interactions/physiology , Macrophages/microbiology , Animals , Chickens , Escherichia coli Infections , Extraintestinal Pathogenic Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Humans , Virulence
18.
Mol Microbiol ; 110(5): 689-706, 2018 12.
Article in English | MEDLINE | ID: mdl-29802751

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) is one of the leading causes of bloodstream infections. Characteristically, these organisms exhibit strong resistance to the bactericidal action of host serum. Although numerous serum resistance factors in ExPEC have been identified, their regulatory mechanisms during in vivo infection remain largely unknown. Here, RNA sequencing analyses together with quantitative reverse-transcription PCR revealed that ExPEC genes involved in the biosynthesis of extracytoplasmic polysaccharides (ECPs) including K-capsule, lipopolysaccharide (LPS), colanic acid, peptidoglycan and Yjb exopolysaccharides were significantly upregulated in response to serum under low oxygen conditions and during bloodstream infection. The oxygen sensor FNR directly activated the expression of K-capsule and colanic acid and also indirectly modulated the expression of colanic acid, Yjb exopolysaccharides and peptidoglycan via the known Rcs regulatory system. The global regulator Fur directly or indirectly repressed the expression ofECP biosynthesis genes in iron replete media, whereas the low iron conditions in the bloodstream could relieve Fur repression. Using in vitro and animal models, FNR, Fur and the Rcs system were confirmed as contributing to ExPEC ECP production, serum resistance and virulence. Altogether, these findings indicated that the global regulators FNR andFur and the signaling transduction system Rcs coordinately regulated the expression of ECP biosynthesis genes leading to increased ExPEC serum resistance in response to low oxygen and low iron levels in the bloodstream.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli , Polysaccharides/metabolism , Serum/metabolism , Serum/microbiology , Animals , Ducks/microbiology , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Virulence
19.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29311232

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) acts as a commensal within the mammalian gut but can induce pathology upon dissemination to other host environments such as the urinary tract and bloodstream. ExPEC genomes are likely shaped by evolutionary forces encountered within the gut, where the bacteria spend much of their time, provoking the question of how their extraintestinal virulence traits arose. The principle of coincidental evolution, in which a gene that evolved in one niche happens to be advantageous in another, has been used to argue that ExPEC virulence factors originated in response to selective pressures within the gut ecosystem. As a test of this hypothesis, the fitness of ExPEC mutants lacking canonical virulence factors was assessed within the intact murine gut in the absence of antibiotic treatment. We found that most of the tested factors, including cytotoxic necrotizing factor type 1 (CNF1), Usp, colibactin, flagella, and plasmid pUTI89, were dispensable for gut colonization. The deletion of genes encoding the adhesin PapG or the toxin HlyA had transient effects but did not interfere with longer-term persistence. In contrast, a mutant missing the type 1 pilus-associated adhesin FimH displayed somewhat reduced persistence within the gut. However, this phenotype varied dependent on the presence of specific competing strains and was partially attributable to aberrant flagellin expression in the absence of fimH These data indicate that FimH and other key ExPEC-associated factors are not strictly required for gut colonization, suggesting that the development of extraintestinal virulence traits is not driven solely by selective pressures within the gut.


Subject(s)
Adhesins, Escherichia coli/metabolism , Escherichia coli Infections/microbiology , Extraintestinal Pathogenic Escherichia coli/metabolism , Fimbriae Proteins/metabolism , Gastrointestinal Tract/microbiology , Virulence Factors/metabolism , Adhesins, Escherichia coli/genetics , Animals , Extraintestinal Pathogenic Escherichia coli/genetics , Female , Fimbriae Proteins/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Virulence Factors/genetics
20.
Microbiol Res ; 196: 69-79, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28164791

ABSTRACT

Extraintestinal pathogenic Escherichia coli (ExPEC) can invade and colonize multiple extraintestinal tissues and can cause a wide range of infections; however the mechanisms of its pathogenicity are not well understood. Flagella contribute to the infection of E. coli strains by mediating adhesion and invasion. Our previous bioinformatic analysis revealed two flagella gene clusters in the genome of an ExPEC isolate, PCN033. One encodes the conventional flagellum system (Flag-1) and the other encodes the Flag-2 system, whose function is uncharacterized. Here we aimed to characterize these two flagellum systems and determine their contributions to the flagellum formation and certain pathogenicity-associated phenotypes. Our observations support the involvement of Flag-1 system, but not Flag-2 system, in the synthesis and maturation of the flagellum structure, and in mediating bacterial swimming and swarming. Moreover, flgD, which encodes a flagellar-hook scaffolding protein in the Flag-1 system, is required for flagellum assembly by influencing the production of FliC (flagellin). Deletion of flgD attenuated ExPEC strain PCN033 invasion and colonization in vivo, probably by affecting bacterial adhesion and invasion, and by reducing resistance to phagocytosis by circulating monocytes. In contrast, these phenotypes were not observed in the strain with deletion of lfgD, encoding the FlgD-like protein in the Flag-2 system. Taken together, these findings indicate that Flag-1 flagellum system is the determinative component of bacterial flagella that contributes to the infection.


Subject(s)
Escherichia coli Infections/microbiology , Escherichia coli Proteins/physiology , Extraintestinal Pathogenic Escherichia coli/physiology , Extraintestinal Pathogenic Escherichia coli/pathogenicity , Flagella/physiology , Animals , Bacterial Adhesion/physiology , Cell Migration Assays , DNA, Bacterial/genetics , Escherichia coli Infections/metabolism , Escherichia coli Proteins/genetics , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/metabolism , Female , Flagella/genetics , Flagellin/chemistry , Flagellin/genetics , Mice , Multigene Family , Phagocytosis , RAW 264.7 Cells , Sequence Deletion , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL