1
|
Shenker BJ, Korostoff J, Walker LP, Zekavat A, Dhingra A, Kim TJ, Boesze-Battaglia K. Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Induces Cellugyrin-(Synaptogyrin 2) Dependent Cellular Senescence in Oral Keratinocytes. Pathogens 2024; 13:155. [PMID: 38392893 PMCID: PMC10892517 DOI: 10.3390/pathogens13020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Recently, we reported that oral-epithelial cells (OE) are unique in their response to Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) in that cell cycle arrest (G2/M) occurs without leading to apoptosis. We now demonstrate that Cdt-induced cell cycle arrest in OE has a duration of at least 7 days with no change in viability. Moreover, toxin-treated OE develops a new phenotype consistent with cellular senescence; this includes increased senescence-associated β-galactosidase (SA-β-gal) activity and accumulation of the lipopigment, lipofuscin. Moreover, the cells exhibit a secretory profile associated with cellular senescence known as the senescence-associated secretory phenotype (SASP), which includes IL-6, IL-8 and RANKL. Another unique feature of Cdt-induced OE senescence is disruption of barrier function, as shown by loss of transepithelial electrical resistance and confocal microscopic assessment of primary gingival keratinocyte structure. Finally, we demonstrate that Cdt-induced senescence is dependent upon the host cell protein cellugyrin, a homologue of the synaptic vesicle protein synaptogyrin. Collectively, these observations point to a novel pathogenic outcome in oral epithelium that we propose contributes to both A. actinomycetemcomitans infection and periodontal disease progression.
Collapse
Affiliation(s)
- Bruce J. Shenker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Jonathan Korostoff
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (T.J.K.)
| | - Lisa P. Walker
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Ali Zekavat
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Anuradha Dhingra
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| | - Taewan J. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (T.J.K.)
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (L.P.W.); (A.Z.); (A.D.); (K.B.-B.)
| |
Collapse
|
2
|
Krsek D, Yara DA, Hrbáčková H, Daniel O, Mančíková A, Schüller S, Bielaszewska M. Translocation of outer membrane vesicles from enterohemorrhagic Escherichia coli O157 across the intestinal epithelial barrier. Front Microbiol 2023; 14:1198945. [PMID: 37303786 PMCID: PMC10248468 DOI: 10.3389/fmicb.2023.1198945] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Outer membrane vesicles (OMVs) carrying virulence factors of enterohemorrhagic Escherichia coli (EHEC) are assumed to play a role in the pathogenesis of life-threatening hemolytic uremic syndrome (HUS). However, it is unknown if and how OMVs, which are produced in the intestinal lumen, cross the intestinal epithelial barrier (IEB) to reach the renal glomerular endothelium, the major target in HUS. We investigated the ability of EHEC O157 OMVs to translocate across the IEB using a model of polarized Caco-2 cells grown on Transwell inserts and characterized important aspects of this process. Using unlabeled or fluorescently labeled OMVs, tests of the intestinal barrier integrity, inhibitors of endocytosis, cell viability assay, and microscopic techniques, we demonstrated that EHEC O157 OMVs translocated across the IEB. OMV translocation involved both paracellular and transcellular pathways and was significantly increased under simulated inflammatory conditions. In addition, translocation was not dependent on OMV-associated virulence factors and did not affect viability of intestinal epithelial cells. Importantly, translocation of EHEC O157 OMVs was confirmed in human colonoids thereby supporting physiological relevance of OMVs in the pathogenesis of HUS.
Collapse
Affiliation(s)
- Daniel Krsek
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | | | - Hana Hrbáčková
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Ondřej Daniel
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Andrea Mančíková
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| | - Stephanie Schüller
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Martina Bielaszewska
- Centre for Epidemiology and Microbiology, National Institute of Public Health, Prague, Czechia
| |
Collapse
|
3
|
Comparative Genomics of Shiga Toxin-Producing Escherichia coli Strains Isolated from Pediatric Patients with and without Hemolytic Uremic Syndrome from 2000 to 2016 in Finland. Microbiol Spectr 2022; 10:e0066022. [PMID: 35730965 PMCID: PMC9430701 DOI: 10.1128/spectrum.00660-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) infection can cause mild to severe illness, such as nonbloody or bloody diarrhea, and the fatal hemolytic uremic syndrome (HUS). The molecular mechanism underlying the variable pathogenicity of STEC infection is not fully defined so far. Here, we performed a comparative genomics study on a large collection of clinical STEC strains collected from STEC-infected pediatric patients with and without HUS in Finland over a 16-year period, aiming to identify the bacterial genetic factors that can predict the risk to cause HUS and poor renal outcome. Of 240 STEC strains included in this study, 52 (21.7%) were from pediatric patients with HUS. Serotype O157:H7 was the main cause of HUS, and Shiga toxin gene subtype stx2a was significantly associated with HUS. Comparative genomics and pangenome-wide association studies identified a number of virulence and accessory genes overrepresented in HUS-associated STEC compared to non-HUS STEC strains, including genes encoding cytolethal distending toxins, type III secretion system effectors, adherence factors, etc. No virulence or accessory gene was significantly associated with risk factors for poor renal outcome among HUS patients assessed in this study, including need for and duration of dialysis, presence and duration of anuria, and leukocyte counts. Whole-genome phylogeny and multiple-correspondence analysis of pangenomes could not separate HUS STEC from non-HUS STEC strains, suggesting that STEC strains with diverse genetic backgrounds may independently acquire genetic elements that determine their varied pathogenicity. Our findings indicate that nonbacterial factors, i.e., characteristics of the host immunity, might affect STEC virulence and clinical outcomes. IMPORTANCE Shiga toxin-producing Escherichia coli (STEC) is a serious public health burden worldwide which causes outbreaks of gastrointestinal diseases and the fatal hemolytic uremic syndrome (HUS) characterized by the triad of mechanical hemolytic anemia, thrombocytopenia, and acute renal failure. Understanding the mechanism underlying the disease severity and patient outcome is of high importance. Using comparative genomics on a large collection of clinical STEC strains from STEC-infected patients with and without HUS, our study provides a reference of STEC genetic factors/variants that can be used as predictors of the development of HUS, which will aid risk assessment at the early stage of STEC infection. Additionally, our findings suggest that nonbacterial factors may play a primary role in the renal outcome in STEC-infected patients with HUS; further studies are needed to validate this.
Collapse
|
4
|
Enterohemorrhagic Escherichia coli and a Fresh View on Shiga Toxin-Binding Glycosphingolipids of Primary Human Kidney and Colon Epithelial Cells and Their Toxin Susceptibility. Int J Mol Sci 2022; 23:ijms23136884. [PMID: 35805890 PMCID: PMC9266556 DOI: 10.3390/ijms23136884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are the human pathogenic subset of Shiga toxin (Stx)-producing E. coli (STEC). EHEC are responsible for severe colon infections associated with life-threatening extraintestinal complications such as the hemolytic-uremic syndrome (HUS) and neurological disturbances. Endothelial cells in various human organs are renowned targets of Stx, whereas the role of epithelial cells of colon and kidneys in the infection process has been and is still a matter of debate. This review shortly addresses the clinical impact of EHEC infections, novel aspects of vesicular package of Stx in the intestine and the blood stream as well as Stx-mediated extraintestinal complications and therapeutic options. Here follows a compilation of the Stx-binding glycosphingolipids (GSLs), globotriaosylceramide (Gb3Cer) and globotetraosylceramide (Gb4Cer) and their various lipoforms present in primary human kidney and colon epithelial cells and their distribution in lipid raft-analog membrane preparations. The last issues are the high and extremely low susceptibility of primary renal and colonic epithelial cells, respectively, suggesting a large resilience of the intestinal epithelium against the human-pathogenic Stx1a- and Stx2a-subtypes due to the low content of the high-affinity Stx-receptor Gb3Cer in colon epithelial cells. The review closes with a brief outlook on future challenges of Stx research.
Collapse
|
5
|
Cytolethal distending toxin: from genotoxin to a potential biomarker and anti-tumor target. World J Microbiol Biotechnol 2021; 37:150. [PMID: 34379213 DOI: 10.1007/s11274-021-03117-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Abstract
Cytolethal Distending Toxin (CDT) belongs to the AB toxin family and is produced by a plethora of Gram-negative bacteria. Eight human-affecting enteropathogens harbor CDT that causes irritable bowel syndrome (IBS), dysentery, chancroid, and periodontitis worldwide. They have a novel molecular mode of action as they interfere in the eukaryotic cell-cycle progression leading to G2/M arrest and apoptosis. CDT, the first bacterial genotoxin described, is encoded in a single operon possessing three proteins, CdtA, CdtB, and CdtC. CdtA and CdtC are needed for the binding of the CDT toxin complex to the cholesterol-rich lipid domains of the host cell while the CdtB is the active moiety. Sequence and 3D structural-based analysis of CdtB showed similarities with nucleases and phosphatases, it was hypothesized that CdtB exercises a biochemical function identical to both these enzymes. CDT is secreted through the outer membrane vesicles from the producing bacteria. It is internalized in the target cells via clathrin-dependent endocytosis and translocated to the host cell nucleus through the Golgi complex and ER. This study discusses the virulence role of CDT, causing pathogenicity by acting as a tri-perditious complex in the CDT-producing species with an emphasis on its potential role as a biomarker and an anti-tumor agent.
Collapse
|
6
|
Galarce N, Sánchez F, Escobar B, Lapierre L, Cornejo J, Alegría-Morán R, Neira V, Martínez V, Johnson T, Fuentes-Castillo D, Sano E, Lincopan N. Genomic Epidemiology of Shiga Toxin-Producing Escherichia coli Isolated from the Livestock-Food-Human Interface in South America. Animals (Basel) 2021; 11:ani11071845. [PMID: 34206206 PMCID: PMC8300192 DOI: 10.3390/ani11071845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Shiga toxin-producing Escherichia coli (STEC) are zoonotic pathogens that cause food-borne diseases in humans, where cattle and derived products play a key role as reservoirs and vehicles. We analyzed the genomic data of STEC strains circulating at the livestock-food-human interface in South America, extracting clinically and epidemiologically relevant information (serotypes, virulome, resistance genes, sequence types, and phylogenomics). This study included 130 STEC genomes obtained from cattle (n = 51), beef (n = 48), and human (n = 31) samples. The successful expansion of O157:H7 (ST11) and non-O157 (ST16, ST21, ST223, ST443, ST677, ST679, ST2388) clones is highlighted, suggesting common activities, such as multilateral trade and travel. Circulating STEC strains analyzed exhibit high genomic diversity and harbor several genetic determinants associated with severe illness in humans, highlighting the need to establish official surveillance of this pathogen that should be focused on detecting molecular determinants of virulence and clonal relatedness, in the whole beef production chain. Abstract Shiga toxin-producing Escherichia coli (STEC) are zoonotic pathogens responsible for causing food-borne diseases in humans. While South America has the highest incidence of human STEC infections, information about the genomic characteristics of the circulating strains is scarce. The aim of this study was to analyze genomic data of STEC strains isolated in South America from cattle, beef, and humans; predicting the antibiotic resistome, serotypes, sequence types (STs), clonal complexes (CCs) and phylogenomic backgrounds. A total of 130 whole genome sequences of STEC strains were analyzed, where 39.2% were isolated from cattle, 36.9% from beef, and 23.8% from humans. The ST11 was the most predicted (20.8%) and included O-:H7 (10.8%) and O157:H7 (10%) serotypes. The successful expansion of non-O157 clones such as ST16/CC29-O111:H8 and ST21/CC29-O26:H11 is highlighted, suggesting multilateral trade and travel. Virulome analyses showed that the predominant stx subtype was stx2a (54.6%); most strains carried ehaA (96.2%), iha (91.5%) and lpfA (77.7%) genes. We present genomic data that can be used to support the surveillance of STEC strains circulating at the livestock-food-human interface in South America, in order to control the spread of critical clones “from farm to table”.
Collapse
Affiliation(s)
- Nicolás Galarce
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
- Correspondence:
| | - Fernando Sánchez
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
- Programa de Doctorado en Ciencias Silvoagropecuarias y Veterinarias, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile
| | - Beatriz Escobar
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
| | - Lisette Lapierre
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
| | - Javiera Cornejo
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
| | - Raúl Alegría-Morán
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
- Facultad de Ciencias Agropecuarias y Ambientales, Universidad Pedro de Valdivia, Santiago 8370007, Chile
| | - Víctor Neira
- Departamento de Medicina Preventiva Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile; (F.S.); (B.E.); (L.L.); (J.C.); (R.A.-M.); (V.N.)
| | - Víctor Martínez
- Departamento de Fomento de la Producción Animal, Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago 8820808, Chile;
| | - Timothy Johnson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA;
| | - Danny Fuentes-Castillo
- Departamento de Patología, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil;
| | - Elder Sano
- Departamento de Microbiología, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brazil; (E.S.); (N.L.)
| | - Nilton Lincopan
- Departamento de Microbiología, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-900, Brazil; (E.S.); (N.L.)
| |
Collapse
|
7
|
Virulence Factor Cargo and Host Cell Interactions of Shiga Toxin-Producing Escherichia coli Outer Membrane Vesicles. Methods Mol Biol 2021; 2291:177-205. [PMID: 33704754 DOI: 10.1007/978-1-0716-1339-9_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Outer membrane vesicles (OMVs), nanoparticles released by Shiga toxin-producing Escherichia coli (STEC), have been identified as novel efficient virulence tools of these pathogens. STEC O157 OMVs carry a cocktail of virulence factors including Shiga toxin 2a (Stx2a), cytolethal distending toxin V (CdtV), EHEC hemolysin, flagellin, and lipopolysaccharide. OMVs are taken up by human intestinal epithelial and microvascular endothelial cells, the major targets during STEC infection, and deliver the virulence factors into host cells. There the toxins separate from OMVs and are trafficked via different pathways to their target compartments, i.e., the cytosol (Stx2a-A subunit), nucleus (CdtV-B subunit), and mitochondria (EHEC hemolysin). This leads to a toxin-specific host cell injury and ultimately apoptotic cell death. Besides their cytotoxic effects, STEC OMVs trigger an inflammatory response via their lipopolysaccharide and flagellin components. In this chapter, we describe methods for the isolation and purification of STEC OMVs, for the detection of OMV-associated virulence factors, and for the analysis of OMV interactions with host cells including OMV cellular uptake and intracellular trafficking of OMVs and OMV-delivered toxins.
Collapse
|
8
|
Pinto G, Almeida C, Azeredo J. Bacteriophages to control Shiga toxin-producing E. coli - safety and regulatory challenges. Crit Rev Biotechnol 2020; 40:1081-1097. [PMID: 32811194 DOI: 10.1080/07388551.2020.1805719] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Shiga toxin-producing Escherichia coli (STEC) are usually found on food products due to contamination from the fecal origin, as their main environmental reservoir is considered to be the gut of ruminants. While this pathogen is far from the incidence of other well-known foodborne bacteria, the severity of STEC infections in humans has triggered global concerns as far as its incidence and control are concerned. Major control strategies for foodborne pathogens in food-related settings usually involve traditional sterilization/disinfection techniques. However, there is an increasing need for the development of further strategies to enhance the antimicrobial outcome, either on food-contact surfaces or directly in food matrices. Phages are considered to be a good alternative to control foodborne pathogens, with some phage-based products already cleared by the Food and Drug Administration (FDA) to be used in the food industry. In European countries, phage-based food decontaminants have already been used. Nevertheless, its broad use in the European Union is not yet possible due to the lack of specific guidelines for the approval of these products. Furthermore, some safety concerns remain to be addressed so that the regulatory requirements can be met. In this review, we present an overview of the main virulence factors of STEC and introduce phages as promising biocontrol agents for STEC control. We further present the regulatory constraints on the approval of phages for food applications and discuss safety concerns that are still impairing their use.
Collapse
Affiliation(s)
- Graça Pinto
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| | - Carina Almeida
- INIAV, IP-National Institute for Agrarian and Veterinary Research, Vairão, Portugal
| | - Joana Azeredo
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Braga, Portugal
| |
Collapse
|
9
|
Pons BJ, Bezine E, Hanique M, Guillet V, Mourey L, Chicher J, Frisan T, Vignard J, Mirey G. Cell transfection of purified cytolethal distending toxin B subunits allows comparing their nuclease activity while plasmid degradation assay does not. PLoS One 2019; 14:e0214313. [PMID: 30921382 PMCID: PMC6438463 DOI: 10.1371/journal.pone.0214313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/11/2019] [Indexed: 12/30/2022] Open
Abstract
The Cytolethal Distending Toxin (CDT) is produced by many pathogenic bacteria. CDT is known to induce genomic DNA damage to host eukaryotic cells through its catalytic subunit, CdtB. CdtB is structurally homologous to DNase I and has a nuclease activity, dependent on several key residues. Yet some differences between various CdtB subunit activities, and discrepancies between biochemical and cellular data, have been observed. To better characterise the role of CdtB in the induction of DNA damage, we affinity-purified wild-type and mutants of CdtB, issued from E. coli and H. ducreyi, under native and denaturing conditions. We then compared their nuclease activity by a classic in vitro assay using plasmid DNA, and two different eukaryotic assays–the first assay where host cells were transfected with a plasmid encoding CdtB, the second assay where host cells were directly transfected with purified CdtB. We show here that in vitro nuclease activities are difficult to quantify, whereas CdtB activities in host cells can be easily interpreted and confirmed the loss of function of the catalytic mutant. Our results highlight the importance of performing multiple assays while studying the effects of bacterial genotoxins, and indicate that the classic in vitro assay should be complemented with cellular assays.
Collapse
Affiliation(s)
- Benoît J. Pons
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
| | - Elisabeth Bezine
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Institut National Polytechnique de Toulouse, Toulouse, France
| | - Mélissa Hanique
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
| | - Valérie Guillet
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse, France
| | - Lionel Mourey
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- Centre National de la Recherche Scientifique (CNRS), Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse, France
| | - Johana Chicher
- Plateforme protéomique Strasbourg Esplanade, Institut de Biologie Moléculaire et Cellulaire (IBMC), FRC1589 Strasbourg, France
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Julien Vignard
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- * E-mail: (GM); (JV)
| | - Gladys Mirey
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, Toulouse, France
- Université Toulouse III–Paul Sabatier (UPS), Toulouse, France
- * E-mail: (GM); (JV)
| |
Collapse
|
10
|
Hernandez LB, Cadona JS, Christensen M, Fernández D, Padola NL, Bustamante AV, Sanso AM. Virulence genes and genetic diversity assessment of Shiga toxin-producing Escherichia coli O91 strains from cattle, beef and poultry products. Microb Pathog 2018; 125:463-467. [PMID: 30300666 DOI: 10.1016/j.micpath.2018.10.009] [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: 08/14/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 11/16/2022]
Abstract
Shiga toxin-producing Escherichia coli (STEC) O91 has ranked in the top five of the non-O157 serogroups most frequently associated with human cases. In order to gain insight into the genetic diversity of O91 Latin American STEC strains, we analyzed their virulence properties and carried out a subtyping assay. A panel of 21 virulence genetic markers associated with human and animal infections was evaluated and the relatedness among strains was determined by a multiple-locus variable-number tandem repeats analysis (MLVA) comprising 9 VNTR loci. Twenty-two STEC O91 isolated from cattle and meat food and belonging to 5 serotypes (O91:H21, O91:H8, O91:H14, O91:H28, O91:H40) were studied. Eight virulence profiles were obtained for the O91 STEC strains: 4 for O91:H21 plus one for O91:H8, O91:H14, O91:H28 and O91:H40. All strains contained ehxA and lpfA0113 genes and only both stx1-positive strains lacked saa, which encodes the STEC autoagglutinating adhesin. Other genes involved in adhesion were detected: ehaA (91%), elfA and espP (86%), ecpA (82%) and, hcpA (77%). The gene encoding the cytolethal distending toxin type-V (CDT-V) was found only in O91:H8 and O91:H21, being present in the majority (89%) of strains of this last serotype. MLVA typing divided the total number of strains into 12 genotypes, and 9 of them were unique to a single strain. No association was observed between the virulence profiles and the source of the strains. Although they lack the eae gene, most of the strains have the genetic potential to adhere to host cells through other structures and possess cdt-V, which has been found in STEC strains involved in serious diseases. The MLVA showed clonal relatedness among strains isolated from cattle belonged to a same dairy farm and suggested that the same clone remains circulating throughout the year and, on the other hand, the need to increase the number of VNTR loci which could allow a higher discrimination among O91:H21 isolates.
Collapse
Affiliation(s)
- Luciana Belén Hernandez
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Jimena Soledad Cadona
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Martín Christensen
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Daniel Fernández
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Nora Lía Padola
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Ana Victoria Bustamante
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina
| | - Andrea Mariel Sanso
- Laboratorio de Inmunoquímica y Biotecnología, Centro de Investigación Veterinaria de Tandil (CIVETAN), CONICET-CIC-UNCPBA, Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Provincia de Buenos Aires, 7000, Tandil, Argentina.
| |
Collapse
|
11
|
Legros N, Pohlentz G, Steil D, Müthing J. Shiga toxin-glycosphingolipid interaction: Status quo of research with focus on primary human brain and kidney endothelial cells. Int J Med Microbiol 2018; 308:1073-1084. [PMID: 30224239 DOI: 10.1016/j.ijmm.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 12/21/2022] Open
Abstract
Shiga toxin (Stx)-mediated injury of the kidneys and the brain represent the major extraintestinal complications in humans upon infection by enterohemorrhagic Escherichia coli (EHEC). Damage of renal and cerebral endothelial cells is the key event in the pathogenesis of the life-threatening hemolytic uremic syndrome (HUS). Stxs are AB5 toxins and the B-pentamers of the two clinically important Stx subtypes Stx1a and Stx2a preferentially bind to the glycosphingolipid globotriaosylceramide (Gb3Cer, Galα4Galβ4Glcβ1Cer) and to less extent to globotetraosylceramide (Gb4Cer, GalNAcβ3Galα4Galβ4Glcβ1), which are expected to reside in lipid rafts in the plasma membrane of the human endothelium. This review summarizes the current knowledge on the Stx glycosphingolipid receptors and their lipid membrane ensemble in primary human brain microvascular endothelial cells (pHBMECs) and primary human renal glomerular endothelial cells (pHRGECs). Increasing knowledge on the precise initial molecular mechanisms by which Stxs interact with cellular targets will help to develop specific therapeutics and/or preventive measures to combat EHEC-caused diseases.
Collapse
Affiliation(s)
- Nadine Legros
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | | | - Daniel Steil
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, D-48149 Münster, Germany; Interdisciplinary Center for Clinical Research (IZKF), University of Münster, D-48149 Münster, Germany.
| |
Collapse
|
12
|
Sanso AM, Bustamante AV, Krüger A, Cadona JS, Alfaro R, Cáceres ME, Fernández D, Lucchesi PMA, Padola NL. Molecular epidemiology of Shiga toxin-producing O113:H21 isolates from cattle and meat. Zoonoses Public Health 2018; 65:569-577. [DOI: 10.1111/zph.12467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Indexed: 12/01/2022]
Affiliation(s)
- A. M. Sanso
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - A. V. Bustamante
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - A. Krüger
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - J. S. Cadona
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - R. Alfaro
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - M. E. Cáceres
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - D. Fernández
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - P. M. A. Lucchesi
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| | - N. L. Padola
- Laboratorio de Inmunoquímica y Biotecnología; Facultad de Ciencias Veterinarias; Centro de Investigación Veterinaria de Tandil; CONICET-CIC-UNCPBA; Tandil Argentina
| |
Collapse
|
13
|
Sorbitol-Fermenting Enterohemorrhagic Escherichia coli O157:H - Isolates from Czech Patients with Novel Plasmid Composition Not Previously Seen in German Isolates. Appl Environ Microbiol 2017; 83:AEM.01454-17. [PMID: 28970221 DOI: 10.1128/aem.01454-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/21/2017] [Indexed: 01/31/2023] Open
Abstract
Sorbitol-fermenting (SF) enterohemorrhagic Escherichia coli (EHEC) O157:H- strains, first identified in Germany, have emerged as important pathogens throughout Europe. Besides chromosomally encoded Shiga toxin 2a (the major virulence factor), several putative virulence loci, including the hly, etp, and sfp operons, encoding EHEC hemolysin, type II secretion system proteins, and Sfp fimbriae, respectively, are located on the 121-kb plasmid pSFO157 in German strains. Here we report novel SF EHEC O157:H- strains isolated from patients in the Czech Republic. These strains share the core genomes and chromosomal virulence loci encoding toxins (stx2a and the cdtV-ABC operon) and adhesins (eae-γ, efa1, lpfAO157OI-141, and lpfAO157OI-154) with German strains but differ essentially in their plasmids. In contrast to all previously detected SF EHEC O157:H- strains, the Czech strains carry two plasmids, of 79 kb and 86 kb. The 79-kb plasmid harbors the sfp operon, but neither of the plasmids contains the hly and etp operons. Sequence analyses demonstrated that the 79-kb plasmid (pSFO157 258/98-1) evolved from pSFO157 of German strains by deletion of a 41,534-bp region via homologous recombination, resulting in loss of the hly and etp operons. The 86-kb plasmid (pSFO157 258/98-2) displays 98% sequence similarity to a 92.7-kb plasmid of an extraintestinal pathogenic E. coli bloodstream isolate. Our finding of this novel plasmid composition in SF EHEC O157:H- strains extends the evolutionary history of EHEC O157 plasmids. Moreover, the unique molecular plasmid characteristics permit the identification of such strains, thereby facilitating further investigations of their geographic distribution, clinical significance, and epidemiology.IMPORTANCE Since their first identification in Germany in 1989, sorbitol-fermenting enterohemorrhagic Escherichia coli O157:H- (nonmotile) strains have emerged as important causes of the life-threatening disease hemolytic-uremic syndrome in Europe. They account for 10 to 20% of sporadic cases of this disease and have caused several large outbreaks. The strains isolated throughout Europe share conserved chromosomal and plasmid characteristics. Here we identified novel sorbitol-fermenting enterohemorrhagic E. coli O157:H- patient isolates in the Czech Republic which differ from all such strains reported previously by their unique plasmid characteristics, including plasmid number, composition of plasmid-carried virulence genes, and plasmid origins. Our findings contribute substantially to understanding the evolution of E. coli O157 strains and their plasmids. In practical terms, they enable the identification of strains with these novel plasmid characteristics in patient stool samples and thus the investigation of their roles as human pathogens in other geographic areas.
Collapse
|
14
|
Li G, Niu H, Zhang Y, Li Y, Xie F, Langford PR, Liu S, Wang C. Haemophilus parasuis cytolethal distending toxin induces cell cycle arrest and p53-dependent apoptosis. PLoS One 2017; 12:e0177199. [PMID: 28545143 PMCID: PMC5436662 DOI: 10.1371/journal.pone.0177199] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/24/2017] [Indexed: 01/20/2023] Open
Abstract
Haemophilus parasuis is the causative agent of Glasser’s disease in pigs. Cytolethal distending toxin (CDT) is an important virulence factor of H. parasuis. It is composed of three subunits: CdtA, CdtB and CdtC and all were successfully expressed in soluble form in Escherichia coli when the signal peptides were removed. Purified CdtB had DNase activity, i.e. caused DNA double strand damage, in vitro and in vivo prior to cell arrest and apoptosis. Flow cytometry analysis showed CdtB alone could induce cell cycle arrest and apoptosis in PK-15 porcine kidney and pulmonary alveolar macrophage (PAM) cells, which could be enhanced by CdtA or/and CdtC. CDT holotoxin could lead to significant cell distension, G2 arrest and apoptotic death in PK-15 and PAM cells. The apoptosis induced by CDT holotoxin was significantly inhibited by pifithrin-α, which indicates that it is p53-dependent. The results suggest that H. parasuis CDT holotoxin is a major virulence factor.
Collapse
Affiliation(s)
- Gang Li
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hui Niu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yanhe Zhang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yanling Li
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Fang Xie
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Paul R. Langford
- Section of Paediatrics, Department of Medicine, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chunlai Wang
- State Key Laboratory of Veterinary Biotechnology, Division of Bacterial Diseases, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- * E-mail:
| |
Collapse
|
15
|
Rubin D, Zhang W, Karch H, Kuczius T. Distinct Expression of Immunoglobulin-Binding Proteins in Shiga Toxin-Producing Escherichia coli Implicates High Protein Stability and a Characteristic Phenotype. Toxins (Basel) 2017; 9:toxins9050153. [PMID: 28468281 PMCID: PMC5450701 DOI: 10.3390/toxins9050153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 11/16/2022] Open
Abstract
Several immunoglobulin-binding proteins of Escherichia coli (Eib) have been isolated from both non-pathogenic and pathogenic E. coli strains. Shiga toxin (Stx)-producing E. coli (STEC) contain eibG either as a single gene or in combination with eibC, while other E. coli strains harbour single or multiple eib genes. The Eib proteins bind human immunoglobulins in a non-immune manner and contribute to bacterial chain-like adherence to human epithelial cells. In this study, the EibG expression in several STEC strains was analysed under different environmental conditions. STEC produced high levels of EibG in complex media and lower levels in low-grade and minimal media under static growth conditions. This characteristic was independent on the Eib subtypes. Microscopically, EibG-expressing STEC exhibited chain formation and aggregation in all employed media, while aggregates were only visible after growth in complex medium. Once expressed, EibG proteins demonstrate high stability during prolonged incubation. Our findings indicate that the regulation of the expression of Eib proteins is highly complex, although the protein levels vary among STEC strains. However, positive upregulation conditions generally result in distinct phenotypes of the isolates.
Collapse
Affiliation(s)
- Dennis Rubin
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Straße 41, 48149 Münster, Germany.
| | - Wenlan Zhang
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Straße 41, 48149 Münster, Germany.
| | - Helge Karch
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Straße 41, 48149 Münster, Germany.
| | - Thorsten Kuczius
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Straße 41, 48149 Münster, Germany.
| |
Collapse
|
16
|
Bielaszewska M, Rüter C, Bauwens A, Greune L, Jarosch KA, Steil D, Zhang W, He X, Lloubes R, Fruth A, Kim KS, Schmidt MA, Dobrindt U, Mellmann A, Karch H. Host cell interactions of outer membrane vesicle-associated virulence factors of enterohemorrhagic Escherichia coli O157: Intracellular delivery, trafficking and mechanisms of cell injury. PLoS Pathog 2017; 13:e1006159. [PMID: 28158302 PMCID: PMC5310930 DOI: 10.1371/journal.ppat.1006159] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 02/15/2017] [Accepted: 12/30/2016] [Indexed: 01/15/2023] Open
Abstract
Outer membrane vesicles (OMVs) are important tools in bacterial virulence but their role in the pathogenesis of infections caused by enterohemorrhagic Escherichia coli (EHEC) O157, the leading cause of life-threatening hemolytic uremic syndrome, is poorly understood. Using proteomics, electron and confocal laser scanning microscopy, immunoblotting, and bioassays, we investigated OMVs secreted by EHEC O157 clinical isolates for virulence factors cargoes, interactions with pathogenetically relevant human cells, and mechanisms of cell injury. We demonstrate that O157 OMVs carry a cocktail of key virulence factors of EHEC O157 including Shiga toxin 2a (Stx2a), cytolethal distending toxin V (CdtV), EHEC hemolysin, and flagellin. The toxins are internalized by cells via dynamin-dependent endocytosis of OMVs and differentially separate from vesicles during intracellular trafficking. Stx2a and CdtV-B, the DNase-like CdtV subunit, separate from OMVs in early endosomes. Stx2a is trafficked, in association with its receptor globotriaosylceramide within detergent-resistant membranes, to the Golgi complex and the endoplasmic reticulum from where the catalytic Stx2a A1 fragment is translocated to the cytosol. CdtV-B is, after its retrograde transport to the endoplasmic reticulum, translocated to the nucleus to reach DNA. CdtV-A and CdtV-C subunits remain OMV-associated and are sorted with OMVs to lysosomes. EHEC hemolysin separates from OMVs in lysosomes and targets mitochondria. The OMV-delivered CdtV-B causes cellular DNA damage, which activates DNA damage responses leading to G2 cell cycle arrest. The arrested cells ultimately die of apoptosis induced by Stx2a and CdtV via caspase-9 activation. By demonstrating that naturally secreted EHEC O157 OMVs carry and deliver into cells a cocktail of biologically active virulence factors, thereby causing cell death, and by performing first comprehensive analysis of intracellular trafficking of OMVs and OMV-delivered virulence factors, we provide new insights into the pathogenesis of EHEC O157 infections. Our data have implications for considering O157 OMVs as vaccine candidates. Enterohemorrhagic Escherichia coli (EHEC) O157, the leading EHEC group causing diarrhea and the life-threatening hemolytic uremic syndrome in humans, produce several virulence factors which play distinct roles in the pathogenesis of these diseases. However, the mechanisms of their secretion and host cell injury are poorly understood. We show here that EHEC O157 strains isolated from patients shed nanostructures termed outer membrane vesicles (OMVs) which contain major EHEC O157 virulence factors including Shiga toxin 2a (Stx2a), cytolethal distending toxin V (CdtV), EHEC hemolysin, and flagellin. The OMVs are taken up by human intestinal epithelial and renal and brain microvascular endothelial cells, which are the major targets during EHEC O157 infections, and deliver the virulence factors intracellularly. Inside cells the virulence factors separate from OMVs and are transported via different pathways to their target compartments including the cytosol (Stx2a), nucleus (CdtV-B subunit), and mitochondria (EHEC hemolysin). Cells exposed to EHEC O157 OMVs develop G2 cell cycle arrest induced by CdtV-B-mediated DNA damage. This is followed by apoptotic cell death triggered by Stx2a and CdtV via caspase-9 activation. OMVs thus serve as novel tools of EHEC O157-mediated host injury and are quite likely involved in the pathogenesis of human diseases.
Collapse
Affiliation(s)
| | - Christian Rüter
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Andreas Bauwens
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | | | - Daniel Steil
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Wenlan Zhang
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Xiaohua He
- Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture (USDA), Albany, California, United States of America
| | - Roland Lloubes
- Laboratoire d'Ingenierie des Systemes Macromoleculaires UMR7255, CNRS-Aix-Marseille Université, Marseille, France
| | - Angelika Fruth
- National Reference Center for Salmonella and Other Enteric Pathogens, Robert Koch Institute, Branch Wernigerode, Wernigerode, Germany
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - M. Alexander Schmidt
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
| | - Helge Karch
- Institute of Hygiene, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
| |
Collapse
|
17
|
Scuron MD, Boesze-Battaglia K, Dlakić M, Shenker BJ. The Cytolethal Distending Toxin Contributes to Microbial Virulence and Disease Pathogenesis by Acting As a Tri-Perditious Toxin. Front Cell Infect Microbiol 2016; 6:168. [PMID: 27995094 PMCID: PMC5136569 DOI: 10.3389/fcimb.2016.00168] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/15/2016] [Indexed: 12/11/2022] Open
Abstract
This review summarizes the current status and recent advances in our understanding of the role that the cytolethal distending toxin (Cdt) plays as a virulence factor in promoting disease by toxin-producing pathogens. A major focus of this review is on the relationship between structure and function of the individual subunits that comprise the AB2 Cdt holotoxin. In particular, we concentrate on the molecular mechanisms that characterize this toxin and which account for the ability of Cdt to intoxicate multiple cell types by utilizing a ubiquitous binding partner on the cell membrane. Furthermore, we propose a paradigm shift for the molecular mode of action by which the active Cdt subunit, CdtB, is able to block a key signaling cascade and thereby lead to outcomes based upon programming and the role of the phosphatidylinositol 3-kinase (PI-3K) in a variety of cells. Based upon the collective Cdt literature, we now propose that Cdt is a unique and potent virulence factor capable of acting as a tri-perditious toxin that impairs host defenses by: (1) disrupting epithelial barriers; (2) suppressing acquired immunity; (3) promoting pro-inflammatory responses. Thus, Cdt plays a key role in facilitating the early stages of infection and the later stages of disease progression by contributing to persistence and impairing host elimination.
Collapse
Affiliation(s)
- Monika D Scuron
- Department of Pathology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| | - Mensur Dlakić
- Department of Microbiology and Immunology, Montana State University Bozeman, MT, USA
| | - Bruce J Shenker
- Department of Pathology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
18
|
Miller R, Wiedmann M. Dynamic Duo-The Salmonella Cytolethal Distending Toxin Combines ADP-Ribosyltransferase and Nuclease Activities in a Novel Form of the Cytolethal Distending Toxin. Toxins (Basel) 2016; 8:E121. [PMID: 27120620 PMCID: PMC4885037 DOI: 10.3390/toxins8050121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/30/2016] [Accepted: 04/15/2016] [Indexed: 01/02/2023] Open
Abstract
The cytolethal distending toxin (CDT) is a well characterized bacterial genotoxin encoded by several Gram-negative bacteria, including Salmonella enterica (S. enterica). The CDT produced by Salmonella (S-CDT) differs from the CDT produced by other bacteria, as it utilizes subunits with homology to the pertussis and subtilase toxins, in place of the traditional CdtA and CdtC subunits. Previously, S-CDT was thought to be a unique virulence factor of S. enterica subspecies enterica serotype Typhi, lending to its classification as the "typhoid toxin." Recently, this important virulence factor has been identified and characterized in multiple nontyphoidal Salmonella (NTS) serotypes as well. The significance of S-CDT in salmonellosis with regards to the: (i) distribution of S-CDT encoding genes among NTS serotypes, (ii) contributions to pathogenicity, (iii) regulation of S-CDT expression, and (iv) the public health implication of S-CDT as it relates to disease severity, are reviewed here.
Collapse
Affiliation(s)
- Rachel Miller
- Department of Food Science, Cornell University, Ithaca, NY 14850 USA.
| | - Martin Wiedmann
- Department of Food Science, Cornell University, Ithaca, NY 14850 USA.
| |
Collapse
|
19
|
Kunsmann L, Rüter C, Bauwens A, Greune L, Glüder M, Kemper B, Fruth A, Wai SN, He X, Lloubes R, Schmidt MA, Dobrindt U, Mellmann A, Karch H, Bielaszewska M. Virulence from vesicles: Novel mechanisms of host cell injury by Escherichia coli O104:H4 outbreak strain. Sci Rep 2015; 5:13252. [PMID: 26283502 PMCID: PMC4539607 DOI: 10.1038/srep13252] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/22/2015] [Indexed: 12/27/2022] Open
Abstract
The highly virulent Escherichia coli O104:H4 that caused the large 2011 outbreak of diarrhoea and haemolytic uraemic syndrome secretes blended virulence factors of enterohaemorrhagic and enteroaggregative E. coli, but their secretion pathways are unknown. We demonstrate that the outbreak strain releases a cocktail of virulence factors via outer membrane vesicles (OMVs) shed during growth. The OMVs contain Shiga toxin (Stx) 2a, the major virulence factor of the strain, Shigella enterotoxin 1, H4 flagellin, and O104 lipopolysaccharide. The OMVs bind to and are internalised by human intestinal epithelial cells via dynamin-dependent and Stx2a-independent endocytosis, deliver the OMV-associated virulence factors intracellularly and induce caspase-9-mediated apoptosis and interleukin-8 secretion. Stx2a is the key OMV component responsible for the cytotoxicity, whereas flagellin and lipopolysaccharide are the major interleukin-8 inducers. The OMVs represent novel ways for the E. coli O104:H4 outbreak strain to deliver pathogenic cargoes and injure host cells.
Collapse
Affiliation(s)
- Lisa Kunsmann
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Christian Rüter
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149 Münster, Germany
| | - Andreas Bauwens
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149 Münster, Germany
| | - Malte Glüder
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Björn Kemper
- Biomedical Technology Center, University of Muenster, 48149 Münster, Germany
| | - Angelika Fruth
- National Reference Center for Salmonella and Other Enteric Pathogens, Robert Koch Institute, Branch Wernigerode, 38855 Wernigerode, Germany
| | - Sun Nyunt Wai
- Department of Molecular Biology, Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, S-90187 Umeå, Sweden
| | - Xiaohua He
- Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture (USDA), Albany, CA 94710, USA
| | - Roland Lloubes
- Laboratoire d'Ingenierie des Systemes Macromoleculaires UMR7255, CNRS-Aix-Marseille Université, 13402 Marseille cedex 20, France
| | - M Alexander Schmidt
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, 48149 Münster, Germany
| | - Ulrich Dobrindt
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Alexander Mellmann
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Helge Karch
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| | - Martina Bielaszewska
- Institute of Hygiene and the National Consulting Laboratory for Hemolytic Uremic Syndrome, University of Münster, 48149 Münster, Germany
| |
Collapse
|
20
|
Kuczius T, Zhang W, Merkel V, Mellmann A, Tarr PI, Karch H. Agitation down-regulates immunoglobulin binding protein EibG expression in Shiga toxin-producing Escherichia coli (STEC). PLoS One 2015; 10:e0119583. [PMID: 25746924 PMCID: PMC4352079 DOI: 10.1371/journal.pone.0119583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 01/23/2015] [Indexed: 11/28/2022] Open
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) carrying eibG synthesize Escherichia coli immunoglobulin binding protein (EibG). EibG nonspecifically binds to immunoglobulins and tends to aggregate in multimers but is poorly expressed in wild-type strains. To study synthesis of the proteins and their regulation in the pathogens, we identified natural growth conditions that increased EibG synthesis. EibG proteins as well as corresponding mRNA were highly expressed under static growth conditions while shearing stress created by agitation during growth repressed protein synthesis. Further regulation effects were driven by reduced oxygen tension, and pH up-regulated EibG expression, but to a lesser extent than growth conditions while decreased temperature down-regulated EibG. Bacteria with increased EibG expression during static growth conditions showed a distinct phenotype with chain formation and biofilm generation, which disappeared with motion. High and low EibG expression was reversible indicating a process with up- and down-regulation of the protein expression. Our findings indicate that shear stress represses EibG expression and might reduce bacterial attachments to cells and surfaces.
Collapse
Affiliation(s)
- Thorsten Kuczius
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Strasse 41, 48149, Münster, Germany
- * E-mail:
| | - Wenlan Zhang
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Strasse 41, 48149, Münster, Germany
| | - Viktor Merkel
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Strasse 41, 48149, Münster, Germany
| | - Alexander Mellmann
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Strasse 41, 48149, Münster, Germany
| | - Phillip I. Tarr
- Department of Pediatrics, Washington University, School of Medicine, Campus Box 8208, 660 S. Euclid, St. Louis, Missouri, 63105, United States of America
| | - Helge Karch
- Institute for Hygiene, Westfälische Wilhelms-University and University Hospital Münster, Robert Koch-Strasse 41, 48149, Münster, Germany
| |
Collapse
|
21
|
Taieb F, Sváb D, Watrin C, Oswald E, Tóth I. Cytolethal distending toxin A, B and C subunit proteins are necessary for the genotoxic effect of Escherichia coli CDT-V. Acta Vet Hung 2015; 63:1-10. [PMID: 25655410 DOI: 10.1556/avet.2015.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cytolethal distending toxins (CDT) are considered the prototype of inhibitory cyclomodulins, and are produced by a wide range of Gram-negative pathogenic bacteria, including Escherichia coli strains of various sero- and pathotypes. CDT is a heterotripartite toxin consisting of three protein subunits, CdtA, CdtB and CdtC. The active subunit, CdtB has DNase activity and causes DNA damage and cell cycle arrest in the target cell. However, several studies have highlighted different roles for CdtA and CdtC subunits. In order to reveal the necessity of CdtA and CdtC subunit proteins in the CDT-specific phenotype, expression clones containing the cdt-V subunit genes were constructed. Using cell culture assays, we demonstrated that clones expressing only the CdtB subunit or in combination with only CdtA or CdtC were unable to trigger the specific cell cycle arrest and changes in cell morphology in HeLa cells. At the same time, the recombinant clone harbouring the whole cdt-V operon caused all the CDT-associated characteristic phenotypes. All these results verify that all the three CDT subunit proteins are necessary for the genotoxic effect caused by CDT-V.
Collapse
Affiliation(s)
- Frederic Taieb
- 1 CHU Purpan USC INRA 1360-CPTP, U1043 INSERM, Pathogénie moléculaire et cellulaire des infections à Escherichia coli Toulouse France
| | - Domonkos Sváb
- 2 Hungarian Academy of Sciences Enteric Bacteriology and Foodborne Zoonoses Group, Institute for Veterinary Medical Research, Centre for Agriculture Research Hungária krt. 21 H-1143 Budapest Hungary
| | - Claude Watrin
- 1 CHU Purpan USC INRA 1360-CPTP, U1043 INSERM, Pathogénie moléculaire et cellulaire des infections à Escherichia coli Toulouse France
| | - Eric Oswald
- 1 CHU Purpan USC INRA 1360-CPTP, U1043 INSERM, Pathogénie moléculaire et cellulaire des infections à Escherichia coli Toulouse France
| | - István Tóth
- 2 Hungarian Academy of Sciences Enteric Bacteriology and Foodborne Zoonoses Group, Institute for Veterinary Medical Research, Centre for Agriculture Research Hungária krt. 21 H-1143 Budapest Hungary
| |
Collapse
|
22
|
Hemolysin of enterohemorrhagic Escherichia coli: Structure, transport, biological activity and putative role in virulence. Int J Med Microbiol 2014; 304:521-9. [DOI: 10.1016/j.ijmm.2014.05.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/09/2014] [Accepted: 05/11/2014] [Indexed: 11/19/2022] Open
|
23
|
Bezine E, Vignard J, Mirey G. The cytolethal distending toxin effects on Mammalian cells: a DNA damage perspective. Cells 2014; 3:592-615. [PMID: 24921185 PMCID: PMC4092857 DOI: 10.3390/cells3020592] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 12/27/2022] Open
Abstract
The cytolethal distending toxin (CDT) is produced by many pathogenic Gram-negative bacteria and is considered as a virulence factor. In human cells, CDT exposure leads to a unique cytotoxicity associated with a characteristic cell distension and induces a cell cycle arrest dependent on the DNA damage response (DDR) triggered by DNA double-strand breaks (DSBs). CDT has thus been classified as a cyclomodulin and a genotoxin. Whereas unrepaired damage can lead to cell death, effective, but improper repair may be detrimental. Indeed, improper repair of DNA damage may allow cells to resume the cell cycle and induce genetic instability, a hallmark in cancer. In vivo, CDT has been shown to induce the development of dysplastic nodules and to lead to genetic instability, defining CDT as a potential carcinogen. It is therefore important to characterize the outcome of the CDT-induced DNA damage and the consequences for intoxicated cells and organisms. Here, we review the latest results regarding the host cell response to CDT intoxication and focus on DNA damage characteristics, cell cycle modulation and cell outcomes.
Collapse
Affiliation(s)
- Elisabeth Bezine
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| | - Julien Vignard
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| | - Gladys Mirey
- INRA, UMR1331, Toxalim, Research Centre in Food Toxicology, F-31027 Toulouse, France.
| |
Collapse
|
24
|
Tyrer PC, Frizelle FA, Keenan JI. Escherichia coli-derived outer membrane vesicles are genotoxic to human enterocyte-like cells. Infect Agent Cancer 2014; 9:2. [PMID: 24405746 PMCID: PMC3898235 DOI: 10.1186/1750-9378-9-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 12/13/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Colorectal cancers are the third most common type in the world. The causes of the disease are poorly understood, but since the discovery of Helicobacter pylori as a causative agent of gastric cancer, attention has turned to bacteria as a possible trigger for colorectal cancer. Recently H. pylori outer membrane vesicles (OMVs) were revealed as potentially genotoxic which can be important first step in carcinogenesis. We therefore investigated whether OMVs from intestinal Escherichia coli could be genotoxic. METHODS OMVs from the avirulent DH5α strain, a pathogenic adherent-invasive E. coli (AIEC) and an enterohaemolytic (EHEC) strain of E. coli were enriched by ultracentrifugation. The effect on the growth and viability of human enterocyte-like Caco-2 cells by OMVs was determined by trypan blue exclusion, MTT and BrdU incorporation assays. The ability of OMVs to induce DNA damage was assayed by single-cell gel electrophoresis, and 8-oxo-dG and γH2Ax immunofluorescence staining. Cytopathological changes were assessed by microscopy. The induction of aneuploidy by the OMVs was measured by flow cytometry in Caco-2 and LoVo cells. RESULTS We found that OMVs derived were internalised by Caco-2 cells, increased cell numbers, induced double-stranded DNA breaks, recruited γH2Ax to the nucleus, initiated DNA rereplication, and produced distended multinucleate cells. DH5α and AIEC OMVs caused free radical generation as indicated by the reduction of glutathione in cells, leading to the development of mutagenic 8-oxo-dG adducts in DNA. Flow cytometry revealed that DH5α and EHEC OMVs increased aneuploidy in p53 mutant Caco-2 cells, but not in p53 wild type LoVo cells. CONCLUSION We conclude that E. coli derived OMVs, whether from avirulent or pathogenic strains are potentially genotoxic.
Collapse
Affiliation(s)
- Peter C Tyrer
- Department of Surgery, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
| | - Frank A Frizelle
- Department of Surgery, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
| | - Jacqueline I Keenan
- Department of Surgery, University of Otago Christchurch, PO Box 4345, Christchurch 8140, New Zealand
| |
Collapse
|
25
|
Bielaszewska M, Schiller R, Lammers L, Bauwens A, Fruth A, Middendorf B, Schmidt MA, Tarr PI, Dobrindt U, Karch H, Mellmann A. Heteropathogenic virulence and phylogeny reveal phased pathogenic metamorphosis in Escherichia coli O2:H6. EMBO Mol Med 2014; 6:347-57. [PMID: 24413188 PMCID: PMC3958309 DOI: 10.1002/emmm.201303133] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Extraintestinal pathogenic and intestinal pathogenic (diarrheagenic) Escherichia coli differ phylogenetically and by virulence profiles. Classic theory teaches simple linear descent in this species, where non-pathogens acquire virulence traits and emerge as pathogens. However, diarrheagenic Shiga toxin-producing E. coli (STEC) O2:H6 not only possess and express virulence factors associated with diarrheagenic and uropathogenic E. coli but also cause diarrhea and urinary tract infections. These organisms are phylogenetically positioned between members of an intestinal pathogenic group (STEC) and extraintestinal pathogenic E. coli. STEC O2:H6 is, therefore, a 'heteropathogen,' and the first such hybrid virulent E. coli identified. The phylogeny of these E. coli and the repertoire of virulence traits they possess compel consideration of an alternate view of pathogen emergence, whereby one pathogroup of E. coli undergoes phased metamorphosis into another. By understanding the evolutionary mechanisms of bacterial pathogens, rational strategies for counteracting their detrimental effects on humans can be developed.
Collapse
|
26
|
Bielaszewska M, Rüter C, Kunsmann L, Greune L, Bauwens A, Zhang W, Kuczius T, Kim KS, Mellmann A, Schmidt MA, Karch H. Enterohemorrhagic Escherichia coli hemolysin employs outer membrane vesicles to target mitochondria and cause endothelial and epithelial apoptosis. PLoS Pathog 2013; 9:e1003797. [PMID: 24348251 PMCID: PMC3861543 DOI: 10.1371/journal.ppat.1003797] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 10/14/2013] [Indexed: 01/08/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) strains cause diarrhea and hemolytic uremic syndrome resulting from toxin-mediated microvascular endothelial injury. EHEC hemolysin (EHEC-Hly), a member of the RTX (repeats-in-toxin) family, is an EHEC virulence factor of increasingly recognized importance. The toxin exists as free EHEC-Hly and as EHEC-Hly associated with outer membrane vesicles (OMVs) released by EHEC during growth. Whereas the free toxin is lytic towards human endothelium, the biological effects of the OMV-associated EHEC-Hly on microvascular endothelial and intestinal epithelial cells, which are the major targets during EHEC infection, are unknown. Using microscopic, biochemical, flow cytometry and functional analyses of human brain microvascular endothelial cells (HBMEC) and Caco-2 cells we demonstrate that OMV-associated EHEC-Hly does not lyse the target cells but triggers their apoptosis. The OMV-associated toxin is internalized by HBMEC and Caco-2 cells via dynamin-dependent endocytosis of OMVs and trafficked with OMVs into endo-lysosomal compartments. Upon endosome acidification and subsequent pH drop, EHEC-Hly is separated from OMVs, escapes from the lysosomes, most probably via its pore-forming activity, and targets mitochondria. This results in decrease of the mitochondrial transmembrane potential and translocation of cytochrome c to the cytosol, indicating EHEC-Hly-mediated permeabilization of the mitochondrial membranes. Subsequent activation of caspase-9 and caspase-3 leads to apoptotic cell death as evidenced by DNA fragmentation and chromatin condensation in the intoxicated cells. The ability of OMV-associated EHEC-Hly to trigger the mitochondrial apoptotic pathway in human microvascular endothelial and intestinal epithelial cells indicates a novel mechanism of EHEC-Hly involvement in the pathogenesis of EHEC diseases. The OMV-mediated intracellular delivery represents a newly recognized mechanism for a bacterial toxin to enter host cells in order to target mitochondria. During the last 30 years, enterohemorrhagic Escherichia coli (EHEC) emerged as worldwide causes of diarrhea and hemolytic uremic syndrome, the most common cause of acute kidney failure in children. EHEC hemolysin (EHEC-Hly) is one of the toxins produced by EHEC during infection that afflict the human host. EHEC-Hly belongs to a large family of toxins, whose members typically kill target cells by inserting themselves into the cell membranes, which results in pore formation and ultimately cell lysis. Here we show that EHEC-Hly associated with outer membrane vesicles (OMVs) secreted by EHEC during growth does not lyse human microvascular endothelial and intestinal epithelial cells, which are the major targets in EHEC-mediated human diseases. Instead, the OMV-associated EHEC-Hly uses the OMVs to enter the cells and acts intracellularly. The toxin separates from its carriers in lysosomes, translocates into mitochondria and triggers apoptotic death of the target cells via the mitochondrial pathway. EHEC-Hly is the first known bacterial toxin, which enters host cells via OMVs in order to attack mitochondria. The apoptotic potential of OMV-associated EHEC-Hly indicates a novel mechanism for this toxin to cause cell death during human EHEC infections.
Collapse
Affiliation(s)
| | - Christian Rüter
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Lisa Kunsmann
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Lilo Greune
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Andreas Bauwens
- Institute of Hygiene, University of Münster, Münster, Germany
| | - Wenlan Zhang
- Institute of Hygiene, University of Münster, Münster, Germany
| | | | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | | | - M. Alexander Schmidt
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Helge Karch
- Institute of Hygiene, University of Münster, Münster, Germany
| |
Collapse
|
27
|
Liyanage NP, Dassanayake RP, Kuszynski CA, Duhamel GE. Contribution of Helicobacter hepaticus cytolethal distending toxin subunits to human epithelial cell cycle arrest and apoptotic death in vitro. Helicobacter 2013; 18:433-43. [PMID: 23895367 PMCID: PMC3808484 DOI: 10.1111/hel.12084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cytolethal distending toxin (CDT) is the only known virulence factor found in H. hepaticus, the cause of chronic typhlocolitis and hepatitis leading to colonic and hepatocellular carcinomas in mice. Interaction of the tripartite polypeptide CdtA, CdtB, and CdtC subunits produced by H. hepaticus CDT (HhepCDT) causes cell cycle arrest and apoptotic death of cultured cells; however, the contribution of individual subunit to these processes has not been investigated. MATERIALS AND METHODS The temporal relationship between cell cycle and apoptotic death of human epithelial HeLa and INT407 cells intoxicated with HhepCDT holotoxin or reconstituted recombinant HhepCDT was compared by flow cytometry. The genotoxic activity of individual and combinations of recombinant HhepCDT protein subunits or increasing concentrations of individual recombinant HhepCDT protein subunits transfected into HeLa cells was assessed at 72 hours post-treatment by flow cytometry. RESULTS Similar time course of HhepCDT-induced G2 /M cell cycle arrest and apoptotic death was found with both cell lines which reached a maximum at 72 hours. The presence of all three HhepCDT subunits was required for maximum cell cycle arrest and apoptosis of both cell lines. Transfection of HeLa cells with HhepCdtB, but not with HhepCdtA or HhepCdtC, resulted in a dose-dependent G2 /M arrest and apoptotic death. CONCLUSION All three subunits of HhepCDT are required for maximum epithelial cell cycle arrest and progression to apoptotic death, and HhepCdtB subunit alone is necessary and sufficient for epithelial cell genotoxicity.
Collapse
Affiliation(s)
- Namal P.M. Liyanage
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, U.S.A
| | - Rohana P. Dassanayake
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, U.S.A
| | - Charles A. Kuszynski
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, U.S.A
| | - Gerald E. Duhamel
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, U.S.A
| |
Collapse
|
28
|
Croxen MA, Law RJ, Scholz R, Keeney KM, Wlodarska M, Finlay BB. Recent advances in understanding enteric pathogenic Escherichia coli. Clin Microbiol Rev 2013; 26:822-80. [PMID: 24092857 PMCID: PMC3811233 DOI: 10.1128/cmr.00022-13] [Citation(s) in RCA: 831] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although Escherichia coli can be an innocuous resident of the gastrointestinal tract, it also has the pathogenic capacity to cause significant diarrheal and extraintestinal diseases. Pathogenic variants of E. coli (pathovars or pathotypes) cause much morbidity and mortality worldwide. Consequently, pathogenic E. coli is widely studied in humans, animals, food, and the environment. While there are many common features that these pathotypes employ to colonize the intestinal mucosa and cause disease, the course, onset, and complications vary significantly. Outbreaks are common in developed and developing countries, and they sometimes have fatal consequences. Many of these pathotypes are a major public health concern as they have low infectious doses and are transmitted through ubiquitous mediums, including food and water. The seriousness of pathogenic E. coli is exemplified by dedicated national and international surveillance programs that monitor and track outbreaks; unfortunately, this surveillance is often lacking in developing countries. While not all pathotypes carry the same public health profile, they all carry an enormous potential to cause disease and continue to present challenges to human health. This comprehensive review highlights recent advances in our understanding of the intestinal pathotypes of E. coli.
Collapse
|
29
|
Sequence variability of P2-like prophage genomes carrying the cytolethal distending toxin V operon in Escherichia coli O157. Appl Environ Microbiol 2013; 79:4958-64. [PMID: 23770900 DOI: 10.1128/aem.01134-13] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cytolethal distending toxins (CDT) are potent cytotoxins of several Gram-negative pathogenic bacteria, including Escherichia coli, in which five types (CDT-I to CDT-V) have been identified so far. CDT-V is frequently associated with Shiga-toxigenic E. coli (STEC), enterohemorrhagic E. coli (EHEC) O157 strains, and strains not fitting any established pathotypes. In this study, we were the first to sequence and annotate a 31.2-kb-long, noninducible P2-like prophage carrying the cdt-V operon from an stx- and eae-negative E. coli O157:H43 strain of bovine origin. The cdt-V operon is integrated in the place of the tin and old phage immunity genes (termed the TO region) of the prophage, and the prophage itself is integrated into the bacterial chromosome between the housekeeping genes cpxP and fieF. The presence of P2-like genes (n = 20) was investigated in a further five CDT-V-positive bovine E. coli O157 strains of various serotypes, three EHEC O157:NM strains, four strains expressing other variants of CDT, and eight CDT-negative strains. All but one CDT-V-positive atypical O157 strain uniformly carried all the investigated genomic regions of P2-like phages, while the EHEC O157 strains missed three regions and the CDT-V-negative strains carried only a few P2-like sequences. Our results suggest that P2-like phages play a role in the dissemination of cdt-V between E. coli O157 strains and that after integration into the bacterial chromosome, they adapted to the respective hosts and became temperate.
Collapse
|
30
|
Cadona JS, Bustamante AV, Parma AE, Lucchesi PMA, Sanso AM. Distribution of additional virulence factors related to adhesion and toxicity in Shiga toxin-producing Escherichia coli isolated from raw products in Argentina. Lett Appl Microbiol 2013; 56:449-55. [PMID: 23488818 DOI: 10.1111/lam.12067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 02/25/2013] [Accepted: 03/06/2013] [Indexed: 12/01/2022]
Abstract
UNLABELLED A total of 73 Shiga toxin-producing Escherichia coli (STEC) isolates, belonging to 25 serotypes and isolated from raw products in Argentina, were examined for the occurrence of genes responsible for bacterial adhesions to intestine, ehaA (EHEC autotransporter), lpfAO113 (long polar fimbriae), sab (STEC autotransporter [AT] contributing to biofilm formation), ecpA (E. coli common pilus), hcpA (haemorrhagic coli pilus), elfA (E. coli laminin-binding fimbriae), sfpA (sorbitol-fermenting EHEC O157 fimbriae plasmid-encoded) and of the toxigenic gene cdt-V (cytolethal distending toxin). Our study showed different adhesin profiles that are not linked to one specific serotype and that all analysed isolates possess, besides stx genes, some adherence genes. Several of the isolates contained also multiple toxin genes. The results of the present work alert the presence of genes coding for additional adhesins and cdt-V toxin in LEE-negative STEC strains that occur in foods, and this traits could increase their pathogenic potential. SIGNIFICANCE AND IMPACT OF THE STUDY Meat products are one of the main vehicles of Shiga toxin-producing E. coli, and the presence of genes coding for additional adhesins and toxins could increase their pathogenic potential. There is a need for a more detailed characterization of the strains in regard to these extra virulence factors.
Collapse
Affiliation(s)
- J S Cadona
- Laboratorio de Inmunoquímica y Biotecnología, CIVETAN-Facultad de Ciencias Veterinarias, Universidad Nacional del Centro de la Pcia. de Buenos Aires, Tandil, Argentina
| | | | | | | | | |
Collapse
|
31
|
Bauwens A, Betz J, Meisen I, Kemper B, Karch H, Müthing J. Facing glycosphingolipid-Shiga toxin interaction: dire straits for endothelial cells of the human vasculature. Cell Mol Life Sci 2013; 70:425-57. [PMID: 22766973 PMCID: PMC11113656 DOI: 10.1007/s00018-012-1060-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 05/25/2012] [Accepted: 06/14/2012] [Indexed: 12/23/2022]
Abstract
The two major Shiga toxin (Stx) types, Stx1 and Stx2, produced by enterohemorrhagic Escherichia coli (EHEC) in particular injure renal and cerebral microvascular endothelial cells after transfer from the human intestine into the circulation. Stxs are AB(5) toxins composed of an enzymatically active A subunit and the pentameric B subunit, which preferentially binds to the glycosphingolipid globotriaosylceramide (Gb3Cer/CD77). This review summarizes the current knowledge on Stx-caused cellular injury and the structural diversity of Stx receptors as well as the initial molecular interaction of Stxs with the human endothelium of different vascular beds. The varying lipoforms of Stx receptors and their spatial organization in lipid rafts suggest a central role in different modes of receptor-mediated endocytosis and intracellular destiny of the toxins. The design and development of tailored Stx neutralizers targeting the oligosaccharide-toxin recognition event has become a very real prospect to ameliorate or prevent life-threatening renal and neurological complications.
Collapse
Affiliation(s)
- Andreas Bauwens
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Josefine Betz
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Iris Meisen
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
- Interdisciplinary Center for Clinical Research, University of Münster, Domagkstr. 3, 48149 Münster, Germany
| | - Björn Kemper
- Center for Biomedical Optics and Photonics, University of Münster, Robert-Koch-Str. 45, 48149 Münster, Germany
| | - Helge Karch
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Str. 41, 48149 Münster, Germany
- Interdisciplinary Center for Clinical Research, University of Münster, Domagkstr. 3, 48149 Münster, Germany
| |
Collapse
|
32
|
Karch H, Müthing J, Dobrindt U, Mellmann A. [Evolution and infection biology of hemolytic-uremic syndrome (HUS) associated E. coli (HUSEC)]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2013; 56:8-14. [PMID: 23275950 DOI: 10.1007/s00103-012-1586-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC), which cause hemolytic-uremic syndrome (HUS), are designated as HUSEC. Their exceptional genome variability driven by evolutionary diversification permits fast adaptation to changed environmental conditions. The HUSEC collection (http://www.ehec.org), which has been established at the Institute for Hygiene in Münster, contains 42 EHEC reference strains (HUSEC001-HUSEC042). It represents a unique repository collection of pathogens and is extremely helpful for the analysis of evolutionary changes and fixed properties in the STEC that cause the most severe host injury. Such genomic attributes include slowly evolving loci, mobile genetic elements that often encode virulence factors and are assimilated via horizontal gene transfer. Current evolutionary models indicate that numerous outbreak strains evolved recently and that highly pathogenic HUSEC descend from less pathogenic progenitors. However, additional data suggest that HUSEC have small effective population sizes. The HUSEC collection is also a valuable resource with which to study important non-Shiga toxin virulence factors.
Collapse
Affiliation(s)
- H Karch
- Institut für Hygiene und Nationales Konsiliarlaboratorium für Hämolytisch-Urämisches Syndrom, Universitätsklinikum Münster, Robert-Koch-Str. 41, 48149, Münster, Deutschland.
| | | | | | | |
Collapse
|
33
|
Fedor Y, Vignard J, Nicolau-Travers ML, Boutet-Robinet E, Watrin C, Salles B, Mirey G. From single-strand breaks to double-strand breaks during S-phase: a new mode of action of theEscherichia coli Cytolethal Distending Toxin. Cell Microbiol 2012; 15:1-15. [DOI: 10.1111/cmi.12028] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Y. Fedor
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - J. Vignard
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - M.-L. Nicolau-Travers
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - E. Boutet-Robinet
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - C. Watrin
- INRA; USC 1360; F-31300 Toulouse France
- Université de Toulouse; UPS; Centre de Physiopathologie de Toulouse Purpan (CPTP); F-31400 Toulouse France
- Inserm; UMR1043; F-31300 Toulouse France
- CNRS; UMR5282; F-31400 Toulouse France
| | - B. Salles
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| | - G. Mirey
- INRA; UMR1331; Toxalim; Research Centre in Food Toxicology; F-31027 Toulouse France
- Université de Toulouse; UPS, UMR1331, Toxalim; F-31062 Toulouse France
| |
Collapse
|
34
|
Molecular characterization reveals three distinct clonal groups among clinical shiga toxin-producing Escherichia coli strains of serogroup O103. J Clin Microbiol 2012; 50:2894-900. [PMID: 22718945 DOI: 10.1128/jcm.00789-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) is one of the most important groups of food-borne pathogens, and STEC strains belonging to the serotype O103:H2 can cause diarrhea, hemorrhagic colitis, and hemolytic-uremic syndrome in humans. STEC O103:non-H2 strains are also sometimes isolated from human patients, but their genetic characteristics and role in significant human enteric disease are not yet understood. Here, we investigated 17 STEC O103:non-H2 strains, including O103:H11, O103:H25, O103:HUT (UT [untypeable]), and O103:H- (nonmotile) isolated in Japan, and their characteristics were compared to those of STEC O103:H2 and other serotype STEC strains. Sequence analyses of fliC and eae genes revealed that strains possessed any of the following combinations: fliC-H2/eae-epsilon, fliC-H11/eae-beta1, and fliC-H25/eae-theta, where fliC-H2, -H11, and -H25 indicate fliC genes encoding H2, H11, and H25 flagella antigens, respectively, and eae-epsilon, -beta1, and -theta indicate eae genes encoding epsilon, beta1, and theta subclass intimins, respectively. Phylogenetic analysis based on the sequences of seven housekeeping genes demonstrated that the O103:H11/[fliC-H11] and O103:H25/[fliC-H25] strains formed two distinct groups, different from that of the O103:H2/[fliC-H2] strains. Interestingly, a group consisting of O103:H11 strains was closely related to STEC O26:H11, which is recognized as a most important non-O157 serotype, suggesting that the STEC O103:H11 and STEC O26:H11 clones evolved from a common ancestor. The multiplex PCR system for the rapid typing of STEC O103 strains described in the present study may aid clinical and epidemiological studies of the STEC O103:H2, O103:H11, and O103:H25 groups. In addition, our data provide further insights into the high variability of STEC stains with emerging new serotypes.
Collapse
|
35
|
Weiss A, Schmidt H, Stöber H. Mechanisms of enterohemorrhagic Escherichia coli spread along the food-chain and precautionary measures. J Verbrauch Lebensm 2011. [DOI: 10.1007/s00003-011-0736-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
36
|
Londoño D, Carvajal J, Strle K, Kim KS, Cadavid D. IL-10 Prevents apoptosis of brain endothelium during bacteremia. THE JOURNAL OF IMMUNOLOGY 2011; 186:7176-86. [PMID: 21602495 DOI: 10.4049/jimmunol.1100060] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-10-deficient mice infected with the relapsing fever bacterium Borrelia turicatae rapidly succumb to a brain hemorrhage if they are unable to clear peak bacteremia. In this study, we investigated the protective role of IL-10 during relapsing-remitting bacteremia and explored the molecular events involved in the protection of brain endothelium by IL-10. Brain endothelial injury was measured with cytotoxicity and diverse apoptotic assays, whereas the signaling pathway analysis was done by quantitative PCR array. The results showed that severe endothelial cell injury leading to hemorrhage in the brain and other organs occurred in IL-10-deficient mice during relapsing-remitting infection. Human brain microvascular endothelial cells (HBMEC) produced abundant proinflammatory mediators upon exposure to whole bacteria or purified bacterial lipoprotein but did not produce any detectable IL-10. Whole bacteria and purified outer membrane lipoprotein rapidly killed HBMEC by apoptosis in a time- and concentration-dependent manner. Exogenous IL-10 protected HBMEC from apoptosis. HBMEC apoptosis during exposure to a low number of bacteria was associated with downregulation of TNF and TNFAIP3 and upregulation of BAX. In contrast, HBMEC apoptosis during exposure to high concentrations of purified outer membrane lipoprotein was associated with marked upregulation of FAS, FAS ligand, and the adaptor molecules RIPK1 and CFLAR. Exogenous IL-10 reversed all the apoptotic signaling changes induced by whole bacteria or its purified lipoprotein. The results indicate that prominent brain endothelial cell apoptosis occurs during relapsing-remitting bacteremia in the absence of IL-10 and point to a prominent role for bacterial lipoprotein-mediated activation of FAS and caspase-3 in this process.
Collapse
Affiliation(s)
- Diana Londoño
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02144, USA
| | | | | | | | | |
Collapse
|
37
|
Jinadasa RN, Bloom SE, Weiss RS, Duhamel GE. Cytolethal distending toxin: a conserved bacterial genotoxin that blocks cell cycle progression, leading to apoptosis of a broad range of mammalian cell lineages. MICROBIOLOGY-SGM 2011; 157:1851-1875. [PMID: 21565933 DOI: 10.1099/mic.0.049536-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cytolethal distending toxin (CDT) is a heterotrimeric AB-type genotoxin produced by several clinically important Gram-negative mucocutaneous bacterial pathogens. Irrespective of the bacterial species of origin, CDT causes characteristic and irreversible cell cycle arrest and apoptosis in a broad range of cultured mammalian cell lineages. The active subunit CdtB has structural homology with the phosphodiesterase family of enzymes including mammalian DNase I, and alone is necessary and sufficient to account for cellular toxicity. Indeed, mammalian cells treated with CDT initiate a DNA damage response similar to that elicited by ionizing radiation-induced DNA double strand breaks resulting in cell cycle arrest and apoptosis. The mechanism of CDT-induced apoptosis remains incompletely understood, but appears to involve both p53-dependent and -independent pathways. While epithelial, endothelial and fibroblast cell lines respond to CDT by undergoing arrest of cell cycle progression resulting in nuclear and cytoplasmic distension that precedes apoptotic cell death, cells of haematopoietic origin display rapid apoptosis following a brief period of cell cycle arrest. In this review, the ecology of pathogens producing CDT, the molecular biology of bacterial CDT and the molecular mechanisms of CDT-induced cytotoxicity are critically appraised. Understanding the contribution of a broadly conserved bacterial genotoxin that blocks progression of the mammalian cell cycle, ultimately causing cell death, should assist with elucidating disease mechanisms for these important pathogens.
Collapse
Affiliation(s)
- Rasika N Jinadasa
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Stephen E Bloom
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Robert S Weiss
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Gerald E Duhamel
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
38
|
Brockmeyer J, Aldick T, Soltwisch J, Zhang W, Tarr PI, Weiss A, Dreisewerd K, Müthing J, Bielaszewska M, Karch H. Enterohaemorrhagic Escherichia coli haemolysin is cleaved and inactivated by serine protease EspPα. Environ Microbiol 2011; 13:1327-41. [PMID: 21352460 PMCID: PMC3472028 DOI: 10.1111/j.1462-2920.2011.02431.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The haemolysin from enterohaemorrhagic Escherichia coli (EHEC-Hly) and the serine protease EspPα are putative virulence factors of EHEC. We investigated the interplay between these secreted factors and demonstrate that EspPα cleaves the 107 kDa large EHEC-Hly. Degradation was observed when purified EspPα was added to a growing culture of an EHEC-Hly-expressing strain, with isolated proteins and with coexpressing strains, and was independent of the EHEC serotype. EHEC-Hly breakdown occurred as a multistage process with the formation of characteristic fragments with relative molecular masses of ∼82 kDa and/or ∼84 kDa and ∼34 kDa. The initial cleavage occurred in the N-terminal hydrophobic domain of EHEC-Hly between Leu235 and Ser236 and abolished its haemolytic activity. In a cellular infection system, the cytolytic potential of EHEC-Hly-secreting recombinant strains was abolished when EspPα was coexpressed. EHEC in contact with human intestinal epithelial cells simultaneously upregulated their EHEC-Hly and EspP indicating that both molecules might interact under physiological conditions. We propose the concept of bacterial effector molecule interference (BEMI), reflecting the concerted interplay of virulence factors. Interference between effector molecules might be an additional way to regulate virulence functions and increases the complexity of monomolecular phenotypes.
Collapse
Affiliation(s)
- Jens Brockmeyer
- Institute of Food Chemistry, University of Münster, Corrensstrasse 45, Münster, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Recently, an unprecedented effort has been directed at understanding the interplay between chronic inflammation and development of cancer, with the case of inflammatory bowel disease (IBD)-associated colorectal cancer at the forefront of this research endeavor. The last decade has been particularly fertile, with the discovery of numerous innovative paradigms linking various inflammatory, proliferative, and innate and adaptive immune signaling pathways to the development of colorectal cancer. Because of the preponderant role of the intestinal microbiota in the initiation and progression of IBD, recent efforts have been directed at understanding the relationship between bacteria and colorectal cancer. The microbiota and its collective genome, the microbiome, form a diverse and complex ecological community that profoundly impacts intestinal homeostasis and disease states. This review will discuss the differential influence of the microbiota on the development of IBD-associated colorectal cancer and highlight the role of innate immune sensor-dependent as well as -independent mechanisms in this pathology.
Collapse
Affiliation(s)
- Janelle C Arthur
- Department of Medicine and the Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina 27599-7080, USA
| | | |
Collapse
|
40
|
Galli L, Miliwebsky E, Irino K, Leotta G, Rivas M. Virulence profile comparison between LEE-negative Shiga toxin-producing Escherichia coli (STEC) strains isolated from cattle and humans. Vet Microbiol 2010; 143:307-13. [DOI: 10.1016/j.vetmic.2009.11.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 11/17/2009] [Accepted: 11/20/2009] [Indexed: 10/20/2022]
|
41
|
Liyanage NPM, Manthey KC, Dassanayake RP, Kuszynski CA, Oakley GG, Duhamel GE. Helicobacter hepaticus cytolethal distending toxin causes cell death in intestinal epithelial cells via mitochondrial apoptotic pathway. Helicobacter 2010; 15:98-107. [PMID: 20402812 DOI: 10.1111/j.1523-5378.2010.00749.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Helicobacter hepaticus, the prototype for enterohepatic Helicobacter species, colonizes the lower intestinal and hepatobiliary tracts of mice and causes typhlocolitis, hepatitis, and hepatocellular carcinoma in susceptible mouse strains. Cytolethal distending toxin (CDT) is the only known virulence factor found in H. hepaticus. CDT of several Gram-negative bacteria is associated with double-stranded DNA breaks resulting in cell cycle arrest and death of a wide range of eukaryotic cells in vitro. We previously observed H. hepaticus CDT (HhCDT) mediated apoptosis in INT407 cells. However, the exact mechanism for the induction of the apoptotic pathway by HhCDT is unknown. The objective of this study was to identify the apoptotic signaling pathway induced by HhCDT in INT407 cells. MATERIALS AND METHODS INT407 cells were incubated with or without recombinant HhCDT for 0-72 hours. H2AX phosphorylation and apoptotic parameters were analyzed. RESULTS H2AX was phosphorylated 24 hours postexposure to HhCDT. Expression of pro-apoptotic Bax protein was upregulated after 24 hours, while Bcl(2) expression decreased. Cytochrome c was released from mitochondria after 12-24 hours of exposure. Concurrently, caspase 3/7 and 9 were activated. However, pretreatment of INT407 cells with caspase inhibitor (Z-VAD-FMK) inhibited the activation of caspase 3/7 and 9. Significant activity of caspase 8 was not observed in toxin treated cells. Activation of caspase 3/7 and caspase 9 confirms the involvement of the mitochondrial apoptotic pathway in HhCDT-treated cells. CONCLUSION These findings show, for the first time, the ability of HhCDT to induce apoptosis via the mitochondrial pathway.
Collapse
Affiliation(s)
- Namal P M Liyanage
- School of Veterinary and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | | | | | | | | |
Collapse
|
42
|
Kalischuk LD, Buret AG. A role for Campylobacter jejuni-induced enteritis in inflammatory bowel disease? Am J Physiol Gastrointest Liver Physiol 2010; 298:G1-9. [PMID: 19875702 DOI: 10.1152/ajpgi.00193.2009] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The inflammatory bowel diseases (IBD), Crohn's disease and ulcerative colitis, are T cell-mediated diseases that are characterized by chronic, relapsing inflammation of the intestinal tract. The pathogenesis of IBD involves the complex interaction between the intestinal microflora, host genetic and immune factors, and environmental stimuli. Epidemiological analyses have implicated acute bacterial enteritis as one of the factors that may incite or exacerbate IBD in susceptible individuals. In this review, we examine how interactions between the common enteric pathogen Campylobacter jejuni (C. jejuni), the host intestinal epithelium, and resident intestinal microflora may contribute to the pathogenesis of IBD. Recent experimental evidence indicates that C. jejuni may permit the translocation of normal, noninvasive microflora via novel processes that implicate epithelial lipid rafts. This breach in intestinal barrier function may, in turn, prime the intestine for chronic inflammatory responses in susceptible individuals. Insights into the interactions between enteric pathogens, the host epithelia, and intestinal microflora will improve our understanding of disease processes that may initiate and/or exacerbate intestinal inflammation in patients with IBD and provide impetus for the development of new therapeutic approaches for the treatment of IBD.
Collapse
|
43
|
Molecular analysis of virulence profiles and Shiga toxin genes in food-borne Shiga toxin-producing Escherichia coli. Appl Environ Microbiol 2009; 75:6187-97. [PMID: 19684176 DOI: 10.1128/aem.00874-09] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In this study, 75 Shiga toxin (Stx)-producing Escherichia coli (STEC) strains originating from foods (n = 73) and drinking water (n = 2) were analyzed for their stx genotype, as well as for further chromosome-, phage-, and plasmid-encoded virulence factors. A broad spectrum of stx genes was detected. Fifty-three strains (70.7%) contained stx(2) or stx(2) variants, including stx(2d), mucus-activatable stx(2d), stx(2e), and stx(2g). Seven strains (9.3%) harbored stx(1) or stx(1c), and 15 strains (20.0%) carried both stx(2) and/or stx(2) variants and stx(1) or stx(1c). Beside stx, the most abundant accessory virulence markers in STEC food isolates were iha (57.3%), ehxA (40.0%), espP (28.0%), and subAB (25.3%). Only four strains were eae positive; three of these belonged to the serogroups O26, O103, and O157 and contained a typical enterohemorrhagic E. coli virulence spectrum. The results of this study show that a number of STEC strains that occur in foods appear to be pathogenic for humans, based on their virulence profiles. Analysis of stx subtypes and detection of additional virulence factors in eae-negative strains may help to better assess the risk of such strains for causing human infection.
Collapse
|
44
|
Brockmeyer J, Spelten S, Kuczius T, Bielaszewska M, Karch H. Structure and function relationship of the autotransport and proteolytic activity of EspP from Shiga toxin-producing Escherichia coli. PLoS One 2009; 4:e6100. [PMID: 19568421 PMCID: PMC2700255 DOI: 10.1371/journal.pone.0006100] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 05/22/2009] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The serine protease autotransporter EspP is a proposed virulence factor of Shiga toxin-producing Escherichia coli (STEC). We recently distinguished four EspP subtypes (EspPalpha, EspPbeta, EspPgamma, and EspPdelta), which display large differences in transport and proteolytic activities and differ widely concerning their distribution within the STEC population. The mechanisms underlying these functional variations in EspP subtypes are, however, unknown. METHODOLOGY/PRINCIPAL FINDINGS The structural basis of proteolytic and autotransport activity was investigated using transposon-based linker scanning mutagenesis, site-directed mutagenesis and structure-function analysis derived from homology modelling of the EspP passenger domain. Transposon mutagenesis of the passenger domain inactivated autotransport when pentapeptide linker insertions occurred in regions essential for overall correct folding or in a loop protruding from the beta-helical core. Loss of proteolytic function was limited to mutations in Domain 1 in the N-terminal third of the EspP passenger. Site-directed mutagenesis demonstrated that His(127), Asp(156) and Ser(263) in Domain 1 form the catalytic triad of EspP. CONCLUSIONS/SIGNIFICANCE Our data indicate that in EspP i) the correct formation of the tertiary structure of the passenger domain is essential for efficient autotransport, and ii) an elastase-like serine protease domain in the N-terminal Domain 1 is responsible for the proteolytic phenotype. Lack of stabilizing interactions of Domain 1 with the core structure of the passenger domain ablates proteolytic activity in subtypes EspPbeta and EspPdelta.
Collapse
Affiliation(s)
- Jens Brockmeyer
- Institute for Hygiene and the National Consulting Laboratory on Hemolytic Uremic Syndrome, University of Münster, Münster, Germany.
| | | | | | | | | |
Collapse
|
45
|
Shiga toxin, cytolethal distending toxin, and hemolysin repertoires in clinical Escherichia coli O91 isolates. J Clin Microbiol 2009; 47:2061-6. [PMID: 19403777 DOI: 10.1128/jcm.00201-09] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shiga toxin (Stx)-producing Escherichia coli (STEC) strains of serogroup O91 are the most common human pathogenic eae-negative STEC strains. To facilitate diagnosis and subtyping of these pathogens, we genotypically and phenotypically characterized 100 clinical STEC O91 isolates. Motile strains expressed flagellar antigens H8 (1 strain), H10 (2 strains), H14 (52 strains), and H21 (20 strains) or were H nontypeable (Hnt) (10 strains); 15 strains were nonmotile. All nonmotile and Hnt strains possessed the fliC gene encoding the flagellin subunit of the H14 antigen (fliC(H14)). Most STEC O91 strains possessed enterohemorrhagic E. coli hlyA and expressed an enterohemolytic phenotype. Among seven stx alleles identified, stx(2dact), encoding mucus- and elastase-activatable Stx2d, was present solely in STEC O91:H21, whereas most strains of the other serotypes possessed stx(1). Moreover, only STEC O91:H21 possessed the cdt-V cluster, encoding cytolethal distending toxin V; the toxin was regularly expressed and was lethal to human microvascular endothelial cells. Infection with STEC O91:H21 was associated with hemolytic-uremic syndrome (P = 0.0015), whereas strains of the other serotypes originated mostly in patients with nonbloody diarrhea. We conclude that STEC O91 clinical isolates belong to at least four lineages that differ by H antigens/fliC types, stx genotypes, and non-stx putative virulence factors, with accumulation of virulence determinants in the O91:H21 lineage. Isolation of STEC O91 from patients' stools on enterohemolysin agar and the rapid initial subtyping of these isolates using fliC genotyping facilitate the identification of these emerging pathogens in clinical and epidemiological studies and enable prediction of the risk of a severe clinical outcome.
Collapse
|
46
|
Aldick T, Bielaszewska M, Uhlin BE, Humpf HU, Wai SN, Karch H. Vesicular stabilization and activity augmentation of enterohaemorrhagicEscherichia colihaemolysin. Mol Microbiol 2009; 71:1496-508. [DOI: 10.1111/j.1365-2958.2009.06618.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
47
|
Boesze-Battaglia K, Brown A, Walker L, Besack D, Zekavat A, Wrenn S, Krummenacher C, Shenker BJ. Cytolethal distending toxin-induced cell cycle arrest of lymphocytes is dependent upon recognition and binding to cholesterol. J Biol Chem 2009; 284:10650-8. [PMID: 19240023 DOI: 10.1074/jbc.m809094200] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Induction of cell cycle arrest in lymphocytes after exposure to the Aggregatibacter actinomycetemcomitans cytolethal distending toxin (Cdt) is dependent upon the integrity of lipid membrane microdomains. In this study we further demonstrate that the association of Cdt with lymphocyte plasma membranes is dependent upon binding to cholesterol. Depletion of cholesterol resulted in reduced toxin binding, whereas repletion of cholesterol-depleted cells restored binding. We employed fluorescence resonance energy transfer and surface plasmon resonance to demonstrate that toxin association with model membranes is dependent upon the concentration of cholesterol; moreover, these interactions were cholesterol-specific as the toxin failed to interact with model membranes containing stigmasterol, ergosterol, or lanosterol. Further analysis of the toxin indicated that the CdtC subunit contains a cholesterol recognition/interaction amino acid consensus (CRAC) region. Mutation of the CRAC site resulted in decreased binding of the holotoxin to cholesterol-containing model membranes as well as to the surface of Jurkat cells. The mutant toxin also exhibited reduced capacity for intracellular transfer of the active toxin subunit, CdtB, as well as reduced toxicity. Collectively, these observations indicate that membrane cholesterol serves as an essential ligand for Cdt and that this association can be blocked by either depleting membranes of cholesterol or mutation of the CRAC site.
Collapse
Affiliation(s)
- Kathleen Boesze-Battaglia
- Departments of Biochemistry and Pathology, University of Pennsylvania School of Dental Medicine, Pennsylvania 19104-6030, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Buvens G, Piérard D, Hachimi-Idrissi S, Lauwers S. First sorbitol-fermenting Verocytotoxin-producing Escherichia coli O157: H- isolated in Belgium. Acta Clin Belg 2009; 64:59-64. [PMID: 19317243 DOI: 10.1179/acb.2009.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We report a case of haemolytic uraemic syndrome (HUS) following an infection with a sorbitol-fermenting Verocytotoxin-producing Escherichia coli (VTEC) O157:H- in a toddler living in the province of East Flanders, Belgium. The patient presented with haemolytic anaemia, haematuria, proteinuria, renal insufficiency, and thrombocytopaenia leading to the diagnosis of HUS. Risk factors for VTEC infection, such as consuming undercooked food of bovine origin and direct contact with farm animals were absent. Also, neither travelling nor contact with travellers were reported. The patient recovered after perfusion with fresh frozen plasma and blood transfusion, and dialysis was not required. This is the first isolation of a sorbitol-fermenting VTEC O157:H- in Belgium. Future research is needed to reveal epidemiologic aspects, such as the main reservoir and transmission routes of this pathogenic E. coli serotype, which has caused outbreaks of HUS in Germany and Scotland.
Collapse
Affiliation(s)
- G Buvens
- Department of Microbiology, (Belgian VTEC Reference Laboratory), Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | | | | |
Collapse
|
49
|
Enterotoxigenic Escherichia coli modulates host intestinal cell membrane asymmetry and metabolic activity. Infect Immun 2008; 77:341-7. [PMID: 18936181 DOI: 10.1128/iai.01097-08] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a common cause of travelers' and postweaning diarrhea in humans and swine, respectively. The extent to which ETEC damages host cells is unclear. Experiments are presented that probe the ability of porcine ETEC isolates to induce apoptosis and cell death in porcine intestinal epithelial cells. Quantification of host phosphatidylserine exposure following ETEC infection suggested that ETEC induced changes in plasma membrane asymmetry, independent of the expression of the heat-labile enterotoxin. Significant host cell death was not observed. ETEC infection also caused a drastic inhibition of host esterase activity, as measured by calcein fluorescence. While ETEC infection resulted in activation of host caspase 3, terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling of DNA double-strand breakage, indicative of late stages of apoptosis, was not observed. Camptothecin-induced apoptosis markedly increased subsequent ETEC adherence. Transfer of cell-free supernatants from apoptotic cells to bacterial inocula prior to infection of naïve cells increased the transcriptional activity of the regulatory region upstream of the K88ac operon and promoted subsequent adherence to host cells.
Collapse
|
50
|
Scheiring J, Andreoli SP, Zimmerhackl LB. Treatment and outcome of Shiga-toxin-associated hemolytic uremic syndrome (HUS). Pediatr Nephrol 2008; 23:1749-60. [PMID: 18704506 PMCID: PMC6901419 DOI: 10.1007/s00467-008-0935-6] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 06/03/2008] [Accepted: 06/09/2008] [Indexed: 12/28/2022]
Abstract
Hemolytic uremic syndrome (HUS) is the most common cause of acute renal failure in childhood and the reason for chronic renal replacement therapy. It leads to significant morbidity and mortality during the acute phase. In addition to acute morbidity and mortality, long-term renal and extrarenal complications can occur in a substantial number of children years after the acute episode of HUS. The most common infectious agents causing HUS are enterohemorrhagic Escherichia coli (EHEC)-producing Shiga toxin (and belonging to the serotype O157:H7) and several non-O157:H7 serotypes. D(+) HUS is an acute disease characterized by prodromal diarrhea followed by acute renal failure. The classic clinical features of HUS include the triad of microangiopathic hemolytic anemia, thrombocytopenia, and acute renal failure. HUS mortality is reported to be between 3% and 5%, and death due to HUS is nearly always associated with severe extrarenal disease, including severe central nervous system (CNS) involvement. Approximately two thirds of children with HUS require dialysis therapy, and about one third have milder renal involvement without the need for dialysis therapy. General management of acute renal failure includes appropriate fluid and electrolyte management, antihypertensive therapy if necessary, and initiation of renal replacement therapy when appropriate. The prognosis of HUS depends on several contributing factors. In general "classic" HUS, induced by EHEC, has an overall better outcome. Totally different is the prognosis in patients with atypical and particularly recurrent HUS. However, patients with severe disease should be screened for genetic disorders of the complement system or other underlying diseases.
Collapse
Affiliation(s)
- Johanna Scheiring
- Department of Pediatrics I, Medical University Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| | | | - Lothar Bernd Zimmerhackl
- Department of Pediatrics I, Medical University Innsbruck, Anichstr. 35, A-6020 Innsbruck, Austria
| |
Collapse
|