Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Epigenomes ; 7(4)2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-38131902

RESUMO

Pathogenic bacteria recognize environmental cues to vary gene expression for host adaptation. Moving from ambient to host temperature, Yersinia enterocolitica responds by immediately repressing flagella synthesis and inducing the virulence plasmid (pYV)-encoded type III secretion system. In contrast, shifting from host to ambient temperature requires 2.5 generations to restore motility, suggesting a link to the cell cycle. We hypothesized that differential DNA methylation contributes to temperature-regulated gene expression. We tested this hypothesis by comparing single-molecule real-time (SMRT) sequencing of Y. enterocolitica DNA from cells growing exponentially at 22 °C and 37 °C. The inter-pulse duration ratio rather than the traditional QV scoring was the kinetic metric to compare DNA from cells grown at each temperature. All 565 YenI restriction sites were fully methylated at both temperatures. Among the 27,118 DNA adenine methylase (Dam) sites, 42 had differential methylation patterns, while 17 remained unmethylated regardless of the temperature. A subset of the differentially methylated Dam sites localized to promoter regions of predicted regulatory genes including LysR-type and PadR-like transcriptional regulators and a cyclic-di-GMP phosphodiesterase. The unmethylated Dam sites localized with a bias to the replication terminus, suggesting they were protected from Dam methylase. No cytosine methylation was detected at Dcm sites.

2.
PLoS One ; 18(9): e0291520, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37699034

RESUMO

Pathogenic Escherichia coli and Salmonella enterica pose serious public health threats due to their ability to cause severe gastroenteritis and life-threatening sequela, particularly in young children. Moreover, the emergence and dissemination of antibiotic resistance in these bacteria have complicated control of infections. Alternative strategies that effectively target these enteric pathogens and negate or reduce the need of antibiotics are urgently needed. Such an alternative is the CRISPR-Cas9 system because it can generate sequence-specific lethal double stranded DNA breaks. In this study, two self-transmissible broad host range conjugative plasmids, pRK24 and pBP136, were engineered to deliver multiplexed CRSIPR-Cas9 systems that specifically target Enterohemorrhagic and Enteropathogenic strains of E. coli (EHEC and EPEC), S. enterica, and blaCMY-2 antibiotic resistance plasmids. Using in vitro mating assays, we show that the conjugative delivery of pRK24-CRISPR-Cas9 carrying guide RNAs to the EPEC/EHEC eae (intimin) gene can selectively kill enterohemorrhagic E. coli O157 eae+ cells (3 log kill at 6 h) but does not kill the isogenic Δeae mutant (P<0.001). Similar results were also obtained with a pBP136 derivative, pTF16, carrying multiplexed guide RNAs targeting E. coli eae and the S. enterica ssaN gene coding for the type III secretion ATPase. Another pBP136 derivative, TF18, carries guide RNAs targeting S. enterica ssaN and the antibiotic resistance gene, blaCMY-2, carried on the multi-drug resistant pAR06302. Introduction of pTF18 into bacteria harboring pAR06302 showed plasmids were cured at an efficiency of 53% (P<0.05). Using a murine neonate EPEC infection model, pTF16 was delivered by a murine derived E. coli strain to EPEC infected mice and showed significant reductions of intestinal EPEC (P<0.05). These results suggest that establishing conjugative CRISPR-Cas9 antimicrobials in the intestinal microbiome may provide protection from enteric pathogens and reduce antibiotic resistance without disrupting the normal microbiota.


Assuntos
Escherichia coli Êntero-Hemorrágica , Escherichia coli O157 , Gastroenterite , Animais , Camundongos , Sistemas CRISPR-Cas/genética , Engenharia , Escherichia coli Êntero-Hemorrágica/genética , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos
4.
Sci Rep ; 13(1): 7902, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-37193760

RESUMO

Understanding transmission dynamics of SARS-CoV-2 in institutions of higher education (IHEs) is important because these settings have potential for rapid viral spread. Here, we used genomic surveillance to retrospectively investigate transmission dynamics throughout the 2020-2021 academic year for the University of Idaho ("University"), a mid-sized IHE in a small rural town. We generated genome assemblies for 1168 SARS-CoV-2 samples collected during the academic year, representing 46.8% of positive samples collected from the University population and 49.8% of positive samples collected from the surrounding community ("Community") at the local hospital during this time. Transmission dynamics differed for the University when compared to the Community, with more infection waves that lasted shorter lengths of time, potentially resulting from high-transmission congregate settings along with mitigation efforts implemented by the University to combat outbreaks. We found evidence for low transmission rates between the University and Community, with approximately 8% of transmissions into the Community originating from the University, and approximately 6% of transmissions into the University originating from the Community. Potential transmission risk factors identified for the University included congregate settings such as sorority and fraternity events and residences, holiday travel, and high caseloads in the surrounding community. Knowledge of these risk factors can help the University and other IHEs develop effective mitigation measures for SARS-CoV-2 and similar pathogens.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/epidemiologia , Estudos Retrospectivos , Genômica , Fatores de Risco
5.
Appl Environ Microbiol ; 89(2): e0124422, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36744930

RESUMO

Ail confers serum resistance in humans and is a critical virulence factor of Y. pestis, the causative agent of plague. Here, the contribution of Ail for Y. pestis survival in the flea vector was examined. Rat or human but not mouse sera were bactericidal against a Y. pestis Δail mutant at 28°C in vitro. Complement components deposited rapidly on the Y. pestis surface as measured by immunofluorescent microscopy. Ail reduced the amount of active C3b on the Y. pestis surface. Human sera retained bactericidal activity against a Y. pestis Δail mutant in the presence of mouse sera. However, in the flea vector, the serum protective properties of Ail were not required. Flea colonization studies using murine sera and Y. pestis KIM6+ wild type, a Δail mutant, and the Δail/ail+ control showed no differences in bacterial prevalence or numbers during the early stage of flea colonization. Similarly, flea studies with human blood showed Ail was not required for serum resistance. Finally, a variant of Ail (AilF100V E108_S109insS) from a human serum-sensitive Y. pestis subsp. microtus bv. Caucasica 1146 conferred resistance to human complement when expressed in the Y. pestis KIM6+ Δail mutant. This indicated that Ail activity was somehow blocked, most likely by lipooligosaccharide, in this serum sensitive strain. IMPORTANCE This work contributes to our understanding of how highly virulent Y. pestis evolved from its innocuous enteric predecessor. Among identified virulence factors is the attachment invasion locus protein, Ail, that is required to protect Y. pestis from serum complement in all mammals tested except mice. Murine sera is not bactericidal. In this study, we asked, is bactericidal sera from humans active in Y. pestis colonized fleas? We found it was not. The importance of this observation is that it identifies a protective niche for the growth of serum sensitive and nonsensitive Y. pestis strains.


Assuntos
Peste , Sifonápteros , Yersinia pestis , Animais , Humanos , Camundongos , Ratos , Antibacterianos/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Mamíferos , Peste/microbiologia , Sifonápteros/metabolismo , Sifonápteros/microbiologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo , Yersinia pestis/genética , Yersinia pestis/metabolismo , Complemento C3b/metabolismo , Complemento C3b/farmacologia
6.
Curr Opin Infect Dis ; 35(3): 188-195, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35665712

RESUMO

PURPOSE OF REVIEW: Pathogenic Yersinia have been a productive model system for studying bacterial pathogenesis. Hallmark contributions of Yersinia research to medical microbiology are legion and include: (i) the first identification of the role of plasmids in virulence, (ii) the important mechanism of iron acquisition from the host, (iii) the first identification of bacterial surface proteins required for host cell invasion, (iv) the archetypical type III secretion system, and (v) elucidation of the role of genomic reduction in the evolutionary trajectory from a fairly innocuous pathogen to a highly virulent species. RECENT FINDINGS: The outer membrane (OM) protein Ail (attachment invasion locus) was identified over 30 years ago as an invasin-like protein. Recent work on Ail continues to provide insights into Gram-negative pathogenesis. This review is a synopsis of the role of Ail in invasion, serum resistance, OM stability, thermosensing, and vaccine development. SUMMARY: Ail is shown to be an essential virulence factor with multiple roles in pathogenesis. The recent adaptation of Yersinia pestis to high virulence, which included genomic reduction to eliminate redundant protein functions, is a model to understand the emergence of new bacterial pathogens.


Assuntos
Yersinia pestis , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Humanos , Virulência , Fatores de Virulência/metabolismo , Yersinia pestis/genética , Yersinia pestis/metabolismo
7.
Curr Opin Infect Dis ; 35(3): 205-214, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35665714

RESUMO

PURPOSE OF REVIEW: This review updates recent findings about Escherichia coli O157:H7 virulence factors and its bovine reservoir. This Shiga toxin (Stx)-producing E. coli belongs to the Enterohemorrhagic E. coli (EHEC) pathotype causing hemorrhagic colitis. Its low infectious dose makes it an efficient, severe, foodborne pathogen. Although EHEC remains in the intestine, Stx can translocate systemically and is cytotoxic to microvascular endothelial cells, especially in the kidney and brain. Disease can progress to life-threatening hemolytic uremic syndrome (HUS) with hemolytic anemia, acute kidney failure, and thrombocytopenia. Young children, the immunocompromised, and the elderly are at the highest risk for HUS. Healthy ruminants are the major reservoir of EHEC and cattle are the primary source of human exposure. RECENT FINDINGS: Advances in understanding E. coli O157:H7 pathogenesis include molecular mechanisms of virulence, bacterial adherence, type three secretion effectors, intestinal microbiome, inflammation, and reservoir maintenance. SUMMARY: Many aspects of E. coli O157:H7 disease remain unclear and include the role of the human and bovine intestinal microbiomes in infection. Therapeutic strategies involve controlling inflammatory responses and/or intestinal barrier function. Finally, elimination/reduction of E. coli O157:H7 in cattle using CRISPR-engineered conjugative bacterial plasmids and/or on-farm management likely hold solutions to reduce infections and increase food safety/security.


Assuntos
Infecções por Escherichia coli , Escherichia coli O157 , Síndrome Hemolítico-Urêmica , Idoso , Animais , Bovinos , Pré-Escolar , Células Endoteliais/patologia , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/genética , Síndrome Hemolítico-Urêmica/epidemiologia , Síndrome Hemolítico-Urêmica/microbiologia , Humanos , Ruminantes , Fatores de Virulência/genética
8.
Adv Physiol Educ ; 45(4): 786-796, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34529538

RESUMO

Idaho Institutional Development Award (IDeA) Network for Biomedical Research Excellence (INBRE) aims to build biomedical research capacity and enhance the scientific and technology knowledge of the Idaho workforce. A key INBRE Program at The College of Idaho, a primarily undergraduate institution of 1,100 students, is a 10-wk summer fellows research experience. This report documents outcomes from 2005 to present, including demographic trends, faculty and student research productivity, self-reported gains, educational attainment, and career outcomes. Of 103 participants, 83.7% were from Idaho, 26.7% from rural areas, and 23.9% first-generation college students. Faculty and student research productivity (conference presentations and peer-reviewed publications) increased threefold. We found that 91.4% of fellows entered a scientific- or healthcare-related career and that 70.7% completed or are currently enrolled in postgraduate training (51.7% doctoral and 19.0% master's level). Anonymous surveys were uniformly positive, with gains in self-confidence and independent laboratory work. Open-ended responses indicated students valued mentoring efforts and improved awareness of scientific opportunities and competitive preparation for postgraduate training. Lastly, we observed that student research involvement increased college-wide during the award period. These data suggest that the summer fellows program is successfully meeting National Institutes of Health IDeA goals and serving as a pipeline to future health research careers and a scientifically trained Idaho workforce.


Assuntos
Pesquisa Biomédica , Estudantes , Humanos , Idaho , Mentores , Universidades
9.
J Bacteriol ; 203(21): e0036121, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34398663

RESUMO

Maintenance of phospholipid (PL) and lipopoly- or lipooligosaccharide (LPS or LOS) asymmetry in the outer membrane (OM) of Gram-negative bacteria is essential but poorly understood. The Yersinia pestis OM Ail protein was required to maintain lipid homeostasis and cell integrity at elevated temperature (37°C). Loss of this protein had pleiotropic effects. A Y. pestis Δail mutant and KIM6+ wild type were systematically compared for (i) growth requirements at 37°C, (ii) cell structure, (iii) antibiotic and detergent sensitivity, (iv) proteins released into supernatants, (v) induction of the heat shock response, and (vi) physiological and genetic suppressors that restored the wild-type phenotype. The Δail mutant grew normally at 28°C but lysed at 37°C when it entered stationary phase, as shown by cell count, SDS-PAGE of cell supernatants, and electron microscopy. Immunofluorescence microscopy showed that the Δail mutant did not assemble Caf1 capsule. Expression of heat shock promoter rpoE or rpoH fused to a lux operon reporter were not induced when the Δail mutant was shifted from 28°C to 37°C (P < 0.001 and P < 0.01, respectively). Mutant lysis was suppressed by addition of 11 mM glucose, 22 or 44 mM glycerol, 2.5 mM Ca2+, or 2.5 mM Mg2+ to the growth medium or by a mutation in the phospholipase A gene (pldA::miniTn5, ΔpldA, or PldAS164A). A model accounting for the temperature-sensitive lysis of the Δail mutant and the Ail-dependent stabilization of the OM tetraacylated LOS at 37°C is presented. IMPORTANCE The Gram-negative pathogen Yersinia pestis transitions between a flea vector (ambient temperature) and a mammalian host (37°C). In response to 37°C, Y. pestis modifies its outer membrane (OM) by reducing the fatty acid content in lipid A, changing the outer leaflet from being predominantly hexaacylated to being predominantly tetraacylated. It also increases the Ail concentration, so it becomes the most prominent OM protein. Both measures are needed for Y. pestis to evade the host innate immune response. Deletion of ail destabilizes the OM at 37°C, causing the cells to lyse. These results show that a protein is essential for maintaining lipid asymmetry and lipid homeostasis in the bacterial OM.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Fatores de Virulência/metabolismo , Yersinia pestis/genética , Yersinia pestis/metabolismo , Cápsulas Bacterianas , Proteínas da Membrana Bacteriana Externa/genética , Cálcio/farmacologia , Carbono/química , Carbono/metabolismo , Regulação para Baixo , Deleção de Genes , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Pleiotropia Genética , Glucose/farmacologia , Fosfolipases/genética , Fosfolipases/metabolismo , Fator sigma/genética , Fator sigma/metabolismo , Temperatura , Fatores de Virulência/genética
10.
FEMS Microbiol Ecol ; 97(8)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34329454

RESUMO

Strictly lytic phages are considered powerful tools for biocontrol of foodborne pathogens. Safety issues needed to be addressed for the biocontrol of Shiga toxin-producing Escherichia coli (STEC) include: lysogenic conversion, Shiga toxin production through phage induction, and emergence/proliferation of bacteriophage insensitive mutants (BIMs). To address these issues, two new lytic phages, vB_EcoS_Ace (Ace) and vB_EcoM_Shy (Shy), were isolated and characterized for life cycle, genome sequence and annotation, pH stability and efficacy at controlling STEC growth. Ace was efficient in controlling host planktonic cells and did not stimulate the production of the Stx prophage or Shiga toxin. A single dose of phage did not lead to the selection of BIMs. However, when reintroduced, BIMs were detected after 24 h of incubation. The gain of resistance was associated with lower virulence, as a subset of BIMs failed to agglutinate with O157-specific antibody and were more sensitive to human serum complement. BIM's biofilm formation capacity and susceptibility to disinfectants was equal to that of the wild-type strain. Overall, this work demonstrated that phage Ace is a safe biocontrol agent against STEC contamination and that the burden of BIM emergence did not represent a greater risk in environmental persistence and human pathogenicity.


Assuntos
Bacteriófagos , Contaminação de Alimentos/prevenção & controle , Escherichia coli Shiga Toxigênica , Bacteriófagos/genética , Agentes de Controle Biológico , Lisogenia , Toxina Shiga/genética , Toxina Shiga II/genética , Escherichia coli Shiga Toxigênica/genética
11.
J Biomol Tech ; 32(2): 63-73, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34121933

RESUMO

The Biomolecular Research Center at Boise State University is a research core facility that supports the study of biomolecules with an emphasis on protein structure and function, molecular interactions, and imaging. The mission of the core is to facilitate access to instrumentation that might otherwise be unavailable because of the cost, training for new users, and scientific staff with specialized skills to support early-stage investigators, as well as more established senior investigators. Data collection and management of users and their research output is essential to understand the impact of the center on the research environment and research productivity. However, challenges are often encountered when trying to fully quantify the impact of a core facility on the institution, as well as on the career success of individual investigators. This challenge can be exacerbated under the conditions of unprecedented growth in biomedical research and shared core facility use that has been experienced at Boise State University, an institution of emerging research excellence. Responding to these challenges required new approaches to information management, reporting, assessment, and evaluation. Our specific data management, evaluation, and assessment challenges included 1) collection and management of annual reporting information from investigators, staff, and students in a streamlined manner that did not lead to reporting fatigue; 2) application of software for analyzing synergy among programs' management strategy and investigator success; and 3) consolidation of core facility management, billing, and reporting capabilities into 1 cohesive system. The data management tools adopted had a beneficial effect by saving time, reducing administrative burden, and streamlining reporting. Practices implemented for data management have facilitated effective evaluation and future program planning. The substantial burden of assessment requirements necessitates early consideration of a strategy for data management to allow assessment of impact.


Assuntos
Pesquisa Biomédica , Gerenciamento de Dados , Humanos , Pesquisadores
12.
Appl Environ Microbiol ; 87(2)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33158889

RESUMO

Subcutaneous vaccination of cattle for enterohemorrhagic Escherichia coli O157:H7 reduces the magnitude and duration of fecal shedding, but the often-required, repeated cattle restraint can increase costs, deterring adoption by producers. In contrast, live oral vaccines may be repeatedly administered in feed, without animal restraint. We investigated whether oral immunization with live stx-negative LEE+E. coli O157:H7 reduced rectoanal junction (RAJ) colonization by wild-type (WT) E. coli O157:H7 strains after challenge. Two groups of cattle were orally dosed twice weekly for 6 weeks with 3 × 109 CFU of a pool of three stx-negative LEE+E. coli O157:H7 strains (vaccine group) or three stx-negative LEE- non-O157:H7 E. coli strains (control group). Three weeks following the final oral dose, animals in both groups were orally challenged with a cocktail of four stx+ LEE+E. coli O157:H7 WT strains. Subsequently, WT strains at the RAJ were enumerated weekly for 4 weeks. Serum antibodies against type III secretion protein (TTSP), the translocated intimin receptor (Tir), and EspA were determined by enzyme-linked immunosorbent assay (ELISA) at day 0 (preimmunization), day 61 (postimmunization, prechallenge), and day 89 (postchallenge). Vaccine group cattle had lower numbers of WT strains at the RAJ than control group cattle on postchallenge days 3 and 7 (P ≤ 0.05). Also, vaccine group cattle shed WT strains for a shorter duration than control group cattle. All cattle seroconverted to TTSP, Tir, and EspA, either following immunization (vaccine group) or following challenge (control group). Increased antibody titers against Tir and TTSP postimmunization were associated with decreased numbers of WT E. coli O157:H7 organisms at the RAJ.IMPORTANCE The bacterium E. coli O157:H7 causes foodborne disease in humans that can lead to bloody diarrhea, kidney failure, vascular damage, and death. Healthy cattle are the main source of this human pathogen. Reducing E. coli O157:H7 in cattle will reduce human disease. Using a randomized comparison, a bovine vaccine to reduce carriage of the human pathogen was tested. A detoxified E. coli O157:H7 strain, missing genes that cause disease, was fed to cattle as an oral vaccine to reduce carriage of pathogenic E. coli O157:H7. After vaccination, the cattle were challenged with disease-causing E. coli O157:H7. The vaccinated cattle had decreased E. coli O157:H7 during the first 7 days postchallenge and shed the bacteria for a shorter duration than the nonvaccinated control cattle. The results support optimization of the approach to cattle vaccination that would reduce human disease.


Assuntos
Doenças dos Bovinos/prevenção & controle , Infecções por Escherichia coli/prevenção & controle , Escherichia coli O157/imunologia , Vacinas contra Escherichia coli , Administração Oral , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Bovinos , Proteínas de Escherichia coli/imunologia , Masculino , Receptores de Superfície Celular/imunologia , Toxina Shiga , Sistemas de Secreção Tipo III/imunologia , Vacinação/veterinária
13.
Microorganisms ; 8(4)2020 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-32316415

RESUMO

Escherichia coli O157:H7 (O157) is noninvasive and a weak biofilm producer; however, a subset of O157 are exceptions. O157 ATCC 43895 forms biofilms and invades epithelial cells. Tn5 mutagenesis identified a mutation responsible for both phenotypes. The insertion mapped within the curli csgB fimbriae locus. Screening of O157 strains for biofilm formation and cell invasion identified a bovine and a clinical isolate with those characteristics. A single base pair A to T transversion, intergenic to the curli divergent operons csgDEFG and csgBAC, was present only in biofilm-producing and invasive strains. Using site-directed mutagenesis, this single base change was introduced into two curli-negative/noninvasive O157 strains and modified strains to form biofilms, produce curli, and gain invasive capability. Transmission electron microscopy (EM) and immuno-EM confirmed curli fibers. EM of bovine epithelial cells (MAC-T) co-cultured with curli-expressing O157 showed intracellular bacteria. The role of curli in O157 persistence in cattle was examined by challenging cattle with curli-positive and -negative O157 and comparing carriage. The duration of bovine colonization with the O157 curli-negative mutant was shorter than its curli-positive isogenic parent. These findings definitively demonstrate that a single base pair stably confers biofilm formation, epithelial cell invasion, and persistence in cattle.

14.
Genome Announc ; 6(25)2018 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-29930034

RESUMO

The Shiga toxin-encoding phage SH2026Stx1 was isolated from Escherichia coli O157:H7 strain 2026. SH2026Stx1 and its detoxified derivative can infect a broad range of E. coli strains, including commensal, enteropathogenic, and enteroaggregative strains. We report here the complete genome sequence of phage SH2026Stx1 and its important features.

15.
Genome Announc ; 6(7)2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449403

RESUMO

Shiga toxin-producing Escherichia coli (STEC) bacteria are zoonotic pathogens. We report here the high-quality complete genome sequences of three STEC O177:H- (fliCH25) strains, SMN152SH1, SMN013SH2, and SMN197SH3. The assembled genomes consisted of one optical map-verified circular chromosome for each strain, plus two plasmids for SMN013SH2 and three plasmids for SMN152SH1 and SMN197SH3, respectively.

16.
Cell Death Discov ; 3: 17055, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28975041

RESUMO

Autophagy is a pivotal innate immune response that not only degrades cytosolic components, but also serves as one of the critical antimicrobial mechanisms eliminating intracellular pathogens. However, its role in host defense against extracellular pathogens is largely unknown. Here we showed that E. coli O157:H7 altered autophagy to evade host defense and facilitate adhesion. Enhancing host cell autophagy with tumor necrosis factor (TNF), host starvation or rapamycin reduced the adherence of E. coli O157:H7 to HT-29 cells. As a key regulator of autophagy, protein kinase A (PKA) was activated by E. coli O157:H7 infection. PKA inhibition by H89 abrogated E. coli O157:H7 inhibition of autophagy and prevented bacterial epithelial adhesion. Thus, PKA had a mediatory role in blocking autophagy and E. coli O157:H7 epithelial adhesion. Furthermore, deletion of translocated intimin receptor (tir) prevented PKA activation, whereas ectopic tir expression in a Δtir mutant strain restored its ability to activate PKA and inhibited autophagy in host cells. This indicated that Tir and PKA played pivotal roles in manipulating host autophagy during infection. Consistent with autophagy inhibition, E. coli O157:H7 infection inhibited endoplasmic reticulum (ER) stress in HT-29 cells, which was reversed by TNF, starvation, or H89 treatment. Additionally, E. coli O157:H7-induced PKA activation suppressed extracellular signal-regulated kinase 1/2 (ERK1/2) activation and enhanced phosphatidylinositol 3-kinase/Akt (PI3K/Akt) signaling, thereby repressing autophagic signaling. Conversely, PKA inhibition prevented downregulation of ERK1/2 signaling due to E. coli O157:H7 infection. In summary, E. coli O157:H7 inhibited host autophagy via Tir-mediated PKA activation that favored bacterial persistence on intestinal epithelial cell surfaces.

17.
J Bacteriol ; 198(7): 1022-34, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26833416

RESUMO

The isolation of aerobic citrate-utilizing Escherichia coli (Cit(+)) in long-term evolution experiments (LTEE) has been termed a rare, innovative, presumptive speciation event. We hypothesized that direct selection would rapidly yield the same class of E. coli Cit(+) mutants and follow the same genetic trajectory: potentiation, actualization, and refinement. This hypothesis was tested with wild-type E. coli strain B and with K-12 and three K-12 derivatives: an E. coli ΔrpoS::kan mutant (impaired for stationary-phase survival), an E. coli ΔcitT::kan mutant (deleted for the anaerobic citrate/succinate antiporter), and an E. coli ΔdctA::kan mutant (deleted for the aerobic succinate transporter). E. coli underwent adaptation to aerobic citrate metabolism that was readily and repeatedly achieved using minimal medium supplemented with citrate (M9C), M9C with 0.005% glycerol, or M9C with 0.0025% glucose. Forty-six independent E. coli Cit(+) mutants were isolated from all E. coli derivatives except the E. coli ΔcitT::kan mutant. Potentiation/actualization mutations occurred within as few as 12 generations, and refinement mutations occurred within 100 generations. Citrate utilization was confirmed using Simmons, Christensen, and LeMaster Richards citrate media and quantified by mass spectrometry. E. coli Cit(+) mutants grew in clumps and in long incompletely divided chains, a phenotype that was reversible in rich media. Genomic DNA sequencing of four E. coli Cit(+) mutants revealed the required sequence of mutational events leading to a refined Cit(+) mutant. These events showed amplified citT and dctA loci followed by DNA rearrangements consistent with promoter capture events for citT. These mutations were equivalent to the amplification and promoter capture CitT-activating mutations identified in the LTEE.IMPORTANCE E. coli cannot use citrate aerobically. Long-term evolution experiments (LTEE) performed by Blount et al. (Z. D. Blount, J. E. Barrick, C. J. Davidson, and R. E. Lenski, Nature 489:513-518, 2012, http://dx.doi.org/10.1038/nature11514 ) found a single aerobic, citrate-utilizing E. coli strain after 33,000 generations (15 years). This was interpreted as a speciation event. Here we show why it probably was not a speciation event. Using similar media, 46 independent citrate-utilizing mutants were isolated in as few as 12 to 100 generations. Genomic DNA sequencing revealed an amplification of the citT and dctA loci and DNA rearrangements to capture a promoter to express CitT, aerobically. These are members of the same class of mutations identified by the LTEE. We conclude that the rarity of the LTEE mutant was an artifact of the experimental conditions and not a unique evolutionary event. No new genetic information (novel gene function) evolved.


Assuntos
Evolução Biológica , Citratos/metabolismo , Transportadores de Ácidos Dicarboxílicos/metabolismo , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Seleção Genética , Meios de Cultura , DNA Bacteriano/genética , Transportadores de Ácidos Dicarboxílicos/genética , Escherichia coli K12/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Mutação , Transportadores de Ânions Orgânicos/genética , Fatores de Tempo
18.
Appl Environ Microbiol ; 82(3): 964-71, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26607594

RESUMO

The increased summertime prevalence of cattle carriage of enterohemorrhagic Shiga toxin-producing Escherichia coli O157:H7 (STEC O157) is associated with the increased summertime incidence of human infection. The mechanism driving the seasonality of STEC O157 carriage among cattle is unknown. We conducted experimental challenge trials to distinguish whether factors extrinsic or intrinsic to cattle underlie the seasonality of STEC O157 colonization. Holstein steers (n = 20) exposed to ambient environmental conditions were challenged with a standardized pool of STEC O157 strains four times at 6-month intervals. The densities and durations of rectoanal junction mucosa (RAJ) colonization with STEC O157 were compared by season (winter versus summer), dose (10(9) CFU versus 10(7) CFU), and route of challenge (oral versus rectal). Following summer challenges, the RAJ STEC O157 colonization density was significantly lower (P = 0.016) and the duration was shorter (P = 0.052) than for winter challenges, a seasonal pattern opposite to that observed naturally. Colonization was unaffected by the challenge route, indicating that passage through the gastrointestinal microbiome did not significantly affect the infectious dose to the RAJ. A 2-log reduction of the challenge doses in the second-year trials was accompanied by similarly reduced RAJ colonization in both seasons (P < 0.001). These results refute the hypothesis that cattle are predisposed to STEC O157 colonization during the summer months, either due to intrinsic factors or indirectly due to gastrointestinal tract microbiome effects. Instead, the data support the hypothesis that the increased summertime STEC O157 colonization results from increased seasonal oral exposure to this pathogen.


Assuntos
Doenças dos Bovinos/microbiologia , Infecções por Escherichia coli/veterinária , Escherichia coli O157/crescimento & desenvolvimento , Escherichia coli O157/isolamento & purificação , Estações do Ano , Animais , Bovinos , Doenças dos Bovinos/epidemiologia , Contagem de Colônia Microbiana , Infecções por Escherichia coli/microbiologia , Escherichia coli O157/genética , Fezes/microbiologia , Genótipo , Interações Hospedeiro-Patógeno , Humanos
19.
Proteomics ; 15(11): 1829-42, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25643951

RESUMO

Building on previous studies, we defined the repertoire of proteins comprising the immunoproteome (IP) of Escherichia coli O157:H7 (O157) cultured in DMEM supplemented with norepinephrine (O157 IP), a ß-adrenergic hormone that regulates E. coli O157 gene expression in the gastrointestinal tract, using a variation of a novel proteomics-based platform proteome mining tool for antigen discovery, called "proteomics-based expression library screening" (PELS; Kudva et al., 2006). The E. coli O157 IP (O157-IP) comprised 91 proteins, and included those identified previously using proteomics-based expression library screening, and also proteins comprising DMEM and bovine rumen fluid proteomes. Outer membrane protein A (OmpA), a common component of the above proteomes, and reportedly a contributor to E. coli O157 adherence to cultured HEp-2 epithelial cells, was interestingly found to be a modulator rather than a contributor to E. coli O157 adherence to bovine rectoanal junction squamous epithelial cells. Our results point to a role for yet to be identified members of the O157-IP in E. coli O157 adherence to rectoanal junction squamous epithelial cells, and additionally implicate a possible role for the outer membrane protein A regulator, TdcA, in the expression of such adhesins. Our observations have implications for the development of efficacious vaccines for preventing E. coli O157 colonization of the bovine gastrointestinal tract.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Células Epiteliais/microbiologia , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/imunologia , Proteínas de Escherichia coli/metabolismo , Animais , Aderência Bacteriana , Bovinos , Células Cultivadas , Células Epiteliais/citologia , Escherichia coli O157/imunologia , Escherichia coli O157/patogenicidade , Proteínas de Escherichia coli/análise , Interações Hospedeiro-Patógeno , Soros Imunes/química , Norepinefrina/farmacologia , Rúmen/citologia , Rúmen/metabolismo , Transativadores/metabolismo
20.
Infect Immun ; 81(11): 4192-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23980115

RESUMO

The human pathogen enterohemorrhagic Escherichia coli (EHEC) O157:H7 colonizes the rectoanal junction (RAJ) in cattle, its natural reservoir. Colonization at the RAJ poses a serious risk for fecal shedding and contamination of the environment. We previously demonstrated that EHEC senses acyl-homoserine lactones (AHLs) produced by the microbiota in the rumen to activate the gad acid resistance genes necessary for survival through the acidic stomachs in cattle and to repress the locus of enterocyte effacement (LEE) genes important for colonization of the RAJ, but unnecessary in the rumen. Devoid of AHLs, the RAJ is the prominent site of colonization of EHEC in cattle. To determine if the presence of AHLs in the RAJ could repress colonization at this site, we engineered EHEC to express the Yersinia enterocolitica AHL synthase gene yenI, which constitutively produces AHLs, to mimic a constant exposure of AHLs in the environment. The yenI(+) EHEC produces oxo-C6-homoserine lactone (oxo-C6-HSL) and had a significant reduction in LEE expression, effector protein secretion, and attaching and effacing (A/E) lesion formation in vitro compared to the wild type (WT). The yenI(+) EHEC also activated expression of the gad genes. To assess whether AHL production, which decreases LEE expression, would decrease RAJ colonization by EHEC, cattle were challenged at the RAJ with WT or yenI(+) EHEC. Although the yenI(+) EHEC colonized the RAJ with efficiency equal to that of the WT, there was a trend for the cattle to shed the WT strain longer than the yenI(+) EHEC.


Assuntos
Acil-Butirolactonas/metabolismo , Proteínas de Bactérias/metabolismo , Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Transativadores/metabolismo , Fatores de Virulência/metabolismo , Canal Anal/microbiologia , Animais , Proteínas de Bactérias/genética , Derrame de Bactérias , Bovinos , Escherichia coli O157/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reto/microbiologia , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA