1
|
Zhao J, Yang W, Gao B, Wang H, Chen L, Shan C, Zhang B, Cha J, Shen J, Xiao J, Wang S, Liu G, Zhao R, Xin A, Xiao P, Gao H. Escherichia coli HPI-induced duodenitis through ubiquitin regulation of the TLR4/NF-κB pathway. BMC Vet Res 2025; 21:66. [PMID: 39953596 PMCID: PMC11829554 DOI: 10.1186/s12917-025-04515-3] [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: 07/31/2023] [Accepted: 01/23/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND The Highly Pathogenic Island (HPI) found in Yersinia pestis can be horizontally transferred to E. coli, enhancing its virulence and pathogenicity. Ubiquitin (Ub) acts as an activator of the NF-κB pathway and plays a critical role in the inflammatory response. However, the precise mechanism by which Ub and the regulated TLR4/NF-κB pathway contribute to HPI-induced intestinal inflammation in E. coli remains unclear. RESULTS In this study, we established Ub overexpression models of small intestinal epithelial cells (in vitro) and BALB/c mice (in vivo) and infected these models with HPI-rich E. coli. We investigated the role of the Ub-regulated TLR4/NF-κB pathway in E. coli HPI-induced intestinal inflammation through qPCR, ELISA, immunofluorescence, immunohistochemistry, and H&E staining. Our findings confirmed that E. coli HPI promoted the expression of Ub, TLR4, and NF-κB in IPEC-J2 cells and induced the translocation of NF-κB p65 protein to the nucleus. Further investigations revealed that Ub overexpression enhanced epithelial cell damage induced by E. coli HPI. This was accompanied by up-regulation of mRNA levels of TLR4, MyD88, NF-κB, IL-1β, and TNF-α, as well as increased release of the inflammatory factors IL-1β and TNF-α. In a mouse model with Ub overexpression infected with E. coli HPI, we observed that Ub overexpression promoted E. coli HPI-induced intestinal inflammation. Mechanistically, E. coli HPI induced intestinal epithelial cell damage by inducing Ub overexpression and modulating the TLR4/NF-κB pathway. CONCLUSIONS In conclusion, this study sheds light on the significant role of the Ub-regulated TLR4/NF-κB pathway in E. coli HPI-induced duodenitis, offering novel insights into the pathogenesis of E. coli infections.
Collapse
Affiliation(s)
- Jingang Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Wei Yang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Bin Gao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Hao Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Liping Chen
- College of Foreign Languages, Yunnan Agricultural University, Kunming, 650201, China
| | - Chunlan Shan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Bo Zhang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jinlong Cha
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Jue Shen
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Jinlong Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Shuai Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Gen Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Aiguo Xin
- Department of Poultry Husbandry and Disease Research, Yunnan Animal Science and Veterinary, Kunming, 650224, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China.
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
2
|
Puentes B, Souto A, Balado M, Rodríguez J, Osorio CR, Jiménez C, Lemos ML. A novel genomic island encodes vibrioferrin synthesis in the marine pathogen Photobacterium damselae subsp. damselae. Microb Pathog 2025; 199:107218. [PMID: 39662786 DOI: 10.1016/j.micpath.2024.107218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/03/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
In this study, we identified and analyzed a novel genomic island (GI), named pddGI-1, located on chromosome II of certain strains of the marine pathogen Photobacterium damselae subsp. damselae (Pdd). This GI shares structural similarities with other GIs found in Vibrio species, such as the Vibrio seventh pandemic island-II (VSP-II) of V. cholerae. The pddGI-1 island is a mosaic of gene blocks that encode functions related to ROS defense, anaerobic energy metabolism, and restriction-modification (RM) systems. Notably, pddGI-1 also includes a complete vibrioferrin siderophore system, enabling the bacteria to thrive in low-iron environments. Vibrioferrin was chemically identified from cell-free supernatants of Pdd RG91. Additionally, a pvsD mutant deficient in vibrioferrin biosynthesis was generated and analyzed. The results suggest that Pdd strains harbouring pddGI-1 gain a distinct growth advantage under iron-limited conditions. These findings, along with previous research, highlight the significant heterogeneity in iron assimilation systems among Pdd strains.
Collapse
Affiliation(s)
- Beatriz Puentes
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Alba Souto
- CICA-Centro Interdisciplinar de Química e Bioloxía, Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, Spain
| | - Miguel Balado
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Jaime Rodríguez
- CICA-Centro Interdisciplinar de Química e Bioloxía, Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, Spain.
| | - Carlos R Osorio
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain.
| | - Carlos Jiménez
- CICA-Centro Interdisciplinar de Química e Bioloxía, Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, 15071, Spain.
| | - Manuel L Lemos
- Department of Microbiology and Parasitology, Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain.
| |
Collapse
|
3
|
Gaborieau B, Delattre R, Adiba S, Clermont O, Denamur E, Ricard JD, Debarbieux L. Variable fitness effects of bacteriophage resistance mutations in Escherichia coli: implications for phage therapy. J Virol 2024; 98:e0111324. [PMID: 39213164 PMCID: PMC11495123 DOI: 10.1128/jvi.01113-24] [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: 06/25/2024] [Accepted: 07/10/2024] [Indexed: 09/04/2024] Open
Abstract
Bacteria exposed to bactericidal treatment, such as antibiotics or bacteriophages (phages), often develop resistance. While phage therapy is proposed as a solution to the antibiotic resistance crisis, the bacterial resistance emerging during phage therapy remains poorly characterized. In this study, we examined a large population of phage-resistant extra-intestinal pathogenic Escherichia coli 536 clones that emerged from both in vitro (non-limited liquid medium) and in vivo (murine pneumonia) conditions. Genome sequencing uncovered a convergent mutational pattern in phage resistance mechanisms under both conditions, particularly targeting two cell-wall components, the K15 capsule and the lipopolysaccharide (LPS). This suggests that their identification in vivo could be predicted from in vitro assays. Phage-resistant clones exhibited a wide range of fitness according to in vitro tests, growth rate, and resistance to amoeba grazing, which could not distinguish between the K15 capsule and LPS mutants. In contrast, K15 capsule mutants retained virulence comparable to the wild-type strain, whereas LPS mutants showed significant attenuation in the murine pneumonia model. Additionally, we observed that resistance to the therapeutic phage through a nonspecific mechanism, such as capsule overproduction, did not systematically lead to co-resistance to other phages that were initially capable or incapable of infecting the wild-type strain. Our findings highlight the importance of incorporating a diverse range of phages in the design of therapeutic cocktails to target potential future phage-resistant clones effectively. IMPORTANCE This study isolated more than 50 phage-resistant mutants from both in vitro and in vivo conditions, exposing an extra-intestinal pathogenic Escherichia coli strain to a single virulent phage. The characterization of these clones revealed several key findings: (1) mutations occurring during phage treatment affect the same pathways as those identified in vitro; (2) the resistance mechanisms are associated with the modification of two cell-wall components, with one involving receptor deletion (phage-specific mechanism) and the other, less frequent, involving receptor masking (phage-nonspecific mechanism); (3) an in vivo virulence assay demonstrated that the absence of the receptor abolishes virulence while masking the receptor preserves it; and (4) clones with a resistance mechanism nonspecific to a particular phage can remain susceptible to other phages. This supports the idea of incorporating diverse phages into therapeutic cocktails designed to collectively target both wild-type and phage-resistant strains, including those with resistance mechanisms nonspecific to a phage.
Collapse
Affiliation(s)
- Baptiste Gaborieau
- Université Paris Cité, INSERM UMR1137, IAME, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
- APHP, Hôpital Louis Mourier, DMU ESPRIT, Service de Médecine Intensive Réanimation, Colombes, France
| | - Raphaëlle Delattre
- Université Paris Cité, INSERM UMR1137, IAME, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Sandrine Adiba
- Institut de Biologie de l'ENS (IBENS), École Normale Supérieure CNRS UMR8197, Paris, France
| | | | - Erick Denamur
- Université Paris Cité, INSERM UMR1137, IAME, Paris, France
- APHP, Hôpital Bichat, Service de Génétique Moléculaire, Paris, France
| | - Jean-Damien Ricard
- Université Paris Cité, INSERM UMR1137, IAME, Paris, France
- APHP, Hôpital Louis Mourier, DMU ESPRIT, Service de Médecine Intensive Réanimation, Colombes, France
| | - Laurent Debarbieux
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| |
Collapse
|
4
|
Bonardi S, Conter M, Andriani L, Bacci C, Magagna G, Rega M, Lamperti L, Loiudice C, Pierantoni M, Filipello V. Emerging of Shiga toxin-producing Escherichia coli O177:H11 and O177:H25 from cattle at slaughter in Italy. Int J Food Microbiol 2024; 423:110846. [PMID: 39079448 DOI: 10.1016/j.ijfoodmicro.2024.110846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/18/2024]
Abstract
Shiga toxin-producing Escherichia coli (STEC) are zoonotic pathogens frequently carried by cattle, responsible in humans of mild to bloody diarrhoea, haemolytic uraemic syndrome (HUS) and even death. In 2023-2024, a study on STEC contamination of hide and carcasses of dairy cattle at slaughter was planned in Emilia-Romagna region (northern Italy). When the study was still in progress and 60 animals were sampled, the detection of STEC O177 isolates reached high rates and gained our attention. A total of five O177 STEC strains were detected, namely four from three carcasses (5.0 %) and one from a hide sample (1.7 %). The isolates were typed by WGS as following: 1) STEC O177:H11 sequence type (ST) 765 (stx2a+, eae+), detected from one carcass; 2) STEC O177:H25 ST659 (stx2c+, eae+) detected from three carcasses and one hide sample. One carcass was contaminated by both STEC serotypes. The isolates carried other virulence determinants often found in STEC strains associated with HUS, namely the exha, astA and espP genes, together with genes for adhesion to the epithelial cells of the gut (lpfA, fdeC, fimH) and non-Locus for Enterocyte Effacement (LEE) effector protein genes (nleA, nleB). The STEC O177:H11 isolate harboured antimicrobial resistance (AMR) genes to β-lactams (blaTEM-1A), aminoglycosides (aadA1, aph(3″)-Ib, aph(6)-Id), trimethoprim (dfrA1), sulphonamides (sul1, sul2), tetracyclines (tetA), (tetB), streptothricin (sat2), and quaternary ammonium compounds (qacEdelta1). On the contrary, the STEC O177:H25 isolates carried no AMR genes. Persistent carriage of STEC O177:H25 ST659 (stx2c+, eae+) at farm level was assessed by testing animals of the same herd sent to slaughter. Interestingly, the colonies of STEC O177:H11 and STEC O177:H25 had different morphology on CHROMagar™ STEC plates, being mauve and colourless, respectively. Since mauve is the colour STEC colonies commonly have on the CHROMagar™ STEC medium, our findings can help microbiologists in the selection of uncommon serotypes. To the best of our knowledge, this is the first detection of STEC O177 from carcasses and hides of dairy cattle at slaughter. Noteworthy, the STEC-positive hide was classified as "very dirty" thus stressing the need of clean animals entering the slaughter chain, as required by Regulation (EC) No 853/2004. Since STEC O177 has been responsible of HUS in Europe, our data could add information on the source of uncommon serogroups in human infections.
Collapse
Affiliation(s)
- Silvia Bonardi
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - Mauro Conter
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - Laura Andriani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - Cristina Bacci
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - Giulia Magagna
- Department of Food Safety, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Via Bianchi 9, 25124 Brescia, Italy
| | - Martina Rega
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - Luca Lamperti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - Carlo Loiudice
- National Veterinary Service, Via Vasari 13/A, 43126 Parma, Italy
| | - Marco Pierantoni
- National Veterinary Service, Via Vasari 13/A, 43126 Parma, Italy
| | - Virginia Filipello
- Department of Food Safety, Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia-Romagna, Via Bianchi 9, 25124 Brescia, Italy
| |
Collapse
|
5
|
Royer G, Clermont O, Marin J, Condamine B, Dion S, Blanquart F, Galardini M, Denamur E. Epistatic interactions between the high pathogenicity island and other iron uptake systems shape Escherichia coli extra-intestinal virulence. Nat Commun 2023; 14:3667. [PMID: 37339949 DOI: 10.1038/s41467-023-39428-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
The intrinsic virulence of extra-intestinal pathogenic Escherichia coli is associated with numerous chromosomal and/or plasmid-borne genes, encoding diverse functions such as adhesins, toxins, and iron capture systems. However, the respective contribution to virulence of those genes seems to depend on the genetic background and is poorly understood. Here, we analyze genomes of 232 strains of sequence type complex STc58 and show that virulence (quantified in a mouse model of sepsis) emerged in a sub-group of STc58 due to the presence of the siderophore-encoding high-pathogenicity island (HPI). When extending our genome-wide association study to 370 Escherichia strains, we show that full virulence is associated with the presence of the aer or sit operons, in addition to the HPI. The prevalence of these operons, their co-occurrence and their genomic location depend on strain phylogeny. Thus, selection of lineage-dependent specific associations of virulence-associated genes argues for strong epistatic interactions shaping the emergence of virulence in E. coli.
Collapse
Affiliation(s)
- Guilhem Royer
- Université Paris Cité, IAME, INSERM, Paris, France
- Département de Prévention, Diagnostic et Traitement des Infections, Hôpital Henri Mondor, Créteil, France
- LABGeM, Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Université Paris-Saclay, Evry, France
- EERA Unit "Ecology and Evolution of Antibiotics Resistance," Institut Pasteur-Assistance Publique/Hôpitaux de Paris-Université Paris-Saclay, Paris, France
- UMR CNRS, 3525, Paris, France
| | | | - Julie Marin
- Université Paris Cité, IAME, INSERM, Paris, France
- Université Sorbonne Paris Nord, IAME, INSERM, Bobigny, France
| | | | - Sara Dion
- Université Paris Cité, IAME, INSERM, Paris, France
| | - François Blanquart
- Center for Interdisciplinary Research in Biology, CNRS, Collège de France, PSL Research University, Paris, France
| | - Marco Galardini
- Institute for Molecular Bacteriology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Hannover, Germany
| | - Erick Denamur
- Université Paris Cité, IAME, INSERM, Paris, France.
- AP-HP, Hôpital Bichat, Laboratoire de Génétique Moléculaire, Paris, France.
| |
Collapse
|
6
|
Li G, Li M, Yu D, Sun W. Effect of high-pathogenicity island (HPI) on TGF-β1/Smad3 pathway in mouse model of E. coli strains causing diarrhea in calf. Res Vet Sci 2023; 156:1-6. [PMID: 36706696 DOI: 10.1016/j.rvsc.2022.09.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/11/2022] [Accepted: 09/18/2022] [Indexed: 01/29/2023]
Abstract
PURPOSE This study evaluated pathogenic effect of TGF-β1/Smad3 pathway in mouse model after infecting them with HPI+ and HPI- strains of Escherichia coli (E. coli) which were isolated from diarrhea in calves. METHODS Kunming mice were randomly divided into 3 groups: a control group, HPI+-infection group and HPI--infection group. After intraperitoneal injection of HPI strains of E. coli (concentration: 3 × 108 cfu/mL) in mice, alanine aminotransferase (ALT) and aspartate aminotransferase (AST), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) contents were detected at 12 h post infection. The sections of liver and kidney were obtained for histopathological observations. Propidium iodide and 4',6-diamidino-2-phenylindole (DAPI) staining was used to analyze the cell apoptosis. The immunohistochemistry staining and quantitative real time PCR (q-PCR) were performed for evaluating the protein and mRNA expression of TGF-β1, Collagen I and Smad3. The histological change and PI staining of liver and kidney showed significant injuries. Compared with the control group, the serum ALT and AST activities and TNF-α and IL-6 contents of mice in the HPI+ and HPI- groups were increased, number of apoptotic cells and expression of TGF-β1, Collagen Iand Smad3 were up-regulated after E. coli infection in liver and kidney, which was significantly increased in HPI+-infected compared to HPI-. CONCLUSION The study concludes that E. coli HPI induced and enhanced the over expression of TGF-β1/Smad3 pathway and ultimately caused pathological anomalies.
Collapse
Affiliation(s)
- Gongmei Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Maohui Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Dan Yu
- Changchun Animal Disease Prevention and Control Center, Changchun, Jilin 130118,China
| | - Wuwen Sun
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin 130118, China.
| |
Collapse
|
7
|
Lages MA, Lemos ML, Balado M. The Temperature-Dependent Expression of the High-Pathogenicity Island Encoding Piscibactin in Vibrionaceae Results From the Combined Effect of the AraC-Like Transcriptional Activator PbtA and Regulatory Factors From the Recipient Genome. Front Microbiol 2021; 12:748147. [PMID: 34867865 PMCID: PMC8639528 DOI: 10.3389/fmicb.2021.748147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
The high-pathogenicity island irp-HPI is widespread among Vibrionaceae encoding the piscibactin siderophore system. The expression of piscibactin genes in the fish pathogen Vibrio anguillarum is favored by low temperatures. However, information about the regulatory mechanism behind irp-HPI gene expression is scarce. In this work, in-frame deletion mutants of V. anguillarum defective in the putative regulators AraC1 and AraC2, encoded by irp-HPI, and in the global regulators H-NS and ToxRS, were constructed and their effect on irp-HPI gene expression was analyzed at 15 and 25°C. The results proved that only AraC1 (renamed as PbtA) is required for the expression of piscibactin biosynthesis and transport genes. PbtA inactivation led to an inability to grow under iron restriction, a loss of the outer membrane piscibactin transporter FrpA, and a significant decrease in virulence for fish. Inactivation of the global repressor H-NS, which is involved in silencing of horizontally acquired genes, also resulted in a lower transcriptional activity of the frpA promoter. Deletion of toxR-S, however, did not have a relevant effect on the expression of the irp-HPI genes. Therefore, while irp-HPI would not be part of the ToxR regulon, H-NS must exert an indirect effect on piscibactin gene expression. Thus, the temperature-dependent expression of the piscibactin-encoding pathogenicity island described in V. anguillarum is the result of the combined effect of the AraC-like transcriptional activator PbtA, harbored in the island, and other not yet defined regulator(s) encoded by the genome. Furthermore, different expression patterns were detected within different irp-HPI evolutionary lineages, which supports a long-term evolution of the irp-HPI genomic island within Vibrionaceae. The mechanism that modulates piscibactin gene expression could also be involved in global regulation of virulence factors in response to temperature changes.
Collapse
Affiliation(s)
- Marta A Lages
- Department of Microbiology and Parasitology, Institute of Aquaculture, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel L Lemos
- Department of Microbiology and Parasitology, Institute of Aquaculture, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Balado
- Department of Microbiology and Parasitology, Institute of Aquaculture, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
8
|
Zhang B, Wang H, Zhao W, Shan C, Liu C, Gao L, Zhao R, Ao P, Xiao P, Lv L, Gao H. New insights into the construction of wild-type Saba pig-derived Escherichia coli irp2 gene deletion strains. 3 Biotech 2021; 11:408. [PMID: 34466347 PMCID: PMC8363713 DOI: 10.1007/s13205-021-02951-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/27/2021] [Indexed: 01/24/2023] Open
Abstract
To construct wild-type E. coli irp2 gene deletion strains, CRISPR/Cas9 gene editing technology was used, and the difficulty and key points of gene editing of wild-type strains were analyzed. Based on the resistance of the CRISPR/Cas9 system expression vector, 4 strains of 41 E. coli strains isolated from Saba pigs were selected as the target strains for the deletion of the irp2 gene, which were sensitive to both ampicillin and kanamycin. Then, CRISPR/Cas9 technology was combined with homologous recombination technology to construct recombinant vectors containing Cas9, sgRNA and donor sequences to knock out the irp2 gene. Finally, the absence of the irp2 gene in E. coli was further verified by iron uptake assays, iron carrier production assays and growth curve measurements. The results showed that three of the selected strains showed single base mutations and deletions (Δirp2-1, Δirp2-2 and Δirp2-3). The deletion of the irp2 gene reduced the ability of E. coli to take up iron ions and produce iron carriers, but not affect the growth characteristics of E. coli. It is shown that the CRISPR/Cas9 knock-out system constructed in this study can successfully knock out the irp2 gene of the wild-type E. coli. Our results providing new insights into genome editing in wild-type strains, which enable further functional studies of the irp2 gene in wild-type E. coli.
Collapse
Affiliation(s)
- Bo Zhang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Hongdan Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Weiwei Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Chunlan Shan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Chaoying Liu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Libo Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Pingxing Ao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| | - Longbao Lv
- Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223 Yunnan China
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201 Yunnan China
| |
Collapse
|
9
|
Hwang SB, Chelliah R, Kang JE, Rubab M, Banan-MwineDaliri E, Elahi F, Oh DH. Role of Recent Therapeutic Applications and the Infection Strategies of Shiga Toxin-Producing Escherichia coli. Front Cell Infect Microbiol 2021; 11:614963. [PMID: 34268129 PMCID: PMC8276698 DOI: 10.3389/fcimb.2021.614963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) is a global foodborne bacterial pathogen that is often accountable for colon disorder or distress. STEC commonly induces severe diarrhea in hosts but can cause critical illnesses due to the Shiga toxin virulence factors. To date, there have been a significant number of STEC serotypes have been evolved. STECs vary from nausea and hemorrhoid (HC) to possible lethal hemolytic-based uremic syndrome (HUS), thrombotic thrombocytopenic purpura (TTP). Inflammation-based STEC is usually a foodborne illness with Shiga toxins (Stx 1 and 2) thought to be pathogenesis. The STEC's pathogenicity depends significantly on developing one or more Shiga toxins, which can constrain host cell protein synthesis leading to cytotoxicity. In managing STEC infections, antimicrobial agents are generally avoided, as bacterial damage and discharge of accumulated toxins are thought the body. It has also been documented that certain antibiotics improve toxin production and the development of these species. Many different groups have attempted various therapies, including toxin-focused antibodies, toxin-based polymers, synbiotic agents, and secondary metabolites remedies. Besides, in recent years, antibiotics' efficacy in treating STEC infections has been reassessed with some encouraging methods. Nevertheless, the primary role of synbiotic effectiveness (probiotic and prebiotic) against pathogenic STEC and other enteropathogens is less recognized. Additional studies are required to understand the mechanisms of action of probiotic bacteria and yeast against STEC infection. Because of the consensus contraindication of antimicrobials for these bacterial pathogens, the examination was focused on alternative remedy strategies for STEC infections. The rise of novel STEC serotypes and approaches employed in its treatment are highlighted.
Collapse
Affiliation(s)
- Su-bin Hwang
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Ji Eun Kang
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Momna Rubab
- School of Food and Agricultural Sciences, University of Management and Technology, Lahore, Pakistan
| | - Eric Banan-MwineDaliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Fazle Elahi
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|
10
|
Two Polyketides Intertwined in Complex Regulation: Posttranscriptional CsrA-Mediated Control of Colibactin and Yersiniabactin Synthesis in Escherichia coli. mBio 2021; 13:e0381421. [PMID: 35100864 PMCID: PMC8805033 DOI: 10.1128/mbio.03814-21] [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] [Indexed: 01/23/2023] Open
Abstract
Bacteria have to process several levels of gene regulation and coordination of interconnected regulatory networks to ensure the most adequate cellular response to specific growth conditions. Especially, expression of complex and costly fitness and pathogenicity-associated traits is coordinated and tightly regulated at multiple levels. We studied the interconnected regulation of the expression of the colibactin and yersiniabactin polyketide biosynthesis machineries, which are encoded by two pathogenicity islands found in many phylogroup B2 Escherichia coli isolates. Comparative phenotypic and genotypic analyses identified the BarA-UvrY two-component system as an important regulatory element involved in colibactin and yersiniabactin expression. The carbon storage regulator (Csr) system controls the expression of a wide range of central metabolic and virulence-associated traits. The availability of CsrA, the key translational regulator of the Csr system, depends on BarA-UvrY activity. We employed reporter gene fusions to demonstrate UvrY- and CsrA-dependent expression of the colibactin and yersiniabactin determinants and confirmed a direct interaction of CsrA with the 5' untranslated leader transcripts of representative genes of the colibactin and yersiniabactin operons by RNA electrophoretic mobility shift assays. This posttranscriptional regulation adds an additional level of complexity to control mechanisms of polyketide expression, which is also orchestrated at the level of ferric uptake regulator (Fur)-dependent regulation of transcription and phosphopantetheinyl transferase-dependent activation of polyketide biosynthesis. Our results emphasize the interconnection of iron- and primary metabolism-responsive regulation of colibactin and yersiniabactin expression by the fine-tuned action of different regulatory mechanisms in response to variable environmental signals as a prerequisite for bacterial adaptability, fitness, and pathogenicity in different habitats. IMPORTANCE Secondary metabolite expression is a widespread strategy among bacteria to improve their fitness in habitats where they constantly compete for resources with other bacteria. The production of secondary metabolites is associated with a metabolic and energetic burden. Colibactin and yersiniabactin are two polyketides, which are expressed in concert and promote the virulence of different enterobacterial pathogens. To maximize fitness, they should be expressed only in microenvironments in which they are required. Accordingly, precise regulation of colibactin and yersiniabactin expression is crucial. We show that the expression of these two polyketides is also interconnected via primary metabolism-responsive regulation at the posttranscriptional level by the CsrA RNA-binding protein. Our findings may help to optimize (over-)expression and further functional characterization of the polyketide colibactin. Additionally, this new aspect of concerted colibactin and yersiniabactin expression extends our knowledge of conditions that favor the expression of these virulence- and fitness-associated factors in different Enterobacterales members.
Collapse
|
11
|
Zhao W, Gao B, Liu C, Zhang B, Shan C, Deng J, Wan Q, Wang X, Zhao R, Gao L, Ao P, Xiao P, Gao H. High pathogenicity island is associated with enhanced autophagy in pathogenic Escherichia coli HPI - infected macrophages. Res Vet Sci 2021; 135:113-120. [PMID: 33465603 DOI: 10.1016/j.rvsc.2021.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 12/31/2020] [Accepted: 01/03/2021] [Indexed: 10/22/2022]
Abstract
High pathogenicity island (HPI), which is widely distributed in Escherichia coli (E. coli), can enhance the pathogenicity of E. coli. Thus the HPI positive E. coli could pose a threat to human and animal health. It remains to be elucidated how HPI affects the virulence of pathogenic E. coli. Autophagy is an important mechanism to maintain cellular homeostasis and an innate immunity responses of organisms against pathogens. The interaction between pathogenic E. coli possessing HPI (E. coli HPI) and host autophagy system has not been reported. In this study, it was demonstrated that pathogenic E. coli induced autophagy in 3D4/21 macrophages and HPI was associated with enhanced autophagy through transmission electron microscopy, immunofluorescence and real-time PCR. The PI3K/Akt/mTOR pathway is an important negative regulatory pathway for autophagy. Through detecting the expression of key genes of PI3K/Akt/mTOR pathway, it was speculated that HPI enhanced the inhibition of the signaling pathway stimulated by pathogenic E. coli. Furthermore, HPI inhibited the secretion of IFN-γ, while the presence of HPI did not significantly affect the secretion of IL-1β. This work is the first attempt to explore the interplay between HPI carried by pathogenic E. coli and host cell autophagy. The findings might enable better understanding of the contribution of HPI to pathogenicity.
Collapse
Affiliation(s)
- Weiwei Zhao
- College of food science and technology, Yunnan Agricultural University, Kunming 650201, China
| | - Bin Gao
- College of food science and technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chang Liu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Bo Zhang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunlan Shan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Jing Deng
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Quan Wan
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Xi Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Ru Zhao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Libo Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Pingxing Ao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China.
| | - Hong Gao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
12
|
Success of Escherichia coli O25b:H4 Sequence Type 131 Clade C Associated with a Decrease in Virulence. Infect Immun 2020; 88:IAI.00576-20. [PMID: 32989036 PMCID: PMC7671891 DOI: 10.1128/iai.00576-20] [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] [Received: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 01/12/2023] Open
Abstract
Escherichia coli O25b:H4 sequence type 131 (ST131), which is resistant to fluoroquinolones and which is a producer of CTX-M-15, is globally one of the major extraintestinal pathogenic E. coli (ExPEC) lineages. Phylogenetic analyses showed that multidrug-resistant ST131 strains belong to clade C, which recently emerged from clade B by stepwise evolution. It has been hypothesized that features other than multidrug resistance could contribute to this dissemination since other major global ExPEC lineages (ST73 and ST95) are mostly antibiotic susceptible. Escherichia coli O25b:H4 sequence type 131 (ST131), which is resistant to fluoroquinolones and which is a producer of CTX-M-15, is globally one of the major extraintestinal pathogenic E. coli (ExPEC) lineages. Phylogenetic analyses showed that multidrug-resistant ST131 strains belong to clade C, which recently emerged from clade B by stepwise evolution. It has been hypothesized that features other than multidrug resistance could contribute to this dissemination since other major global ExPEC lineages (ST73 and ST95) are mostly antibiotic susceptible. To test this hypothesis, we compared early biofilm production, presence of ExPEC virulence factors (VFs), and in vivo virulence in a mouse sepsis model in 19 and 20 epidemiologically relevant strains of clades B and C, respectively. Clade B strains were significantly earlier biofilm producers (P < 0.001), carriers of more VFs (P = 4e−07), and faster killers of mice (P = 2e−10) than clade C strains. Gene inactivation experiments showed that the H30-fimB and ibeART genes were associated with in vivo virulence. Competition assays in sepsis, gut colonization, and urinary tract infection models between the most anciently diverged strain (B1 subclade), one C1 subclade strain, and a B4 subclade recombining strain harboring some clade C-specific genetic events showed that the B1 strain always outcompeted the C1 strain, whereas the B4 strain outcompeted the C1 strain, depending on the mouse niches. All these findings strongly suggest that clade C evolution includes a progressive loss of virulence involving multiple genes, possibly enhancing overall strain fitness by avoiding severe infections, even if it comes at the cost of a lower colonization ability.
Collapse
|
13
|
Galardini M, Clermont O, Baron A, Busby B, Dion S, Schubert S, Beltrao P, Denamur E. Major role of iron uptake systems in the intrinsic extra-intestinal virulence of the genus Escherichia revealed by a genome-wide association study. PLoS Genet 2020; 16:e1009065. [PMID: 33112851 PMCID: PMC7592755 DOI: 10.1371/journal.pgen.1009065] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/20/2020] [Indexed: 11/18/2022] Open
Abstract
The genus Escherichia is composed of several species and cryptic clades, including E. coli, which behaves as a vertebrate gut commensal, but also as an opportunistic pathogen involved in both diarrheic and extra-intestinal diseases. To characterize the genetic determinants of extra-intestinal virulence within the genus, we carried out an unbiased genome-wide association study (GWAS) on 370 commensal, pathogenic and environmental strains representative of the Escherichia genus phylogenetic diversity and including E. albertii (n = 7), E. fergusonii (n = 5), Escherichia clades (n = 32) and E. coli (n = 326), tested in a mouse model of sepsis. We found that the presence of the high-pathogenicity island (HPI), a ~35 kbp gene island encoding the yersiniabactin siderophore, is highly associated with death in mice, surpassing other associated genetic factors also related to iron uptake, such as the aerobactin and the sitABCD operons. We confirmed the association in vivo by deleting key genes of the HPI in E. coli strains in two phylogenetic backgrounds. We then searched for correlations between virulence, iron capture systems and in vitro growth in a subset of E. coli strains (N = 186) previously phenotyped across growth conditions, including antibiotics and other chemical and physical stressors. We found that virulence and iron capture systems are positively correlated with growth in the presence of numerous antibiotics, probably due to co-selection of virulence and resistance. We also found negative correlations between virulence, iron uptake systems and growth in the presence of specific antibiotics (i.e. cefsulodin and tobramycin), which hints at potential “collateral sensitivities” associated with intrinsic virulence. This study points to the major role of iron capture systems in the extra-intestinal virulence of the genus Escherichia. Bacterial isolates belonging to the genus Escherichia can be human commensals but also opportunistic pathogens, with the ability to cause extra-intestinal infection. There is therefore the need to identify the genetic elements that favour extra-intestinal virulence, so that virulent bacterial isolates can be identified through genome analysis and potential treatment strategies be developed. To reduce the influence of host variability on virulence, we have used a mouse model of sepsis to characterize the virulence of 370 strains belonging to the genus Escherichia, for which whole genome sequences were also available. We have used a statistical approach called Genome-Wide Association Study (GWAS) to show how the presence of genes that encode for iron scavenging are significantly associated with the propensity of a bacterial isolate to cause extra-intestinal infections. Taking advantage of previously generated growth data on a subset of the strains and its correlation to virulence we generated hypothesis on the relationship between iron scavenging and growth in the presence of various antimicrobials, which could have implications for developing new treatment strategies.
Collapse
Affiliation(s)
- Marco Galardini
- EMBL-EBI, Wellcome Genome Campus, Cambridge, United Kingdom
- * E-mail: (MG); (ED)
| | | | | | - Bede Busby
- Genome Biology Unit, EMBL, Heidelberg, Germany
| | - Sara Dion
- Université de Paris, IAME, UMR1137, INSERM, Paris, France
| | - Sören Schubert
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU Munich, Germany
| | - Pedro Beltrao
- EMBL-EBI, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Erick Denamur
- Université de Paris, IAME, UMR1137, INSERM, Paris, France
- AP-HP, Laboratoire de Génétique Moléculaire, Hôpital Bichat, Paris, France
- * E-mail: (MG); (ED)
| |
Collapse
|
14
|
IurV, Encoded by ORF VCA0231, Is Involved in the Regulation of Iron Uptake Genes in Vibrio cholerae. Genes (Basel) 2020; 11:genes11101184. [PMID: 33053678 PMCID: PMC7600106 DOI: 10.3390/genes11101184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/16/2022] Open
Abstract
The pathogen Vibrio cholerae has multiple iron acquisition systems which allow bacteria to exploit a variety of iron sources across the different environments on which it thrives. The expression of such iron uptake systems is highly regulated, mainly by the master iron homeostasis regulator Fur but also by other mechanisms. Recently, we documented that the expression of many of the iron-responsive genes is also modulated by riboflavin. Among them, the open reading frame VCA0231, repressed both by riboflavin and iron, encodes a putative transcriptional regulator of the AraC/XylS family. Nonetheless, the genes or functions affected by this factor are unknown. In the present study, a series of in silico analyses was performed in order to identify the putative functions associated with the product of VCA0231. The STRING database predicted many iron uptake genes as functional partners for the product of VCA0231. In addition, a genomic neighborhood analysis with the Enzyme Function Initiative tools detected many Pfam families involved in iron homeostasis genetically associated with VCA0231. Moreover, a phylogenetic tree showed that other AraC/XylS members known to regulate siderophore utilization in bacteria clustered together and the product of VCA0231 localized in this cluster. This suggested that the product of VCA0231, here named IurV, is involved in the regulation of iron uptake processes. RNAseq was performed to determine the transcriptional effects of a deletion in VCA0231. A total of 52 genes were overexpressed and 21 genes were downregulated in response to the iurV deletion. Among these, several iron uptake genes and other iron homeostasis-related genes were found. Six gene ontology (GO) functional terms were enriched in the upregulated genes, of which five were related to iron metabolism. The regulatory pattern observed in the transcriptomics of a subset of genes was independently confirmed by quantitative real time PCR analysis. The results indicate that IurV is a novel regulator of the AraC/XylS family involved in the repression of iron uptake genes. Whether this effect is direct or indirect remains to be determined.
Collapse
|
15
|
Abstract
Escherichia coli is a commensal of the vertebrate gut that is increasingly involved in various intestinal and extra-intestinal infections as an opportunistic pathogen. Numerous pathotypes that represent groups of strains with specific pathogenic characteristics have been described based on heterogeneous and complex criteria. The democratization of whole-genome sequencing has led to an accumulation of genomic data that render possible a population phylogenomic approach to the emergence of virulence. Few lineages are responsible for the pathologies compared with the diversity of commensal strains. These lineages emerged multiple times during E. coli evolution, mainly by acquiring virulence genes located on mobile elements, but in a specific chromosomal phylogenetic background. This repeated emergence of stable and cosmopolitan lineages argues for an optimization of strain fitness through epistatic interactions between the virulence determinants and the remaining genome.
Collapse
|
16
|
Metabolomics Study on Pathogenic and Non-pathogenic E. coli with Closely Related Genomes with a Focus on Yersiniabactin and Its Known and Novel Derivatives. Metabolites 2020; 10:metabo10060221. [PMID: 32481767 PMCID: PMC7344775 DOI: 10.3390/metabo10060221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 11/18/2022] Open
Abstract
The Escherichia coli (E. coli) strains Nissle 1917 (EcN), 83972 and CFT073 are closely related but differ in their phenotypes and pathogenicity. The aim of this study was to compare the metabolome of these strains based on metabolomic data analysis of bacterial samples using liquid chromatography-high resolution mass spectrometry (LC-HRMS). The strains were cultivated in minimum essential medium at 37 °C for 6 h. The sterilized culture supernatant was analyzed, followed by data processing to create feature lists, and statistical analysis to identify discriminating features in the metabolomes of the three strains. Metabolites were identified using the exact masses, isotope patterns, and fragmentation spectra. The results showed that the metabolome of EcN differs significantly from the metabolomes of E. coli 83972 and CFT073. Based on the analysis, yersiniabactin (Ybt), its metal complexes, and its known structural derivatives escherichelin and ulbactin B were identified as discriminating features; the latter has not been described for E. coli before. Additionally, novel Ytb derivatives were found and tentatively identified by LC-MS/HRMS. All these metabolites were determined in significantly higher levels in the metabolome of EcN compared to E. coli 83972, which may explain a large part of the observed differences of the metabolomes.
Collapse
|
17
|
Abstract
Host iron restriction is a common mechanism for limiting the growth of pathogens. We compared the regulatory network controlled by Fur in uropathogenic E. coli (UPEC) to that of nonpathogenic E. coli K-12 to uncover strategies that pathogenic bacteria use to overcome iron limitation. Although iron homeostasis functions were regulated by Fur in the uropathogen as expected, a surprising finding was the activation of the stringent and general stress responses in the uropathogen fur mutant, which was rescued by amino acid addition. This coordinated global response could be important in controlling growth and survival under nutrient-limiting conditions and during transitions from the nutrient-rich environment of the lower gastrointestinal (GI) tract to the more restrictive environment of the urinary tract. The coupling of the response of iron limitation to increased demand for amino acids could be a critical attribute that sets UPEC apart from other E. coli pathotypes. Pathogenicity islands and plasmids bear genes for pathogenesis of various Escherichia coli pathotypes. Although there is a basic understanding of the contribution of these virulence factors to disease, less is known about variation in regulatory networks in determining disease phenotypes. Here, we dissected a regulatory network directed by the conserved iron homeostasis regulator, ferric uptake regulator (Fur), in uropathogenic E. coli (UPEC) strain CFT073. Comparing anaerobic genome-scale Fur DNA binding with Fur-dependent transcript expression and protein levels of the uropathogen to that of commensal E. coli K-12 strain MG1655 showed that the Fur regulon of the core genome is conserved but also includes genes within the pathogenicity/genetic islands. Unexpectedly, regulons indicative of amino acid limitation and the general stress response were also indirectly activated in the uropathogen fur mutant, suggesting that induction of the Fur regulon increases amino acid demand. Using RpoS levels as a proxy, addition of amino acids mitigated the stress. In addition, iron chelation increased RpoS to the same levels as in the fur mutant. The increased amino acid demand of the fur mutant or iron chelated cells was exacerbated by aerobic conditions, which could be partly explained by the O2-dependent synthesis of the siderophore aerobactin, encoded by an operon within a pathogenicity island. Taken together, these data suggest that in the iron-poor environment of the urinary tract, amino acid availability could play a role in the proliferation of this uropathogen, particularly if there is sufficient O2 to produce aerobactin.
Collapse
|
18
|
Johnson JR, Magistro G, Clabots C, Porter S, Manges A, Thuras P, Schubert S. Contribution of yersiniabactin to the virulence of an Escherichia coli sequence type 69 ("clonal group A") cystitis isolate in murine models of urinary tract infection and sepsis. Microb Pathog 2018; 120:128-131. [PMID: 29702209 DOI: 10.1016/j.micpath.2018.04.048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/30/2018] [Accepted: 04/23/2018] [Indexed: 11/15/2022]
Abstract
Escherichia coli sequence type 69 (ST69; "clonal group A") is an important extraintestinal pathogen. To clarify the yersiniabactin siderophore system's role in ST69's extraintestinal virulence we compared a wild-type ST69 cystitis isolate, isogenic irp2 (yersiniabactin) mutants, and irp2-complemented mutants in murine models of sepsis and urinary tract infection (UTI). irp2 mutants were attenuated mildly in the UTI model and profoundly in the sepsis model. In both models, complementation with a functional copy of irp2 restored full parental virulence. These findings suggest that in ST69 the yersiniabactin system has a minor role in urovirulence and a major role in sepsis causation.
Collapse
Affiliation(s)
- James R Johnson
- Veterans Affairs Medical Center, 1 Veterans Drive, Minneapolis, MN, USA; University of Minnesota, 420 Delaware St. SE, MMC 250, Minneapolis, MN 55455, USA.
| | - Giuseppe Magistro
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377 Munich, Germany
| | - Connie Clabots
- Veterans Affairs Medical Center, 1 Veterans Drive, Minneapolis, MN, USA
| | - Stephen Porter
- Veterans Affairs Medical Center, 1 Veterans Drive, Minneapolis, MN, USA; University of Minnesota, 420 Delaware St. SE, MMC 250, Minneapolis, MN 55455, USA
| | - Amee Manges
- University of British Columbia, School of Population and Public Health, 137-2206 East Mall, Vancouver, BC V6T 1Z3, Canada
| | - Paul Thuras
- Veterans Affairs Medical Center, 1 Veterans Drive, Minneapolis, MN, USA; University of Minnesota, 420 Delaware St. SE, MMC 250, Minneapolis, MN 55455, USA
| | - Sören Schubert
- Max von Pettenkofer-Institut, Ludwig-Maximilians-University of Munich, Marchioninistrasse 17, 81377 Munich, Germany
| |
Collapse
|
19
|
In vivo screening platform for shiga toxin-producing Escherichia coli (STEC) using Caenorhabditis elegans as a model. PLoS One 2018; 13:e0193277. [PMID: 29489863 PMCID: PMC5831388 DOI: 10.1371/journal.pone.0193277] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 02/07/2018] [Indexed: 01/02/2023] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) strains are the main cause of bacillary dysentery, although STEC strains generally induce milder disease symptoms compared to Shigella species. This study aimed to determine the virulence of STEC using the nematode Caenorhabditis elegans as a model host. Worm killing, fertility and bacterial colonisation assays were performed to examine the potential difference in the virulence of STEC strains compared to that of the control E. coli OP50 strains on which worms were fed. A statistically significant difference in the survival rates of C. elegans was observed in that the STEC strains caused death in 8–10 days and the E. coli OP50 strains caused death in 15 days. STEC strains severely reduced the fertility of the worms. The intestinal load of bacteria in the adult stage nematodes harbouring the E. coli OP50 strains was found to be 3.5 log CFU mL-1. In contrast, the STEC strains E15, E18 and E22 harboured 4.1, 4.2 and 4.7 log CFU ml−1 per nematode, respectively. The heat-killed STEC strains significantly increased the longevity of the worms compared to the non-heated STEC strains. In addition, PCR-based genomic profiling of shiga toxin genes, viz., stx1 and stx2, identified in selected STEC strains revealed that these toxins may be associated with the virulence of the STEC strains. This study demonstrated that C. elegans is an effective model to examine and compare the pathogenicity and virulence variation of STEC strains to that of E. coli OP50 strains.
Collapse
|
20
|
Stork C, Kovács B, Rózsai B, Putze J, Kiel M, Dorn Á, Kovács J, Melegh S, Leimbach A, Kovács T, Schneider G, Kerényi M, Emödy L, Dobrindt U. Characterization of Asymptomatic Bacteriuria Escherichia coli Isolates in Search of Alternative Strains for Efficient Bacterial Interference against Uropathogens. Front Microbiol 2018; 9:214. [PMID: 29491858 PMCID: PMC5817090 DOI: 10.3389/fmicb.2018.00214] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 01/30/2018] [Indexed: 12/16/2022] Open
Abstract
Asymptomatic bacterial colonization of the urinary bladder (asymptomatic bacteriuria, ABU) can prevent bladder colonization by uropathogens and thus symptomatic urinary tract infection (UTI). Deliberate bladder colonization with Escherichia coli ABU isolate 83972 has been shown to outcompete uropathogens and prevent symptomatic UTI by bacterial interference. Many ABU isolates evolved from uropathogenic ancestors and, although attenuated, may still be able to express virulence-associated factors. Our aim was to screen for efficient and safe candidate strains that could be used as alternatives to E. coli 83972 for preventive and therapeutic bladder colonization. To identify ABU E. coli strains with minimal virulence potential but maximal interference efficiency, we compared nine ABU isolates from diabetic patients regarding their virulence- and fitness-associated phenotypes in vitro, their virulence in a murine model of sepsis and their genome content. We identified strains in competitive growth experiments, which successfully interfere with colonization of ABU isolate 83972 or uropathogenic E. coli strain 536. Six isolates were able to outcompete E. coli 83972 and two of them also outcompeted UPEC 536 during growth in urine. Superior competitiveness was not simply a result of better growth abilities in urine, but seems also to involve expression of antagonistic factors. Competitiveness in urine did not correlate with the prevalence of determinants coding for adhesins, iron uptake, toxins, and antagonistic factors. Three ABU strains (isolates 61, 106, and 123) with superior competitiveness relative to ABU model strain 83972 display low in vivo virulence in a murine sepsis model, and susceptibility to antibiotics. They belong to different phylogroups and differ in the presence of ExPEC virulence- and fitness-associated genes. Importantly, they all lack marked cytotoxic activity and exhibit a high LD50 value in the sepsis model. These strains represent promising candidates for a more detailed assessment of relevant fitness traits in urine and their suitability for therapeutic bladder colonization.
Collapse
Affiliation(s)
- Christoph Stork
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Beáta Kovács
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary.,First Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Barnabás Rózsai
- Department of Paediatrics, University of Pécs, Pécs, Hungary
| | - Johannes Putze
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Matthias Kiel
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Ágnes Dorn
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Judit Kovács
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Szilvia Melegh
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | | | | | - György Schneider
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Monika Kerényi
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Levente Emödy
- Department of Medical Microbiology and Immunology, University of Pécs, Pécs, Hungary
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster, Germany
| |
Collapse
|