1
|
Rodriguez-Valverde D, Leon-Montes N, Belmont-Monroy L, Ruiz-Perez F, Santiago AE. Lipoprotein Lpp and L, D-transpeptidases regulate the master regulator of virulence AggR in EAEC. Sci Rep 2025; 15:13988. [PMID: 40263412 PMCID: PMC12015436 DOI: 10.1038/s41598-025-96373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a diarrheagenic pathotype associated with traveler's diarrhea, foodborne outbreaks, and sporadic diarrhea in industrialized and developing countries. Regulation of virulence factors in EAEC is mediated by the master regulator AggR, an AraC/XylS family member controlling the expression of more than 44 genes associated with metabolism and virulence. Although the AggR regulon is well-characterized, the mechanism and upstream signaling cascades that regulate its activation are poorly understood. This study demonstrates that Lpp (Braun's lipoprotein) and L, D-transpeptidases are required for AggR activation. We found that deletion lpp in EAEC resulted in the downregulation of more than 100 genes involved in transport, metabolism, and virulence. Among the genes, fourteen transcriptional factors, including AggR, were differentially expressed in 042Δlpp. Our findings also showed that Lpp anchoring to the peptidoglycan is a requisite for AggR-activation. Hence, chemical inhibition or genetic deletion of L, D-transpeptidases encoding genes involved in the crosslink of Lpp to the peptidoglycan abolished AggR activation. Moreover, the 042Δlpp mutant exhibited reduced biofilm formation on abiotic surfaces and reduced colonization of human intestinal colonoids. This is the first study to demonstrate the tight regulation of the AraC/XylS transcriptional regulator AggR, essential in EAEC virulence and intestinal colonization by components of the bacterial cell envelope.
Collapse
Affiliation(s)
- Diana Rodriguez-Valverde
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Nancy Leon-Montes
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Laura Belmont-Monroy
- Laboratorio de Microbiología Molecular, Instituto Nacional de Pediatría, Mexico City, Mexico
| | - Fernando Ruiz-Perez
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA
| | - Araceli E Santiago
- University of Virginia, School of Medicine, Department of Pediatrics, Child Health Research Center, 409 Lane Road, MR-4 Building, P.O Box 801326, Charlottesville, VA, 22908, USA.
| |
Collapse
|
2
|
Pick K, Stadel L, Raivio TL. Escherichia coli phage-inducible chromosomal island aids helper phage replication and represses the locus of enterocyte effacement pathogenicity island. THE ISME JOURNAL 2025; 19:wrae258. [PMID: 39745890 PMCID: PMC11773190 DOI: 10.1093/ismejo/wrae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/29/2024] [Accepted: 01/01/2025] [Indexed: 01/04/2025]
Abstract
In this study, we identify and characterize a novel phage-inducible chromosomal island (PICI) found in commensal Escherichia coli MP1. This novel element, EcCIMP1, is induced and mobilized by the temperate helper phage vB_EcoP_Kapi1. EcCIMP1 contributes to superinfection immunity against its helper phage, impacting bacterial competition outcomes. Genetic analysis of EcCIMP1 led us to uncover a putative transcriptional repressor, which silences virulence gene expression in the murine pathogen Citrobacter rodentium. We also found a putative excisionase encoded by EcCIMP1 which paradoxically does not promote excision of EcCIMP1 but rather supports excision of the helper phage. Another putative excisionase encoded by a presumed integrative conjugative element can also support the excision of vB_EcoP_Kapi1, demonstrating crosstalk between excisionases from multiple classes of mobile genetic elements within the same cell. Although phylogenetically distant from other characterized PICIs, EcCIMP1 and EcCIMP1-like elements are prevalent in both pathogenic and commensal isolates of E. coli from around the world, underscoring the importance of characterizing these abundant genetic elements.
Collapse
Affiliation(s)
- Kat Pick
- Department of Biological Sciences, CW 405 Biological Sciences Building, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Lauren Stadel
- Department of Biological Sciences, CW 405 Biological Sciences Building, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - Tracy L Raivio
- Department of Biological Sciences, CW 405 Biological Sciences Building, University of Alberta, Edmonton, AB T6G 2E9, Canada
| |
Collapse
|
3
|
Berger P, Dumevi RM, Berger M, Hastor I, Treffon J, Kouzel IU, Kehl A, Scherff N, Dobrindt U, Mellmann A. RpoS Acts as a Global Repressor of Virulence Gene Expression in Escherichia coli O104:H4 and Enteroaggregative E coli. J Infect Dis 2024; 230:840-851. [PMID: 38526342 DOI: 10.1093/infdis/jiae151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/08/2024] [Accepted: 03/22/2024] [Indexed: 03/26/2024] Open
Abstract
In 2011, in Germany, Escherichia coli O104:H4 caused the enterohemorrhagic E coli (EHEC) outbreak with the highest incidence rate of hemolytic uremic syndrome. This pathogen carries an exceptionally potent combination of EHEC- and enteroaggregative E coli (EAEC)-specific virulence factors. Here, we identified an E coli O104:H4 isolate that carried a single-nucleotide polymorphism (SNP) in the start codon (ATG > ATA) of rpoS, encoding the alternative sigma factor S. The rpoS ATG > ATA SNP was associated with enhanced EAEC-specific virulence gene expression. Deletion of rpoS in E coli O104:H4 Δstx2 and typical EAEC resulted in a similar effect. Both rpoS ATG > ATA and ΔrpoS strains exhibited stronger virulence-related phenotypes in comparison to wild type. Using promoter-reporter gene fusions, we demonstrated that wild-type RpoS repressed aggR, encoding the main regulator of EAEC virulence. In summary, our work demonstrates that RpoS acts as a global repressor of E coli O104:H4 virulence, primarily through an AggR-dependent mechanism.
Collapse
Affiliation(s)
- Petya Berger
- Institute of Hygiene, University of Münster
- National Consulting Laboratory for Hemolytic Uremic Syndrome, Institute of Hygiene, University of Münster
| | | | | | | | | | - Ian U Kouzel
- Department of Biology, University of Konstanz, Germany
| | | | | | | | - Alexander Mellmann
- Institute of Hygiene, University of Münster
- National Consulting Laboratory for Hemolytic Uremic Syndrome, Institute of Hygiene, University of Münster
| |
Collapse
|
4
|
Izquierdo-Vega JA, Castillo-Juarez RJ, Sánchez-Gutiérrez M, Ares MA, De La Cruz MA. A Mini-Review of Enteroaggregative Escherichia coli with a Specific Target on the Virulence Factors Controlled by the AggR Master Regulator. Pol J Microbiol 2023; 72:347-354. [PMID: 37875068 PMCID: PMC10725161 DOI: 10.33073/pjm-2023-037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/08/2023] [Indexed: 10/26/2023] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) strains have been linked to several outbreaks of severe diarrhea around the world, and this bacterium is now commonly resistant to antibiotics. As part of the pathophysiology of EAEC, the characteristic pattern of adherence looks like stacked bricks on the intestinal epithelium. This phenotype depends on an aggregative adhesion plasmid (pAA), which codes for a regulatory protein named AggR. The AggR protein is a master regulator that transcriptionally actives the main virulence genes in this E. coli pathotype, such as those that encode the aggregative adhesion fimbriae, dispersin and its secretion apparatus, Aar regulatory protein, and type VI secretion system. Several reports have shown that AggR positively affects most EAEC virulence genes, functioning as a classic transcriptional activator in the promoter region of these genes, interacting with the RNA polymerase. This minireview article integrates the information about virulence determinants of EAEC controlled by the AggR regulator.
Collapse
Affiliation(s)
| | | | | | - Miguel A. Ares
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, México
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, México
| | | |
Collapse
|
5
|
Rodriguez-Valverde D, Giron JA, Hu Y, Nataro JP, Ruiz-Perez F, Santiago AE. Highly-conserved regulatory activity of the ANR family in the virulence of diarrheagenic bacteria through interaction with master and global regulators. Sci Rep 2023; 13:7024. [PMID: 37120613 PMCID: PMC10148876 DOI: 10.1038/s41598-023-33997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023] Open
Abstract
ANR (AraC negative regulators) are a novel class of small regulatory proteins commonly found in enteric pathogens. Aar (AggR-activated regulator), the best-characterized member of the ANR family, regulates the master transcriptional regulator of virulence AggR and the global regulator HNS in enteroaggregative Escherichia coli (EAEC) by protein-protein interactions. On the other hand, Rnr (RegA-negative regulator) is an ANR homolog identified in attaching and effacing (AE) pathogens, including Citrobacter rodentium and enteropathogenic Escherichia coli (EPEC), sharing only 25% identity with Aar. We previously found that C. rodentium lacking Rnr exhibits prolonged shedding and increased gut colonization in mice compared to the parental strain. To gain mechanistic insights into this phenomenon, we characterized the regulatory role of Rnr in the virulence of prototype EPEC strain E2348/69 by genetic, biochemical, and human organoid-based approaches. Accordingly, RNA-seq analysis revealed more than 500 genes differentially regulated by Rnr, including the type-3 secretion system (T3SS). The abundance of EspA and EspB in whole cells and bacterial supernatants confirmed the negative regulatory activity of Rnr on T3SS effectors. We found that besides HNS and Ler, twenty-six other transcriptional regulators were also under Rnr control. Most importantly, the deletion of aar in EAEC or rnr in EPEC increases the adherence of these pathogens to human intestinal organoids. In contrast, the overexpression of ANR drastically reduces bacterial adherence and the formation of AE lesions in the intestine. Our study suggests a conserved regulatory mechanism and a central role of ANR in modulating intestinal colonization by these enteropathogens despite the fact that EAEC and EPEC evolved with utterly different virulence programs.
Collapse
Affiliation(s)
- Diana Rodriguez-Valverde
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, MR-4 Building, P.O. Box 801326, Charlottesville, VA, 22908, USA
| | - Jorge A Giron
- Centro de Detección Biomolecular, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Translational Genomics Research Institute, 445 N. 5th St, Phoenix, AZ, 85004, USA
| | - Yang Hu
- CD Genomics, Shirley, NY, USA
| | - James P Nataro
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, MR-4 Building, P.O. Box 801326, Charlottesville, VA, 22908, USA
| | - Fernando Ruiz-Perez
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, MR-4 Building, P.O. Box 801326, Charlottesville, VA, 22908, USA
| | - Araceli E Santiago
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, 409 Lane Road, MR-4 Building, P.O. Box 801326, Charlottesville, VA, 22908, USA.
| |
Collapse
|
6
|
Prieto A, Bernabeu M, Sánchez-Herrero JF, Pérez-Bosque A, Miró L, Bäuerl C, Collado C, Hüttener M, Juárez A. Modulation of AggR levels reveals features of virulence regulation in enteroaggregative E. coli. Commun Biol 2021; 4:1295. [PMID: 34785760 PMCID: PMC8595720 DOI: 10.1038/s42003-021-02820-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) strains are one of the diarrheagenic pathotypes. EAEC strains harbor a virulence plasmid (pAA2) that encodes, among other virulence determinants, the aggR gene. The expression of the AggR protein leads to the expression of several virulence determinants in both plasmids and chromosomes. In this work, we describe a novel mechanism that influences AggR expression. Because of the absence of a Rho-independent terminator in the 3'UTR, aggR transcripts extend far beyond the aggR ORF. These transcripts are prone to PNPase-mediated degradation. Structural alterations in the 3'UTR result in increased aggR transcript stability, leading to increased AggR levels. We therefore investigated the effect of increased AggR levels on EAEC virulence. Upon finding the previously described AggR-dependent virulence factors, we detected novel AggR-regulated genes that may play relevant roles in EAEC virulence. Mutants exhibiting high AggR levels because of structural alterations in the aggR 3'UTR show increased mobility and increased pAA2 conjugation frequency. Furthermore, among the genes exhibiting increased fold change values, we could identify those of metabolic pathways that promote increased degradation of arginine, fatty acids and gamma-aminobutyric acid (GABA), respectively. In this paper, we discuss how the AggR-dependent increase in specific metabolic pathways activity may contribute to EAEC virulence.
Collapse
Affiliation(s)
- Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | | | - Anna Pérez-Bosque
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain
| | - Lluïsa Miró
- Department of Biochemistry and Physiology, Universitat de Barcelona, Barcelona, Spain
- Institut de Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain
| | - Christine Bäuerl
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Paterna, Valencia, Spain
| | - Mário Hüttener
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.
| | - Antonio Juárez
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain.
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
7
|
Occurrence of Escherichia coli and Salmonella species in Some Livestock (Poultry) Feeds in Mando, Kaduna, Nigeria. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.60] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The study aimed at assessing the proximate composition, isolation, characterization of some Enterobacteriaceae from two (2) brands of poultry feeds marketed in Mando, Kaduna, Nigeria. A total of sixteen (16) samples of two (2) different poultry feeds (starter and finisher) from four (4) poultry farms in mando were collected and subjected to proximate and microbiological analysis. The proximate analysis was carried out using standard techniques and procedures. All the feed samples were cultured on separate media which include Eosin methylene blue (EMB), nutrient Agar (NA) and Salmonella-Shigella Agar (SSA) media using standard procedures. The antibiogram of the selected antibiotics was evaluated against the test isolates. The result of proximate analysis of the starter and finisher feeds indicated that the Dry matter of starter feed had the highest percentage composition of 95.02% and crude fiber of the finisher feed had the lowest composition of 3.78%. The highest number of bacterial load was recorded to be 10.0×104 CFU/g for the feed sample A (starter feed) and 12.0x104CFU/g was recorded for the feed sample B (finisher feed) which had the highest number of bacterial load recorded among the two (2) different poultry feeds analyzed. The bacteria isolates were identified as Salmonella species and Escherichia coli. Total viable count (TVC) of Salmonella species and E. coli in the feed samples (starter and finisher) ranges from 3.0×104CFU/g to 12.0×104CFU/g. Both organisms (Salmonella species, E. coli) were found as 37.5% and 25% of the analyzed feeds (Broiler starter and broiler finisher) samples, respectively. There was no level of significant (p>0.05) difference between the level of contamination of Salmonella species and E. coli in the two different feeds analyzed, as p=0.06 and p=0.13 for Salmonella species, and E. coli respectively. Sample A and B (Starter and Finisher) feeds had the highest number of Salmonella species occurrence with six ( 6) positive samples while E. coli was recorded in four (4) samples of A and B (Starter and Finisher) feeds. The result of the antibiogram indicated that ciprofloxacin (30 µg), Gentamycin (30µg), Perfloxacin (30µg) and Tarvid (30µg) was effective against Salmonella species and Escherichia coli. The significant of spread of the species of the Enterobacteriaceae in livestock feeds requires the need for effective quality assurance and control, good hygiene practices in production and proper handling of the poultry feeds.
Collapse
|
8
|
Icke C, Hodges FJ, Pullela K, McKeand SA, Bryant JA, Cunningham AF, Cole JA, Henderson IR. Glycine acylation and trafficking of a new class of bacterial lipoprotein by a composite secretion system. eLife 2021; 10:63762. [PMID: 33625358 PMCID: PMC7943197 DOI: 10.7554/elife.63762] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
Protein acylation is critical for many cellular functions across all domains of life. In bacteria, lipoproteins have important roles in virulence and are targets for the development of antimicrobials and vaccines. Bacterial lipoproteins are secreted from the cytosol via the Sec pathway and acylated on an N-terminal cysteine residue through the action of three enzymes. In Gram-negative bacteria, the Lol pathway transports lipoproteins to the outer membrane. Here, we demonstrate that the Aat secretion system is a composite system sharing similarity with elements of a type I secretion systems and the Lol pathway. During secretion, the AatD subunit acylates the substrate CexE on a highly conserved N-terminal glycine residue. Mutations disrupting glycine acylation interfere with membrane incorporation and trafficking. Our data reveal CexE as the first member of a new class of glycine-acylated lipoprotein, while Aat represents a new secretion system that displays the substrate lipoprotein on the cell surface.
Collapse
Affiliation(s)
- Christopher Icke
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Freya J Hodges
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Karthik Pullela
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | | | | | - Adam F Cunningham
- Institute of Microbiology and Infection, Birmingham, United Kingdom.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Jeff A Cole
- Institute of Microbiology and Infection, Birmingham, United Kingdom
| | - Ian R Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| |
Collapse
|
9
|
The Multiomics Analyses of Fecal Matrix and Its Significance to Coeliac Disease Gut Profiling. Int J Mol Sci 2021; 22:ijms22041965. [PMID: 33671197 PMCID: PMC7922330 DOI: 10.3390/ijms22041965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
Gastrointestinal (GIT) diseases have risen globally in recent years, and early detection of the host’s gut microbiota, typically through fecal material, has become a crucial component for rapid diagnosis of such diseases. Human fecal material is a complex substance composed of undigested macromolecules and particles, and the processing of such matter is a challenge due to the unstable nature of its products and the complexity of the matrix. The identification of these products can be used as an indication for present and future diseases; however, many researchers focus on one variable or marker looking for specific biomarkers of disease. Therefore, the combination of genomics, transcriptomics, proteomics and metabonomics can give a detailed and complete insight into the gut environment. The proper sample collection, sample preparation and accurate analytical methods play a crucial role in generating precise microbial data and hypotheses in gut microbiome research, as well as multivariate data analysis in determining the gut microbiome functionality in regard to diseases. This review summarizes fecal sample protocols involved in profiling coeliac disease.
Collapse
|
10
|
Abdelwahab R, Yasir M, Godfrey RE, Christie GS, Element SJ, Saville F, Hassan EA, Ahmed EH, Abu-Faddan NH, Daef EA, Busby SJW, Browning DF. Antimicrobial resistance and gene regulation in Enteroaggregative Escherichia coli from Egyptian children with diarrhoea: Similarities and differences. Virulence 2020; 12:57-74. [PMID: 33372849 PMCID: PMC7781526 DOI: 10.1080/21505594.2020.1859852] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a common diarrhoeagenic human pathogen, isolated from patients in both developing and industrialized countries, that is becoming increasingly resistant to many frontline antibiotics. In this study, we screened 50 E. coli strains from children presenting with diarrhea at the outpatients clinic of Assiut University Children’s Hospital, Egypt. We show that all of these isolates were resistant to multiple classes of antibiotics and identified two as being typical EAEC strains. Using whole genome sequencing, we determined that both isolates carried, amongst others, blaCTX-M and blaTEM antibiotic resistance genes, as well as many classical EAEC virulence determinants, including the transcriptional regulator, AggR. We demonstrate that the expression of these virulence determinants is dependent on AggR, including aar, which encodes for a repressor of AggR, Aar. Since biofilm formation is the hallmark of EAEC infection, we examined the effect of Aar overexpression on both biofilm formation and AggR-dependent gene expression. We show that whilst Aar has a minimal effect on AggR-dependent transcription it is able to completely disrupt biofilm formation, suggesting that Aar affects these two processes differently. Taken together, our results suggest a model for the induction of virulence gene expression in EAEC that may explain the ubiquity of EAEC in both sick and healthy individuals.
Collapse
Affiliation(s)
- Radwa Abdelwahab
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK.,Faculty of Medicine, Assiut University , Assiut, Egypt
| | - Muhammad Yasir
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK.,Quadram Institute Bioscience, Norwich Research Park , Norwich, UK
| | - Rita E Godfrey
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| | - Gabrielle S Christie
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| | - Sarah J Element
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| | - Faye Saville
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| | | | | | | | - Enas A Daef
- Faculty of Medicine, Assiut University , Assiut, Egypt
| | - Stephen J W Busby
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham , Birmingham, UK
| |
Collapse
|
11
|
Boisen N, Østerlund MT, Joensen KG, Santiago AE, Mandomando I, Cravioto A, Chattaway MA, Gonyar LA, Overballe-Petersen S, Stine OC, Rasko DA, Scheutz F, Nataro JP. Redefining enteroaggregative Escherichia coli (EAEC): Genomic characterization of epidemiological EAEC strains. PLoS Negl Trop Dis 2020; 14:e0008613. [PMID: 32898134 PMCID: PMC7500659 DOI: 10.1371/journal.pntd.0008613] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/18/2020] [Accepted: 07/20/2020] [Indexed: 11/19/2022] Open
Abstract
Although enteroaggregative E. coli (EAEC) has been implicated as a common cause of diarrhea in multiple settings, neither its essential genomic nature nor its role as an enteric pathogen are fully understood. The current definition of this pathotype requires demonstration of cellular adherence; a working molecular definition encompasses E. coli which do not harbor the heat-stable or heat-labile toxins of enterotoxigenic E. coli (ETEC) and harbor the genes aaiC, aggR, and/or aatA. In an effort to improve the definition of this pathotype, we report the most definitive characterization of the pan-genome of EAEC to date, applying comparative genomics and functional characterization on a collection of 97 EAEC strains isolated in the course of a multicenter case-control diarrhea study (Global Enteric Multi-Center Study, GEMS). Genomic analysis revealed that the EAEC strains mapped to all phylogenomic groups of E. coli. Circa 70% of strains harbored one of the five described AAF variants; there were no additional AAF variants identified, and strains that lacked an identifiable AAF generally did not have an otherwise complete AggR regulon. An exception was strains that harbored an ETEC colonization factor (CF) CS22, like AAF a member of the chaperone-usher family of adhesins, but not phylogenetically related to the AAF family. Of all genes scored, sepA yielded the strongest association with diarrhea (P = 0.002) followed by the increased serum survival gene, iss (p = 0.026), and the outer membrane protease gene ompT (p = 0.046). Notably, the EAEC genomes harbored several genes characteristically associated with other E. coli pathotypes. Our data suggest that a molecular definition of EAEC could comprise E. coli strains harboring AggR and a complete AAF(I-V) or CS22 gene cluster. Further, it is possible that strains meeting this definition could be both enteric bacteria and urinary/systemic pathogens.
Collapse
Affiliation(s)
- Nadia Boisen
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Mark T. Østerlund
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Katrine G. Joensen
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - Araceli E. Santiago
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| | - Inacio Mandomando
- Centro de Investigação em Saúde da Manhiça (CISM), Maputo, Mozambique
| | - Alejandro Cravioto
- Universidad Nacional Autónoma de México, Faculty of Medicine, Mexico City, Mexico
| | - Marie A. Chattaway
- Public Health England, Gastrointestinal Bacteria Reference Unit (GBRU), Colindale, United Kingdom
| | - Laura A. Gonyar
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| | | | - O. Colin Stine
- University of Maryland School of Medicine, Department of Epidemiology and Public Health, Baltimore, Maryland, United States of America
| | - David A. Rasko
- University of Maryland School of Medicine, Institute for Genome Sciences, Department of Microbiology and Immunology, Baltimore, Maryland, United States of America
| | - Flemming Scheutz
- Statens Serum Institut, Department of Bacteria, Parasites and Fungi, Copenhagen, Denmark
| | - James P. Nataro
- University of Virginia School of Medicine, Department of Pediatrics, Charlottesville, Virginia, United States of America
| |
Collapse
|
12
|
Belmont-Monroy L, Saitz-Rojas W, Soria-Bustos J, Mickey AS, Sherman NE, Orsburn BC, Ruiz-Perez F, Santiago AE. Characterization of a novel AraC/XylS-regulated family of N-acyltransferases in pathogens of the order Enterobacterales. PLoS Pathog 2020; 16:e1008776. [PMID: 32845938 PMCID: PMC7478709 DOI: 10.1371/journal.ppat.1008776] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 09/08/2020] [Accepted: 07/04/2020] [Indexed: 11/18/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is a diarrheagenic pathotype associated with traveler’s diarrhea, foodborne outbreaks and sporadic diarrhea in industrialized and developing countries. Regulation of virulence in EAEC is mediated by AggR and its negative regulator Aar. Together, they control the expression of at least 210 genes. On the other hand, we observed that about one third of Aar-regulated genes are related to metabolism and transport. In this study we show the AggR/Aar duo controls the metabolism of lipids. Accordingly, we show that AatD, encoded in the AggR-regulated aat operon (aatPABCD) is an N-acyltransferase structurally similar to the essential Apolipoprotein N-acyltransferase Lnt and is required for the acylation of Aap (anti-aggregation protein). Deletion of aatD impairs post-translational modification of Aap and causes its accumulation in the bacterial periplasm. trans-complementation of 042aatD mutant with the AatD homolog of ETEC or with the N-acyltransferase Lnt reestablished translocation of Aap. Site-directed mutagenesis of the E207 residue in the putative acyltransferase catalytic triad disrupted the activity of AatD and caused accumulation of Aap in the periplasm due to reduced translocation of Aap at the bacterial surface. Furthermore, Mass spectroscopy revealed that Aap is acylated in a putative lipobox at the N-terminal of the mature protein, implying that Aap is a lipoprotein. Lastly, deletion of aatD impairs bacterial colonization of the streptomycin-treated mouse model. Our findings unveiled a novel N-acyltransferase family associated with bacterial virulence, and that is tightly regulated by AraC/XylS regulators in the order Enterobacterales. Although the regulatory scheme of AggR is well understood, the biological relevance of half of AggR-regulated proteins remains unknown. In this study we provide experimental evidence that the AggR-regulated AatD is a novel N-acyltransferase restricted to pathogens of the order Enterobacterales, including EAEC, ETEC, Yersinia sp., and C. rodentium. AatD is structurally similar to Lnt. However, unlike Lnt which is essential for cellular functions, AatD is a dedicated N-acyltransferase required for post-translational modification of virulence factors. Aap was identified as a lipoprotein acylated by AatD. Lipid modification in Aap provides an important post-translational mechanism to regulate the trafficking, stability and subcellular localization of Aap. In the absence of AatD, Aap is retained in the periplasmic space and cannot be translocated to the bacterial surface, presumably, restricting the biological function of the protein. Our data suggest that AggR and Aar virulence regulators, not only regulate the expression of Aap virulence factor at the transcriptional level, but also regulate translocation of Aap to the bacterial surface, which is required for full virulence of EAEC, unveiling an important mechanism of virulence regulation.
Collapse
Affiliation(s)
- Laura Belmont-Monroy
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
- Department of Public Health, UNAM School of Medicine and Federico Gomez Children’s Hospital, Mexico City, Mexico
| | - Waleska Saitz-Rojas
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
| | - Jorge Soria-Bustos
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
| | - Abigail S. Mickey
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
| | - Nicholas E. Sherman
- W. M. Keck Biomedical Mass Spectrometry Lab. University of Virginia, Charlottesville, Virginia, United States of America
| | - Benjamin C. Orsburn
- W. M. Keck Biomedical Mass Spectrometry Lab. University of Virginia, Charlottesville, Virginia, United States of America
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
| | - Araceli E. Santiago
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children’s Hospital, Charlottesville, Virginia, United States of America
- * E-mail: .
| |
Collapse
|
13
|
Dual Function of Aar, a Member of the New AraC Negative Regulator Family, in Escherichia coli Gene Expression. Infect Immun 2020; 88:IAI.00100-20. [PMID: 32253248 DOI: 10.1128/iai.00100-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/30/2020] [Indexed: 11/20/2022] Open
Abstract
Enteroaggregative Escherichia coli (EAEC) is an E. coli pathotype associated with diarrhea and growth faltering. EAEC virulence gene expression is controlled by the autoactivated AraC family transcriptional regulator, AggR. AggR activates transcription of a large number of virulence genes, including Aar, which in turn acts as a negative regulator of AggR itself. Aar has also been shown to affect expression of E. coli housekeeping genes, including H-NS, a global regulator that acts at multiple promoters and silences AT-rich genes (such as those in the AggR regulon). Although Aar has been shown to bind both AggR and H-NS in vitro, functional significance of these interactions has not been shown in vivo In order to dissect this regulatory network, we removed the complex interdependence of aggR and aar by placing the genes under the control of titratable promoters. We measured phenotypic and genotypic changes on downstream genes in EAEC strain 042 and E. coli K-12 strain DH5α, which lacks the AggR regulon. In EAEC, we found that low expression of aar increases aafA fimbrial gene expression via H-NS; however, when aar is more highly expressed, it acts as a negative regulator via AggR. In DH5α, aar affected expression of E. coli genes in some cases via H-NS and in some cases independent of H-NS. Our data support the model that Aar interacts in concert with AggR, H-NS, and possibly other regulators and that these interactions are likely to be functionally significant in vivo.
Collapse
|
14
|
Petro CD, Duncan JK, Seldina YI, Allué-Guardia A, Eppinger M, Riddle MS, Tribble DR, Johnson RC, Dalgard CL, Sukumar G, Connor P, Boisen N, Melton-Celsa AR. Genetic and Virulence Profiles of Enteroaggregative Escherichia coli (EAEC) Isolated From Deployed Military Personnel (DMP) With Travelers' Diarrhea. Front Cell Infect Microbiol 2020; 10:200. [PMID: 32509590 PMCID: PMC7251025 DOI: 10.3389/fcimb.2020.00200] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/16/2020] [Indexed: 02/01/2023] Open
Abstract
To discern if there was a particular genotype associated with clinical enteroaggregative Escherichia coli (EAEC) strains isolated from deployed military personnel (DMP) with travelers' diarrhea (TD), we characterized a collection of EAEC from DMP deployed to Afghanistan, Djibouti, Kenya, or Honduras. Although we did not identify a specific EAEC genotype associated with TD in DMP, we found that EAEC isolated at the first clinic visit were more likely to encode the dispersin gene aap than EAEC collected at follow-up visits. A majority of the EAEC isolates were typical EAEC that adhered to HEp-2 cells, formed biofilms, and harbored genes for aggregative adherence fimbriae (AAF), AggR, and serine protease autotransporters of Enterobacteriaceae (SPATEs). A separate subset of the EAEC had aggR and genes for SPATEs but encoded a gene highly homologous to that for CS22, a fimbriae more commonly found in enterotoxigenic E. coli. None of these CS22-encoding EAEC formed biofilms in vitro or adhered to HEp-2 cells. Whole genome sequence and single nucleotide polymorphism analyses demonstrated that most of the strains were genetically diverse, but that a few were closely related. Isolation of these related strains occurred within days to more than a year apart, a finding that suggests a persistent source and genomic stability. In an ampicillin-treated mouse model we found that an agg4A+ aar- isolate formed a biofilm in the intestine and caused reduced weight gain in mice, whereas a strain that did not form an in vivo biofilm caused no morbidity. Our diverse strain collection from DMP displays the heterogeneity of EAEC strains isolated from human patients, and our mouse model of infection indicated the genotype agg4A+ aar– and/or capacity to form biofilm in vivo may correlate to disease severity.
Collapse
Affiliation(s)
- Courtney D Petro
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Jeffrey K Duncan
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Yuliya I Seldina
- Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Anna Allué-Guardia
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States.,South Texas Center for Emerging Infectious Diseases, San Antonio, TX, United States
| | - Mark Eppinger
- Department of Biology, The University of Texas at San Antonio, San Antonio, TX, United States.,South Texas Center for Emerging Infectious Diseases, San Antonio, TX, United States
| | - Mark S Riddle
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - David R Tribble
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Ryan C Johnson
- Department of Preventative Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Gauthaman Sukumar
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,Collaborative Health Initiative Research Program, Henry Jackson Foundation, Bethesda, MD, United States
| | - Patrick Connor
- Military Enteric Disease Group, Academic Department of Military Medicine, Birmingham, United Kingdom
| | - Nadia Boisen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Angela R Melton-Celsa
- Department of Microbiology and Immunolgy, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
15
|
Emergence of Enteroaggregative Escherichia coli within the ST131 Lineage as a Cause of Extraintestinal Infections. mBio 2020; 11:mBio.00353-20. [PMID: 32430467 PMCID: PMC7240153 DOI: 10.1128/mbio.00353-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
E. coli ST131 is an important extraintestinal pathogenic lineage. A signature characteristic of ST131 is its ability to asymptomatically colonize the gastrointestinal tract and then opportunistically cause extraintestinal infections, such as cystitis, pyelonephritis, and urosepsis. In this study, we identified an ST131 H27 sublineage that has acquired the enteroaggregative diarrheagenic phenotype, spread across multiple continents, and caused multiple outbreaks of community-acquired ESBL-associated bloodstream infections in Denmark. The strain’s ability to both cause diarrhea and innocuously colonize the human gastrointestinal tract may facilitate its dissemination and establishment in the community. Escherichia coli sequence type 131 (ST131) is a major cause of urinary and bloodstream infections. Its association with extended-spectrum β-lactamases (ESBLs) significantly complicates treatment. Its best-described component is the rapidly expanding H30Rx clade, containing allele 30 of the type 1 fimbrial adhesin gene fimH. This lineage appears to have emerged in the United States and spread around the world in part due to the acquisition of the ESBL-encoding blaCTX-M-15 gene and resistance to fluoroquinolones. However, non-H30 ST131 sublineages with other acquired CTX-M-type resistance genes are also emerging. Based on whole-genome analyses, we describe here the presence of an (fimH) H27 E. coli ST131 sublineage that has recently caused an outbreak of community-acquired bacteremia and recurrent urinary tract infections (UTIs) in Denmark. This sublineage has acquired both a virulence plasmid (pAA) that defines the enteroaggregative E. coli (EAEC) diarrheagenic pathotype and multiple genes associated with extraintestinal E. coli (ExPEC); combined, these traits have made this particular ST131 sublineage successful at colonizing its human host and causing recurrent UTI. Moreover, using a historic World Health Organization (WHO) E. coli collection and publicly available genome sequences, we identified a global H27 EAEC ST131 sublineage that dates back as far as 1998. Most H27 EAEC ST131 isolates harbor pAA or pAA-like plasmids, and our analysis strongly implies a single ancestral acquisition among these isolates. These findings illustrate both the profound plasticity of this important pathogenic E. coli ST131 H27 sublineage and genetic acquisitions of EAEC-specific virulence traits that likely confer an enhanced ability to cause intestinal colonization.
Collapse
|
16
|
Escherichia coli ST131 clones harbouring AggR and AAF/V fimbriae causing bacteremia in Mozambican children: Emergence of new variant of fimH27 subclone. PLoS Negl Trop Dis 2020; 14:e0008274. [PMID: 32357189 PMCID: PMC7219792 DOI: 10.1371/journal.pntd.0008274] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 05/13/2020] [Accepted: 04/07/2020] [Indexed: 11/25/2022] Open
Abstract
Multidrug-resistant Escherichia coli ST131 fimH30 responsible for extra-intestinal pathogenic (ExPEC) infections is globally distributed. However, the occurrence of a subclone fimH27 of ST131 harboring both ExPEC and enteroaggregative E. coli (EAEC) related genes and belonging to commonly reported O25:H4 and other serotypes causing bacteremia in African children remain unknown. We characterized 325 E. coli isolates causing bacteremia in Mozambican children between 2001 and 2014 by conventional multiplex polymerase chain reaction and whole genome sequencing. Incidence rate of EAEC bacteremia was calculated among cases from the demographic surveillance study area. Approximately 17.5% (57/325) of isolates were EAEC, yielding an incidence rate of 45.3 episodes/105 children-years-at-risk among infants; and 44 of isolates were sequenced. 72.7% (32/44) of sequenced strains contained simultaneously genes associated with ExPEC (iutA, fyuA and traT); 88.6% (39/44) harbored the aggregative adherence fimbriae type V variant (AAF/V). Sequence type ST-131 accounted for 84.1% (37/44), predominantly belonging to serotype O25:H4 (59% of the 37); 95.6% (35/44) harbored fimH27. Approximately 15% (6/41) of the children died, and five of the six yielded ST131 strains (83.3%) mostly (60%; 3/5) due to serotypes other than O25:H4. We report the emergence of a new subclone of ST-131 E. coli strains belonging to O25:H4 and other serotypes harboring both ExPEC and EAEC virulence genes, including agg5A, associated with poor outcome in bacteremic Mozambican children, suggesting the need for prompt recognition for appropriate management. Escherichia coli ST131 has emerged as a globally disseminated multi-drug resistant clone associated with extra-intestinal infections acquired in the community or hospital. In Manhiça district, E. coli is among the top five leading bloodstream pathogens in children. We characterized E. coli strains causing bacteremia in young children in a rural hospital of Mozambique, providing novel information on the occurrence of a new subclone of ST131 harboring both ExPEC and EAEC related genes and belonging to commonly reported O25:H4 and other serotypes. These data suggest the need for further understanding of pathogenesis and clinical impact of this new entity to inform prompt recognition and appropriate treatment.
Collapse
|
17
|
Peñil-Celis A, Garcillán-Barcia MP. Crosstalk Between Type VI Secretion System and Mobile Genetic Elements. Front Mol Biosci 2019; 6:126. [PMID: 31799257 PMCID: PMC6863884 DOI: 10.3389/fmolb.2019.00126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Many bacterial processes require cell-cell contacts. Such are the cases of bacterial conjugation, one of the main horizontal gene transfer mechanisms that physically spreads DNA, and the type VI secretion systems (T6SSs), which deploy antibacterial activity. Bacteria depend on conjugation to adapt to changing environments, while T6SS killing activity could pose a threat to mating partners. Here we review the experimental evidences of overlapping and interaction between the T6SSs, bacterial conjugation, and conjugative genetic elements.
Collapse
Affiliation(s)
- Arancha Peñil-Celis
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, Santander, Spain
| | - M Pilar Garcillán-Barcia
- Instituto de Biomedicina y Biotecnología de Cantabria, Universidad de Cantabria-Consejo Superior de Investigaciones Científicas, Santander, Spain
| |
Collapse
|
18
|
Boisen N, Melton-Celsa AR, Hansen AM, Zangari T, Smith MA, Russo LM, Scheutz F, O'Brien AD, Nataro JP. The Role of the AggR Regulon in the Virulence of the Shiga Toxin-Producing Enteroaggregative Escherichia coli Epidemic O104:H4 Strain in Mice. Front Microbiol 2019; 10:1824. [PMID: 31456767 PMCID: PMC6700298 DOI: 10.3389/fmicb.2019.01824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/24/2019] [Indexed: 12/04/2022] Open
Abstract
An O104:H4 Shiga toxin (Stx)-producing enteroaggregative Escherichia coli (EAEC) strain caused a large outbreak of bloody diarrhea and the hemolytic uremic syndrome in 2011. We previously developed an ampicillin (Amp)-treated C57BL/6 mouse model to measure morbidity (weight loss) and mortality of mice orally infected with the prototype Stx-EAEC strain C227-11. Here, we hypothesized that mice fed C227-11 cured of the pAA plasmid or deleted for individual genes on that plasmid would display reduced virulence compared to animals given the wild-type (wt) strain. C227-11 cured of the pAA plasmid or deleted for the known pAA-encoded virulence genes aggR, aggA, sepA, or aar were fed to Amp-treated C57BL/6 mice at doses of 1010–1011CFU. Infected animals were then either monitored for morbidity and lethality for 28 days or euthanized to determine intestinal pathology and colonization levels at selected times. The pAA-cured, aggR, and aggA mutants of strain C227-11 all showed reduced colonization at various intestinal sites. However, the aggR mutant was the only mutant attenuated for virulence as it showed both reduced morbidity and mortality. The aar mutant showed increased expression of the aggregative adherence fimbriae (AAF) and caused greater systemic effects in infected mice when compared to the C227-11 wt strain. However, unexpectedly, both the aggA and aar mutants displayed increased weight loss compared to wt. The sepA mutant did not exhibit altered morbidity or mortality in the Amp-treated mouse model compared to wt. Our data suggest that the increased morbidity due to the aar mutant could possibly be via an effect on expression of an as yet unknown virulence-associated factor under AggR control.
Collapse
Affiliation(s)
- Nadia Boisen
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark.,Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Angela R Melton-Celsa
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Anne-Marie Hansen
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonia Zangari
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Mark A Smith
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Lisa M Russo
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Flemming Scheutz
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Alison D O'Brien
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW The current review is to update the results on epidemiology, pathobiology, and genes related to virulence, clinical presentation, molecular diagnosis, antimicrobial resistance, and extraintestinal infection of enteroaggregative Escherichia coli (EAEC). RECENT FINDINGS EAEC subclinical infection was significantly associated with reduced length at 2 years of age and EAEC and coinfections were associated with reduced delta weight-for-length and weight-for-age z-scores in the first 6 months of age in the MAL-ED birth cohort study. EAEC was associated with malnutrition in children 6-24 months of age in prospective case-control studies in Bangladesh and Brazil. Virulence gene-based studies have suggested aggregative fimbriae II may be a major contributor to disease, whereas AggR-activated regulator a marker of less severe disease. The high ability of EAEC colonization likely exacerbates effects of other microbial virulence strategies. Molecular diagnosis has been useful for understanding EAEC burden, although different criteria may relate to different pathogenic outcomes. SUMMARY EAEC gained special interest in the past few years, especially due to association with growth decrements in children with subclinical infections and its important role as a copathogen. Understanding of EAEC pathogenesis advanced but further research is needed for elucidating both microbial and host factors influencing infection outcomes.
Collapse
|
20
|
The bile salt glycocholate induces global changes in gene and protein expression and activates virulence in enterotoxigenic Escherichia coli. Sci Rep 2019; 9:108. [PMID: 30643184 PMCID: PMC6331568 DOI: 10.1038/s41598-018-36414-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 11/20/2018] [Indexed: 12/23/2022] Open
Abstract
Pathogenic bacteria use specific host factors to modulate virulence and stress responses during infection. We found previously that the host factor bile and the bile component glyco-conjugated cholate (NaGCH, sodium glycocholate) upregulate the colonization factor CS5 in enterotoxigenic Escherichia coli (ETEC). To further understand the global regulatory effects of bile and NaGCH, we performed Illumina RNA-Seq and found that crude bile and NaGCH altered the expression of 61 genes in CS5 + CS6 ETEC isolates. The most striking finding was high induction of the CS5 operon (csfA-F), its putative transcription factor csvR, and the putative ETEC virulence factor cexE. iTRAQ-coupled LC-MS/MS proteomic analyses verified induction of the plasmid-borne virulence proteins CS5 and CexE and also showed that NaGCH affected the expression of bacterial membrane proteins. Furthermore, NaGCH induced bacteria to aggregate, increased their adherence to epithelial cells, and reduced their motility. Our results indicate that CS5 + CS6 ETEC use NaGCH present in the small intestine as a signal to initiate colonization of the epithelium.
Collapse
|
21
|
Yasir M, Icke C, Abdelwahab R, Haycocks JR, Godfrey RE, Sazinas P, Pallen MJ, Henderson IR, Busby SJW, Browning DF. Organization and architecture of AggR-dependent promoters from enteroaggregative Escherichia coli. Mol Microbiol 2018; 111:534-551. [PMID: 30485564 PMCID: PMC6392122 DOI: 10.1111/mmi.14172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2018] [Indexed: 11/27/2022]
Abstract
Enteroaggregative Escherichia coli (EAEC), is a diarrhoeagenic human pathogen commonly isolated from patients in both developing and industrialized countries. Pathogenic EAEC strains possess many virulence determinants, which are thought to be involved in causing disease, though, the exact mechanism by which EAEC causes diarrhoea is unclear. Typical EAEC strains possess the transcriptional regulator, AggR, which controls the expression of many virulence determinants, including the attachment adherence fimbriae (AAF) that are necessary for adherence to human gut epithelial cells. Here, using RNA‐sequencing, we have investigated the AggR regulon from EAEC strain 042 and show that AggR regulates the transcription of genes on both the bacterial chromosome and the large virulence plasmid, pAA2. Due to the importance of fimbriae, we focused on the two AAF/II fimbrial gene clusters in EAEC 042 (afaB‐aafCB and aafDA) and identified the promoter elements and AggR‐binding sites required for fimbrial expression. In addition, we examined the organization of the fimbrial operon promoters from other important EAEC strains to understand the rules of AggR‐dependent activation. Finally, we generated a series of semi‐synthetic promoters to define the minimal sequence required for AggR‐mediated activation and show that the correct positioning of a single AggR‐binding site is sufficient to confer AggR‐dependence.
Collapse
Affiliation(s)
- Muhammad Yasir
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.,Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Christopher Icke
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Radwa Abdelwahab
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK.,Faculty of Medicine, Assiut University, Assiut, Egypt
| | - James R Haycocks
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Rita E Godfrey
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Pavelas Sazinas
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs Lyngby, Denmark
| | - Mark J Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UA, UK
| | - Ian R Henderson
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Stephen J W Busby
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
22
|
Krishna A, Holden MTG, Peacock SJ, Edwards AM, Wigneshweraraj S. Naturally occurring polymorphisms in the virulence regulator Rsp modulate Staphylococcus aureus survival in blood and antibiotic susceptibility. MICROBIOLOGY (READING, ENGLAND) 2018; 164:1189-1195. [PMID: 30028663 PMCID: PMC6230762 DOI: 10.1099/mic.0.000695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/03/2018] [Indexed: 01/20/2023]
Abstract
Nasal colonization by the pathogen Staphylococcus aureus is a risk factor for subsequent infection. Loss of function mutations in the gene encoding the virulence regulator Rsp are associated with the transition of S. aureus from a colonizing isolate to one that causes bacteraemia. Here, we report the identification of several novel activity-altering mutations in rsp detected in clinical isolates, including for the first time, mutations that enhance agr operon activity. We assessed how these mutations affected infection-relevant phenotypes and found loss and enhancement of function mutations to have contrasting effects on S. aureus survival in blood and antibiotic susceptibility. These findings add to the growing body of evidence that suggests S. aureus 'trades off' virulence for the acquisition of traits that benefit survival in the host, and indicates that infection severity and treatment options can be significantly affected by mutations in the virulence regulator rsp.
Collapse
Affiliation(s)
- Aishwarya Krishna
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Matthew T. G. Holden
- Wellcome Trust Sanger Institute, Hinxton, UK
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Sharon J. Peacock
- Wellcome Trust Sanger Institute, Hinxton, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Andrew M. Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | | |
Collapse
|
23
|
Borgersen Q, Bolick DT, Kolling GL, Aijuka M, Ruiz-Perez F, Guerrant RL, Nataro JP, Santiago AE. Abundant production of exopolysaccharide by EAEC strains enhances the formation of bacterial biofilms in contaminated sprouts. Gut Microbes 2018; 9:264-278. [PMID: 29543544 PMCID: PMC6219584 DOI: 10.1080/19490976.2018.1429877] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 01/02/2018] [Accepted: 01/12/2018] [Indexed: 02/03/2023] Open
Abstract
Enteroaggregative E. coli (EAEC) is associated with food-borne outbreaks of diarrhea and growth faltering among children in developing countries. A Shiga toxin-producing EAEC strain of serotype O104:H4 strain caused one of the largest outbreaks of a food-borne infection in Europe in 2011. The outbreak was traced to contaminated fenugreek sprouts, yet the mechanisms whereby such persistent contamination of sprouts could have occurred are not clear. We found that under ambient conditions of temperature and in minimal media, pathogenic Shiga toxin-producing EAEC O104:H4 227-11 and non-Shiga toxin-producing 042 strains both produce high levels of exopolysaccharide structures (EPS) that are released to the external milieu. The exopolysaccharide was identified as colanic acid (CA). Unexpectedly, Shiga-toxin producing EAEC strain 227-11 produced 3-6-fold higher levels of CA than the 042 strain, suggesting differential regulation of the CA in the two strains. The presence of CA was accompanied by the formation of large biofilm structures on the surface of sprouts. The wcaF-wza chromosomal locus was required for the synthesis of CA in EAEC 042. Deletion in the glycosyltransferase wcaE gene abolished the production of CA in 042, and resulted in diminished adherence to sprouts when co-cultured at ambient temperature. In conclusion, this work suggests that copious production of CA may contribute to persistence of EAEC in the environment and suggests a potential explanation for the large Shiga toxin-producing EAEC outbreak in 2011.
Collapse
Affiliation(s)
- Quintin Borgersen
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children's Hospital, Charlottesville, Virginia
| | - David T. Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA , USA
| | - Glynis L. Kolling
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA , USA
| | - Matthew Aijuka
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children's Hospital, Charlottesville, Virginia
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children's Hospital, Charlottesville, Virginia
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA , USA
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children's Hospital, Charlottesville, Virginia
| | - Araceli E. Santiago
- Department of Pediatrics, University of Virginia School of Medicine and University of Virginia Children's Hospital, Charlottesville, Virginia
| |
Collapse
|
24
|
Hüttener M, Prieto A, Espelt J, Bernabeu M, Juárez A. Stringent Response and AggR-Dependent Virulence Regulation in the Enteroaggregative Escherichia coli Strain 042. Front Microbiol 2018; 9:717. [PMID: 29692772 PMCID: PMC5902536 DOI: 10.3389/fmicb.2018.00717] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/27/2018] [Indexed: 12/22/2022] Open
Abstract
Virulence expression in the enteroaggregative Escherichia coli strain 042 requires the transcriptional activator AggR. We show in this report that, as reported for other virulence factors, the nucleotide second messenger (p)ppGpp is needed for a high expression level of AggR. As expected from these findings, expression of AggR-activated genes such as the AafA pilin subunit is downregulated in the absence of (p)ppGpp. Considering the fact that biofilm formation in strain 042 requires the AafA protein, biofilm development in strain 042 is impaired in derivatives that lack either the AggR protein, the virulence plasmid that encodes AggR (pAA2) or the ability to synthesize (p)ppGpp. These results show a direct correlation between (p)ppGpp, expression of AggR and biofilm development in strain 042.
Collapse
Affiliation(s)
- Mário Hüttener
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Joan Espelt
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Antonio Juárez
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| |
Collapse
|
25
|
Affiliation(s)
- Bente Olesen
- Department of Clinical Microbiology, Herlev and Gentofte Hospital, University of Copenhagen, Denmark
| |
Collapse
|
26
|
Kampmeier S, Berger M, Mellmann A, Karch H, Berger P. The 2011 German Enterohemorrhagic Escherichia Coli O104:H4 Outbreak-The Danger Is Still Out There. Curr Top Microbiol Immunol 2018; 416:117-148. [PMID: 30062592 DOI: 10.1007/82_2018_107] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are Shiga toxin (Stx) producing bacteria causing a disease characterized by bloody (or non-bloody) diarrhea, which might progress to hemolytic uremic syndrome (HUS). EHEC O104:H4 caused the largest ever recorded EHEC outbreak in Germany in 2011, which in addition showed the so far highest incidence rate of EHEC-related HUS worldwide. The aggressive outbreak strain carries an unusual combination of virulence traits characteristic to both EHEC-a chromosomally integrated Stx-encoding bacteriophage, and enteroaggregative Escherichia coli-pAA plasmid-encoded aggregative adherence fimbriae mediating its tight adhesion to epithelia cells. There are currently still open questions regarding the 2011 EHEC outbreak, e.g., with respect to the exact molecular mechanisms resulting in the hypervirulence of the strain, the natural reservoir of EHEC O104:H4, and suitable therapeutic strategies. Nevertheless, our knowledge on these issues has substantially expanded since 2011. Here, we present an overview of the epidemiological, clinical, microbiological, and molecular biological data available on the 2011 German EHEC O104:H4 outbreak.
Collapse
Affiliation(s)
| | - Michael Berger
- Institute of Hygiene, University of Münster, Münster, Germany
| | | | - Helge Karch
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Petya Berger
- Institute of Hygiene, University of Münster, Münster, Germany.
| |
Collapse
|
27
|
Santiago AE, Yan MB, Hazen TH, Sauder B, Meza-Segura M, Rasko DA, Kendall MM, Ruiz-Perez F, Nataro JP. The AraC Negative Regulator family modulates the activity of histone-like proteins in pathogenic bacteria. PLoS Pathog 2017; 13:e1006545. [PMID: 28806780 PMCID: PMC5570504 DOI: 10.1371/journal.ppat.1006545] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 08/24/2017] [Accepted: 07/20/2017] [Indexed: 02/04/2023] Open
Abstract
The AraC Negative Regulators (ANR) comprise a large family of virulence regulators distributed among diverse clinically important Gram-negative pathogens, including Vibrio spp., Salmonella spp., Shigella spp., Yersinia spp., Citrobacter spp., and pathogenic E. coli strains. We have previously reported broad effects of the ANR members on regulators of the AraC/XylS family. Here, we interrogate possible broader effects of the ANR members on the bacterial transcriptome. Our studies focused on Aar (AggR-activated regulator), an ANR family archetype in enteroaggregative E. coli (EAEC) isolate 042. Transcriptome analysis of EAEC strain 042, 042aar and 042aar(pAar) identified more than 200 genes that were differentially expressed (+/- 1.5 fold, p<0.05). Most of those genes are located on the bacterial chromosome (195 genes, 92.85%), and are associated with regulation, transport, metabolism, and pathogenesis, based on the predicted annotation; a considerable number of Aar-regulated genes encoded for hypothetical proteins (46 genes, 21.9%) and regulatory proteins (25, 11.9%). Notably, the transcriptional expression of three histone-like regulators, H-NS (orf1292), H-NS homolog (orf2834) and StpA, was down-regulated in the absence of aar and may explain some of the effects of Aar on gene expression. By employing a bacterial two-hybrid system, LacZ reporter assays, pull-down and electrophoretic mobility shift assay (EMSA) analysis, we demonstrated that Aar binds directly to H-NS and modulates H-NS-induced gene silencing. Importantly, Aar was highly expressed in the mouse intestinal tract and was found to be necessary for maximal H-NS expression. In conclusion, this work further extends our knowledge of genes under the control of Aar and its biological relevance in vivo. The AraC Negative Regulators (ANR) is a large family of negative regulators distributed in several clinically relevant Gram-negative pathogens, including Vibrio spp., Salmonella spp., Shigella spp., Yersinia spp., Citrobacter spp., pathogenic E. coli, and members of the Pasteurellaceae. Previously, we showed that the ANR family suppresses transcriptional expression of virulence factors such as fimbriae, toxins, and the type VI secretion system by directly down-regulating AraC/XylS master regulators. Transcriptome and biochemical analysis of Aar (AggR-activated regulator), an ANR family archetype in enteroaggregative E. coli (EAEC) 042, demonstrated that Aar binds directly to H-NS and modulates the H-NS-induced gene expression. Accordingly, mutation of aar decreased expression of the H-NS-regulated Lpf fimbriae, LPS-related enzymes, GadXW operon and porins. Importantly, Aar was highly expressed in the mouse intestinal tract and was found to be necessary for maximal H-NS expression. These findings unveil an exquisite regulatory network in pathogenic bacteria, which operates by concomitant control of master transcriptional regulators of the AraC family and global negative H-NS regulators.
Collapse
Affiliation(s)
- Araceli E. Santiago
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- * E-mail:
| | - Michael B. Yan
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Tracy H. Hazen
- Institute for Genome Sciences, Department of Microbiology and Immunology. University of Maryland, Baltimore, Maryland, United States of America
| | - Brooke Sauder
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Mario Meza-Segura
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - David A. Rasko
- Institute for Genome Sciences, Department of Microbiology and Immunology. University of Maryland, Baltimore, Maryland, United States of America
| | - Melissa M. Kendall
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - James P. Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
28
|
Hodson C, Yang J, Hocking DM, Azzopardi K, Chen Q, Holien JK, Parker MW, Tauschek M, Robins-Browne RM. Control of Virulence Gene Expression by the Master Regulator, CfaD, in the Prototypical Enterotoxigenic Escherichia coli Strain, H10407. Front Microbiol 2017; 8:1525. [PMID: 28848532 PMCID: PMC5554520 DOI: 10.3389/fmicb.2017.01525] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/28/2017] [Indexed: 12/24/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the most common bacterial cause of diarrhea in children in developing countries, as well as in travelers to these countries. To cause disease, ETEC needs to produce a series of virulence proteins including enterotoxins, colonization factors and secretion pathways, which enable this pathogen to colonize the human small intestine and deliver enterotoxins to epithelial cells. Previously, a number of studies have demonstrated that CfaD, an AraC-like transcriptional regulator, plays a key role in virulence gene expression by ETEC. In this study, we carried out a transcriptomic analysis of ETEC strain, H10407, grown under different conditions, and determined the complete set of genes that are regulated by CfaD. In this way, we identified a number of new target genes, including rnr-1, rnr-2, etpBAC, agn43, flu, traM and ETEC_3214, whose expression is strongly activated by CfaD. Using promoter-lacZ reporters, primer extension and electrophoretic mobility shift assays, we characterized the CfaD-mediated activation of several selected target promoters. We also showed that the gut-associated environmental signal, sodium bicarbonate, stimulates CfaD-mediated upregulation of its virulence target operons. Finally, we screened a commercial small molecule library and identified a compound (CH-1) that specifically inhibited the regulatory function of CfaD, and by 2-D analoging, we identified a second inhibitor (CH-2) with greater potency.
Collapse
Affiliation(s)
- Carla Hodson
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia
| | - Ji Yang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia
| | - Dianna M Hocking
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia
| | - Kristy Azzopardi
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia.,Murdoch Childrens Research Institute, The Royal Children's Hospital, ParkvilleVIC, Australia
| | - Qianyu Chen
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia
| | - Jessica K Holien
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, FitzroyVIC, Australia
| | - Michael W Parker
- Australian Cancer Research Foundation Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, FitzroyVIC, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, ParkvilleVIC, Australia
| | - Marija Tauschek
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia
| | - Roy M Robins-Browne
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, ParkvilleVIC, Australia.,Murdoch Childrens Research Institute, The Royal Children's Hospital, ParkvilleVIC, Australia
| |
Collapse
|
29
|
Havt A, Lima IF, Medeiros PH, Clementino MA, Santos AK, Amaral MS, Veras HN, Prata MM, Lima NL, Di Moura A, Leite ÁM, Soares AM, Filho JQ, Houpt ER, Nataro JP, Guerrant RL, Lima AA. Prevalence and virulence gene profiling of enteroaggregative Escherichia coli in malnourished and nourished Brazilian children. Diagn Microbiol Infect Dis 2017; 89:98-105. [PMID: 28780245 PMCID: PMC5608016 DOI: 10.1016/j.diagmicrobio.2017.06.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 01/07/2023]
Abstract
The impact of enteroaggregative E. coli (EAEC) infection on childhood malnutrition and inflammation has been suggested, regardless of diarrhea. We investigated whether EAEC and its virulence-related genes (VRGs) are associated with malnutrition in a case-control study. Children aged 6-24 months from Brazil were enrolled as malnourished if weight-for-age Z-score (WAZ) ≤ -2 and nourished if WAZ > -1. Stools were cultured and examined for E. coli. DNA was extracted from fecal isolates and tested for EAEC by polymerase chain reaction (PCR). Positive samples were analyzed by 5 multiplex PCRs to identify 20 EAEC VRGs. Biomarkers of intestinal barrier function and inflammation were measured. The prevalence of EAEC was 39.94%. Samples that presented both aaiC and aatA genes were associated with malnutrition (P = 0.045). A high prevalence of VRGs was observed and the aafC gene was significantly associated with malnourished (P = 0.0101). Strains lacking aar and pic genes were associated with malnutrition (P = 0.018), while the concomitant presence of aar, pic, agg4A, and capU genes was associated with nourished (P = 0.031). These data reinforce the EAEC impact on malnutrition, the importance of aar as negative regulator and the great contribution of AAF/II fimbria for the pathobiology of EAEC.
Collapse
Affiliation(s)
- Alexandre Havt
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil.
| | - Ila Fn Lima
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Pedro Hqs Medeiros
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Marco Af Clementino
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Ana Ks Santos
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Marília Smg Amaral
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Herlice N Veras
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Mara Mg Prata
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Noélia L Lima
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Alessandra Di Moura
- Institute for the Promotion of Nutrition and Human Development, 15 Professor Carlos Lobo, 60281-740, Fortaleza, Ceará, Brazil
| | - Álvaro M Leite
- Institute for the Promotion of Nutrition and Human Development, 15 Professor Carlos Lobo, 60281-740, Fortaleza, Ceará, Brazil
| | - Alberto M Soares
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - José Q Filho
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil
| | - Eric R Houpt
- Center for Global Health & Division of Infectious Diseases and International Health and Department of Pediatrics, University of Virginia, 1400 W Main Street, 22908-1379, Charlottesville, VA, USA
| | - James P Nataro
- Center for Global Health & Division of Infectious Diseases and International Health and Department of Pediatrics, University of Virginia, 1400 W Main Street, 22908-1379, Charlottesville, VA, USA
| | - Richard L Guerrant
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil; Center for Global Health & Division of Infectious Diseases and International Health and Department of Pediatrics, University of Virginia, 1400 W Main Street, 22908-1379, Charlottesville, VA, USA
| | - Aldo Am Lima
- Institute of Biomedicine for Brazilian Semiarid, Federal University of Ceará, 1315 Coronel Nunes de Melo, 60430-270, Fortaleza, Brazil; Center for Global Health & Division of Infectious Diseases and International Health and Department of Pediatrics, University of Virginia, 1400 W Main Street, 22908-1379, Charlottesville, VA, USA
| |
Collapse
|
30
|
Berger P, Knödler M, Förstner KU, Berger M, Bertling C, Sharma CM, Vogel J, Karch H, Dobrindt U, Mellmann A. The primary transcriptome of the Escherichia coli O104:H4 pAA plasmid and novel insights into its virulence gene expression and regulation. Sci Rep 2016; 6:35307. [PMID: 27748404 PMCID: PMC5066232 DOI: 10.1038/srep35307] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/27/2016] [Indexed: 11/30/2022] Open
Abstract
Escherichia coli O104:H4 (E. coli O104:H4), which caused a massive outbreak of acute gastroenteritis and hemolytic uremic syndrome in 2011, carries an aggregative adherence fimbriae I (AAF/I) encoding virulence plasmid, pAA. The importance of pAA in host-pathogen interaction and disease severity has been demonstrated, however, not much is known about its transcriptional organization and gene regulation. Here, we analyzed the pAA primary transcriptome using differential RNA sequencing, which allows for the high-throughput mapping of transcription start site (TSS) and non-coding RNA candidates. We identified 248 TSS candidates in the 74-kb pAA and only 21% of them could be assigned as TSS of annotated genes. We detected TSS for the majority of pAA-encoded virulence factors. Interestingly, we mapped TSS, which could allow for the transcriptional uncoupling of the AAF/I operon, and potentially regulatory antisense RNA candidates against the genes encoding dispersin and the serine protease SepA. Moreover, a computational search for transcription factor binding sites suggested for AggR-mediated activation of SepA expression, which was additionally experimentally validated. This work advances our understanding of the molecular basis of E. coli O104:H4 pathogenicity and provides a valuable resource for further characterization of pAA virulence gene regulation.
Collapse
Affiliation(s)
- Petya Berger
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Michael Knödler
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Konrad U Förstner
- Core Unit Systems Medicine, University of Würzburg, Würzburg, Germany.,Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany.,Research Center for Infectious Diseases, University of Würzburg, Würzburg, Germany
| | - Michael Berger
- Institute of Hygiene, University of Münster, Münster, Germany
| | | | - Cynthia M Sharma
- Research Center for Infectious Diseases, University of Würzburg, Würzburg, Germany
| | - Jörg Vogel
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, Germany
| | - Helge Karch
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster, Germany
| | | |
Collapse
|
31
|
Santiago AE, Yan MB, Tran M, Wright N, Luzader DH, Kendall MM, Ruiz-Perez F, Nataro JP. A large family of anti-activators accompanying XylS/AraC family regulatory proteins. Mol Microbiol 2016; 101:314-32. [PMID: 27038276 PMCID: PMC4983702 DOI: 10.1111/mmi.13392] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2016] [Indexed: 11/29/2022]
Abstract
AraC Negative Regulators (ANR) suppress virulence genes by directly down‐regulating AraC/XylS members in Gram‐negative bacteria. In this study, we sought to investigate the distribution and molecular mechanisms of regulatory function for ANRs among different bacterial pathogens. We identified more than 200 ANRs distributed in diverse clinically important gram negative pathogens, including Vibrio spp., Salmonella spp., Shigella spp., Yersinia spp., Citrobacter spp., enterotoxigenic (ETEC) and enteroaggregative E. coli (EAEC), and members of the Pasteurellaceae. By employing a bacterial two hybrid system, pull down assays and surface plasmon resonance (SPR) analysis, we demonstrate that Aar (AggR‐activated regulator), a prototype member of the ANR family in EAEC, binds with high affinity to the central linker domain of AraC‐like member AggR. ANR‐AggR binding disrupted AggR dimerization and prevented AggR‐DNA binding. ANR homologs of Vibrio cholerae, Citrobacter rodentium, Salmonella enterica and ETEC were capable of complementing Aar activity by repressing aggR expression in EAEC strain 042. ANR homologs of ETEC and Vibrio cholerae bound to AggR as well as to other members of the AraC family, including Rns and ToxT. The predicted proteins of all ANR members exhibit three highly conserved predicted α‐helices. Site‐directed mutagenesis studies suggest that at least predicted α‐helices 2 and 3 are required for Aar activity. In sum, our data strongly suggest that members of the novel ANR family act by directly binding to their cognate AraC partners.
Collapse
Affiliation(s)
- Araceli E Santiago
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Michael B Yan
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Minh Tran
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nathan Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA, USA
| | - Deborah H Luzader
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Melissa M Kendall
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Fernando Ruiz-Perez
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James P Nataro
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
32
|
Wassenaar TM, Gunzer F. The prediction of virulence based on presence of virulence genes in E. coli may not always be accurate. Gut Pathog 2015; 7:15. [PMID: 26089987 PMCID: PMC4471914 DOI: 10.1186/s13099-015-0062-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/04/2015] [Indexed: 11/15/2022] Open
Abstract
Now that microbial whole genome sequencing is in reach of many researchers, it is common to infer virulent properties of a given bacterial isolate based on the presence of virulence genes. However, this may lead to inaccurate presumptions of virulence. Using the findings of a recent publication (Da Silva Santos et al. Gut Pathog 7:2, 2015) where virulence was inferred from a genome sequence and subsequently confirmed by in vitro analysis, we present an alternative view on the case described in that publication. Our alternative view point, which is further substantiated by whole genome sequencing of probiotic E. coli strains, may contribute to a more balanced vision on the interactions between pathogens and host.
Collapse
Affiliation(s)
- Trudy M Wassenaar
- Molecular Microbiology and Genomics Consultants, Tannenstrasse 7, 55576 Zotzenheim, Germany
| | - Florian Gunzer
- Institute of Medical Microbiology and Hygiene, TU Dresden, Dresden, Germany
| |
Collapse
|
33
|
Boisen N, Melton-Celsa AR, Scheutz F, O'Brien AD, Nataro JP. Shiga toxin 2a and Enteroaggregative Escherichia coli--a deadly combination. Gut Microbes 2015; 6:272-8. [PMID: 26039753 PMCID: PMC4615819 DOI: 10.1080/19490976.2015.1054591] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In 2011, a Shiga toxin (Stx) type 2a-producing enteroaggregative E. coli (EAEC) strain of serotype O104:H4 caused a large lethal outbreak in Northern Europe. Until recently, the pathogenic mechanisms explaining the high virulence of the strain have remained unclear. Our laboratories have shown that EAEC genes encoded on the pAA virulence plasmid, particularly the AggR-regulated AAF/I fimbriae, enhance inflammation and enable the outbreak strain to both adhere to epithelial cells and translocate Stx2a across the intestinal epithelium, possibly explaining the high incidence of the life threatening post-diarrheal sequelae of hemolytic uremic syndrome. Epidemiologic evidence supports a model of EAEC pathogenesis comprising the concerted action of multiple virulence factors along with induction of inflammation. Here, we suggest a model for the pathogenesis of the O104:H4 outbreak strain that includes contributions from EAEC alone, but incorporating additional injury induced by Stx2a.
Collapse
Affiliation(s)
- Nadia Boisen
- Statens Serum Institut; Department of Microbiology and Clinical Control; Copenhagen, Denmark,Correspondence to: Nadia Boisen;
| | - Angela R Melton-Celsa
- Statens Serum Institut; Department of Microbiology and Clinical Control; Copenhagen, Denmark
| | - Flemming Scheutz
- Statens Serum Institut; Department of Microbiology and Clinical Control; Copenhagen, Denmark
| | - Alison D O'Brien
- Department of Microbiology and Immunology; Uniformed Services University of the Health Sciences; Bethesda, MD USA
| | - James P Nataro
- Department of Pediatrics; University of Virginia School of Medicine; Charlottesville, VA USA
| |
Collapse
|