1
|
Li F, Li M, Nie L, Zuo J, Fan W, Lian L, Hu J, Chen S, Jiang W, Han X, Wang H. Molecular Epidemiology and Antibiotic Resistance Associated with Avian Pathogenic Escherichia coli in Shanxi Province, China, from 2021 to 2023. Microorganisms 2025; 13:541. [PMID: 40142434 PMCID: PMC11946381 DOI: 10.3390/microorganisms13030541] [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: 01/25/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Avian Pathogenic Escherichia coli (APEC) constitutes a major etiological agent of avian colibacillosis, which significantly hinders the development of the poultry industry. Conducting molecular epidemiological studies of APEC plays a crucial role in its prevention and control. This study aims to elucidate the molecular epidemiological characteristics of Avian Pathogenic Escherichia coli in Shanxi Province. In this study, 135 APEC strains were isolated and identified from 150 liver samples of diseased and deceased chickens exhibiting clinical symptoms, which were collected from farms in Shanxi Province between 2021 and 2023. The isolates were then analyzed for phylogenetic clustering, drug resistance, resistance genes, virulence genes, and biofilm formation capabilities. The results revealed that the proportions of the A, B1, B2, and D evolutionary subgroups were 26.67%, 32.59%, 17.78%, and 15.56%, respectively. The drug resistance testing results indicated that 92% of the isolates exhibited resistance to cotrimoxazole, kanamycin, chloramphenicol, amoxicillin, tetracycline, and other antibiotics. In contrast, 95% of the strains were sensitive to ofloxacin, amikacin, and ceftazidime. The most prevalent resistance genes included tetracycline-related (tetA) at 88.15%, followed by beta-lactam-related (bla-TEM) at 85.19%, and peptide-related (mcr1) at 12.59%. The virulence gene analysis revealed that ibeB, ompA, iucD, and mat were present in more than 90% of the isolates. The results revealed that 110 strains were biofilm-positive, corresponding to a detection rate of 81.48%. No significant correlation was found between the drug resistance genes, virulence genes, and the drug resistance phenotype. A moderate negative correlation was observed between the adhesion-related gene tsh and biofilm formation ability (r = -0.38). This study provides valuable insights into the prevention and control of avian colibacillosis in Shanxi Province.
Collapse
Affiliation(s)
- Fangfang Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Mengya Li
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Lianhua Nie
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Jiakun Zuo
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Wenyan Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Liyan Lian
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Jiangang Hu
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Shuming Chen
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
| | - Wei Jiang
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Xiangan Han
- Shanghai Veterinary Research Institute, The Chinese Academy of Agricultural Sciences (CAAS), 518 Ziyue Road, Shanghai 200241, China; (J.Z.); (L.L.); (J.H.); (W.J.)
| | - Haidong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China; (F.L.); (M.L.); (L.N.); (W.F.); (S.C.)
| |
Collapse
|
2
|
Brek T, Gohal GA, Yasir M, Azhar EI, Al-Zahrani IA. Meningitis and Bacteremia by Unusual Serotype of Salmonella enterica Strain: A Whole Genome Analysis. Interdiscip Perspect Infect Dis 2024; 2024:3554734. [PMID: 38558876 PMCID: PMC10980553 DOI: 10.1155/2024/3554734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/27/2023] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Background Although meningitis caused by Salmonella species is relatively rare and accounts for <1% of the confirmed cases in neonates, it is associated with case complications and fatality rates up to 50-70% when compared to other forms of Gram-negative bacilli meningitis. Objectives We conducted an investigation into the first reported case of neonatal meningitis caused by nontyphoidal S. enterica in Jazan, a region in the southwestern part of Saudi Arabia. Methods CSF and blood culture were collected from a female neonate patient to confirm the presence of bacterial meningitis. WGS was conducted to find out the comprehensive genomic characterization of S. enterica isolate. Results A 3-week-old infant was admitted to a local hospital with fever, poor feeding, and hypoactivity. She was diagnosed with Salmonella meningitis and bacteremia caused by S. enterica, which was sensitive to all antimicrobials tested. WGS revealed the specific strain to be S. enterica serotype Johannesburg JZ01, belonging to ST515 and cgMLST 304742. Conclusions We presented a genomic report of rare case of NTS meningitis in an infant who is living in a rural town in Jazan region, Saudi Arabia. Further research is required to understand the impact of host genetic factors on invasive nontyphoidal Salmonella infection.
Collapse
Affiliation(s)
- Thamer Brek
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Public Health Laboratory, The Regional Laboratory and the Central Blood Bank, Jazan Health Directorate, Jazan, Saudi Arabia
| | - Gassem A. Gohal
- Department of Pediatrics, Faculty of Medicine, Jazan University, Jazan, Saudi Arabia
| | - Muhammad Yasir
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Esam I. Azhar
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibrahim A. Al-Zahrani
- Medical Laboratory Sciences Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit-Biosafety Level-3, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Rauti R, Navok S, Biran D, Tadmor K, Leichtmann-Bardoogo Y, Ron EZ, Maoz BM. Insight on Bacterial Newborn Meningitis Using a Neurovascular-Unit-on-a-Chip. Microbiol Spectr 2023; 11:e0123323. [PMID: 37222614 PMCID: PMC10269748 DOI: 10.1128/spectrum.01233-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Understanding the pathogenesis of bacterial infections is critical for combatting them. For some infections, animal models are inadequate and functional genomic studies are not possible. One example is bacterial meningitis, a life-threatening infection with high mortality and morbidity. Here, we used the newly developed, physiologically relevant, organ-on-a-chip platform integrating the endothelium with neurons, closely mimicking in vivo conditions. Using high-magnification microscopy, permeability measurements, electrophysiological recordings, and immunofluorescence staining, we studied the dynamic by which the pathogens cross the blood-brain barrier and damage the neurons. Our work opens up possibilities for performing large-scale screens with bacterial mutant libraries for identifying the virulence genes involved in meningitis and determining the role of these genes, including various capsule types, in the infection process. These data are essential for understanding and therapy of bacterial meningitis. Moreover, our system offers possibilities for the study of additional infections-bacterial, fungal, and viral. IMPORTANCE The interactions of newborn meningitis (NBM) with the neurovascular unit are very complex and are hard to study. This work presents a new platform to study NBM in a system that enables monitoring of multicellular interactions and identifies processes that were not observed before.
Collapse
Affiliation(s)
- Rossana Rauti
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sharon Navok
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Dvora Biran
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Keshet Tadmor
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | | | - Eliora Z. Ron
- The Shmunis School of Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
| | - Ben M. Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Yun J, Mao L, Li J, Hao F, Yang L, Zhang W, Sun M, Liu M, Wang S, Li W. Molecular characterization and antimicrobial resistance profile of pathogenic Escherichia coli from goats with respiratory disease in eastern China. Microb Pathog 2022; 166:105501. [DOI: 10.1016/j.micpath.2022.105501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/15/2022] [Accepted: 03/21/2022] [Indexed: 11/16/2022]
|
5
|
The MCR-3 inside linker appears as a facilitator of colistin resistance. Cell Rep 2021; 35:109135. [PMID: 34010644 DOI: 10.1016/j.celrep.2021.109135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 12/23/2020] [Accepted: 04/23/2021] [Indexed: 01/08/2023] Open
Abstract
An evolving family of mobile colistin resistance (MCR) enzymes is threatening public health. However, the molecular mechanism by which the MCR enzyme as a rare member of lipid A-phosphoethanolamine (PEA) transferases gains the ability to confer phenotypic colistin resistance remains enigmatic. Here, we report an unusual example that genetic duplication and amplification produce a functional variant (Ah762) of MCR-3 in certain Aeromonas species. The lipid A-binding cavity of Ah762 is functionally defined. Intriguingly, we locate a hinge linker of Ah762 (termed Linker 59) that determines the MCR. Genetic and biochemical characterization reveals that Linker 59 behaves as a facilitator to render inactive MCR variants to regain the ability of colistin resistance. Along with molecular dynamics (MD) simulation, isothermal titration calorimetry (ITC) suggests that this facilitator guarantees the formation of substrate phosphatidylethanolamine (PE)-accessible pocket within MCR-3-like enzymes. Therefore, our finding defines an MCR-3 inside facilitator for colistin resistance.
Collapse
|
6
|
Desvaux M, Dalmasso G, Beyrouthy R, Barnich N, Delmas J, Bonnet R. Pathogenicity Factors of Genomic Islands in Intestinal and Extraintestinal Escherichia coli. Front Microbiol 2020; 11:2065. [PMID: 33101219 PMCID: PMC7545054 DOI: 10.3389/fmicb.2020.02065] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli is a versatile bacterial species that includes both harmless commensal strains and pathogenic strains found in the gastrointestinal tract in humans and warm-blooded animals. The growing amount of DNA sequence information generated in the era of "genomics" has helped to increase our understanding of the factors and mechanisms involved in the diversification of this bacterial species. The pathogenic side of E. coli that is afforded through horizontal transfers of genes encoding virulence factors enables this bacterium to become a highly diverse and adapted pathogen that is responsible for intestinal or extraintestinal diseases in humans and animals. Many of the accessory genes acquired by horizontal transfers form syntenic blocks and are recognized as genomic islands (GIs). These genomic regions contribute to the rapid evolution, diversification and adaptation of E. coli variants because they are frequently subject to rearrangements, excision and transfer, as well as to further acquisition of additional DNA. Here, we review a subgroup of GIs from E. coli termed pathogenicity islands (PAIs), a concept defined in the late 1980s by Jörg Hacker and colleagues in Werner Goebel's group at the University of Würzburg, Würzburg, Germany. As with other GIs, the PAIs comprise large genomic regions that differ from the rest of the genome by their G + C content, by their typical insertion within transfer RNA genes, and by their harboring of direct repeats (at their ends), integrase determinants, or other mobility loci. The hallmark of PAIs is their contribution to the emergence of virulent bacteria and to the development of intestinal and extraintestinal diseases. This review summarizes the current knowledge on the structure and functional features of PAIs, on PAI-encoded E. coli pathogenicity factors and on the role of PAIs in host-pathogen interactions.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRAE, MEDiS, Clermont-Ferrand, France
| | - Guillaume Dalmasso
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Racha Beyrouthy
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Nicolas Barnich
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Julien Delmas
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Richard Bonnet
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
7
|
Zhu N, Liu W, Prakash A, Zhang C, Kim KS. Targeting E. coli invasion of the blood-brain barrier for investigating the pathogenesis and therapeutic development of E. coli meningitis. Cell Microbiol 2020; 22:e13231. [PMID: 32447809 DOI: 10.1111/cmi.13231] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/15/2022]
Abstract
Escherichia coli is the most common Gram-negative bacillary organism causing neonatal meningitis. Escherichia coli meningitis remains an important cause of mortality and morbidity, but the pathogenesis of E. coli penetration of the blood-brain barrier remains incompletely understood. Escherichia coli entry into the brain occurs in the meningeal and cortex capillaries, not in the choroid plexus, and exploits epidermal growth factor receptor (EGFR) and cysteinyl leukotrienes (CysLTs) for invasion of the blood-brain barrier. The present study examined whether EGFR and CysLTs are inter-related in their contribution to E. coli invasion of the blood-brain barrier and whether counteracting EGFR and CysLTs is a beneficial adjunct to antibiotic therapy of E. coli meningitis. We showed that (a) meningitis isolates of E. coli exploit EGFR and CysLTs for invasion of the blood-brain barrier, (b) the contribution of EGFR is upstream of that of CysLTs, and (c) counteracting EGFR and CysLTs as an adjunctive therapy improved the outcome (survival, neuronal injury and memory impairment) of animals with E. coli meningitis. These findings suggest that investigation of host factors contributing to E. coli invasion of the blood-brain barrier will help in enhancing the pathogenesis and development of new therapeutic targets for E. coli meningitis in the era of increasing resistance to conventional antibiotics.
Collapse
Affiliation(s)
- Ningyu Zhu
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wei Liu
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Atish Prakash
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chengxian Zhang
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Genomic and Proteomic Characterization of the Extended-Spectrum β-Lactamase (ESBL)-Producing Escherichia coli Strain CCUG 73778: A Virulent, Nosocomial Outbreak Strain. Microorganisms 2020; 8:microorganisms8060893. [PMID: 32545759 PMCID: PMC7355845 DOI: 10.3390/microorganisms8060893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/23/2023] Open
Abstract
Escherichia coli strain CCUG 78773 is a virulent extended-spectrum β-lactamase (ESBL)-producing ST131-O25b type strain isolated during an outbreak at a regional university hospital. The complete and closed genome sequence, comprising one chromosome (5,076,638 bp) and six plasmids (1718–161,372 bp), is presented. Characterization of the genomic features detected the presence of 59 potential antibiotic resistance factors, including three prevalent β-lactamases. Several virulence associated elements were determined, mainly related with adherence, invasion, biofilm formation and antiphagocytosis. Twenty-eight putative type II toxin-antitoxin systems were found. The plasmids were characterized, through in silico analyses, confirming the two β-lactamase-encoding plasmids to be conjugative, while the remaining plasmids were mobilizable. BLAST analysis of the plasmid sequences showed high similarity with plasmids in E. coli from around the world. Expression of many of the described virulence and AMR factors was confirmed by proteomic analyses, using bottom-up, liquid chromatography-tandem mass spectrometry (LC-MS/MS). The detailed characterization of E. coli strain CCUG 78773 provides a reference for the relevance of genetic elements, as well as the characterization of antibiotic resistance and the spread of bacteria harboring ESBL genes in the hospital environment.
Collapse
|
9
|
Kim KS. Investigating Bacterial Penetration of the Blood-Brain Barrier for the Pathogenesis, Prevention, and Therapy of Bacterial Meningitis. ACS Infect Dis 2020; 6:34-42. [PMID: 31805229 DOI: 10.1021/acsinfecdis.9b00319] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The most distressing aspect of bacterial meningitis is limited improvement in the mortality and morbidity despite attributable advances in antimicrobial chemotherapy and supportive care. A major contributing factor to such mortality and morbidity is our incomplete understanding of the pathogenesis of this disease. Microbial penetration of the blood-brain barrier, a prerequisite for the development of bacterial meningitis, exploits specific host and bacterial factors as well as host cell signaling molecules. Determination and characterization of such host and bacterial factors have been instrumental for developing our current knowledge on the pathogenesis of bacterial meningitis. In addition, counteracting such host and microbial factors has been shown to be efficacious in the prevention of bacterial meningitis. Antimicrobial therapy alone has limited efficacy in improving the outcome of bacterial meningitis. Recent studies suggest that counteracting targets contributing to bacterial penetration of the blood-brain barrier are a beneficial therapeutic adjunct to antimicrobial therapy in improving the outcome of bacterial meningitis. Taken together, these findings indicate that the elucidation of host and bacterial factors contributing to microbial penetration of the blood-brain barrier provides a novel strategy for investigating the pathogenesis, prevention, and therapy of bacterial meningitis.
Collapse
Affiliation(s)
- Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Room 3157, Baltimore, Maryland 21287, United States
| |
Collapse
|
10
|
Hirose Y, Yamaguchi M, Goto K, Sumitomo T, Nakata M, Kawabata S. Competence-induced protein Ccs4 facilitates pneumococcal invasion into brain tissue and virulence in meningitis. Virulence 2019; 9:1576-1587. [PMID: 30251911 PMCID: PMC6177246 DOI: 10.1080/21505594.2018.1526530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Streptococcus pneumoniae is a major pathogen that causes pneumonia, sepsis, and meningitis. The candidate combox site 4 (ccs4) gene has been reported to be a pneumococcal competence-induced gene. Such genes are involved in development of S. pneumoniae competence and virulence, though the functions of ccs4 remain unknown. In the present study, the role of Ccs4 in the pathogenesis of pneumococcal meningitis was examined. We initially constructed a ccs4 deletion mutant and complement strains, then examined their association with and invasion into human brain microvascular endothelial cells. Wild-type and Ccs4-complemented strains exhibited significantly higher rates of association and invasion as compared to the ccs4 mutant strain. Deletion of ccs4 did not change bacterial growth activity or expression of NanA and CbpA, known brain endothelial pneumococcal adhesins. Next, mice were infected either intravenously or intranasally with pneumococcal strains. In the intranasal infection model, survival rates were comparable between wild-type strain-infected and ccs4 mutant strain-infected mice, while the ccs4 mutant strain exhibited a lower level of virulence in the intravenous infection model. In addition, at 24 hours after intravenous infection, the bacterial burden in blood was comparable between the wild-type and ccs4 mutant strain-infected mice, whereas the wild-type strain-infected mice showed a significantly higher bacterial burden in the brain. These results suggest that Ccs4 contributes to pneumococcal invasion of host brain tissues and functions as a virulence factor.
Collapse
Affiliation(s)
- Yujiro Hirose
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masaya Yamaguchi
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Kana Goto
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Tomoko Sumitomo
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masanobu Nakata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Shigetada Kawabata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| |
Collapse
|
11
|
Feng Y, Chen CS, Ho J, Pearce D, Hu S, Wang B, Desai P, Kim KS, Zhu H. High-Throughput Chip Assay for Investigating Escherichia coli Interaction with the Blood-Brain Barrier Using Microbial and Human Proteome Microarrays (Dual-Microarray Technology). Anal Chem 2018; 90:10958-10966. [PMID: 30106562 DOI: 10.1021/acs.analchem.8b02513] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Bacterial meningitis in neonates and infants is an acute lethal disease and occurs in response to microbial exploitation of the blood-brain barrier (BBB), resulting in the intracranial inflammation. Several pathogens, such as Escherichia coli ( E. coli), can cause this devastating disease; however, the underlying molecular mechanisms by which these pathogens exploit the BBB remain incompletely understood. To identify important players on both the pathogen and host sides that govern the E. coli-BBB cell interactions, we took advantage of the E. coli and human proteome microarrays (i.e., HuProt) as an unbiased, proteome-wide tool for identification of important players on both sides. Using the E. coli proteome microarrays, we developed a unique high throughput chip-based cell probing assay to probe with fluorescent live human brain microvascular endothelial cells (HBMEC, which constitute the BBB). We identified several transmembrane proteins, which effectively bound to live HBMEC. We focused on YojI protein for further study. By probing the HuProt arrays with YojI, interferon-alpha receptor (IFNAR2) was identified as one of its binding proteins. The importance of YojI and IFNAR2 involved in E. coli-HBMEC interactions was characterized using the YojI knockout bacteria and IFNAR2-knock down HBMEC and further confirmed by E. coli binding assay in HBMEC. This study represents a new paradigm (dual-microarray technology) that enables rapid, unbiased discovery of both pathogen and host players that are involved in pathogen-host interactions for human infectious diseases in a high throughput manner.
Collapse
Affiliation(s)
- Yingzhu Feng
- Key Laboratory of Bio-theological Science and Technology of Ministry of Education, College of Bioengineering , Chongqing University , Chongqing 400030 , PR China.,Department of Pharmacology and Molecular Sciences, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21205 , United States.,School of Life Sciences , Sun Yat-Sen University , Guangzhou 510275 , China
| | - Chien-Sheng Chen
- Department of Food Safety/Hygiene and Risk Management , Tainan City 701 , Taiwan.,Department of Pharmacology and Molecular Sciences, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21205 , United States.,Department of Biomedical Science and Engineering , National Central University , Taoyuan City 32001 , Taiwan
| | - Jessica Ho
- Department of Pharmacology and Molecular Sciences, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21205 , United States
| | - Donna Pearce
- Division of Pediatric Infectious Diseases, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21287 , United States
| | - Shaohui Hu
- Department of Pharmacology and Molecular Sciences, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21205 , United States
| | - Bochu Wang
- Key Laboratory of Bio-theological Science and Technology of Ministry of Education, College of Bioengineering , Chongqing University , Chongqing 400030 , PR China
| | - Prashant Desai
- The Sidney Kimmel Comprehensive Cancer Center, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21231 , United States
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21287 , United States
| | - Heng Zhu
- Department of Pharmacology and Molecular Sciences, School of Medicine , Johns Hopkins University , Baltimore , Maryland 21205 , United States
| |
Collapse
|
12
|
Zhao WD, Liu DX, Wei JY, Miao ZW, Zhang K, Su ZK, Zhang XW, Li Q, Fang WG, Qin XX, Shang DS, Li B, Li QC, Cao L, Kim KS, Chen YH. Caspr1 is a host receptor for meningitis-causing Escherichia coli. Nat Commun 2018; 9:2296. [PMID: 29895952 PMCID: PMC5997682 DOI: 10.1038/s41467-018-04637-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 05/03/2018] [Indexed: 12/17/2022] Open
Abstract
Escherichia coli is the leading cause of neonatal Gram-negative bacterial meningitis, but the pathogenesis of E. coli meningitis remains elusive. E. coli penetration of the blood–brain barrier (BBB) is the critical step for development of meningitis. Here, we identify Caspr1, a single-pass transmembrane protein, as a host receptor for E. coli virulence factor IbeA to facilitate BBB penetration. Genetic ablation of endothelial Caspr1 and blocking IbeA–Caspr1 interaction effectively prevent E. coli penetration into the brain during meningitis in rodents. IbeA interacts with extracellular domain of Caspr1 to activate focal adhesion kinase signaling causing E. coli internalization into the brain endothelial cells of BBB. E. coli can invade hippocampal neurons causing apoptosis dependent on IbeA–Caspr1 interaction. Our results indicate that E. coli exploits Caspr1 as a host receptor for penetration of BBB resulting in meningitis, and that Caspr1 might be a useful target for prevention or therapy of E. coli meningitis. Penetration of the blood–brain barrier (BBB) is crucial for development of E. coli-caused meningitis. Here, the authors show that a host membrane protein, Caspr1, acts as a receptor for a bacterial virulence factor to facilitate BBB penetration and entry of E. coli into brain neurons.
Collapse
Affiliation(s)
- Wei-Dong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China.
| | - Dong-Xin Liu
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Jia-Yi Wei
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Zi-Wei Miao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Zheng-Kang Su
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Xue-Wei Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Qiang Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Wen-Gang Fang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Xiao-Xue Qin
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - De-Shu Shang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Bo Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Qing-Chang Li
- Department of Pathology, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Liu Cao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 200 North Wolfe St, Room 3157, Baltimore, MD, 21287, USA
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, 110122, Shenyang, China.
| |
Collapse
|
13
|
Abstract
Escherichia coli is the most common Gram-negative bacillary organism causing meningitis, and E. coli meningitis continues to be an important cause of mortality and morbidity throughout the world. Our incomplete knowledge of its pathogenesis contributes to such mortality and morbidity. Recent reports of E. coli strains producing CTX-M-type or TEM-type extended-spectrum β-lactamases create a challenge. Studies using in vitro and in vivo models of the blood-brain barrier have shown that E. coli meningitis follows a high degree of bacteremia and invasion of the blood-brain barrier. E. coli invasion of the blood-brain barrier, the essential step in the development of E. coli meningitis, requires specific microbial and host factors as well as microbe- and host-specific signaling molecules. Blockade of such microbial and host factors contributing to E. coli invasion of the blood-brain barrier is shown to be efficient in preventing E. coli penetration into the brain. The basis for requiring a high degree of bacteremia for E. coli penetration of the blood-brain barrier, however, remains unclear. Continued investigation on the microbial and host factors contributing to a high degree of bacteremia and E. coli invasion of the blood-brain barrier is likely to identify new targets for prevention and therapy of E. coli meningitis.
Collapse
|
14
|
Diversity and distribution of nuclease bacteriocins in bacterial genomes revealed using Hidden Markov Models. PLoS Comput Biol 2017; 13:e1005652. [PMID: 28715501 PMCID: PMC5536347 DOI: 10.1371/journal.pcbi.1005652] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/31/2017] [Accepted: 06/21/2017] [Indexed: 11/28/2022] Open
Abstract
Bacteria exploit an arsenal of antimicrobial peptides and proteins to compete with each other. Three main competition systems have been described: type six secretion systems (T6SS); contact dependent inhibition (CDI); and bacteriocins. Unlike T6SS and CDI systems, bacteriocins do not require contact between bacteria but are diffusible toxins released into the environment. Identified almost a century ago, our understanding of bacteriocin distribution and prevalence in bacterial populations remains poor. In the case of protein bacteriocins, this is because of high levels of sequence diversity and difficulties in distinguishing their killing domains from those of other competition systems. Here, we develop a robust bioinformatics pipeline exploiting Hidden Markov Models for the identification of nuclease bacteriocins (NBs) in bacteria of which, to-date, only a handful are known. NBs are large (>60 kDa) toxins that target nucleic acids (DNA, tRNA or rRNA) in the cytoplasm of susceptible bacteria, usually closely related to the producing organism. We identified >3000 NB genes located on plasmids or on the chromosome from 53 bacterial species distributed across different ecological niches, including human, animals, plants, and the environment. A newly identified NB predicted to be specific for Pseudomonas aeruginosa (pyocin Sn) was produced and shown to kill P. aeruginosa thereby validating our pipeline. Intriguingly, while the genes encoding the machinery needed for NB translocation across the cell envelope are widespread in Gram-negative bacteria, NBs are found exclusively in γ-proteobacteria. Similarity network analysis demonstrated that NBs fall into eight groups each with a distinct arrangement of protein domains involved in import. The only structural feature conserved across all groups was a sequence motif critical for cell-killing that is generally not found in bacteriocins targeting the periplasm, implying a specific role in translocating the nuclease to the cytoplasm. Finally, we demonstrate a significant association between nuclease colicins, NBs specific for Escherichia coli, and virulence factors, suggesting NBs play a role in infection processes, most likely by enabling pathogens to outcompete commensal bacteria. Bacteria deploy a variety of antimicrobials to kill competing bacteria. Nuclease bacteriocins are a miscellaneous group of protein toxins that target closely related species, cleaving nucleic acids in the cytoplasm. It has proved difficult to establish how widespread bacteriocins are in bacterial populations due to the high diversity of bacteriocin-encoding genes. Here, we describe an in silico approach to identify nuclease bacteriocin genes in bacterial genomes and to distinguish them from other competition toxins. Bacteria that contain nuclease bacteriocin genes are found in many different types of environment but are prevalent in niches where interbacterial competition is likely to be high. Nuclease bacteriocins are found exclusively in γ-proteobacteria and are particularly abundant in the Enterobacteriaceae and Pseudomonadaceae families. Although the sequences we identify are indeed diverse (<20% sequence identity between protein families) we show that all nuclease bacteriocins contain an invariant motif, usually within a common structural scaffold, that is implicated in translocating the cytotoxic nuclease to the cytoplasm. Finally, we show that nuclease bacteriocins in pathogenic E. coli are strongly associated with virulence factors suggesting they play a role in pathogenicity mechanisms.
Collapse
|
15
|
Kim S, Kim YT, Yoon H, Lee JH, Ryu S. The complete genome sequence of Cronobacter sakazakii ATCC 29544 T, a food-borne pathogen, isolated from a child's throat. Gut Pathog 2017; 9:2. [PMID: 28053670 PMCID: PMC5209807 DOI: 10.1186/s13099-016-0150-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/07/2016] [Indexed: 11/10/2022] Open
Abstract
Background Cronobacter sakazakii is an emerging opportunistic pathogen that is associated with rare but life-threatening cases of severe diseases: meningitis, necrotizing enterocolitis, and sepsis in premature and full-term infants. However, the pathogenesis mechanism of this pathogen remains largely unknown. To determine its pathogenesis at the genomic level, the genome of C. sakazakii ATCC 29544T was completely sequenced and analyzed. Results The genomic DNA, containing a circular chromosome and three plasmids, is composed of 4,511,265 bp with a GC content of 56.71%, containing 4380 predicted open reading frames (ORFs), 22 rRNA genes, and 83 tRNA genes. The plasmids, designated pCSK29544_p1, pCSK29544_p2, and pCSK29544_p3, were 93,905-bp, 4938-bp, and 53,457-bp with GC contents of 57.02, 54.88, and 50.07%, respectively. They were also predicted to have 72, 6, and 57 ORFs without RNA genes. Conclusions The strain ATCC 29544T genome has ompA and ibeB-homologous cusC genes, probably associated with the invasion of human brain microvascular endothelial cells (BMECs). In addition, gene clusters for siderophore production (iucABCD/iutA) and the related transport system (eitCBAD) were detected in pCSK29544_p1 plasmid, indicating better iron uptake ability for survival. Furthermore, to survive under extremely dry condition like milk powder, this genome has gene clusters for biosynthesis of capsular proteins (CSK29544_00281-00284) and cellulose (CSK29544_01124-01127) for biofilm formation and a gene cluster for utilization of sialic acid in the milk (nanKTAR). The genome information of C. sakazakii ATCC 29544T would provide further understanding of its pathogenesis at the molecular level for the regulation of pathogenicity and the development of a rapid detection method using biomarkers. Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0150-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Seongok Kim
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea.,Department of Applied Chemistry and Biological Engineering, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| | - You-Tae Kim
- Department of Food Science and Biotechnology and Institute of Life Science and Resources, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 17104 Republic of Korea
| | - Hyunjin Yoon
- Department of Applied Chemistry and Biological Engineering, Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon, 16499 Republic of Korea
| | - Ju-Hoon Lee
- Department of Food Science and Biotechnology and Institute of Life Science and Resources, Kyung Hee University, 1732 Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 17104 Republic of Korea
| | - Sangryeol Ryu
- Department of Food and Animal Biotechnology, Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, and Center for Food and Bioconvergence, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826 Republic of Korea
| |
Collapse
|
16
|
Amigo N, Zhang Q, Amadio A, Zhang Q, Silva WM, Cui B, Chen Z, Larzabal M, Bei J, Cataldi A. Overexpressed Proteins in Hypervirulent Clade 8 and Clade 6 Strains of Escherichia coli O157:H7 Compared to E. coli O157:H7 EDL933 Clade 3 Strain. PLoS One 2016; 11:e0166883. [PMID: 27880834 PMCID: PMC5120812 DOI: 10.1371/journal.pone.0166883] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 11/04/2016] [Indexed: 11/19/2022] Open
Abstract
Escherichia coli O157:H7 is responsible for severe diarrhea and hemolytic uremic syndrome (HUS), and predominantly affects children under 5 years. The major virulence traits are Shiga toxins, necessary to develop HUS and the Type III Secretion System (T3SS) through which bacteria translocate effector proteins directly into the host cell. By SNPs typing, E. coli O157:H7 was separated into nine different clades. Clade 8 and clade 6 strains were more frequently associated with severe disease and HUS. In this study, we aimed to identify differentially expressed proteins in two strains of E. coli O157:H7 (clade 8 and clade 6), obtained from cattle and compared them with the well characterized reference EDL933 strain (clade 3). Clade 8 and clade 6 strains show enhanced pathogenicity in a mouse model and virulence-related properties. Proteins were extracted and analyzed using the TMT-6plex labeling strategy associated with two dimensional liquid chromatography and mass spectrometry in tandem. We detected 2241 proteins in the cell extract and 1787 proteins in the culture supernatants. Attention was focused on the proteins related to virulence, overexpressed in clade 6 and 8 strains compared to EDL933 strain. The proteins relevant overexpressed in clade 8 strain were the curli protein CsgC, a transcriptional activator (PchE), phage proteins, Stx2, FlgM and FlgD, a dienelactone hydrolase, CheW and CheY, and the SPATE protease EspP. For clade 6 strain, a high overexpression of phage proteins was detected, mostly from Stx2 encoding phage, including Stx2, flagellin and the protease TagA, EDL933_p0016, dienelactone hydrolase, and Haemolysin A, amongst others with unknown function. Some of these proteins were analyzed by RT-qPCR to corroborate the proteomic data. Clade 6 and clade 8 strains showed enhanced transcription of 10 out of 12 genes compared to EDL933. These results may provide new insights in E. coli O157:H7 mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Natalia Amigo
- Institute of Biotechnology, CICVyA, National Institute of Agricultural Technology. Hurlingham, Buenos Aires, Argentina
| | - Qi Zhang
- AGRO-Biological Gene Research Center, Guangdong `Academy of Agricultural Sciences (GDAAS), Guangzhou, China
| | - Ariel Amadio
- Rafaela Experimental Station, National Institute of Agricultural Technology. Rafaela, Santa Fe, Argentina
| | - Qunjie Zhang
- AGRO-Biological Gene Research Center, Guangdong `Academy of Agricultural Sciences (GDAAS), Guangzhou, China
| | - Wanderson M. Silva
- Institute of Biotechnology, CICVyA, National Institute of Agricultural Technology. Hurlingham, Buenos Aires, Argentina
| | - Baiyuan Cui
- AGRO-Biological Gene Research Center, Guangdong `Academy of Agricultural Sciences (GDAAS), Guangzhou, China
| | - Zhongjian Chen
- AGRO-Biological Gene Research Center, Guangdong `Academy of Agricultural Sciences (GDAAS), Guangzhou, China
| | - Mariano Larzabal
- Institute of Biotechnology, CICVyA, National Institute of Agricultural Technology. Hurlingham, Buenos Aires, Argentina
| | - Jinlong Bei
- AGRO-Biological Gene Research Center, Guangdong `Academy of Agricultural Sciences (GDAAS), Guangzhou, China
- * E-mail:
| | - Angel Cataldi
- Institute of Biotechnology, CICVyA, National Institute of Agricultural Technology. Hurlingham, Buenos Aires, Argentina
| |
Collapse
|
17
|
Sphingosine 1-Phosphate Activation of EGFR As a Novel Target for Meningitic Escherichia coli Penetration of the Blood-Brain Barrier. PLoS Pathog 2016; 12:e1005926. [PMID: 27711202 PMCID: PMC5053521 DOI: 10.1371/journal.ppat.1005926] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 09/11/2016] [Indexed: 12/24/2022] Open
Abstract
Central nervous system (CNS) infection continues to be an important cause of mortality and morbidity, necessitating new approaches for investigating its pathogenesis, prevention and therapy. Escherichia coli is the most common Gram-negative bacillary organism causing meningitis, which develops following penetration of the blood-brain barrier (BBB). By chemical library screening, we identified epidermal growth factor receptor (EGFR) as a contributor to E. coli invasion of the BBB in vitro. Here, we obtained the direct evidence that CNS-infecting E. coli exploited sphingosine 1-phosphate (S1P) for EGFR activation in penetration of the BBB in vitro and in vivo. We found that S1P was upstream of EGFR and participated in EGFR activation through S1P receptor as well as through S1P-mediated up-regulation of EGFR-related ligand HB-EGF, and blockade of S1P function through targeting sphingosine kinase and S1P receptor inhibited EGFR activation, and also E. coli invasion of the BBB. We further found that both S1P and EGFR activations occurred in response to the same E. coli proteins (OmpA, FimH, NlpI), and that S1P and EGFR promoted E. coli invasion of the BBB by activating the downstream c-Src. These findings indicate that S1P and EGFR represent the novel host targets for meningitic E. coli penetration of the BBB, and counteracting such targets provide a novel approach for controlling E. coli meningitis in the era of increasing resistance to conventional antibiotics.
Collapse
|
18
|
Huang SH, Chi F, Peng L, Bo T, Zhang B, Liu LQ, Wu X, Mor-Vaknin N, Markovitz DM, Cao H, Zhou YH. Vimentin, a Novel NF-κB Regulator, Is Required for Meningitic Escherichia coli K1-Induced Pathogen Invasion and PMN Transmigration across the Blood-Brain Barrier. PLoS One 2016; 11:e0162641. [PMID: 27657497 PMCID: PMC5033352 DOI: 10.1371/journal.pone.0162641] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 08/25/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND NF-κB activation, pathogen invasion, polymorphonuclear leukocytes (PMN) transmigration (PMNT) across the blood-brain barrier (BBB) are the pathogenic triad hallmark features of bacterial meningitis, but the mechanisms underlying these events remain largely unknown. Vimentin, which is a novel NF-κB regulator, is the primary receptor for the major Escherichia coli K1 virulence factor IbeA that contributes to the pathogenesis of neonatal bacterial sepsis and meningitis (NSM). We have previously shown that IbeA-induced NF-κB signaling through its primary receptor vimentin as well as its co-receptor PTB-associated splicing factor (PSF) is required for pathogen penetration and leukocyte transmigration across the BBB. This is the first in vivo study to demonstrate how vimentin and related factors contributed to the pathogenic triad of bacterial meningitis. METHODOLOGY/PRINCIPAL FINDINGS The role of vimentin in IbeA+ E. coli K1-induced NF-κB activation, pathogen invasion, leukocyte transmigration across the BBB has now been demonstrated by using vimentin knockout (KO) mice. In the in vivo studies presented here, IbeA-induced NF-κB activation, E. coli K1 invasion and polymorphonuclear neutrophil (PMN) transmigration across the BBB were significantly reduced in Vim-/- mice. Decreased neuronal injury in the hippocampal dentate gyrus was observed in Vim-/- mice with meningitis. The major inflammatory regulator α7 nAChR and several signaling molecules contributing to NF-κB activation (p65 and p-CamKII) were significantly reduced in the brain tissues of the Vim-/- mice with E. coli meningitis. Furthermore, Vim KO resulted in significant reduction in neuronal injury and in α7 nAChR-mediated calcium signaling. CONCLUSION/SIGNIFICANCE Vimentin, a novel NF-κB regulator, plays a detrimental role in the host defense against meningitic infection by modulating the NF-κB signaling pathway to increase pathogen invasion, PMN recruitment, BBB permeability and neuronal inflammation. Our findings provide the first evidence for Vim-dependent mechanisms underlying the pathogenic triad of bacterial meningitis.
Collapse
Affiliation(s)
- Sheng-He Huang
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Microbiology, School of Public Health and Tropocal Medicine, Southern Medical University, Guangzhou 510515, China
- * E-mail: (YHZ); (SHH)
| | - Feng Chi
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Pathology, Southern California Research Center for ALPD and Cirrhosis, the Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Liang Peng
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Clinic Laboratory, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Tao Bo
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Pediatrics, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Bao Zhang
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Microbiology, School of Public Health and Tropocal Medicine, Southern Medical University, Guangzhou 510515, China
| | - Li-Qun Liu
- Saban Research Institute of Childrens Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, California, United States of America
- Department of Pediatrics, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Nirit Mor-Vaknin
- Department of Internal Medicine, Division of Infectious Diseases, 5220 MSRB III, 1150 West Medical Center Drive, University of Michigan, Ann Arbor, MI, United States of America
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, 5220 MSRB III, 1150 West Medical Center Drive, University of Michigan, Ann Arbor, MI, United States of America
| | - Hong Cao
- Department of Microbiology, School of Public Health and Tropocal Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yan-Hong Zhou
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (YHZ); (SHH)
| |
Collapse
|
19
|
Dando SJ, Mackay-Sim A, Norton R, Currie BJ, St John JA, Ekberg JAK, Batzloff M, Ulett GC, Beacham IR. Pathogens penetrating the central nervous system: infection pathways and the cellular and molecular mechanisms of invasion. Clin Microbiol Rev 2014; 27:691-726. [PMID: 25278572 PMCID: PMC4187632 DOI: 10.1128/cmr.00118-13] [Citation(s) in RCA: 300] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The brain is well protected against microbial invasion by cellular barriers, such as the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB). In addition, cells within the central nervous system (CNS) are capable of producing an immune response against invading pathogens. Nonetheless, a range of pathogenic microbes make their way to the CNS, and the resulting infections can cause significant morbidity and mortality. Bacteria, amoebae, fungi, and viruses are capable of CNS invasion, with the latter using axonal transport as a common route of infection. In this review, we compare the mechanisms by which bacterial pathogens reach the CNS and infect the brain. In particular, we focus on recent data regarding mechanisms of bacterial translocation from the nasal mucosa to the brain, which represents a little explored pathway of bacterial invasion but has been proposed as being particularly important in explaining how infection with Burkholderia pseudomallei can result in melioidosis encephalomyelitis.
Collapse
Affiliation(s)
- Samantha J Dando
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Alan Mackay-Sim
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Robert Norton
- Townsville Hospital, Townsville, Queensland, Australia
| | - Bart J Currie
- Menzies School of Health Research and Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - James A St John
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia
| | - Jenny A K Ekberg
- Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland, Australia School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Michael Batzloff
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| | - Glen C Ulett
- School of Medical Science and Griffith Health Institute, Griffith University, Gold Coast, Queensland, Australia
| | - Ifor R Beacham
- Institute for Glycomics, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
20
|
Wang S, Shi Z, Xia Y, Li H, Kou Y, Bao Y, Dai J, Lu C. IbeB is involved in the invasion and pathogenicity of avian pathogenic Escherichia coli. Vet Microbiol 2012; 159:411-9. [DOI: 10.1016/j.vetmic.2012.04.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 04/09/2012] [Accepted: 04/10/2012] [Indexed: 10/28/2022]
|
21
|
Yan QQ, Condell O, Power K, Butler F, Tall BD, Fanning S. Cronobacter species (formerly known as Enterobacter sakazakii) in powdered infant formula: a review of our current understanding of the biology of this bacterium. J Appl Microbiol 2012; 113:1-15. [PMID: 22420458 DOI: 10.1111/j.1365-2672.2012.05281.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cronobacter species (formerly known as Enterobacter sakazakii) are opportunistic pathogens that can cause necrotizing enterocolitis, bacteraemia and meningitis, predominantly in neonates. Infection in these vulnerable infants has been linked to the consumption of contaminated powdered infant formula (PIF). Considerable research has been undertaken on this organism in the past number of years which has enhanced our understanding of this neonatal pathogen leading to improvements in its control within the PIF production environment. The taxonomy of the organism resulted in the recognition of a new genus, Cronobacter, which consists of seven species. This paper presents an up-to-date review of our current knowledge of Cronobacter species. Taxonomy, genome sequencing, current detection protocols and epidemiology are all discussed. In addition, consideration is given to the control of this organism in the manufacturing environment, as a first step towards reducing the occurrence of this pathogen in PIF.
Collapse
Affiliation(s)
- Q Q Yan
- UCD Centre for Food Safety, WHO Collaborating Centre for Research, Reference & Training on Cronobacter, School of Public Health, Physiotherapy & Population Science, University College Dublin, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
22
|
Bencurova E, Mlynarcik P, Bhide M. An insight into the ligand-receptor interactions involved in the translocation of pathogens across blood-brain barrier. ACTA ACUST UNITED AC 2011; 63:297-318. [PMID: 22092557 DOI: 10.1111/j.1574-695x.2011.00867.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2011] [Revised: 08/09/2011] [Accepted: 09/02/2011] [Indexed: 01/01/2023]
Abstract
Traversal of pathogen across the blood-brain barrier (BBB) is an essential step for central nervous system (CNS) invasion. Pathogen traversal can occur paracellularly, transcellularly, and/or in infected phagocytes (Trojan horse mechanism). To trigger the translocation processes, mainly through paracellular and transcellular ways, interactions between protein molecules of pathogen and BBB are inevitable. Simply, it takes two to tango: both host receptors and pathogen ligands. Underlying molecular basis of BBB translocation of various pathogens has been revealed in the last decade, and a plethora of experimental data on protein-protein interactions has been created. This review compiles these data and should give insights into the ligand-receptor interactions that occur during BBB translocation. Further, it sheds light on cell signaling events triggered in response to ligand-receptor interaction. Understanding of the molecular principles of pathogen-host interactions that are involved in traversal of the BBB should contribute to develop new vaccine and drug strategies to prevent CNS infections.
Collapse
Affiliation(s)
- Elena Bencurova
- Laboratory of Biomedical Microbiology and Immunology, Department of Microbiology and Immunology, University of Veterinary Medicine and Pharmacy, Kosice, Slovakia
| | | | | |
Collapse
|
23
|
Maruvada R, Kim KS. Extracellular loops of the Eschericia coli outer membrane protein A contribute to the pathogenesis of meningitis. J Infect Dis 2011; 203:131-40. [PMID: 21148506 DOI: 10.1093/infdis/jiq009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neonatal meningitis by Eschericia coli RS218 occurs due to bacteremia and its transmigration across the blood-brain barrier. Although the outer membrane protein A (OmpA), a molecule with extracellular loops has been shown to contribute to the above phenomenon, we do not know the exact the role of these individual loops. Using bacterial strains whose individual loops have been removed, we demonstrated that whereas Loops1 and 2 contribute to 70%-80% bacterial survival in serum, bacterial entry into human brain microvascular endothelial cells (HBMEC) is governed by Loops1, 2, and 3. Cellular invasion was shown to require activation of host cytosolic phospholipase A2 (cPLA2α) by Loops1 and 2 but not 3. This suggests 2 distinct pathways for bacterial entry into host cells. Loop 4 played no role in either serum survival, cellular entry, or cPLA2α signaling. These findings demonstrate for the first time the different contributions of extracellular loops of OmpA to the pathogenesis of E. coli meningitis.
Collapse
Affiliation(s)
- Ravi Maruvada
- Division of Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| | | |
Collapse
|
24
|
Effects of ibeA deletion on virulence and biofilm formation of avian pathogenic Escherichia coli. Infect Immun 2010; 79:279-87. [PMID: 20974831 DOI: 10.1128/iai.00821-10] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The ibeA gene is located on a genomic island, GimA, which is involved in the pathogenesis of neonatal meningitis Escherichia coli (NMEC) and avian pathogenic E. coli (APEC). The prevalence of ibeA in the APEC collection in China was investigated, and 20 of 467 strains (4.3%) were positive. In addition, analysis of the association of the E. coli reference (ECOR) groups with positive strains revealed that ibeA was linked to group B2. The ibeA gene in DE205B was analyzed and compared to those of APEC and NMEC, which indicated that the specificity of ibeA was not consistent along pathotypes. The invasion of chicken embryo fibroblast DF-1 cells by APEC DE205B and RS218 was observed, which suggested that DF-1 cells could be a model to study the mechanism of APEC invasion. The inactivation of ibeA in APEC DE205B led to the reduced capacity to invade DF-1 cells, defective virulence in vivo, and decreased biofilm formation compared to the wild-type strain. In addition, strain AAEC189 expressing ibeA exhibited enhanced invasion capacity and biofilm formation. The results of the quantitative real-time reverse transcription-PCR (qRT-PCR) analysis and animal system infection experiments indicated that the loss of ibeA decreased the colonization and proliferation capacities of APEC in the brain during system infection.
Collapse
|
25
|
Zhu L, Maruvada R, Sapirstein A, Malik KU, Peters-Golden M, Kim KS. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood-brain barrier. Infect Immun 2010; 78:4302-10. [PMID: 20696828 PMCID: PMC2950368 DOI: 10.1128/iai.00624-10] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 07/09/2010] [Accepted: 07/30/2010] [Indexed: 01/29/2023] Open
Abstract
Escherichia coli K1 meningitis occurs following penetration of the blood-brain barrier, but the underlying mechanisms involved in E. coli penetration of the blood-brain barrier remain incompletely understood. We have previously shown that host cytosolic phospholipase A(2)α (cPLA(2)α) contributes to E. coli invasion of human brain microvascular endothelial cells (HBMEC), which constitute the blood-brain barrier, but the underlying mechanisms remain unclear. cPLA(2)α selectively liberates arachidonic acid from membrane phospholipids. Here, we provide the first direct evidence that host 5-lipoxygenase and lipoxygenase products of arachidonic acid, cysteinyl leukotrienes (LTs), contribute to E. coli K1 invasion of HBMEC and penetration into the brain, and their contributions involve protein kinase C alpha (PKCα). These findings demonstrate that arachidonic acid metabolism regulates E. coli penetration of the blood-brain barrier, and studies are needed to further elucidate the mechanisms involved with metabolic products of arachidonic acid for their contribution to E. coli invasion of the blood-brain barrier.
Collapse
Affiliation(s)
- Longkun Zhu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| | - Ravi Maruvada
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| | - Adam Sapirstein
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| | - Kafait U. Malik
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| | - Marc Peters-Golden
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| | - Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, 200 North Wolfe St., Room 3157, Baltimore, Maryland 21287, Department of Anesthesiology and Critical Care Medicine, 600 North Wolfe Street, Meyer 297-A, Baltimore, Maryland 21287, Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, Michigan 48109
| |
Collapse
|
26
|
The GimA locus of extraintestinal pathogenic E. coli: does reductive evolution correlate with habitat and pathotype? PLoS One 2010; 5:e10877. [PMID: 20526361 PMCID: PMC2878320 DOI: 10.1371/journal.pone.0010877] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Accepted: 05/06/2010] [Indexed: 11/19/2022] Open
Abstract
IbeA (invasion of brain endothelium), which is located on a genomic island termed GimA, is involved in the pathogenesis of several extraintestinal pathogenic E. coli (ExPEC) pathotypes, including newborn meningitic E. coli (NMEC) and avian pathogenic E. coli (APEC). To unravel the phylogeny of GimA and to investigate its island character, the putative insertion locus of GimA was determined via Long Range PCR and DNA-DNA hybridization in 410 E. coli isolates, including APEC, NMEC, uropathogenic (UPEC), septicemia-associated E. coli (SEPEC), and human and animal fecal isolates as well as in 72 strains of the E. coli reference (ECOR) collection. In addition to a complete GimA (∼20.3 kb) and a locus lacking GimA we found a third pattern containing a 342 bp remnant of GimA in this strain collection. The presence of GimA was almost exclusively detected in strains belonging to phylogenetic group B2. In addition, the complete GimA was significantly more frequent in APEC and NMEC strains while the GimA remnant showed a higher association with UPEC strains. A detailed analysis of the ibeA sequences revealed the phylogeny of this gene to be consistent with that obtained by Multi Locus Sequence Typing of the strains. Although common criteria for genomic islands are partially fulfilled, GimA rather seems to be an ancestral part of phylogenetic group B2, and it would therefore be more appropriate to term this genomic region GimA locus instead of genomic island. The existence of two other patterns reflects a genomic rearrangement in a reductive evolution-like manner.
Collapse
|
27
|
Kucerova E, Clifton SW, Xia XQ, Long F, Porwollik S, Fulton L, Fronick C, Minx P, Kyung K, Warren W, Fulton R, Feng D, Wollam A, Shah N, Bhonagiri V, Nash WE, Hallsworth-Pepin K, Wilson RK, McClelland M, Forsythe SJ. Genome sequence of Cronobacter sakazakii BAA-894 and comparative genomic hybridization analysis with other Cronobacter species. PLoS One 2010; 5:e9556. [PMID: 20221447 PMCID: PMC2833190 DOI: 10.1371/journal.pone.0009556] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2009] [Accepted: 02/14/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The genus Cronobacter (formerly called Enterobacter sakazakii) is composed of five species; C. sakazakii, C. malonaticus, C. turicensis, C. muytjensii, and C. dublinensis. The genus includes opportunistic human pathogens, and the first three species have been associated with neonatal infections. The most severe diseases are caused in neonates and include fatal necrotizing enterocolitis and meningitis. The genetic basis of the diversity within the genus is unknown, and few virulence traits have been identified. METHODOLOGY/PRINCIPAL FINDINGS We report here the first sequence of a member of this genus, C. sakazakii strain BAA-894. The genome of Cronobacter sakazakii strain BAA-894 comprises a 4.4 Mb chromosome (57% GC content) and two plasmids; 31 kb (51% GC) and 131 kb (56% GC). The genome was used to construct a 387,000 probe oligonucleotide tiling DNA microarray covering the whole genome. Comparative genomic hybridization (CGH) was undertaken on five other C. sakazakii strains, and representatives of the four other Cronobacter species. Among 4,382 annotated genes inspected in this study, about 55% of genes were common to all C. sakazakii strains and 43% were common to all Cronobacter strains, with 10-17% absence of genes. CONCLUSIONS/SIGNIFICANCE CGH highlighted 15 clusters of genes in C. sakazakii BAA-894 that were divergent or absent in more than half of the tested strains; six of these are of probable prophage origin. Putative virulence factors were identified in these prophage and in other variable regions. A number of genes unique to Cronobacter species associated with neonatal infections (C. sakazakii, C. malonaticus and C. turicensis) were identified. These included a copper and silver resistance system known to be linked to invasion of the blood-brain barrier by neonatal meningitic strains of Escherichia coli. In addition, genes encoding for multidrug efflux pumps and adhesins were identified that were unique to C. sakazakii strains from outbreaks in neonatal intensive care units.
Collapse
Affiliation(s)
- Eva Kucerova
- School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Bacterial meningitis continues to be an important cause of mortality and morbidity in neonates and children throughout the world. The introduction of the protein conjugate vaccines against Haemophilus influenzae type b, Streptococcus pneumoniae, and Neisseria meningitidis has changed the epidemiology of bacterial meningitis. Suspected bacterial meningitis is a medical emergency and needs empirical antimicrobial treatment without delay, but recognition of pathogens with increasing resistance to antimicrobial drugs is an important factor in the selection of empirical antimicrobial regimens. At present, strategies to prevent and treat bacterial meningitis are compromised by incomplete understanding of the pathogenesis. Further research on meningitis pathogenesis is thus needed. This Review summarises information on the epidemiology, pathogenesis, new diagnostic methods, empirical antimicrobial regimens, and adjunctive treatment of acute bacterial meningitis in infants and children.
Collapse
Affiliation(s)
- Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
29
|
Characterization of BNT2, an intrinsically curved DNA of Escherichia coli O157:H7. Biochem Biophys Res Commun 2010; 391:1792-7. [PMID: 20051226 DOI: 10.1016/j.bbrc.2009.12.160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 12/28/2009] [Indexed: 01/27/2023]
Abstract
The gene regulation by intrinsically curved DNA is one way for bacterial sensing of and response to environmental changes. Previously, we showed that the genetic element BNT2 upstream of the ecf (eae-positive conserved fragment) operon in the Escherichia coli O157:H7 virulence plasmid (pO157) has characteristics typical of intrinsically curved DNA, including the presence of multi-homopolymeric adenine:thymine tracts (AT tracts) and electrophoretic anomaly at 4 degrees C. Here we report that a local intrinsic curvature induced by the two phased AT tracts within the unusual promoter sequence of BNT2 played a major role for its temperature-dependent promoter activity. The base substitution of the AT tract in the spacer DNA between the -35 and the unusual -10 regions of the BNT2 promoter with non-AT tract sequence reduced intrinsic curvature slightly at 4 degrees C, but greatly affected its transcriptional activity. This implies that such a local intrinsic curvature within the unusual promoter of BNT2 is important for thermoregulation of the ecf operon.
Collapse
|
30
|
Lim JY, Yoon JW, Hovde CJ. A brief overview of Escherichia coli O157:H7 and its plasmid O157. J Microbiol Biotechnol 2010; 20:5-14. [PMID: 20134227 PMCID: PMC3645889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Enterohemorrhagic Escherichia coli O157:H7 is a major food-borne pathogen causing severe disease in humans worldwide. Healthy cattle are a reservoir of E. coli O157:H7 and bovine food products and fresh produce contaminated with bovine waste are the most common sources for disease outbreaks in the United States. E. coli O157:H7 also survives well in the environment. The ability to cause human disease, colonize the bovine gastrointestinal tract, and survive in the environment, requires that E. coli O157:H7 adapt to a wide variety of conditions. Three major virulence factors of E. coli O157:H7 have been identified including Shiga toxins, a pathogenicity island called the locus of enterocyte effacement, and an F-like plasmid, pO157. Among these virulence factors, the role of the pO157 is least understood. This review provides a board overview of E. coli O157:H7 with an emphasis on the pO157.
Collapse
Affiliation(s)
- Ji Youn Lim
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, Idaho 83844, U.S.A
| | - Jang W. Yoon
- Advanced Human Resource and Research Group for Medical Science (BK21), Konkuk University School of Medicine, Seoul 143-701, Korea
| | - Carolyn J. Hovde
- Department of Microbiology, Molecular Biology, and Biochemistry, University of Idaho, Moscow, Idaho 83844, U.S.A
| |
Collapse
|
31
|
Daneman R, Rescigno M. The gut immune barrier and the blood-brain barrier: are they so different? Immunity 2009; 31:722-35. [PMID: 19836264 DOI: 10.1016/j.immuni.2009.09.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Accepted: 09/29/2009] [Indexed: 12/18/2022]
Abstract
In order to protect itself from a diverse set of environmental pathogens and toxins, the body has developed a number of barrier mechanisms to limit the entry of potential hazards. Here, we compare two such barriers: the gut immune barrier, which is the primary barrier against pathogens and toxins ingested in food, and the blood-brain barrier, which protects the central nervous system from pathogens and toxins in the blood. Although each barrier provides defense in very different environments, there are many similarities in their mechanisms of action. In both cases, there is a physical barrier formed by a cellular layer that tightly regulates the movement of ions, molecules, and cells between two tissue spaces. These barrier cells interact with different cell types, which dynamically regulate their function, and with a different array of immune cells that survey the physical barrier and provide innate and adaptive immunity.
Collapse
Affiliation(s)
- Richard Daneman
- University of California, San Francisco, Department of Anatomy, San Francisco, CA 94143-0452, USA.
| | | |
Collapse
|
32
|
Identification of IbeR as a stationary-phase regulator in meningitic Escherichia coli K1 that carries a loss-of-function mutation in rpoS. J Biomed Biotechnol 2009; 2009:520283. [PMID: 19300523 PMCID: PMC2655632 DOI: 10.1155/2009/520283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Accepted: 12/01/2008] [Indexed: 11/23/2022] Open
Abstract
IbeR is a regulator present in meningitic Escherichia coli strain E44 that carries a loss-of-function mutation in the stationary-phase (SP) regulatory gene rpoS. In order to determine whether IbeR is an SP regulator in E44, two-dimensional gel electrophoresis and LC-MS were used to compare the proteomes of a noninvasive ibeR deletion mutant BR2 and its parent strain E44 in the SP. Four up-regulated (TufB, GapA, OmpA, AhpC) and three down-regulated (LpdA, TnaA, OpmC) proteins in BR2 were identified when compared to E44. All these proteins contribute to energy metabolism or stress resistance, which is related to SP regulation. One of the down-regulated proteins, tryptophanase (TnaA), which is regulated by RpoS in other E. coli strains, is associated with SP regulation via production of a signal molecule indole. Our studies demonstrated that TnaA was required for E44 invasion, and that indole was able to restore the noninvasive phenotype of the tnaA mutant. The production of indole was significantly reduced in BR2, indicating that ibeR is required for the indole production via tnaA. Survival studies under different stress conditions indicated that IbeR contributed to bacteria stress resistance in the SP. Taken together, IbeR is a novel regulator contributing to the SP regulation.
Collapse
|
33
|
Cortes MAM, Gibon J, Chanteloup NK, Moulin-Schouleur M, Gilot P, Germon P. Inactivation of ibeA and ibeT results in decreased expression of type 1 fimbriae in extraintestinal pathogenic Escherichia coli strain BEN2908. Infect Immun 2008; 76:4129-36. [PMID: 18591231 PMCID: PMC2519445 DOI: 10.1128/iai.00334-08] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2008] [Revised: 04/25/2008] [Accepted: 06/21/2008] [Indexed: 12/21/2022] Open
Abstract
IbeA in extraintestinal pathogenic Escherichia coli (ExPEC) strains was previously described for its role in invasion. Here we investigated the role of IbeA and IbeT, encoded by a gene located downstream of ibeA, in the adhesion of the avian ExPEC strain BEN2908 to human brain microvascular endothelial cells (HBMEC). The DeltaibeA mutant was less adhesive to HBMEC than the wild-type strain BEN2908 was. Because strain BEN2908 also expresses type 1 fimbriae, we measured the adhesion specifically due to IbeA by comparing the adhesive properties of a Deltafim derivative of strain BEN2908 to those of a double Deltafim DeltaibeA mutant. No differences were observed, indicating that the reduction of adhesion in BEN2908 DeltaibeA could be due to a decrease in type 1 fimbria expression. We indeed showed that the decreased adhesion of BEN2908 DeltaibeA was correlated with a decrease in type 1 fimbria expression. Accordingly, more bacteria had a fim promoter orientated in the off position in a culture of BEN2908 DeltaibeA than in a culture of BEN2908. Expression of fimB and fimE, two genes encoding recombinases participating in controlling the orientation of the fim promoter, was decreased in BEN2908 DeltaibeA. A reduction of type 1 fimbria expression due to a preferential orientation of the fim promoter in the off position was also seen in an ibeT mutant of strain BEN2908. We finally suggest a role for IbeA and IbeT in modulating the expression of type 1 fimbriae through an as yet unknown mechanism.
Collapse
Affiliation(s)
- Mélanie A M Cortes
- INRA, UR 1282 Infectiologie Animale et Santé Publique, Laboratoire de Pathogénie Bactérienne, Nouzilly, France
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Central nervous system (CNS) infections continue to be an important cause of morbidity and mortality. Microbial invasion and traversal of the blood-brain barrier is a prerequisite for CNS infections. Pathogens can cross the blood-brain barrier transcellularly, paracellularly and/or in infected phagocytes (the so-called Trojan-horse mechanism). Consequently, pathogens can cause blood-brain barrier dysfunction, including increased permeability, pleocytosis and encephalopathy. A more complete understanding of the microbial-host interactions that are involved in microbial traversal of the blood-brain barrier and the associated barrier dysfunction should help to develop new strategies to prevent CNS infections.
Collapse
|
35
|
Zou Y, He L, Chi F, Jong A, Huang SH. Involvement of Escherichia coli K1 ibeT in bacterial adhesion that is associated with the entry into human brain microvascular endothelial cells. Med Microbiol Immunol 2007; 197:337-44. [PMID: 18040715 DOI: 10.1007/s00430-007-0065-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Indexed: 01/06/2023]
Abstract
IbeT is a downstream gene of the invasion determinant ibeA in the chromosome of a clinical isolate of Escherichia coli K1 strain RS218 (serotype 018:K1:H7). Both ibeT and ibeA are in the same operon. Our previous mutagenesis and complementation studies suggested that ibeT may coordinately contribute to E. coli K1 invasion with ibeA. An isogenic in-frame deletion mutant of ibeT has been made by chromosomal gene replacement with a recombinant suicide vector carrying a fragment with an ibeT internal deletion. The characteristics of the mutant in meningitic E. coli infection were examined in vitro [cell culture of human brain microvascular endothelial cells (HBMEC)] and in vivo (infant rat model of E. coli meningitis) in comparison with the parent strain. The ibeT deletion mutant was significantly less adhesive and invasive than its parent strain E. coli E44 in vitro, and the adhesion- and invasion-deficient phenotypes of the mutant can be complemented by the ibeT gene. Recombinant IbeT protein is able to block E. coli E44 invasion of HBMEC. Furthermore, the ibeT deletion mutant is less capable of colonizing intestine and less virulent in bacterial translocation across the blood-brain barrier (BBB) than its parent E. coli E44 in vivo. These data suggest that ibeT-mediated E. coli K1 adhesion is associated with the bacterial invasion process.
Collapse
Affiliation(s)
- Yanming Zou
- Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | | | | | | | | |
Collapse
|
36
|
Townsend SM, Hurrell E, Gonzalez-Gomez I, Lowe J, Frye JG, Forsythe S, Badger JL. Enterobacter sakazakii invades brain capillary endothelial cells, persists in human macrophages influencing cytokine secretion and induces severe brain pathology in the neonatal rat. MICROBIOLOGY-SGM 2007; 153:3538-3547. [PMID: 17906151 DOI: 10.1099/mic.0.2007/009316-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Enterobacter sakazakii is an opportunistic pathogen associated with contaminated powdered infant formula and a rare cause of Gram-negative sepsis that can develop into meningitis and brain abscess formation in neonates. Bacterial pathogenesis remains to be fully elucidated. In this study, the host inflammatory response was evaluated following intracranial inoculation of Ent. sakazakii into infant rats. Infiltrating macrophages and neutrophils composed multiple inflammatory foci and contained phagocytosed bacteria. Several genotypically distinct Ent. sakazakii strains (16S cluster groups 1-4) were shown to invade rat capillary endothelial brain cells (rBCEC4) in vitro. Further, the persistence of Ent. sakazakii in macrophages varied between strains. The presence of putative sod genes and SOD activity may influence the survival of acidic conditions and macrophage oxidase and contribute to Ent. sakazakii intracellular persistence. The influence of macrophage uptake of Ent. sakazakii on immunoregulatory cytokine expression was assessed by ELISA. This demonstrated that the IL-10/IL-12 ratio is high after 24 h. This is suggestive of a type 2 immune response which is inefficient in fighting intracellular infections. These findings may help explain how the diversity in virulence traits among Ent. sakazakii isolates and an unsuccessful immune response contribute to the opportunistic nature of this infection.
Collapse
Affiliation(s)
- Stacy M Townsend
- School of Biomedical and Natural Sciences, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Edward Hurrell
- School of Biomedical and Natural Sciences, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Ignacio Gonzalez-Gomez
- University of Southern California Keck School of Medicine Los Angeles, CA 90027, USA
- Department of Pathology, Children's Hospital Los Angeles, CA 90027, USA
| | - James Lowe
- Department of Neurology, Queen's Medical Centre NHS Trust, Nottingham NG7 2UH, UK
| | - Jonathan G Frye
- Bacterial Epidemiology and Antimicrobial Resistance Research Unit, USDA, Agricultural Research Service, 950 College Station Road, Athens, GA 30605, USA
| | - Stephen Forsythe
- School of Biomedical and Natural Sciences, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Julie L Badger
- University of Southern California Keck School of Medicine Los Angeles, CA 90027, USA
- Department of Pathology, Children's Hospital Los Angeles, CA 90027, USA
| |
Collapse
|
37
|
Zou Y, He L, Wu CH, Cao H, Xie ZH, Ouyang Y, Wang Y, Jong A, Huang SH. PSF is an IbeA-binding protein contributing to meningitic Escherichia coli K1 invasion of human brain microvascular endothelial cells. Med Microbiol Immunol 2007; 196:135-43. [PMID: 17318576 DOI: 10.1007/s00430-006-0034-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Indexed: 11/29/2022]
Abstract
During the development of Escherichia coli K1 meningitis, interaction between E. coli invasion protein IbeA and surface protein(s) on human brain microvascular endothelial cells (HBMEC) is required for invasion and IbeA-mediated signaling. Here, an IbeA-binding protein was identified as polypyrimidine tract-binding protein (PTB)-associated splicing factor (PSF). The specific binding was confirmed by ligand overlay assay. The cell surface-expressed PSF was verified by the confocal microscopy. Recombinant PSF blocked E. coli K1 invasion of HBMEC effectively. Overexpression of PSF in the lentivirus-transducted HBMEC significantly enhanced E. coli K1 invasion. These results suggest that IbeA interacts with PSF for the E. coli K1 invasion of HBMEC.
Collapse
Affiliation(s)
- Yanming Zou
- Saban Research Institute, Childrens Hospital Los Angeles, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim KS. Microbial translocation of the blood–brain barrier. Int J Parasitol 2006; 36:607-14. [PMID: 16542662 DOI: 10.1016/j.ijpara.2006.01.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Revised: 01/19/2006] [Accepted: 01/30/2006] [Indexed: 11/23/2022]
Abstract
A major contributing factor to high mortality and morbidity associated with CNS infection is the incomplete understanding of the pathogenesis of this disease. Relatively small numbers of pathogens account for most cases of CNS infections in humans, but it is unclear how such pathogens cross the blood-brain barrier (BBB) and cause infections. The development of the in vitro BBB model using human brain microvascular endothelial cells has facilitated our understanding of the microbial translocation of the BBB, a key step for the acquisition of CNS infections. Recent studies have revealed that microbial translocation of the BBB involves host cell actin cytoskeletal rearrangements, most likely as the result of specific microbial-host interactions. A better understanding of microbial-host interactions that are involved in microbial translocation of the BBB should help in developing new strategies to prevent CNS infections. This review summarises our current understanding of the pathogenic mechanisms involved in translocation of the BBB by meningitis-causing bacteria, fungi and parasites.
Collapse
Affiliation(s)
- Kwang Sik Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 256, Baltimore, MD 21287, USA.
| |
Collapse
|
39
|
Abstract
Escherichia coliis the most common Gram-negative organism causing neonatal meningitis. Neonatal E. colimeningitis continues to be an important cause of mortality and morbidity throughout the world. Our incomplete knowledge of its pathogenesis and pathophysiology contributes to such mortality and morbidity. Recent reports of neonatal meningitis caused by E. coli strains producing CTX-M-type or TEM-type extended-spectrum β-lactamases create a challenge. E. colipenetration into the brain, the essential step in the development of E. coli meningitis, requires a high-degree of bacteremia and penetration of the blood-brain barrier as live bacteria, but the underlying mechanisms remain incompletely understood. Recent functional genomic approaches of meningitis-causing E. coli in both in vitro and in vivo models of the blood-brain barrier (e.g., human brain microvascular endothelial cells and animal models of experimental hematogenousE. colimeningitis, respectively) have identified several E. coli factors contributing to a high-degree of bacteremia, as well as specific microbial factors contributing to E. coli invasion of the blood-brain barrier. In addition, E. coli penetration of the blood-brain barrier involves specific host factors as well as microbe- and host-specific signaling molecules. Blockade of such microbial and host factors and host cell signaling molecules is efficient in preventing E. coli penetration into the brain. Continued investigation of the microbial and host factors contributing to E. colibacteremia andinvasion of the blood-brain barrier is likely to identify new targets for prevention and therapy of E. coli meningitis, thereby limiting the exposure to emerging antimicrobial-resistant E. coli.
Collapse
|
40
|
Abstract
Pathogenic Escherichia coli causes extraintestinal infections such as urinary tract infection and meningitis, which are prevalent and associated with considerable morbidity. Previous investigations have identified common strategies evolved by pathogenic E. coli to exploit host cell function and cause extraintestinal infections, which include the invasion into non-phagocytic eukaryotic cells such as epithelial and endothelial cells and associated host cell actin cytoskeletal rearrangements. However, the mechanisms involved in pathogenic E. coli invasion of eukaryotic cells are shown to differ depending upon types of host tissues and microbial determinants. In this mini-review, invasion processes of pathogenic E. coli are discussed using E. coli K1 invasion of human brain microvascular endothelial cells (HBMEC) as a paradigm. E. coli K1 is the most common Gram-negative organism causing neonatal meningitis, and E. coli invasion of HBMEC is shown to be a prerequisite for E. coli traversal of the blood-brain barrier in vivo. Previous studies have demonstrated that E. coli translocation of the blood-brain barrier is the result of specific E. coli host interactions including specific signal transduction pathways and modulation of endocytic pathways. Recent studies using functional genomics have identified additional microbial determinants contributing to E. coli K1 invasion of HBMEC. Complete understanding of microbial-host interactions that are involved in E. coli K1 invasion of HBMEC should help in the development of new strategies to prevent E. coli meningitis.
Collapse
Affiliation(s)
- Brian Y Kim
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, 600 North Wolfe Street/Park 256, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
41
|
Hasebe R, Kimura T, Nakamura K, Ochiai K, Okazaki K, Wada R, Umemura T. Differential susceptibility of equine and mouse brain microvascular endothelial cells to equine herpesvirus 1 infection. Arch Virol 2005; 151:775-86. [PMID: 16328147 DOI: 10.1007/s00705-005-0653-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 08/16/2005] [Indexed: 10/25/2022]
Abstract
Equine herpesvirus 1 (EHV-1) shows endotheliotropism in the central nervous system (CNS) of infected horses. However, infection of endothelial cells has not been observed in the CNS of infected mice. To explore the basis for this difference in endotheliotropism, we compared the susceptibility of equine brain microvascular endothelial cells (EBMECs) and mouse brain microvascular endothelial cells (MBMECs) to EHV-1 infection. The kinetics of viral growth in EBMECs was typical of a fully productive infection whereas viral infection in MBMECs seemed to be nonproductive. Immunofluorescence microscopy using anti-EHV-1 polyclonal antibody demonstrated viral antigen in infected EBMECs, but not infected MBMECs. EHV-1 immediate early (IE), early (ICP0), and late (gB, gD and gK) transcripts were expressed in infected EBMECs. However, none of these genes was detected in infected MBMECs by reverse transcription-polymerase chain reaction. Electron microscopic examination at the stage of viral entry showed that viral particles were present within uncoated vesicles in the cytoplasm of EBMECs, but absent from those of MBMECs. These results suggest that viral entry is an important determinant of the susceptibility of EBMECs and MBMECs to EHV-1 infection.
Collapse
Affiliation(s)
- R Hasebe
- Laboratory of Comparative Pathology, Hokkaido University, Sapporo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Stenberg F, Chovanec P, Maslen SL, Robinson CV, Ilag LL, von Heijne G, Daley DO. Protein Complexes of the Escherichia coli Cell Envelope. J Biol Chem 2005; 280:34409-19. [PMID: 16079137 DOI: 10.1074/jbc.m506479200] [Citation(s) in RCA: 174] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein complexes are an intrinsic aspect of life in the membrane. Knowing which proteins are assembled in these complexes is therefore essential to understanding protein function(s). Unfortunately, recent high throughput protein interaction studies have failed to deliver any significant information on proteins embedded in the membrane, and many membrane protein complexes remain ill defined. In this study, we have optimized the blue native-PAGE technique for the study of membrane protein complexes in the inner and outer membranes of Escherichia coli. In combination with second dimension SDS-PAGE and mass spectrometry, we have been able to identify 43 distinct protein complexes. In addition to a number of well characterized complexes, we have identified known and orphan proteins in novel oligomeric states. For two orphan proteins, YhcB and YjdB, our findings enable a tentative functional assignment. We propose that YhcB is a hitherto unidentified additional subunit of the cytochrome bd oxidase and that YjdB, which co-localizes with the ZipA protein, is involved in cell division. Our reference two-dimensional blue native-SDS-polyacrylamide gels will facilitate future studies of the assembly and composition of E. coli membrane protein complexes during different growth conditions and in different mutant backgrounds.
Collapse
Affiliation(s)
- Filippa Stenberg
- Department of Biochemistry and Stockholm University, Stockholm SE-106 91, Sweden
| | | | | | | | | | | | | |
Collapse
|
43
|
Germon P, Chen YH, He L, Blanco JE, Brée A, Schouler C, Huang SH, Moulin-Schouleur M. ibeA, a virulence factor of avian pathogenic Escherichia coli. MICROBIOLOGY-SGM 2005; 151:1179-1186. [PMID: 15817785 DOI: 10.1099/mic.0.27809-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The presence of ibeA, a gene encoding a known virulence factor of Escherichia coli strains responsible for neonatal meningitis in humans, was investigated in the genome of 213 avian pathogenic E. coli (APEC) strains and 55 non-pathogenic E. coli strains of avian origin. Fifty-three strains were found to be ibeA(+), all of which belonged to the APEC group. The ibeA gene is therefore positively linked to the pathogenicity of strains (P<0.0001). Analysis of the serogroup of strains revealed a positive association of ibeA with serogroups O18, O88 and O2. On the contrary, only 1/59 O78 strains are ibeA(+), indicating a negative association of ibeA with this serogroup (P<0.0001). The role of ibeA in the virulence of the APEC strain BEN 2908 was investigated by constructing an ibeA mutant. Challenge assays on 3-week-old chickens showed a reduced virulence for the ibeA mutant. Furthermore, the APEC strain BEN 2908 was able to invade brain microvascular epithelial cells, this invasion being significantly reduced upon inactivation of ibeA. Altogether, these results suggest a role of ibeA in the pathogenicity of some APEC strains and confirm the close relationship between APEC and other human extraintestinal pathogenic E. coli isolates.
Collapse
Affiliation(s)
- Pierre Germon
- INRA - Centre de Tours, UR 86, Pathologie Bactérienne, 37380 Nouzilly, France
| | - Yu-Hua Chen
- Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA
| | - Lina He
- Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA
| | - Jesús E Blanco
- Laboratorio de Referencia de E. coli, Departamento de Microbiología y Parasitología, Faculdad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | - Annie Brée
- INRA - Centre de Tours, UR 86, Pathologie Bactérienne, 37380 Nouzilly, France
| | - Catherine Schouler
- INRA - Centre de Tours, UR 86, Pathologie Bactérienne, 37380 Nouzilly, France
| | - Sheng-He Huang
- Children's Hospital Los Angeles and University of Southern California Keck School of Medicine, Los Angeles, CA 90027, USA
| | | |
Collapse
|
44
|
Yoon JW, Lim JY, Park YH, Hovde CJ. Involvement of the Escherichia coli O157:H7(pO157) ecf operon and lipid A myristoyl transferase activity in bacterial survival in the bovine gastrointestinal tract and bacterial persistence in farm water troughs. Infect Immun 2005; 73:2367-78. [PMID: 15784583 PMCID: PMC1087426 DOI: 10.1128/iai.73.4.2367-2378.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli O157:H7 is an important food-borne pathogen that causes hemorrhagic colitis and the hemolytic-uremic syndrome in humans. Recently, we reported that the pO157 ecf (E. coli attaching and effacing gene-positive conserved fragments) operon is thermoregulated by an intrinsically curved DNA and contains the genes for bacterial surface-associated proteins, including a second copy of lipid A myristoyl transferase, whose chromosomal copy is the lpxM gene product. E. coli O157:H7 survives and persists well in diverse environments from the human and bovine gastrointestinal tracts (GIT) to nutrient-dilute farm water troughs. Transcriptional regulation of the ecf operon by intrinsic DNA curvature and the genetic redundancy of lpxM that is associated with lipid A modification led us to hypothesize that the pO157 ecf operon and lpxM are associated with bacterial survival and persistence in various in vivo and ex vivo environments by optimizing bacterial membrane structure and/or integrity. To test this hypothesis, three isogenic ecf operon and/or lpxM deletion mutants of E. coli O157:H7 ATCC 43894 were constructed and analyzed in vitro and in vivo. The results showed that a double mutant carrying deletions in the ecf and lpxM genes had an altered lipid A structure and membrane fatty acid composition, did not survive passage through the bovine GIT, did not persist well in farm water troughs, had increased susceptibility to a broad spectrum of antibiotics and detergents, and had impaired motility. Electron microscopic analyses showed gross changes in bacterial membrane structure.
Collapse
Affiliation(s)
- Jang W Yoon
- Department of Microbiology, Molecular Biology and Biochemistry, University of Idaho, Moscow, ID 83844-3052, USA
| | | | | | | |
Collapse
|
45
|
Xie Y, Kim KJ, Kim KS. Current concepts on Escherichia coli K1 translocation of the blood-brain barrier. ACTA ACUST UNITED AC 2005; 42:271-9. [PMID: 15477040 DOI: 10.1016/j.femsim.2004.09.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2004] [Accepted: 09/02/2004] [Indexed: 11/19/2022]
Abstract
The mortality and morbidity associated with neonatal gram-negative meningitis have remained significant despite advances in antimicrobial chemotherapy. Escherichia coli K1 is the most common gram-negative organism causing neonatal meningitis. Our incomplete knowledge of the pathogenesis of this disease is one of the main reasons for this high mortality and morbidity. We have previously established both in vitro and in vivo models of the blood-brain barrier (BBB) using human brain microvascular endothelial cells (HBMEC) and hematogenous meningitis in neonatal rats, respectively. With these in vitro and in vivo models, we have shown that successful crossing of the BBB by circulating E. coli requires a high-degree of bacteremia, E. coli binding to and invasion of HBMEC, and E. coli traversal of the BBB as live bacteria. Our previous studies using TnphoA, signature-tagged mutagenesis and differential fluorescence induction identified several E. coli K1 determinants such as OmpA, Ibe proteins, AslA, TraJ and CNF1 contributing to invasion of HBMEC in vitro and traversal of the blood-brain barrier in vivo. We have shown that some of these determinants interact with specific receptors on HBMEC, suggesting E. coli translocation of the BBB is the result of specific pathogen-host cell interactions. Recent studies using functional genomics techniques have identified additional E. coli K1 factors that contribute to the high degree of bacteremia and HBMEC binding/invasion/transcytosis. In this review, we summarize the current knowledge on the mechanisms underlying the successful E. coli translocation of the BBB.
Collapse
Affiliation(s)
- Yi Xie
- Division of Pediatric Infectious Diseases, School of Medicine, Johns Hopkins University, 600 North Wolfe Street, Park 256, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
46
|
Hess P, Altenhöfer A, Khan AS, Daryab N, Kim KS, Hacker J, Oelschlaeger TA. A Salmonella fim homologue in Citrobacter freundii mediates invasion in vitro and crossing of the blood-brain barrier in the rat pup model. Infect Immun 2004; 72:5298-307. [PMID: 15322026 PMCID: PMC517473 DOI: 10.1128/iai.72.9.5298-5307.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
From the invasive Citrobacter freundii strain 3009, an invasion determinant was cloned, sequenced, and expressed. Sequence analysis of the determinant showed high homology with the fim determinant from Salmonella enterica serovar Typhimurium. The genes of the invasion determinant directed invasion of recombinant Escherichia coli K-12 strains into human epithelial cell lines of the bladder and gut as well as mannose-sensitive yeast agglutination and were termed fim(Cf) genes. Expression of the Fim(Cf) proteins was shown by (35)S labeling and/or Western blotting. In the infant rat model of experimental hematogenous meningitis, C. freundii strain 3009 and the in vitro invasive recombinant E. coli K-12 strain harboring the fim(Cf) determinant reached the cerebrospinal fluid, in contrast to the case for the control strain. The fim determinant was also necessary for efficient in vitro invasion by C. freundii, because a deletion mutant was strongly reduced in its invasion efficiency. The mutation could be complemented in trans by the corresponding genes. Invasion by C. freundii could be blocked only by d-mannose, GlcNAc, and chitin hydrolysate and not by other carbohydrates tested. In contrast, yeast agglutination was not affected by GlcNAc or chitin hydrolysate. This finding indicated mannose residues to be essential for both yeast agglutination and invasion, whereas GlcNAc (oligomer) residues of host cells are involved exclusively in invasion. These results showed the fim determinant of C. freundii to be responsible for d-mannose- and GlcNAc-dependent in vitro invasion without being assembled into pili and for crossing of the blood-brain barrier in the infant rat model.
Collapse
Affiliation(s)
- Petra Hess
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - Artur Altenhöfer
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - A. Salam Khan
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - Neda Daryab
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - Kwang Sik Kim
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - Jörg Hacker
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
| | - Tobias A. Oelschlaeger
- Institut für Molekulare Infektionsbiologie, University of Würzburg, Würzburg, Germany, Division of Infectious Diseases, Children's Hospital of Los Angeles and University of Southern California, Los Angeles, California
- Corresponding author. Mailing address: Institut für Molekulare Infektionsbiologie, Röntgenring 11, 97070 Würzburg, Germany. Phone: 931 312150. Fax: 931 312578. E-mail:
| |
Collapse
|
47
|
Kaniuk NA, Vinogradov E, Li J, Monteiro MA, Whitfield C. Chromosomal and plasmid-encoded enzymes are required for assembly of the R3-type core oligosaccharide in the lipopolysaccharide of Escherichia coli O157:H7. J Biol Chem 2004; 279:31237-50. [PMID: 15155763 DOI: 10.1074/jbc.m401879200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type R3 core oligosaccharide predominates in the lipopolysaccharides from enterohemorrhagic Escherichia coli isolates including O157:H7. The R3 core biosynthesis (waa) genetic locus contains two genes, waaD and waaJ, that are predicted to encode glycosyltransferases involved in completion of the outer core. Through determination of the structures of the lipopolysaccharide core in precise mutants and biochemical analyses of enzyme activities, WaaJ was shown to be a UDP-glucose:(galactosyl) lipopolysaccharide alpha-1,2-glucosyltransferase, and WaaD was shown to be a UDP-glucose:(glucosyl)lipopolysaccharide alpha-1,2-glucosyltransferase. The residue added by WaaJ was identified as the ligation site for O polysaccharide, and this was confirmed by determination of the structure of the linkage region in serotype O157 lipopolysaccharide. The initial O157 repeat unit begins with an N-acetylgalactosamine residue in a beta-anomeric configuration, whereas the biological repeat unit for O157 contains alpha-linked N-acetylgalactosamine residues. With the characterization of WaaJ and WaaD, the activities of all of the enzymes encoded by the R3 waa locus are either known or predicted from homology data with a high level of confidence. However, when core oligosaccharide structure is considered, the origin of an additional alpha-1,3-linked N-acetylglucosamine residue in the outer core is unknown. The gene responsible for a nonstoichiometric alpha-1,7-linked N-acetylglucosamine substituent in the heptose (inner core) region was identified on the large virulence plasmids of E. coli O157 and Shigella flexneri serotype 2a. This is the first plasmid-encoded core oligosaccharide biosynthesis enzyme reported in E. coli.
Collapse
Affiliation(s)
- Natalia A Kaniuk
- Department of Microbiology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
48
|
Yoon JW, Minnich SA, Ahn JS, Park YH, Paszczynski A, Hovde CJ. Thermoregulation of the Escherichia coli O157:H7 pO157 ecf operon and lipid A myristoyl transferase activity involves intrinsically curved DNA. Mol Microbiol 2004; 51:419-35. [PMID: 14756783 DOI: 10.1046/j.1365-2958.2003.03827.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Escherichia coli O157:H7 survives in diverse environments from the ruminant gastrointestinal tract to cool nutrient-dilute water. We hypothesized that the gene regulation required for this flexibility includes intrinsically curved DNA that responds to environmental changes. Three intrinsically curved DNAs were cloned from the E. coli O157:H7 virulence plasmid (pO157), sequenced and designated Bent 1 through Bent 3 (BNT1, BNT2 and BNT3). Compared to BNT1 and BNT3, BNT2 had characteristics typical of intrinsically curved DNA including electrophoretic gel retardation at 4 degrees C, six partially phased adenine:thymine tracts and transcriptional activation. BNT2::lacZ operon fusions showed that BNT2 activated transcription at 24 degrees C compared to 37 degrees C and was partially repressed by a bacterial nucleoid-associated protein H-NS. BNT2 regulated the E. coli attaching and effacing gene-positive conserved fragments 1-4 (ecf1-4) that are conserved in Shiga toxin-producing E. coli associated with human disease. Experimental analyses showed that ecf1-4 formed an operon. ecf1, 2 and 3 encoded putative proteins associated with bacterial surface polysaccharide biosynthesis and invasion and ecf4 complemented a chromosomal deletion of lpxM encoding lipid A myristoyl transferase. Mass spectrometric analysis of lipid A from ecf and lpxM single and double mutants showed that myristoylation was altered at lower temperature.
Collapse
Affiliation(s)
- Jang W Yoon
- Department of Microbiology, Molecular Biology and Biochemistry, University of Idaho, Moscow, ID 83844-3052, USA
| | | | | | | | | | | |
Collapse
|
49
|
Kim SH, Jia W, Bishop RE, Gyles C. An msbB homologue carried in plasmid pO157 encodes an acyltransferase involved in lipid A biosynthesis in Escherichia coli O157:H7. Infect Immun 2004; 72:1174-80. [PMID: 14742570 PMCID: PMC321574 DOI: 10.1128/iai.72.2.1174-1180.2004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Escherichia coli O157:H7 carries a chromosomal msbB1 and a plasmid-encoded msbB2 gene. We characterized msbB2 function as a homologue of msbB1 by examination of wild-type organisms and mutant strains that lacked functional msbB1, msbB2, and both msbB1 and msbB2. The msbB double-mutant strain generated pentaacyl lipid A, while the single-mutant strains synthesized hexaacyl lipid A. Complementation with overexpressed msbB2 converted pentaacyl into hexaacyl lipid A in the double-mutant strain. The transcription of both msbB genes occurred simultaneously. Lack of MsbB2 activity slightly increased the microheterogeneity of the lipid A species. These results suggest that the msbB2 gene plays a role not only in the routine generation of fully hexaacylated lipid A but also in suppressing the microheterogeneity of lipid A species, the endotoxic determinant of the organism.
Collapse
Affiliation(s)
- Sang-Hyun Kim
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada N1G 2W1
| | | | | | | |
Collapse
|
50
|
Khan NA, Shin S, Chung JW, Kim KJ, Elliott S, Wang Y, Kim KS. Outer membrane protein A and cytotoxic necrotizing factor-1 use diverse signaling mechanisms for Escherichia coli K1 invasion of human brain microvascular endothelial cells. Microb Pathog 2003; 35:35-42. [PMID: 12860457 DOI: 10.1016/s0882-4010(03)00090-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Escherichia coli K1 invasion of brain microvascular endothelial cells (BMEC) is a prerequisite for penetration into the central nervous system. We previously have shown that outer membrane protein A (OmpA) and cytotoxic necrotizing factor-1 (CNF1) contribute to E. coli K1 invasion of BMEC. In this study we constructed a double-knockout mutant by deleting ompA and cnf1. We demonstrated that the double-knockout mutant was significantly less invasive in human BMEC as compared with its individual Delta ompA and Delta cnf1 mutants, suggesting that the contributions of OmpA and CNF1 to BMEC invasion are independent of each other. In addition, we showed that OmpA treatment of human BMEC resulted in phosphatidylinositol 3-kinase (PI3K) activation with no effect on RhoA, while CNF1 treatment resulted in RhoA activation with no effect on PI3K, supporting the concept that OmpA and CNF1 contribute to E. coli K1 invasion of BMEC using different mechanisms. This concept was further confirmed by using both PI3K inhibitor (LY294002) and Rho kinase inhibitor (Y27632), which exhibited additive effects on inhibiting E. coli K1 invasion of BMEC. We isolated a 96KD OmpA interacting human BMEC protein by affinity chromatography using purified OmpA, which was identified as gp96 protein, a member of the HSP90 family. This receptor differed from the CNF1 receptor (37LRP) identified from human BMEC. Taken together, these data indicate that OmpA and CNF1 contribute to E. coli K1 invasion of BMEC in an additive manner by interacting with different BMEC receptors and using diverse host cell signaling mechanisms.
Collapse
Affiliation(s)
- Naveed Ahmed Khan
- Division of Pediatric Infectious Diseases, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | |
Collapse
|