1
|
Pagliuca C, Colicchio R, Resta SC, Talà A, Scaglione E, Mantova G, Continisio L, Pagliarulo C, Bucci C, Alifano P, Salvatore P. Neisseria meningitidis activates pyroptotic pathways in a mouse model of meningitis: role of a two-partner secretion system. Front Cell Infect Microbiol 2024; 14:1384072. [PMID: 39376663 PMCID: PMC11456522 DOI: 10.3389/fcimb.2024.1384072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024] Open
Abstract
There is evidence that in infected cells in vitro the meningococcal HrpA/HrpB two-partner secretion system (TPS) mediates the exit of bacteria from the internalization vacuole and the docking of bacteria to the dynein motor resulting in the induction of pyroptosis. In this study we set out to study the role of the HrpA/HrpB TPS in establishing meningitis and activating pyroptotic pathways in an animal model of meningitis using a reference serogroup C meningococcal strain, 93/4286, and an isogenic hrpB knockout mutant, 93/4286ΩhrpB. Survival experiments confirmed the role of HrpA/HrpB TPS in the invasive meningococcal disease. In fact, the ability of the hrpB mutant to replicate in brain and spread systemically was impaired in mice infected with hrpB mutant. Furthermore, western blot analysis of brain samples during the infection demonstrated that: i. N. meningitidis activated canonical and non-canonical inflammasome pyroptosis pathways in the mouse brain; ii. the activation of caspase-11, caspase-1, and gasdermin-D was markedly reduced in the hrpB mutant; iii. the increase in the amount of IL-1β and IL-18, which are an important end point of pyroptosis, occurs in the brains of mice infected with the wild-type strain 93/4286 and is strongly reduced in those infected with 93/4286ΩhrpB. In particular, the activation of caspase 11, which is triggered by cytosolic lipopolysaccharide, indicates that during meningococcal infection pyroptosis is induced by intracellular infection after the exit of the bacteria from the internalizing vacuole, a process that is hindered in the hrpB mutant. Overall, these results confirm, in an animal model, that the HrpA/HrpB TPS plays a role in the induction of pyroptosis and suggest a pivotal involvement of pyroptosis in invasive meningococcal disease, paving the way for the use of pyroptosis inhibitors in the adjuvant therapy of the disease.
Collapse
Affiliation(s)
- Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Mantova
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
| | - Leonardo Continisio
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Caterina Pagliarulo
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Pietro Alifano
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotecnologies, University of Naples "Federico II", Naples, Italy
- The Institute CEINGE-Biotecnologie Avanzate Franco Salvatore s.c.ar.l., Naples, Italy
- Task Force on Microbiome Studies, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
2
|
Talà A, Guerra F, Calcagnile M, Romano R, Resta SC, Paiano A, Chiariello M, Pizzolante G, Bucci C, Alifano P. HrpA anchors meningococci to the dynein motor and affects the balance between apoptosis and pyroptosis. J Biomed Sci 2022; 29:45. [PMID: 35765029 PMCID: PMC9241232 DOI: 10.1186/s12929-022-00829-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In Neisseria meningitidis the HrpA/HrpB two-partner secretion system (TPS) was implicated in diverse functions including meningococcal competition, biofilm formation, adherence to epithelial cells, intracellular survival and vacuolar escape. These diverse functions could be attributed to distinct domains of secreted HrpA. METHODS A yeast two-hybrid screening, in vitro pull-down assay and immunofluorescence microscopy experiments were used to investigate the interaction between HrpA and the dynein light-chain, Tctex-type 1 (DYNLT1). In silico modeling was used to analyze HrpA structure. Western blot analysis was used to investigate apoptotic and pyroptotic markers. RESULTS The HrpA carboxy-terminal region acts as a manganese-dependent cell lysin, while the results of a yeast two-hybrid screening demonstrated that the HrpA middle region has the ability to bind the dynein light-chain, Tctex-type 1 (DYNLT1). This interaction was confirmed by in vitro pull-down assay and immunofluorescence microscopy experiments showing co-localization of N. meningitidis with DYNLT1 in infected epithelial cells. In silico modeling revealed that the HrpA-M interface interacting with the DYNLT1 has similarity with capsid proteins of neurotropic viruses that interact with the DYNLT1. Indeed, we found that HrpA plays a key role in infection of and meningococcal trafficking within neuronal cells, and is implicated in the modulation of the balance between apoptosis and pyroptosis. CONCLUSIONS Our findings revealed that N. meningitidis is able to effectively infect and survive in neuronal cells, and that this ability is dependent on HrpA, which establishes a direct protein-protein interaction with DYNLTI in these cells, suggesting that the HrpA interaction with dynein could be fundamental for N. meningitidis spreading inside the neurons. Moreover, we found that the balance between apoptotic and pyroptotic pathways is heavily affected by HrpA.
Collapse
Affiliation(s)
- Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Matteo Calcagnile
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Silvia Caterina Resta
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Aurora Paiano
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Mario Chiariello
- Core Research Laboratory-Siena, Institute for Cancer Research and Prevention (ISPRO), 53100, Siena, Italy.,Institute of Clinical Physiology (IFC), National Research Council (CNR), 53100, Siena, Italy
| | - Graziano Pizzolante
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy.
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100, Lecce, Italy.
| |
Collapse
|
3
|
Surface architecture of Neisseria meningitidis capsule and outer membrane as revealed by Atomic Force Microscopy. Res Microbiol 2021; 172:103865. [PMID: 34284091 DOI: 10.1016/j.resmic.2021.103865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 11/21/2022]
Abstract
An extensive morphological analysis of the Neisseria meningitidis cell envelope, including serogroup B capsule and outer membrane, based on atomic force microscopy (AFM) together with mechanical characterization by force spectroscopic measurements, has been carried out. Three meningococcal strains were used: the encapsulated serogroup B strain B1940, and the isogenic mutants B1940 siaD(+C) (lacking capsule), and B1940 cps (lacking both capsule and lipooligosaccharide outer core). regularly structured AFM experiments with the encapsulated strain B1940 provided unprecedented images of the meningococcal capsule, which seems to be characterized by protrusions ("bumps") with the lateral dimensions of about 30 nm. Measurement of the Young's modulus provided quantitative assessment of the property of the capsule to confer resistance to mechanical stress. Moreover, Raman spectroscopy gave a fingerprint by which it was possible to identify the specific molecular species of the three strains analyzed, and to highlight major differences between them.
Collapse
|
4
|
Al Suwayyid BA, Rankine-Wilson L, Speers DJ, Wise MJ, Coombs GW, Kahler CM. Meningococcal Disease-Associated Prophage-Like Elements Are Present in Neisseria gonorrhoeae and Some Commensal Neisseria Species. Genome Biol Evol 2020; 12:3938-3950. [PMID: 32031617 PMCID: PMC7058167 DOI: 10.1093/gbe/evaa023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2020] [Indexed: 12/14/2022] Open
Abstract
Neisseria spp. possess four genogroups of filamentous prophages, termed Nf1 to 4. A filamentous bacteriophage from the Nf1 genogroup termed meningococcal disease-associated phage (MDA φ) is associated with clonal complexes of Neisseria meningitidis that cause invasive meningococcal disease. Recently, we recovered an isolate of Neisseria gonorrhoeae (ExNg63) from a rare case of gonococcal meningitis, and found that it possessed a region with 90% similarity to Nf1 prophages, specifically, the meningococcal MDA φ. This led to the hypothesis that the Nf1 prophage may be more widely distributed amongst the genus Neisseria. An analysis of 92 reference genomes revealed the presence of intact Nf1 prophages in the commensal species, Neisseria lactamica and Neisseria cinerea in addition to the pathogen N. gonorrhoeae. In N. gonorrhoeae, Nf1 prophages had a restricted distribution but were present in all representatives of MLST ST1918. Of the 160 phage integration sites identified, only one common insertion site was found between one isolate of N. gonorrhoeae and N. meningitidis. There was an absence of any obvious conservation of the receptor for prophage entry, PilE, suggesting that the phage may have been obtained by natural transformation. An examination of the restriction modification systems and mutated mismatch repair systems with prophage presence suggested that there was no obvious preference for these hosts. A timed phylogeny inferred that N. meningitidis was the donor of the Nf1 prophages in N. lactamica and N. gonorrhoeae. Further work is required to determine whether Nf1 prophages are active and can act as accessory colonization factors in these species.
Collapse
Affiliation(s)
- Barakat A Al Suwayyid
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,School of Biomedical Sciences, The University of Western Australia, Crawley, Australia.,Ministry of Education, Riyadh, Saudi Arabia
| | - Leah Rankine-Wilson
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
| | - David J Speers
- Department of Microbiology, PathWest Laboratory Medicine WA, Queen Elizabeth II Medical Centre, Nedlands, Australia.,School of Medicine and Pharmacology, University of Western Australia, Crawley, Australia
| | - Michael J Wise
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,Computer Science and Software Engineering, The University of Western Australia, Crawley, Australia
| | - Geoffrey W Coombs
- Department of Microbiology, PathWest Laboratory Medicine WA, Fiona Stanley Hospital, Murdoch, Australia.,Antimicrobial Resistance and Infectious Diseases Research Laboratory, School of Veterinary Life Sciences, Murdoch University, Australia
| | - Charlene M Kahler
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Australia.,School of Biomedical Sciences, The University of Western Australia, Crawley, Australia
| |
Collapse
|
5
|
Sánchez-Busó L, Golparian D, Parkhill J, Unemo M, Harris SR. Genetic variation regulates the activation and specificity of Restriction-Modification systems in Neisseria gonorrhoeae. Sci Rep 2019; 9:14685. [PMID: 31605008 PMCID: PMC6789123 DOI: 10.1038/s41598-019-51102-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/25/2019] [Indexed: 01/14/2023] Open
Abstract
Restriction-Modification systems (RMS) are one of the main mechanisms of defence against foreign DNA invasion and can have an important role in the regulation of gene expression. The obligate human pathogen Neisseria gonorrhoeae carries one of the highest loads of RMS in its genome; between 13 to 15 of the three main types. Previous work has described their organization in the reference genome FA1090 and has inferred the associated methylated motifs. Here, we studied the structure of RMS and target methylated motifs in 25 gonococcal strains sequenced with Single Molecule Real-Time (SMRT) technology, which provides data on DNA modification. The results showed a variable picture of active RMS in different strains, with phase variation switching the activity of Type III RMS, and both the activity and specificity of a Type I RMS. Interestingly, the Dam methylase was found in place of the NgoAXI endonuclease in two of the strains, despite being previously thought to be absent in the gonococcus. We also identified the real methylation target of NgoAXII as 5′-GCAGA-3′, different from that previously described. Results from this work give further insights into the diversity and dynamics of RMS and methylation patterns in N. gonorrhoeae.
Collapse
Affiliation(s)
- Leonor Sánchez-Busó
- Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK. .,Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Daniel Golparian
- WHO Collaborating Centre for Gonorrhoea and other Sexually Transmitted Infections, National Reference Laboratory for Sexually Transmitted Infections, Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and other Sexually Transmitted Infections, National Reference Laboratory for Sexually Transmitted Infections, Department of Laboratory Medicine, Clinical Microbiology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Simon R Harris
- Microbiotica Ltd, Biodata Innovation Centre, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
6
|
Colicchio R, Pagliuca C, Ricci S, Scaglione E, Grandgirard D, Masouris I, Farina F, Pagliarulo C, Mantova G, Paragliola L, Leib SL, Koedel U, Pozzi G, Alifano P, Salvatore P. Virulence Traits of a Serogroup C Meningococcus and Isogenic cssA Mutant, Defective in Surface-Exposed Sialic Acid, in a Murine Model of Meningitis. Infect Immun 2019; 87:e00688-18. [PMID: 30718288 PMCID: PMC6434112 DOI: 10.1128/iai.00688-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022] Open
Abstract
In serogroup C Neisseria meningitidis, the cssA (siaA) gene codes for an UDP-N-acetylglucosamine 2-epimerase that catalyzes the conversion of UDP-N-acetyl-α-d-glucosamine into N-acetyl-d-mannosamine and UDP in the first step in sialic acid biosynthesis. This enzyme is required for the biosynthesis of the (α2→9)-linked polysialic acid capsule and for lipooligosaccharide (LOS) sialylation. In this study, we have used a reference serogroup C meningococcal strain and an isogenic cssA knockout mutant to investigate the pathogenetic role of surface-exposed sialic acids in a model of meningitis based on intracisternal inoculation of BALB/c mice. Results confirmed the key role of surface-exposed sialic acids in meningococcal pathogenesis. The 50% lethal dose (LD50) of the wild-type strain 93/4286 was about four orders of magnitude lower than that of the cssA mutant. Compared to the wild-type strain, the ability of this mutant to replicate in brain and spread systemically was severely impaired. Evaluation of brain damage evidenced a significant reduction in cerebral hemorrhages in mice infected with the mutant in comparison with the levels in those challenged with the wild-type strain. Histological analysis showed the typical features of bacterial meningitis, including inflammatory cells in the subarachnoid, perivascular, and ventricular spaces especially in animals infected with the wild type. Noticeably, 80% of mice infected with the wild-type strain presented with massive bacterial localization and accompanying inflammatory infiltrate in the corpus callosum, indicating high tropism of meningococci exposing sialic acids toward this brain structure and a specific involvement of the corpus callosum in the mouse model of meningococcal meningitis.
Collapse
Affiliation(s)
- Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Susanna Ricci
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Denis Grandgirard
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Ilias Masouris
- Department of Neurology, Ludwig Maximilians University of Munich, Munich, Germany
| | - Fabrizio Farina
- Department of Law, Economics, Management and Quantitative Methods, University of Sannio, Benevento, Italy
| | | | - Giuseppe Mantova
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Laura Paragliola
- Department of Integrated Activity of Laboratory Medicine and Transfusion, Complex Operative Unit of Clinical Microbiology, University Hospital Federico II, Naples, Italy
| | - Stephen L Leib
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Uwe Koedel
- Department of Neurology, Ludwig Maximilians University of Munich, Munich, Germany
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
- Department of Integrated Activity of Laboratory Medicine and Transfusion, Complex Operative Unit of Clinical Microbiology, University Hospital Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate s.c.ar.l., Naples, Italy
| |
Collapse
|
7
|
Wang H, Xing X, Wang J, Pang B, Liu M, Larios-Valencia J, Liu T, Liu G, Xie S, Hao G, Liu Z, Kan B, Zhu J. Hypermutation-induced in vivo oxidative stress resistance enhances Vibrio cholerae host adaptation. PLoS Pathog 2018; 14:e1007413. [PMID: 30376582 PMCID: PMC6226196 DOI: 10.1371/journal.ppat.1007413] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/09/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023] Open
Abstract
Bacterial pathogens are highly adaptable organisms, a quality that enables them to overcome changing hostile environments. For example, Vibrio cholerae, the causative agent of cholera, is able to colonize host small intestines and combat host-produced reactive oxygen species (ROS) during infection. To dissect the molecular mechanisms utilized by V. cholerae to overcome ROS in vivo, we performed a whole-genome transposon sequencing analysis (Tn-seq) by comparing gene requirements for colonization using adult mice with and without the treatment of the antioxidant, N-acetyl cysteine. We found that mutants of the methyl-directed mismatch repair (MMR) system, such as MutS, displayed significant colonization advantages in untreated, ROS-rich mice, but not in NAC-treated mice. Further analyses suggest that the accumulation of both catalase-overproducing mutants and rugose colony variants in NAC- mice was the leading cause of mutS mutant enrichment caused by oxidative stress during infection. We also found that rugose variants could revert back to smooth colonies upon aerobic, in vitro culture. Additionally, the mutation rate of wildtype colonized in NAC- mice was significantly higher than that in NAC+ mice. Taken together, these findings support a paradigm in which V. cholerae employs a temporal adaptive strategy to battle ROS during infection, resulting in enriched phenotypes. Moreover, ΔmutS passage and complementation can be used to model hypermuation in diverse pathogens to identify novel stress resistance mechanisms. Cholera is a devastating diarrheal disease that is still endemic to many developing nations, with the worst outbreak in history having occurred recently in Yemen. Vibrio cholerae, the causative agent of cholera, transitions from aquatic reservoirs to the human gastrointestinal tract, where it expresses virulence factors to facilitate colonization of the small intestines and to combat host innate immune effectors, such as reactive oxygen species (ROS). We applied a genome-wide transposon screen (Tn-seq) and identified that deletion of mutS, which is part of DNA mismatch repair system, drastically increased colonization in ROS-rich mice. The deletion of mutS led to the accumulation of catalase-overproducing mutants and a high frequency rugose phenotype when exposed to ROS selective pressures in vivo. Additionally, ROS elevated mutation frequency in wildtype, both in vitro and in vivo. Our data imply that V. cholerae may modulate mutation frequency as a temporal adaptive strategy to overcome oxidative stress and to enhance infectivity.
Collapse
Affiliation(s)
- Hui Wang
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
- * E-mail: (HW); (JH)
| | - Xiaolin Xing
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Jipeng Wang
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Bo Pang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ming Liu
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Jessie Larios-Valencia
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
| | - Tao Liu
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Ge Liu
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Saijun Xie
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Guijuan Hao
- Department of Microbiology, Nanjing Agricultural University, Nanjing, China
| | - Zhi Liu
- Department of Biotechnology, Huazhong University of Science and Technology, Wuhan, China
| | - Biao Kan
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jun Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States of America
- * E-mail: (HW); (JH)
| |
Collapse
|
8
|
Ortiz-Velez L, Ortiz-Villalobos J, Schulman A, Oh JH, van Pijkeren JP, Britton RA. Genome alterations associated with improved transformation efficiency in Lactobacillus reuteri. Microb Cell Fact 2018; 17:138. [PMID: 30176942 PMCID: PMC6122466 DOI: 10.1186/s12934-018-0986-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022] Open
Abstract
Background Lactic acid bacteria (LAB) are one of the microorganisms of choice for the development of protein delivery systems for therapeutic purposes. Although there are numerous tools to facilitate genome engineering of lactobacilli; transformation efficiency still limits the ability to engineer their genomes. While genetically manipulating Lactobacillus reuteri ATCC PTA 6475 (LR 6475), we noticed that after an initial transformation, several LR 6475 strains significantly improved their ability to take up plasmid DNA via electroporation. Our goal was to understand the molecular basis for how these strains acquired the ability to increase transformation efficiency. Results Strains generated after transformation of plasmids pJP067 and pJP042 increased their ability to transform plasmid DNA about one million fold for pJP067, 100-fold for pSIP411 and tenfold for pNZ8048. Upon sequencing of the whole genome from these strains, we identified several genomic mutations and rearrangements, with all strains containing mutations in the transformation related gene A (trgA). To evaluate the role of trgA in transformation of DNA, we generated a trgA null that improved the transformation efficiency of LR 6475 to transform pSIP411 and pJP067 by at least 100-fold, demonstrating that trgA significantly impairs the ability of LR 6475 to take-up plasmid DNA. We also identified genomic rearrangements located in and around two prophages inserted in the LR 6475 genome that included deletions, insertions and an inversion of 336 Kb. A second group of rearrangements was observed in a Type I restriction modification system, in which the specificity subunits underwent several rearrangements in the target recognition domain. Despite the magnitude of these rearrangements in the prophage genomes and restriction modification systems, none of these genomic changes impacted transformation efficiency to the level induced by trgA. Conclusions Our findings demonstrate how genetic manipulation of LR 6475 with plasmid DNA leads to genomic changes that improve their ability to transform plasmid DNA; highlighting trgA as the primary driver of this phenotype. Additionally, this study also underlines the importance of characterizing genetic changes that take place after genome engineering of strains for therapeutic purposes. Electronic supplementary material The online version of this article (10.1186/s12934-018-0986-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Ortiz-Velez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | | | - Abby Schulman
- Department of Cognitive Sciences, Rice University, Houston, TX, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| |
Collapse
|
9
|
Lees JA, Kremer PHC, Manso AS, Croucher NJ, Ferwerda B, Serón MV, Oggioni MR, Parkhill J, Brouwer MC, van der Ende A, van de Beek D, Bentley SD. Large scale genomic analysis shows no evidence for pathogen adaptation between the blood and cerebrospinal fluid niches during bacterial meningitis. Microb Genom 2017; 3:e000103. [PMID: 28348877 PMCID: PMC5361624 DOI: 10.1099/mgen.0.000103] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 12/18/2016] [Indexed: 12/26/2022] Open
Abstract
Recent studies have provided evidence for rapid pathogen genome diversification, some of which could potentially affect the course of disease. We have previously described such variation seen between isolates infecting the blood and cerebrospinal fluid (CSF) of a single patient during a case of bacterial meningitis. Here, we performed whole-genome sequencing of paired isolates from the blood and CSF of 869 meningitis patients to determine whether such variation frequently occurs between these two niches in cases of bacterial meningitis. Using a combination of reference-free variant calling approaches, we show that no genetic adaptation occurs in either invaded niche during bacterial meningitis for two major pathogen species, Streptococcus pneumoniae and Neisseria meningitidis. This study therefore shows that the bacteria capable of causing meningitis are already able to do this upon entering the blood, and no further sequence change is necessary to cross the blood–brain barrier. Our findings place the focus back on bacterial evolution between nasopharyngeal carriage and invasion, or diversity of the host, as likely mechanisms for determining invasiveness.
Collapse
Affiliation(s)
- John A Lees
- 1Pathogen Genomics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Philip H C Kremer
- 2Department of Neurology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Ana S Manso
- 3Department of Genetics, University of Leicester, Leicester, UK
| | - Nicholas J Croucher
- 4Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Bart Ferwerda
- 2Department of Neurology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Mercedes Valls Serón
- 2Department of Neurology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Marco R Oggioni
- 3Department of Genetics, University of Leicester, Leicester, UK
| | - Julian Parkhill
- 1Pathogen Genomics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Matthijs C Brouwer
- 2Department of Neurology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Arie van der Ende
- 5Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands.,6Netherlands Reference Laboratory for Bacterial Meningitis, Academic Medical Center, Amsterdam, The Netherlands
| | - Diederik van de Beek
- 2Department of Neurology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
10
|
Esson D, Mather AE, Scanlan E, Gupta S, de Vries SPW, Bailey D, Harris SR, McKinley TJ, Méric G, Berry SK, Mastroeni P, Sheppard SK, Christie G, Thomson NR, Parkhill J, Maskell DJ, Grant AJ. Genomic variations leading to alterations in cell morphology of Campylobacter spp. Sci Rep 2016; 6:38303. [PMID: 27910897 PMCID: PMC5133587 DOI: 10.1038/srep38303] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/07/2016] [Indexed: 12/14/2022] Open
Abstract
Campylobacter jejuni, the most common cause of bacterial diarrhoeal disease, is normally helical. However, it can also adopt straight rod, elongated helical and coccoid forms. Studying how helical morphology is generated, and how it switches between its different forms, is an important objective for understanding this pathogen. Here, we aimed to determine the genetic factors involved in generating the helical shape of Campylobacter. A C. jejuni transposon (Tn) mutant library was screened for non-helical mutants with inconsistent results. Whole genome sequence variation and morphological trends within this Tn library, and in various C. jejuni wild type strains, were compared and correlated to detect genomic elements associated with helical and rod morphologies. All rod-shaped C. jejuni Tn mutants and all rod-shaped laboratory, clinical and environmental C. jejuni and Campylobacter coli contained genetic changes within the pgp1 or pgp2 genes, which encode peptidoglycan modifying enzymes. We therefore confirm the importance of Pgp1 and Pgp2 in the maintenance of helical shape and extended this to a wide range of C. jejuni and C. coli isolates. Genome sequence analysis revealed variation in the sequence and length of homopolymeric tracts found within these genes, providing a potential mechanism of phase variation of cell shape.
Collapse
Affiliation(s)
- Diane Esson
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Alison E. Mather
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Eoin Scanlan
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Srishti Gupta
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Stefan P. W. de Vries
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - David Bailey
- Department of Chemical Engineering and Biotechnology, University of Cambridge, New Museums Site, Pembroke Street, Cambridge, UK
| | - Simon R. Harris
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Trevelyan J. McKinley
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Guillaume Méric
- The Milner Centre for Evolution, Department of Biology and Biotechnology, University of Bath, Claverton Down, Bath, UK
| | - Sophia K. Berry
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Samuel K. Sheppard
- The Milner Centre for Evolution, Department of Biology and Biotechnology, University of Bath, Claverton Down, Bath, UK
| | - Graham Christie
- Department of Chemical Engineering and Biotechnology, University of Cambridge, New Museums Site, Pembroke Street, Cambridge, UK
| | - Nicholas R. Thomson
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
- The London School of Hygiene and Tropical Medicine, London, UK
| | - Julian Parkhill
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Duncan J. Maskell
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| | - Andrew J. Grant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, UK
| |
Collapse
|
11
|
Siena E, D’Aurizio R, Riley D, Tettelin H, Guidotti S, Torricelli G, Moxon ER, Medini D. In-silico prediction and deep-DNA sequencing validation indicate phase variation in 115 Neisseria meningitidis genes. BMC Genomics 2016; 17:843. [PMID: 27793092 PMCID: PMC5084427 DOI: 10.1186/s12864-016-3185-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 10/22/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The Neisseria meningitidis (Nm) chromosome shows a high abundance of simple sequence DNA repeats (SSRs) that undergo stochastic, reversible mutations at high frequency. This mechanism is reflected in an extensive phenotypic diversity that facilitates Nm adaptation to dynamic environmental changes. To date, phase-variable phenotypes mediated by SSRs variation have been experimentally confirmed for 26 Nm genes. RESULTS Here we present a population-scale comparative genomic analysis that identified 277 genes and classified them into 52 strong, 60 moderate and 165 weak candidates for phase variation. Deep-coverage DNA sequencing of single colonies grown overnight under non-selective conditions confirmed the presence of high-frequency, stochastic variation in 115 of them, providing circumstantial evidence for their phase variability. We confirmed previous observations of a predominance of variable SSRs within genes for components located on the cell surface or DNA metabolism. However, in addition we identified an unexpectedly broad spectrum of other metabolic functions, and most of the variable SSRs were predicted to induce phenotypic changes by modulating gene expression at a transcriptional level or by producing different protein isoforms rather than mediating on/off translational switching through frameshifts. Investigation of the evolutionary history of SSR contingency loci revealed that these loci were inherited from a Nm ancestor, evolved independently within Nm, or were acquired by Nm through lateral DNA exchange. CONCLUSIONS Overall, our results have identified a broader and qualitatively different phenotypic diversification of SSRs-mediated stochastic variation than previously documented, including its impact on central Nm metabolism.
Collapse
Affiliation(s)
| | - Romina D’Aurizio
- GSK Vaccines, 53100 Siena, Italy
- Present address: Institute of Informatics and Telematics and Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| | - David Riley
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
- Present address: Personal Genome Disgnostics inc., Baltimore, MD 21224 USA
| | - Hervé Tettelin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | | | | | - E. Richard Moxon
- Medical Sciences Division, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, OX3 9DS UK
| | | |
Collapse
|
12
|
D'Argenio V, Casaburi G, Precone V, Pagliuca C, Colicchio R, Sarnataro D, Discepolo V, Kim SM, Russo I, Del Vecchio Blanco G, Horner DS, Chiara M, Pesole G, Salvatore P, Monteleone G, Ciacci C, Caporaso GJ, Jabrì B, Salvatore F, Sacchetti L. Metagenomics Reveals Dysbiosis and a Potentially Pathogenic N. flavescens Strain in Duodenum of Adult Celiac Patients. Am J Gastroenterol 2016; 111:879-90. [PMID: 27045926 PMCID: PMC4897008 DOI: 10.1038/ajg.2016.95] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Celiac disease (CD)-associated duodenal dysbiosis has not yet been clearly defined, and the mechanisms by which CD-associated dysbiosis could concur to CD development or exacerbation are unknown. In this study, we analyzed the duodenal microbiome of CD patients. METHODS The microbiome was evaluated in duodenal biopsy samples of 20 adult patients with active CD, 6 CD patients on a gluten-free diet, and 15 controls by DNA sequencing of 16S ribosomal RNA libraries. Bacterial species were cultured, isolated and identified by mass spectrometry. Isolated bacterial species were used to infect CaCo-2 cells, and to stimulate normal duodenal explants and cultured human and murine dendritic cells (DCs). Inflammatory markers and cytokines were evaluated by immunofluorescence and ELISA, respectively. RESULTS Proteobacteria was the most abundant and Firmicutes and Actinobacteria the least abundant phyla in the microbiome profiles of active CD patients. Members of the Neisseria genus (Betaproteobacteria class) were significantly more abundant in active CD patients than in the other two groups (P=0.03). Neisseria flavescens (CD-Nf) was the most abundant Neisseria species in active CD duodenum. Whole-genome sequencing of CD-Nf and control-Nf showed genetic diversity of the iron acquisition systems and of some hemoglobin-related genes. CD-Nf was able to escape the lysosomal compartment in CaCo-2 cells and to induce an inflammatory response in DCs and in ex-vivo mucosal explants. CONCLUSIONS Marked dysbiosis and an abundance of a peculiar CD-Nf strain characterize the duodenal microbiome in active CD patients thus suggesting that the CD-associated microbiota could contribute to the many inflammatory signals in this disorder.
Collapse
Affiliation(s)
- Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | | | - Vincenza Precone
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Chiara Pagliuca
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Daniela Sarnataro
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Valentina Discepolo
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Medical Translational Sciences and European Laboratory for the Investigation of Food Induced Diseases, University of Naples Federico II, Naples, Italy
| | - Sangman M Kim
- Department of Medicine and the University of Chicago Celiac Disease Center, University of Chicago, Chicago, Illinois, USA
| | - Ilaria Russo
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | | | - David S Horner
- Department of Biosciences, University of Milan, Milan, Italy
| | - Matteo Chiara
- Department of Biosciences, University of Milan, Milan, Italy
| | - Graziano Pesole
- Institute of Biomembranes and Bioenergetics, National Research Council, Bari, Italy.,Department of Biochemistry and Molecular Biology, University of Bari A. Moro, Bari, Italy
| | - Paola Salvatore
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | | | - Carolina Ciacci
- Department of Medicine and Surgery, University of Salerno, Salerno, Italy
| | | | - Bana Jabrì
- Department of Medicine and the University of Chicago Celiac Disease Center, University of Chicago, Chicago, Illinois, USA
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.,IRCCS-Fondazione SDN, Naples, Italy
| | | |
Collapse
|
13
|
Colicchio R, Pagliuca C, Pastore G, Cicatiello AG, Pagliarulo C, Talà A, Scaglione E, Sammartino JC, Bucci C, Alifano P, Salvatore P. Fitness Cost of Rifampin Resistance in Neisseria meningitidis: In Vitro Study of Mechanisms Associated with rpoB H553Y Mutation. Antimicrob Agents Chemother 2015; 59:7637-49. [PMID: 26416867 PMCID: PMC4649176 DOI: 10.1128/aac.01746-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/23/2015] [Indexed: 12/14/2022] Open
Abstract
Rifampin chemoprophylaxis against Neisseria meningitidis infections led to the onset of rifampin resistance in clinical isolates harboring point mutations in the rpoB gene, coding for the RNA polymerase β chain. These resistant strains are rare in medical practice, suggesting their decreased fitness in the human host. In this study, we isolated rifampin-resistant rpoB mutants from hypervirulent serogroup C strain 93/4286 and analyzed their different properties, including the ability to grow/survive in different culture media and in differentiated THP-1 human monocytes and to compete with the wild-type strain in vitro. Our results demonstrate that different rpoB mutations (H553Y, H553R, and S549F) may have different effects, ranging from low- to high-cost effects, on bacterial fitness in vitro. Moreover, we found that the S549F mutation confers temperature sensitivity, possibly explaining why it is observed very rarely in clinical isolates. Comparative high-throughput RNA sequencing analysis of bacteria grown in chemically defined medium demonstrated that the low-cost H553Y substitution resulted in global transcriptional changes that functionally mimic the stringent response. Interestingly, many virulence-associated genes, including those coding for meningococcal type IV pili, porin A, adhesins/invasins, IgA protease, two-partner secretion system HrpA/HrpB, enzymes involved in resistance to oxidative injury, lipooligosaccharide sialylation, and capsular polysaccharide biosynthesis, were downregulated in the H553Y mutant compared to their level of expression in the wild-type strain. These data might account for the reduced capacity of this mutant to grow/survive in differentiated THP-1 cells and explain the rarity of H553Y mutants among clinical isolates.
Collapse
Affiliation(s)
- Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology, Federico II University Medical School, Naples, Italy SDN-Foundation, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology, Federico II University Medical School, Naples, Italy Ceinge Advanced Biotechnologies, Naples, Italy
| | - Gabiria Pastore
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | | | - Caterina Pagliarulo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Adelfia Talà
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Elena Scaglione
- Department of Molecular Medicine and Medical Biotechnology, Federico II University Medical School, Naples, Italy
| | - Josè Camilla Sammartino
- Department of Molecular Medicine and Medical Biotechnology, Federico II University Medical School, Naples, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology, Federico II University Medical School, Naples, Italy Ceinge Advanced Biotechnologies, Naples, Italy
| |
Collapse
|
14
|
Bessen DE, McShan WM, Nguyen SV, Shetty A, Agrawal S, Tettelin H. Molecular epidemiology and genomics of group A Streptococcus. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2015; 33:393-418. [PMID: 25460818 PMCID: PMC4416080 DOI: 10.1016/j.meegid.2014.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/11/2014] [Accepted: 10/13/2014] [Indexed: 12/15/2022]
Abstract
Streptococcus pyogenes (group A Streptococcus; GAS) is a strict human pathogen with a very high prevalence worldwide. This review highlights the genetic organization of the species and the important ecological considerations that impact its evolution. Recent advances are presented on the topics of molecular epidemiology, population biology, molecular basis for genetic change, genome structure and genetic flux, phylogenomics and closely related streptococcal species, and the long- and short-term evolution of GAS. The application of whole genome sequence data to addressing key biological questions is discussed.
Collapse
Affiliation(s)
- Debra E Bessen
- Department of Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA.
| | - W Michael McShan
- University of Oklahoma Health Sciences Center, Department of Pharmaceutical Sciences, College of Pharmacy, Oklahoma City, OK 73117, USA.
| | - Scott V Nguyen
- University of Oklahoma Health Sciences Center, Department of Pharmaceutical Sciences, College of Pharmacy, Oklahoma City, OK 73117, USA.
| | - Amol Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Sonia Agrawal
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Hervé Tettelin
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
15
|
Leca M, Bornet C, Montana M, Curti C, Vanelle P. Meningococcal vaccines: Current state and future outlook. ACTA ACUST UNITED AC 2015; 63:144-51. [DOI: 10.1016/j.patbio.2015.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/17/2015] [Indexed: 10/23/2022]
|
16
|
Kwiatek A, Bacal P, Wasiluk A, Trybunko A, Adamczyk-Poplawska M. The dam replacing gene product enhances Neisseria gonorrhoeae FA1090 viability and biofilm formation. Front Microbiol 2014; 5:712. [PMID: 25566225 PMCID: PMC4269198 DOI: 10.3389/fmicb.2014.00712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 11/29/2014] [Indexed: 12/23/2022] Open
Abstract
Many Neisseriaceae do not exhibit Dam methyltransferase activity and, instead of the dam gene, possess drg (dam replacing gene) inserted in the leuS/dam locus. The drg locus in Neisseria gonorrhoeae FA1090 has a lower GC-pairs content (40.5%) compared to the whole genome of N. gonorrhoeae FA1090 (52%). The gonococcal drg gene encodes a DNA endonuclease Drg, with GmeATC specificity. Disruption of drg or insertion of the dam gene in gonococcal genome changes the level of expression of genes as shown by transcriptome analysis. For the drg-deficient N. gonorrhoeae mutant, a total of 195 (8.94% of the total gene pool) genes exhibited an altered expression compared to the wt strain by at least 1.5 fold. In dam-expressing N. gonorrhoeae mutant, the expression of 240 genes (11% of total genes) was deregulated. Most of these deregulated genes were involved in translation, DNA repair, membrane biogenesis and energy production as shown by cluster of orthologous group analysis. In vivo, the inactivation of drg gene causes the decrease of the number of live neisserial cells and long lag phase of growth. The insertion of dam gene instead of drg locus restores cell viability. We have also shown that presence of the drg gene product is important for N. gonorrhoeae FA1090 in adhesion, including human epithelial cells, and biofilm formation. Biofilm produced by drg-deficient strain is formed by more dispersed cells, compared to this one formed by parental strain as shown by scanning electron and confocal microscopy. Also adherence assays show a significantly smaller biomass of formed biofilm (OD570 = 0.242 ± 0.038) for drg-deficient strain, compared to wild-type strain (OD570 = 0.378 ± 0.057). Dam-expressing gonococcal cells produce slightly weaker biofilm with cells embedded in an extracellular matrix. This strain has also a five times reduced ability for adhesion to human epithelial cells. In this context, the presence of Drg is more advantageous for N. gonorrhoeae biology than Dam presence.
Collapse
Affiliation(s)
- Agnieszka Kwiatek
- Department of Virology, Institute of Microbiology, Faculty of Biology, University of Warsaw Warsaw, Poland
| | - Pawel Bacal
- Laboratory of Theory and Applications of Electrodes, Faculty of Chemistry, University of Warsaw Warsaw, Poland
| | - Adrian Wasiluk
- Department of Virology, Institute of Microbiology, Faculty of Biology, University of Warsaw Warsaw, Poland
| | - Anastasiya Trybunko
- Department of Virology, Institute of Microbiology, Faculty of Biology, University of Warsaw Warsaw, Poland
| | - Monika Adamczyk-Poplawska
- Department of Virology, Institute of Microbiology, Faculty of Biology, University of Warsaw Warsaw, Poland
| |
Collapse
|
17
|
Recombination drives genome evolution in outbreak-related Legionella pneumophila isolates. Nat Genet 2014; 46:1205-11. [PMID: 25282102 DOI: 10.1038/ng.3114] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 09/12/2014] [Indexed: 11/09/2022]
Abstract
Legionella pneumophila is a strictly environmental pathogen and the etiological agent of legionellosis. It is known that non-vertical processes have a major role in the short-term evolution of pathogens, but little is known about the relevance of these and other processes in environmental bacteria. We report the whole-genome sequencing of 69 L. pneumophila strains linked to recurrent outbreaks in a single location (Alcoy, Spain) over 11 years. We found some examples where the genome sequences of isolates of the same sequence type and outbreak did not cluster together and were more closely related to sequences from different outbreaks. Our analyses identify 16 recombination events responsible for almost 98% of the SNPs detected in the core genome and an apparent acceleration in the evolutionary rate. These results have profound implications for the understanding of microbial populations and for public health interventions in Legionella outbreak investigations.
Collapse
|
18
|
Alfsnes K, Raynaud X, Tønjum T, Ambur OH. Mathematical and live meningococcal models for simple sequence repeat dynamics - coherent predictions and observations. PLoS One 2014; 9:e101637. [PMID: 24999629 PMCID: PMC4085013 DOI: 10.1371/journal.pone.0101637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 06/10/2014] [Indexed: 11/18/2022] Open
Abstract
Evolvability by means of simple sequence repeat (SSR) instability is a feature under the constant influence of opposing selective pressures to expand and compress the repeat tract and is mechanistically influenced by factors that affect genetic instability. In addition to direct selection for protein expression and structural integrity, other factors that influence tract length evolution were studied. The genetic instability of SSRs that switch the expression of antibiotic resistance ON and OFF was modelled mathematically and monitored in a panel of live meningococcal strains. The mathematical model showed that the SSR length of a theoretical locus in an evolving population may be shaped by direct selection of expression status (ON or OFF), tract length dependent (α) and tract length independent factors (β). According to the model an increase in α drives the evolution towards shorter tracts. An increase in β drives the evolution towards a normal distribution of tract lengths given that an upper and a lower limit are set. Insertion and deletion biases were shown to skew allelic distributions in both directions. The meningococcal SSR model was tested in vivo by monitoring the frequency of spectinomycin resistance OFF→ON switching in a designed locus. The instability of a comprehensive panel of the homopolymeric SSRs, constituted of a range of 5-13 guanine nucleotides, was monitored in wildtype and mismatch repair deficient backgrounds. Both the repeat length itself and mismatch repair deficiency were shown to influence the genetic instability of the homopolymeric tracts. A possible insertion bias was observed in tracts ≤G10. Finally, an inverse correlation between the number of tract-encoded amino acids and growth in the presence of ON-selection illustrated a limitation to SSR expansion in an essential gene associated with the designed model locus and the protein function mediating antibiotic resistance.
Collapse
Affiliation(s)
- Kristian Alfsnes
- Department of Microbiology, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital (Rikshospitalet), Oslo, Norway
| | - Xavier Raynaud
- Department of Mathematics, University of Oslo, Oslo, Norway
| | - Tone Tønjum
- Department of Microbiology, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital (Rikshospitalet), Oslo, Norway
| | - Ole Herman Ambur
- Department of Microbiology, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital (Rikshospitalet), Oslo, Norway
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
- * E-mail:
| |
Collapse
|
19
|
Experimental evolution and the dynamics of genomic mutation rate modifiers. Heredity (Edinb) 2014; 113:375-80. [PMID: 24849169 DOI: 10.1038/hdy.2014.49] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 04/11/2014] [Accepted: 04/15/2014] [Indexed: 01/01/2023] Open
Abstract
Because genes that affect mutation rates are themselves subject to mutation, mutation rates can be influenced by natural selection and other evolutionary forces. The population genetics of mutation rate modifier alleles has been a subject of theoretical interest for many decades. Here, we review experimental contributions to our understanding of mutation rate modifier dynamics. Numerous evolution experiments have shown that mutator alleles (modifiers that elevate the genomic mutation rate) can readily rise to high frequencies via genetic hitchhiking in non-recombining microbial populations. Whereas these results certainly provide an explanatory framework for observations of sporadically high mutation rates in pathogenic microbes and in cancer lineages, it is nonetheless true that most natural populations have very low mutation rates. This raises the interesting question of how mutator hitchhiking is suppressed or its phenotypic effect reversed in natural populations. Very little experimental work has addressed this question; with this in mind, we identify some promising areas for future experimental investigation.
Collapse
|
20
|
Mohawk KL, Poly F, Sahl JW, Rasko DA, Guerry P. High frequency, spontaneous motA mutations in Campylobacter jejuni strain 81-176. PLoS One 2014; 9:e88043. [PMID: 24558375 PMCID: PMC3928116 DOI: 10.1371/journal.pone.0088043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 01/02/2014] [Indexed: 12/28/2022] Open
Abstract
Campylobacter jejuni is an important cause of bacterial diarrhea worldwide. The pathogenesis of C. jejuni is poorly understood and complicated by phase variation of multiple surface structures including lipooligosaccharide, capsule, and flagellum. When C. jejuni strain 81-176 was plated on blood agar for single colonies, the presence of translucent, non-motile colonial variants was noted among the majority of opaque, motile colonies. High-throughput genomic sequencing of two flagellated translucent and two opaque variants as well as the parent strain revealed multiple genetic changes compared to the published genome. However, the only mutated open reading frame common between the two translucent variants and absent from the opaque variants and the parent was motA, encoding a flagellar motor protein. A total of 18 spontaneous motA mutations were found that mapped to four distinct sites in the gene, with only one class of mutation present in a phase variable region. This study exemplifies the mutative/adaptive properties of C. jejuni and demonstrates additional variability in C. jejuni beyond phase variation.
Collapse
Affiliation(s)
- Krystle L. Mohawk
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Frédéric Poly
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Jason W. Sahl
- Institute for Genome Sciences, Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - David A. Rasko
- Institute for Genome Sciences, Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Patricia Guerry
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
21
|
Shea MW. The Long Road to an Effective Vaccine for Meningococcus Group B (MenB). Ann Med Surg (Lond) 2013; 2:53-6. [PMID: 25628885 PMCID: PMC4306095 DOI: 10.1016/s2049-0801(13)70037-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/15/2013] [Indexed: 12/20/2022] Open
Abstract
Neisseria meningitidis infection can cause life-threatening meningitis and meningococcal septicaemia. Over the past 40 years, vaccines against most of the main meningococcal serogroups have offered increasingly good protection from disease, with one major exception in the developed world: serogroup B meningococcus (MenB). In the United States, MenB accounts for about a quarter of cases of meningococcal meningitis, with the bulk of the rest caused by meningococcus serogroups C (MenC) and Y (MenY). In the UK, where a vaccine against MenC is widely used, MenB is now responsible for nearly 90% of cases of invasive meningococcal disease. Recent attempts to create a universal MenB vaccine have been thwarted by the variability of the surface proteins of MenB and by the similarity of the MenB capsule to human glycoproteins. This review discusses current meningococcal vaccine strategies and their limitations with regard to MenB, and examines a promising new strategy for the rational design of a MenB vaccine. Thanks to a fusion of a rational reverse genetics approach and a membrane vesicle approach, a MenB vaccine, 4CMenB (Bexsero(®)), has finally gained regulatory approval in Europe and could be in clinical use by the end of 2013.
Collapse
Affiliation(s)
- Michael W Shea
- St. Hugh's College, University of Oxford, Oxford, OX2 6LE, UK
| |
Collapse
|
22
|
Serogroup-specific interaction of Neisseria meningitidis capsular polysaccharide with host cell microtubules and effects on tubulin polymerization. Infect Immun 2013; 82:265-74. [PMID: 24166951 DOI: 10.1128/iai.00501-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
We have previously shown that during late stages of the infectious process, serogroup B meningococci (MenB) are able to escape the phagosome of in vitro-infected human epithelial cells. They then multiply in the cytosolic environment and spread intracellularly and to surrounding cells by exploiting the microtubule cytoskeleton, as suggested by results of infections in the presence of microtubule inhibitors and evidence of nanotubes connecting neighboring cells. In this study, by using microtubule binding assays with purified microtubule asters and bundles and microtubule bundles synthesized in vitro, we demonstrate that the MenB capsule directly mediates the interaction between bacteria and microtubules. The direct interaction between the microtubules and the MenB capsular polysaccharide was confirmed by coimmunoprecipitation experiments. Unexpectedly, serogroup C meningococci (MenC), which have a capsular polysaccharide that differs from that of MenB only by its anomeric linkage, α(2→9) instead of α(2→8), were not able to interact with the microtubules, and the lack of interaction was not due to capsular polysaccharide O-acetylation that takes place in most MenC strains but not in MenB strains. Moreover, we demonstrate that the MenB capsular polysaccharide inhibits tubulin polymerization in vitro. Thus, at variance with MenC, MenB may interfere with microtubule dynamics during cell infection.
Collapse
|
23
|
|
24
|
Abstract
In prokaryotes, alteration in gene expression was observed with the modification of DNA, especially DNA methylation. Such changes are inherited from generation to generation with no alterations in the DNA sequence and represent the epigenetic signal in prokaryotes. DNA methyltransferases are enzymes involved in DNA modification and thus in epigenetic regulation of gene expression. DNA methylation not only affects the thermodynamic stability of DNA, but also changes its curvature. Methylation of specific residues on DNA can affect the protein-DNA interactions. DNA methylation in prokaryotes regulates a number of physiological processes in the bacterial cell including transcription, DNA mismatch repair and replication initiation. Significantly, many reports have suggested a role of DNA methylation in regulating the expression of a number of genes in virulence and pathogenesis thus, making DNA methlytransferases novel targets for the designing of therapeutics. Here, we summarize the current knowledge about the influence of DNA methylation on gene regulation in different bacteria, and on bacterial virulence.
Collapse
Affiliation(s)
- Ritesh Kumar
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India,
| | | |
Collapse
|
25
|
Kumar R, Sabareesh V, Mukhopadhyay AK, Rao DN. Mutations in hpyAVIBM, C⁵ cytosine DNA methyltransferase from Helicobacter pylori result in relaxed specificity. FEBS J 2012; 279:1080-92. [PMID: 22269034 DOI: 10.1111/j.1742-4658.2012.08502.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The genome of Helicobacter pylori is rich in restriction-modification (RM) systems. Approximately 4% of the genome codes for components of RM systems. hpyAVIBM, which codes for a phase-variable C(5) cytosine methyltransferase (MTase) from H. pylori, lacks a cognate restriction enzyme. Over-expression of M.HpyAVIB in Escherichia coli enhances the rate of mutations. However, when the catalytically inactive F9N or C82W mutants of M.HpyAVIB were expressed in E. coli, mutations were not observed. The M.HpyAVIB gene itself was mutated to give rise to different variants of the MTase. M.HpyAVIB variants were purified and differences in kinetic properties and specificity were observed. Intriguingly, purified MTase variants showed relaxed substrate specificity. Homologues of hpyAVIBM homologues amplified and sequenced from different clinical isolates showed similar variations in sequence. Thus, hpyAVIBM presents an interesting example of allelic variations in H. pylori where changes in the nucleotide sequence result in proteins with new properties.
Collapse
Affiliation(s)
- Ritesh Kumar
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | | | | | | |
Collapse
|
26
|
Malygin EG, Hattman S. DNA methyltransferases: mechanistic models derived from kinetic analysis. Crit Rev Biochem Mol Biol 2012; 47:97-193. [PMID: 22260147 DOI: 10.3109/10409238.2011.620942] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The sequence-specific transfer of methyl groups from donor S-adenosyl-L-methionine (AdoMet) to certain positions of DNA-adenine or -cytosine residues by DNA methyltransferases (MTases) is a major form of epigenetic modification. It is virtually ubiquitous, except for some notable exceptions. Site-specific methylation can be regarded as a means to increase DNA information capacity and is involved in a large spectrum of biological processes. The importance of these functions necessitates a deeper understanding of the enzymatic mechanism(s) of DNA methylation. DNA MTases fall into one of two general classes; viz. amino-MTases and [C5-cytosine]-MTases. Amino-MTases, common in prokaryotes and lower eukaryotes, catalyze methylation of the exocyclic amino group of adenine ([N6-adenine]-MTase) or cytosine ([N4-cytosine]-MTase). In contrast, [C5-cytosine]-MTases methylate the cyclic carbon-5 atom of cytosine. Characteristics of DNA MTases are highly variable, differing in their affinity to their substrates or reaction products, their kinetic parameters, or other characteristics (order of substrate binding, rate limiting step in the overall reaction). It is not possible to present a unifying account of the published kinetic analyses of DNA methylation because different authors have used different substrate DNAs and/or reaction conditions. Nevertheless, it would be useful to describe those kinetic data and the mechanistic models that have been derived from them. Thus, this review considers in turn studies carried out with the most consistently and extensively investigated [N6-adenine]-, [N4-cytosine]- and [C5-cytosine]-DNA MTases.
Collapse
Affiliation(s)
- Ernst G Malygin
- Institute of Molecular Biology, State Research Center of Virology and Biotechnology Vector, Novosibirsk, Russia
| | | |
Collapse
|
27
|
Characterization of type II and III restriction-modification systems from Bacillus cereus strains ATCC 10987 and ATCC 14579. J Bacteriol 2011; 194:49-60. [PMID: 22037402 DOI: 10.1128/jb.06248-11] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The genomes of two Bacillus cereus strains (ATCC 10987 and ATCC 14579) have been sequenced. Here, we report the specificities of type II/III restriction (R) and modification (M) enzymes. Found in the ATCC 10987 strain, BceSI is a restriction endonuclease (REase) with the recognition and cut site CGAAG 24-25/27-28. BceSII is an isoschizomer of AvaII (G/GWCC). BceSIII cleaves at ACGGC 12/14. The BceSIII C terminus resembles the catalytic domains of AlwI, MlyI, and Nt.BstNBI. BceSIV is composed of two subunits and cleaves on both sides of GCWGC. BceSIV activity is strongly stimulated by the addition of cofactor ATP or GTP. The large subunit (R1) of BceSIV contains conserved motifs of NTPases and DNA helicases. The R1 subunit has no endonuclease activity by itself; it strongly stimulates REase activity when in complex with the R2 subunit. BceSIV was demonstrated to hydrolyze GTP and ATP in vitro. BceSIV is similar to CglI (GCSGC), and homologs of R1 are found in 11 sequenced bacterial genomes, where they are paired with specificity subunits. In addition, homologs of the BceSIV R1-R2 fusion are found in many sequenced microbial genomes. An orphan methylase, M.BceSV, was found to modify GCNGC, GGCC, CCGG, GGNNCC, and GCGC sites. A ParB-methylase fusion protein appears to nick DNA nonspecifically. The ATCC 14579 genome encodes an active enzyme Bce14579I (GCWGC). BceSIV and Bce14579I belong to the phospholipase D (PLD) family of endonucleases that are widely distributed among Bacteria and Archaea. A survey of type II and III restriction-modification (R-M) system genes is presented from sequenced B. cereus, Bacillus anthracis, and Bacillus thuringiensis strains.
Collapse
|
28
|
Genotypic and phenotypic modifications of Neisseria meningitidis after an accidental human passage. PLoS One 2011; 6:e17145. [PMID: 21386889 PMCID: PMC3046118 DOI: 10.1371/journal.pone.0017145] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Accepted: 01/11/2011] [Indexed: 01/27/2023] Open
Abstract
A scientist in our laboratory was accidentally infected while working with Z5463, a Neisseria meningitidis serogroup A strain. She developed severe symptoms (fever, meningism, purpuric lesions) that fortunately evolved with antibiotic treatment to complete recovery. Pulse-field gel electrophoresis confirmed that the isolate obtained from the blood culture (Z5463BC) was identical to Z5463, more precisely to a fourth subculture of this strain used the week before the contamination (Z5463PI). In order to get some insights into genomic modifications that can occur in vivo, we sequenced these three isolates. All the strains contained a mutated mutS allele and therefore displayed an hypermutator phenotype, consistent with the high number of mutations (SNP, Single Nucleotide Polymorphism) detected in the three strains. By comparing the number of SNP in all three isolates and knowing the number of passages between Z5463 and Z5463PI, we concluded that around 25 bacterial divisions occurred in the human body. As expected, the in vivo passage is responsible for several modifications of phase variable genes. This genomic study has been completed by transcriptomic and phenotypic studies, showing that the blood strain used a different haemoglobin-linked iron receptor (HpuA/B) than the parental strains (HmbR). Different pilin variants were found after the in vivo passage, which expressed different properties of adhesion. Furthermore the deletion of one gene involved in LOS biosynthesis (lgtB) results in Z5463BC expressing a different LOS than the L9 immunotype of Z2491. The in vivo passage, despite the small numbers of divisions, permits the selection of numerous genomic modifications that may account for the high capacity of the strain to disseminate.
Collapse
|
29
|
Nyamjav D, Rozhok S, Holz RC. Immobilization of motile bacterial cells via dip-pen nanolithography. NANOTECHNOLOGY 2010; 21:235105. [PMID: 20472944 DOI: 10.1088/0957-4484/21/23/235105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
A strategy to bind bacterial cells to surfaces in a directed fashion via dip-pen nanolithography (DPN) is presented. Cellular attachment to pre-designed DPN generated microarrays was found to be dependent on the shape and size of the surface feature. While this observation is likely due in part to a dense, well formed mercaptohexadecanoic acid (MHA) monolayer generated via DPN, it may also simply be due to the physical shape of the surface structure. Motile Pseudomonas aeruginosa bacterial cells were observed to bind to DPN generated mercaptohexadecanoic acid/poly-L-lysine (MHA/PLL) line patterns, 'blocks' made up of eight lines with 100 nm spacings, with approximately 80% occupancy. Cellular binding to these 'block' surface structures occurs via an electrostatic interaction between negatively charged groups on the bacterial cell surface and positively charged poly-L-lysine (PLL) assemblies. These data indicate that these DPN generated 'block' surface structures provide a promising footprint for the attachment of motile bacterial cells that may find utility in cell based biosensors or single cell studies.
Collapse
Affiliation(s)
- Dorjderem Nyamjav
- The Department of Chemistry, Loyola University-Chicago, Chicago, IL 60626, USA
| | | | | |
Collapse
|
30
|
Sun K, Jiao XD, Zhang M, Sun L. DNA adenine methylase is involved in the pathogenesis of Edwardsiella tarda. Vet Microbiol 2010; 141:149-54. [DOI: 10.1016/j.vetmic.2009.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 08/23/2009] [Accepted: 09/04/2009] [Indexed: 12/24/2022]
|
31
|
Colicchio R, Ricci S, Lamberti F, Pagliarulo C, Pagliuca C, Braione V, Braccini T, Talà A, Montanaro D, Tripodi S, Cintorino M, Troncone G, Bucci C, Pozzi G, Bruni CB, Alifano P, Salvatore P. The meningococcal ABC-Type L-glutamate transporter GltT is necessary for the development of experimental meningitis in mice. Infect Immun 2009; 77:3578-87. [PMID: 19528209 PMCID: PMC2737999 DOI: 10.1128/iai.01424-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 01/21/2009] [Accepted: 06/05/2009] [Indexed: 11/20/2022] Open
Abstract
Experimental animal models of bacterial meningitis are useful to study the host-pathogen interactions occurring at the cerebral level and to analyze the pathogenetic mechanisms behind this life-threatening disease. In this study, we have developed a mouse model of meningococcal meningitis based on the intracisternal inoculation of bacteria. Experiments were performed with mouse-passaged serogroup C Neisseria meningitidis. Survival and clinical parameters of infected mice and microbiological and histological analysis of the brain demonstrated the establishment of meningitis with features comparable to those of the disease in humans. When using low bacterial inocula, meningococcal replication in the brain was very efficient, with a 1,000-fold increase of viable counts in 18 h. Meningococci were also found in the blood, spleens, and livers of infected mice, and bacterial loads in different organs were dependent on the infectious dose. As glutamate uptake from the host has been implicated in meningococcal virulence, mice were infected intracisternally with an isogenic strain deficient in the ABC-type L-glutamate transporter GltT. Noticeably, the mutant was attenuated in virulence in mixed infections, indicating that wild-type bacteria outcompeted the GltT-deficient meningococci. The data show that the GltT transporter plays a role in meningitis and concomitant systemic infection, suggesting that meningococci may use L-glutamate as a nutrient source and as a precursor to synthesize the antioxidant glutathione.
Collapse
|
32
|
Functional characterization of excision repair and RecA-dependent recombinational DNA repair in Campylobacter jejuni. J Bacteriol 2009; 191:3785-93. [PMID: 19376866 DOI: 10.1128/jb.01817-08] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The presence and functionality of DNA repair mechanisms in Campylobacter jejuni are largely unknown. In silico analysis of the complete translated genome of C. jejuni NCTC 11168 suggests the presence of genes involved in methyl-directed mismatch repair (MMR), nucleotide excision repair, base excision repair (BER), and recombinational repair. To assess the functionality of these putative repair mechanisms in C. jejuni, mutS, uvrB, ung, and recA knockout mutants were constructed and analyzed for their ability to repair spontaneous point mutations, UV irradiation-induced DNA damage, and nicked DNA. Inactivation of the different putative DNA repair genes did not alter the spontaneous mutation frequency. Disruption of the UvrB and RecA orthologues, but not the putative MutS or Ung proteins, resulted in a significant reduction in viability after exposure to UV irradiation. Assays performed with uracil-containing plasmid DNA showed that the putative uracil-DNA glycosylase (Ung) protein, important for initiation of the BER pathway, is also functional in C. jejuni. Inactivation of recA also resulted in a loss of natural transformation. Overall, the data indicate that C. jejuni has multiple functional DNA repair systems that may protect against DNA damage and limit the generation of genetic diversity. On the other hand, the apparent absence of a functional MMR pathway may enhance the frequency of on-and-off switching of phase variable genes typical for C. jejuni and may contribute to the genetic heterogeneity of the C. jejuni population.
Collapse
|
33
|
The biofilm life cycle and virulence of Pseudomonas aeruginosa are dependent on a filamentous prophage. ISME JOURNAL 2008; 3:271-82. [PMID: 19005496 DOI: 10.1038/ismej.2008.109] [Citation(s) in RCA: 247] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Mature Pseudomonas aeruginosa biofilms undergo specific developmental events. Using a bacteriophage mutant, generated by deletion of the entire filamentous Pf4 prophage, we show that the phage is essential for several stages of the biofilm life cycle and that it significantly contributes to the virulence of P. aeruginosa in vivo. Here, we show for the first time that biofilms of the Pf4 phage-deficient mutant did not develop hollow centres or undergo cell death, typical of the differentiation process of wild-type (WT) P. aeruginosa PAO1 biofilms. Furthermore, microcolonies of the Pf4 mutant were significantly smaller in size and less stable compared with the WT biofilm. Small colony variants (SCVs) were detectable in the dispersal population of the WT biofilm at the time of dispersal and cell death, whereas no SCVs were detected in the effluent of the Pf4 mutant biofilm. This study shows that at the time when cell death occurs in biofilms of the WT, the Pf4 phage converts into a superinfective form, which correlates with the appearance of variants in the dispersal population. Unexpectedly, mice infected with the Pf4 mutant survived significantly longer than those infected with its isogenic WT strain, showing that Pf4 contributes to the virulence of P. aeruginosa. Hence, a filamentous prophage is a major contributor to the life cycle and adaptive behaviour of P. aeruginosa and offers an explanation for the prevalence of phage in this organism.
Collapse
|
34
|
Talà A, Progida C, De Stefano M, Cogli L, Spinosa MR, Bucci C, Alifano P. The HrpB-HrpA two-partner secretion system is essential for intracellular survival of Neisseria meningitidis. Cell Microbiol 2008; 10:2461-82. [PMID: 18680551 DOI: 10.1111/j.1462-5822.2008.01222.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
In this study we used HeLa cells to investigate the role of the HrpB-HrpA two-partner secretion (TPS) system in the meningococcal infection cycle. Although there is evidence that several pathogenic microorganisms may use TPS systems to colonize epithelial surfaces, the meningococcal HrpB-HrpA TPS system was not primarily involved in adhesion to or invasion of HeLa cells. Instead, this system was essential for intracellular survival and escape from infected cells. Gentamicin protection assays, immunofluorescence and transmission electron microscopy analyses demonstrated that, in contrast to the wild-type strain, HrpB-HrpA-deficient mutants were primarily confined to late endocytic vacuoles and trapped in HeLa cells. Haemolytic tests using human erythrocytes suggested that the secreted HrpA proteins could act as manganese-dependent lysins directly involved in mediating vacuole escape. In addition, we demonstrated that escape of wild-type meningococci from infected cells required the use of an intact tubulin cytoskeleton and that the hrpB-hrpA genes, which are absent in other Neisseria spp., were upregulated during infection.
Collapse
Affiliation(s)
- Adelfia Talà
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali (DiSTeBA), Università del Salento, Via Provinciale Monteroni, 73100 Lecce, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Talà A, De Stefano M, Bucci C, Alifano P. Reverse transcriptase-PCR differential display analysis of meningococcal transcripts during infection of human cells: up-regulation of priA and its role in intracellular replication. BMC Microbiol 2008; 8:131. [PMID: 18664272 PMCID: PMC2527323 DOI: 10.1186/1471-2180-8-131] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Accepted: 07/29/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In vitro studies with cell line infection models are beginning to disclose the strategies that Neisseria meningitidis uses to survive and multiply inside the environment of the infected host cell. The goal of this study was to identify novel virulence determinants that are involved in this process using an in vitro infection system. RESULTS By using reverse transcriptase-PCR differential display we have identified a set of meningococcal genes significantly up-regulated during residence of the bacteria in infected HeLa cells including genes involved in L-glutamate transport (gltT operon), citrate metabolism (gltA), disulfide bond formation (dsbC), two-partner secretion (hrpA-hrpB), capsulation (lipA), and DNA replication/repair (priA). The role of PriA, a protein that in Escherichia coli plays a central role in replication restart of collapsed or arrested DNA replication forks, has been investigated. priA inactivation resulted in a number of growth phenotypes that were fully complemented by supplying a functional copy of priA. The priA-defective mutant exhibited reduced viability during late logarithmic growth phase. This defect was more severe when it was incubated under oxygen-limiting conditions using nitrite as terminal electron acceptors in anaerobic respiration. When compared to wild type it was more sensitive to hydrogen peroxide and the nitric oxide generator sodium nitroprusside. The priA-defective strain was not affected in its ability to invade HeLa cells, but, noticeably, exhibited severely impaired intracellular replication and, at variance with wild type and complemented strains, it co-localized with lysosomal associated membrane protein 1. CONCLUSION In conclusion, our study i.) demonstrates the efficacy of the experimental strategy that we describe for discovering novel virulence determinants of N. meningitidis and ii.) provides evidence for a role of priA in preventing both oxidative and nitrosative injury, and in intracellular meningococcal replication.
Collapse
Affiliation(s)
- Adelfia Talà
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università degli Studi del Salento, Via Monteroni, 73100 Lecce, Italy.
| | | | | | | |
Collapse
|
36
|
Telford JL. Bacterial genome variability and its impact on vaccine design. Cell Host Microbe 2008; 3:408-16. [PMID: 18541217 DOI: 10.1016/j.chom.2008.05.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 05/16/2008] [Accepted: 05/16/2008] [Indexed: 11/16/2022]
Abstract
The majority of currently available successful vaccines induce host responses against antigens that are highly conserved in the targeted pathogens. The diphtheria, tetanus, and pertussis vaccines confer protection by inducing neutralizing antibodies to the conserved bacterial toxins that are the major virulence factors. The Hemophilus influenzae B vaccine induces responses to conserved epitopes in the sugar structure of the bacterial capsular polysaccharide. However, the efficacy of more recently developed vaccines is limited by antigen variation, which also presents a challenge for future vaccine development. This review will explore bacterial genome variability and its impact on vaccine development.
Collapse
Affiliation(s)
- John L Telford
- Novartis Vaccines and Diagnostics Srl, Via Fiorentina 1, 53100 Siena, Italy.
| |
Collapse
|
37
|
Treangen TJ, Ambur OH, Tonjum T, Rocha EPC. The impact of the neisserial DNA uptake sequences on genome evolution and stability. Genome Biol 2008; 9:R60. [PMID: 18366792 PMCID: PMC2397512 DOI: 10.1186/gb-2008-9-3-r60] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/13/2008] [Accepted: 03/26/2008] [Indexed: 12/03/2022] Open
Abstract
A study of the origin and distribution of the abundant short DNA uptake sequence (DUS) in six genomes of Neisseria suggests that transformation and recombination are tightly linked in evolution and that recombination has a key role in the establishment of DUS. Background Efficient natural transformation in Neisseria requires the presence of short DNA uptake sequences (DUSs). Doubts remain whether DUSs propagate by pure selfish molecular drive or are selected for 'safe sex' among conspecifics. Results Six neisserial genomes were aligned to identify gene conversion fragments, DUS distribution, spacing, and conservation. We found a strong link between recombination and DUS: DUS spacing matches the size of conversion fragments; genomes with shorter conversion fragments have more DUSs and more conserved DUSs; and conversion fragments are enriched in DUSs. Many recent and singly occurring DUSs exhibit too high divergence with homologous sequences in other genomes to have arisen by point mutation, suggesting their appearance by recombination. DUSs are over-represented in the core genome, under-represented in regions under diversification, and absent in both recently acquired genes and recently lost core genes. This suggests that DUSs are implicated in genome stability rather than in generating adaptive variation. DUS elements are most frequent in the permissive locations of the core genome but are themselves highly conserved, undergoing mutation selection balance and/or molecular drive. Similar preliminary results were found for the functionally analogous uptake signal sequence in Pasteurellaceae. Conclusion As do many other pathogens, Neisseria and Pasteurellaceae have hyperdynamic genomes that generate deleterious mutations by intrachromosomal recombination and by transient hypermutation. The results presented here suggest that transformation in Neisseria and Pasteurellaceae allows them to counteract the deleterious effects of genome instability in the core genome. Thus, rather than promoting hypervariation, bacterial sex could be regenerative.
Collapse
Affiliation(s)
- Todd J Treangen
- Algorithms and Genetics Group, Department of Computer Science, Technical University of Catalonia, Jordi Girona Salgado, 1-3, E-08034 Barcelona, Spain.
| | | | | | | |
Collapse
|
38
|
Spinosa MR, Progida C, Talà A, Cogli L, Alifano P, Bucci C. The Neisseria meningitidis capsule is important for intracellular survival in human cells. Infect Immun 2007; 75:3594-603. [PMID: 17470547 PMCID: PMC1932921 DOI: 10.1128/iai.01945-06] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
While much data exist in the literature about how Neisseria meningitidis adheres to and invades human cells, its behavior inside the host cell is largely unknown. One of the essential meningococcal attributes for pathogenesis is the polysaccharide capsule, which has been shown to be important for bacterial survival in extracellular fluids. To investigate the role of the meningococcal capsule in intracellular survival, we used B1940, a serogroup B strain, and its isogenic derivatives, which lack either the capsule or both the capsule and the lipooligosaccharide outer core, to infect human phagocytic and nonphagocytic cells and monitor invasion and intracellular growth. Our data indicate that the capsule, which negatively affects bacterial adhesion and, consequently, entry, is, in contrast, fundamental for the intracellular survival of this microorganism. The results of in vitro assays suggest that an increased resistance to cationic antimicrobial peptides (CAMPs), important components of the host innate defense system against microbial infections, is a possible mechanism by which the capsule protects the meningococci in the intracellular environment. Indeed, unencapsulated bacteria were more susceptible than encapsulated bacteria to defensins, cathelicidins, protegrins, and polymyxin B, which has long been used as a model compound to define the mechanism of action of CAMPs. We also demonstrate that both the capsular genes (siaD and lipA) and those encoding an efflux pump involved in resistance to CAMPs (mtrCDE) were up-regulated during the intracellular phase of the infectious cycle.
Collapse
Affiliation(s)
- Maria Rita Spinosa
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università degli Studi del Salento, Via Monteroni, 73100 Lecce, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Colicchio R, Pagliarulo C, Lamberti F, Vigliotta G, Bruni CB, Alifano P, Salvatore P. RecB-dependent mutator phenotype in Neisseria meningitidis strains naturally defective in mismatch repair. DNA Repair (Amst) 2006; 5:1428-38. [PMID: 16911877 DOI: 10.1016/j.dnarep.2006.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Revised: 07/03/2006] [Accepted: 07/05/2006] [Indexed: 11/15/2022]
Abstract
Several invasive serogroup B meningococcal strains phylogenetically related to the lineage III (ET-24) exhibited a mutator phenotype as shown by mutagenicity assay using rifampicin-resistance as a selection marker. Hypermutation was associated to the presence of defective mutL alleles that were genetically characterized. Interestingly, the mutator phenotype was suppressed when a non-functional recB(ET-37) allele, derived from ET-37 meningococcal strains, replaced the functional recB allele in a lineage III strain. In contrast, the same gene replacement did not affect mutation frequencies in a mismatch repair-proficient strain. These results suggested that in MutL-deficient strains spontaneous mutations mostly arise from post-replicative DNA synthesis associated to the activity of the RecBCD recombination pathway.
Collapse
Affiliation(s)
- Roberta Colicchio
- Dipartimento di Biologia e Patologia Cellulare e Molecolare L. Califano, Università di Napoli Federico II, and Istituto di Endocrinologia ed Oncologia Sperimentale G. Salvatore of the CNR, Via S. Pansini 5, 80131 Napoli, Italy
| | | | | | | | | | | | | |
Collapse
|
40
|
Hall LMC, Henderson-Begg SK. Hypermutable bacteria isolated from humans – a critical analysis. Microbiology (Reading) 2006; 152:2505-2514. [PMID: 16946246 DOI: 10.1099/mic.0.29079-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hypermutable bacteria of several species have been described among isolates recovered from humans over the last decade. Interpretation of the literature in this area is complicated by diversity in the determination and definition of hypermutability, and this review outlines the different methods used. Inactivation of the mismatch repair genemutSis often implicated in the mutator phenotype; the reported effect ofmutSinactivation on mutation frequency varies widely between species, from under 10-fold to nearly 1000-fold, but also varies among different reports on the same species. Particularly high proportions of mutators have been reported amongPseudomonas aeruginosaand other species in the cystic fibrosis lung, epidemic serogroup ANeisseria meningitidis, andHelicobacter pylori. Aspects of the biology of these infections that could be relevant to hypermutability are discussed, and some future directions that may increase our understanding of mutators among bacteria isolated from humans are considered.
Collapse
Affiliation(s)
- Lucinda M C Hall
- Centre for Infectious Disease, Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, 4 Newark Street, London E1 2AT, UK
| | - Stephanie K Henderson-Begg
- Centre for Infectious Disease, Institute of Cell and Molecular Science, Barts and The London School of Medicine and Dentistry, Queen Mary, University of London, 4 Newark Street, London E1 2AT, UK
| |
Collapse
|
41
|
Monaco C, Talà A, Spinosa MR, Progida C, De Nitto E, Gaballo A, Bruni CB, Bucci C, Alifano P. Identification of a meningococcal L-glutamate ABC transporter operon essential for growth in low-sodium environments. Infect Immun 2006; 74:1725-40. [PMID: 16495545 PMCID: PMC1418650 DOI: 10.1128/iai.74.3.1725-1740.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GdhR is a meningococcal transcriptional regulator that was previously shown to positively control the expression of gdhA, encoding the NADP-specific L-glutamate dehydrogenase (NADP-GDH), in response to the growth phase and/or to the carbon source. In this study we used reverse transcriptase-PCR-differential display (to identify additional GdhR-regulated genes. The results indicated that GdhR, in addition to NADP-GDH, controls the expression of a number of genes involved in glucose catabolism by the Entner-Doudoroff pathway and in l-glutamate import by an unknown ABC transport system. The genes encoding the putative periplasmic substrate-binding protein (NMB1963) and the permease (NMB1965) of the ABC transporter were genetically inactivated. Uptake experiments demonstrated an impairment of L-glutamate import in the NMB1965-defective mutant in the absence or in the presence of a low sodium ion concentration. In contrast, at a sodium ion concentration above 60 mM, the uptake defect disappeared, possibly because the activity of a sodium-driven secondary transporter became predominant. Indeed, the NMB1965-defective mutant was unable to grow at a low sodium ion concentration (<20 mM) in a chemically defined medium containing L-glutamate and four other amino acids that supported meningococcal growth, but it grew when the sodium ion concentration was raised to higher values (>60 mM). The same growth phenotype was observed in the NMB1963-defective mutant. Cell invasion and intracellular persistence assays and expression data during cell invasion provided evidence that the l-glutamate ABC transporter, tentatively named GltT, was critical for meningococcal adaptation in the low-sodium intracellular environment.
Collapse
Affiliation(s)
- Caterina Monaco
- Dipartimento di Scienze e Tecnologie Biologiche ed Ambientali, Università di Lecce, Via Monteroni, 73100 Lecce, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Neisseria meningitidis (the meningococcus) is an important commensal, pathogen and model organism that faces up to the environment in its exclusive human host with a small but hyperdynamic genome. Compared with Escherichia coli, several DNA-repair genes are absent in N. meningitidis, whereas the gene products of others interact differently. Instead of responding to external stimuli, the meningococcus spontaneously produces a plethora of genetic variants. The frequent genomic alterations and polymorphisms have profound consequences for the interaction of this microorganism with its host, impacting structural and antigenic changes in crucial surface components that are relevant for adherence and invasion as well as antibiotic resistance and vaccine development.
Collapse
Affiliation(s)
- Tonje Davidsen
- Centre for Molecular Biology and Neuroscience and Institute of Microbiology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway
| | | |
Collapse
|
43
|
Joseph N, Duppatla V, Rao DN. Prokaryotic DNA Mismatch Repair. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 2006; 81:1-49. [PMID: 16891168 DOI: 10.1016/s0079-6603(06)81001-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Nimesh Joseph
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | | |
Collapse
|
44
|
Stabler RA, Marsden GL, Witney AA, Li Y, Bentley SD, Tang CM, Hinds J. Identification of pathogen-specific genes through microarray analysis of pathogenic and commensal Neisseria species. MICROBIOLOGY-SGM 2005; 151:2907-2922. [PMID: 16151203 DOI: 10.1099/mic.0.28099-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The release of the complete genome sequences of Neisseria meningitidis MC58 and Z2491 along with access to the sequences of N. meningitidis FAM18 and Neisseria gonorrhoeae FA1090 allowed the construction of a pan-Neisseria microarray, with every gene in all four genomes represented. The microarray was used to analyse a selection of strains including all N. meningitidis serogroups and commensal Neisseria species. For each strain, genes were defined as present, divergent or absent using gack analysis software. Comparison of the strains identified genes that were conserved within N. meningitidis serogroup B strains but absent from all commensal strains tested, consisting of mainly virulence-associated genes and transmissible elements. The microarray was able to distinguish between pilin genes, pilC orthologues and serogroup-specific capsule biosynthetic genes, and to identify dam and drg genotypes. Previously described N. meningitidis genes involved in iron response, adherence to epithelial cells, and pathogenicity were compared to the microarray analysis. The microarray data correlated with other genetic typing methods and were able to predict genotypes for uncharacterized strains and thus offer the potential for a rapid typing method. The subset of pathogen-specific genes identified represents potential drug or vaccine targets that would not eliminate commensal neisseriae and the associated naturally acquired immunity.
Collapse
Affiliation(s)
- Richard A Stabler
- Bacterial Microarray Group, St George's Hospital Medical School, London SW7 0RE, UK
| | - Gemma L Marsden
- Bacterial Microarray Group, St George's Hospital Medical School, London SW7 0RE, UK
| | - Adam A Witney
- Bacterial Microarray Group, St George's Hospital Medical School, London SW7 0RE, UK
| | - Yanwen Li
- Centre for Molecular Microbiology and Infection, Department of Infectious Diseases, Imperial College London, London SW7 2AZ, UK
| | - Stephen D Bentley
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, UK
| | - Christoph M Tang
- Centre for Molecular Microbiology and Infection, Department of Infectious Diseases, Imperial College London, London SW7 2AZ, UK
| | - Jason Hinds
- Bacterial Microarray Group, St George's Hospital Medical School, London SW7 0RE, UK
| |
Collapse
|
45
|
Taylor VL, Titball RW, Oyston PCF. Oral immunization with a dam mutant of Yersinia pseudotuberculosis protects against plague. MICROBIOLOGY-SGM 2005; 151:1919-1926. [PMID: 15941999 DOI: 10.1099/mic.0.27959-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Inactivation of the gene encoding DNA adenine methylase (dam) has been shown to attenuate some pathogens such as Salmonella enterica serovar Typhimurium and is a lethal mutation in others such as Yersinia pseudotuberculosis strain YPIII. In this study the dam methylase gene in Yersinia pseudotuberculosis strain IP32953 was inactivated. Unlike the wild-type, DNA isolated from the mutant could be digested with MboI, which is consistent with an altered pattern of DNA methylation. The mutant was sensitive to bile salts but not to 2-aminopurine. The effect of dam inactivation on gene expression was examined using a DNA microarray. In BALB/c mice inoculated orally or intravenously with the dam mutant, the median lethal dose (MLD) was at least 10(6)-fold higher than the MLD of the wild-type. BALB/c mice inoculated with the mutant were protected against a subcutaneous challenge with 100 MLDs of Yersinia pestis strain GB and an intravenous challenge with 300 MLDs of Y. pseudotuberculosis IP32953.
Collapse
Affiliation(s)
- Victoria L Taylor
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Richard W Titball
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel St, London WC1E 7HT, UK
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Petra C F Oyston
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| |
Collapse
|
46
|
Horton JR, Liebert K, Hattman S, Jeltsch A, Cheng X. Transition from nonspecific to specific DNA interactions along the substrate-recognition pathway of dam methyltransferase. Cell 2005; 121:349-61. [PMID: 15882618 PMCID: PMC2656680 DOI: 10.1016/j.cell.2005.02.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Revised: 02/04/2005] [Accepted: 02/17/2005] [Indexed: 10/25/2022]
Abstract
DNA methyltransferases methylate target bases within specific nucleotide sequences. Three structures are described for bacteriophage T4 DNA-adenine methyltransferase (T4Dam) in ternary complexes with partially and fully specific DNA and a methyl-donor analog. We also report the effects of substitutions in the related Escherichia coli DNA methyltransferase (EcoDam), altering residues corresponding to those involved in specific interaction with the canonical GATC target sequence in T4Dam. We have identified two types of protein-DNA interactions: discriminatory contacts, which stabilize the transition state and accelerate methylation of the cognate site, and antidiscriminatory contacts, which do not significantly affect methylation of the cognate site but disfavor activity at noncognate sites. These structures illustrate the transition in enzyme-DNA interaction from nonspecific to specific interaction, suggesting that there is a temporal order for formation of specific contacts.
Collapse
Affiliation(s)
- John R. Horton
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, Georgia 30322
| | - Kirsten Liebert
- School of Engineering and Science, International University Bremen, Campus Ring 1, 28759 Bremen, Germany
| | - Stanley Hattman
- Department of Biology, University of Rochester, Rochester, New York 14627
| | - Albert Jeltsch
- School of Engineering and Science, International University Bremen, Campus Ring 1, 28759 Bremen, Germany
- Correspondence: ;
| | - Xiaodong Cheng
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, Georgia 30322
- Correspondence: ;
| |
Collapse
|
47
|
Zaleski P, Wojciechowski M, Piekarowicz A. The role of Dam methylation in phase variation of Haemophilus influenzae genes involved in defence against phage infection. Microbiology (Reading) 2005; 151:3361-3369. [PMID: 16207918 DOI: 10.1099/mic.0.28184-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Haemophilus influenzae uses phase variation (PV) to modulate the activity of its defence systems against phage infection. The PV of the restriction–modification (R-M) system HindI, the main defence system against phage infection and incoming chromosomal and phage DNA in H. influenzae Rd, is driven by changes of the pentanucleotide repeat tract within the coding sequence of the hsdM gene and is influenced by lack of Dam methylation. Phase-variable resistance/sensitivity to phage infection correlates with changes in lipooligosaccharide (LOS) structure and occurs by slippage of tetranucleotide repeats within the gene lic2A, coding for a step in the biosynthesis of LOS. The lack of Dam activity destabilizes the tetranuclotide (5′-CAAT) repeat tract and increases the frequency of switching from sensitivity to resistance to phage infection more than in the opposite direction. The PV of the lgtC gene does not influence resistance or sensitivity to phage infection. Insertional inactivation of lic2A, but not lgtC or lgtF, leads to resistance to phage infection and to the same structure of the LOS as observed among phase-variable phage-resistant variants. This indicates that in the H. influenzae Rd LOS only the first two sugars (Glc-Gal) extending from the third heptose are part of bacterial phage receptors.
Collapse
Affiliation(s)
- Piotr Zaleski
- Institute of Microbiology, Warsaw University, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Marek Wojciechowski
- Institute of Microbiology, Warsaw University, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Andrzej Piekarowicz
- Institute of Microbiology, Warsaw University, Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
48
|
Snyder LAS, Davies JK, Ryan CS, Saunders NJ. Comparative overview of the genomic and genetic differences between the pathogenic Neisseria strains and species. Plasmid 2005; 54:191-218. [PMID: 16024078 DOI: 10.1016/j.plasmid.2005.04.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 04/18/2005] [Accepted: 04/21/2005] [Indexed: 01/19/2023]
Abstract
The availability of complete genome sequences from multiple pathogenic Neisseria strains and species has enabled a comprehensive survey of the genomic and genetic differences occurring within these species. In this review, we describe the chromosomal rearrangements that have occurred, and the genomic islands and prophages that have been identified in the various genomes. We also describe instances where specific genes are present or absent, other instances where specific genes have been inactivated, and situations where there is variation in the version of a gene that is present. We also provide an overview of mosaic genes present in these genomes, and describe the variation systems that allow the expression of particular genes to be switched ON or OFF. We have also described the presence and location of mobile non-coding elements in the various genomes. Finally, we have reviewed the incidence and properties of various extra-chromosomal elements found within these species. The overall impression is one of genomic variability and instability, resulting in increased functional flexibility within these species.
Collapse
Affiliation(s)
- Lori A S Snyder
- Bacterial Pathogenesis and Functional Genomics Group, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| | | | | | | |
Collapse
|
49
|
Davidsen T, Bjørås M, Seeberg EC, Tønjum T. Antimutator role of DNA glycosylase MutY in pathogenic Neisseria species. J Bacteriol 2005; 187:2801-9. [PMID: 15805527 PMCID: PMC1070393 DOI: 10.1128/jb.187.8.2801-2809.2005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Genome alterations due to horizontal gene transfer and stress constantly generate strain on the gene pool of Neisseria meningitidis, the causative agent of meningococcal (MC) disease. The DNA glycosylase MutY of the base excision repair pathway is involved in the protection against oxidative stress. MC MutY expressed in Escherichia coli exhibited base excision activity towards DNA substrates containing A:7,8-dihydro-8-oxo-2'-deoxyguanosine and A:C mismatches. Expression in E. coli fully suppressed the elevated spontaneous mutation rate found in the E. coli mutY mutant. An assessment of MutY activity in lysates of neisserial wild-type and mutY mutant strains showed that both MC and gonococcal (GC) MutY is expressed and active in vivo. Strikingly, MC and GC mutY mutants exhibited 60- to 140-fold and 20-fold increases in mutation rates, respectively, compared to the wild-type strains. Moreover, the differences in transitions and transversions in rpoB conferring rifampin resistance observed with the wild type and mutants demonstrated that the neisserial MutY enzyme works in preventing GC-->AT transversions. These findings are important in the context of models linking mutator phenotypes of disease isolates to microbial fitness.
Collapse
Affiliation(s)
- T Davidsen
- Centre for Molecular Biology and Neuroscience and Institute of Microbiology, University of Oslo, Rikshospitalet, N-0027 Oslo, Norway
| | | | | | | |
Collapse
|
50
|
Watson ME, Burns JL, Smith AL. Hypermutable Haemophilus influenzae with mutations in mutS are found in cystic fibrosis sputum. MICROBIOLOGY-SGM 2005; 150:2947-2958. [PMID: 15347753 DOI: 10.1099/mic.0.27230-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Hypermutable bacterial pathogens exist at surprisingly high prevalence and benefit bacterial populations by promoting adaptation to selective environments, including resistance to antibiotics. Five hundred Haemophilus influenzae isolates were screened for an increased frequency of mutation to resistance to rifampicin, nalidixic acid and spectinomycin: of the 14 hypermutable isolates identified, 12 were isolated from cystic fibrosis (CF) sputum. Analysis by enterobacterial repetitive intergenic consensus (ERIC)-PCR and ribotyping identified eight distinct genetic fingerprints. The hypermutable phenotype of seven of the eight unique isolates was associated with polymorphisms in conserved sites of mutS. Four of the mutant mutS alleles were cloned and failed to complement the mutator phenotype of a mutS : : TSTE mutant of H. influenzae strain Rd KW20. Antibiotic susceptibility testing of the hypermutators identified one beta-lactamase-negative ampicillin-resistant (BLNAR) isolate with two isolates producing beta-lactamase. Six isolates from the same patient with CF, with the same genetic fingerprint, were clonal by multilocus sequence typing (MLST). In this clone, there was an evolution to higher MIC values for the antibiotics administered to the patient during the period in which the strains were isolated. Hypermutable H. influenzae with mutations in mutS are prevalent, particularly in the CF lung environment, and may be selected for and maintained by antibiotic pressure.
Collapse
MESH Headings
- Adenosine Triphosphatases/genetics
- Adenosine Triphosphatases/physiology
- Ampicillin Resistance
- Anti-Bacterial Agents/pharmacology
- Bacterial Proteins/genetics
- Bacterial Proteins/physiology
- Cloning, Molecular
- Cystic Fibrosis/microbiology
- DNA Fingerprinting
- DNA, Bacterial/analysis
- DNA, Bacterial/chemistry
- DNA, Bacterial/isolation & purification
- DNA, Intergenic
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Drug Resistance, Bacterial/genetics
- Genes, Bacterial
- Genetic Complementation Test
- Haemophilus influenzae/drug effects
- Haemophilus influenzae/genetics
- Haemophilus influenzae/isolation & purification
- Humans
- Molecular Sequence Data
- MutS DNA Mismatch-Binding Protein
- Mutation
- Nalidixic Acid/pharmacology
- Polymorphism, Genetic
- Repetitive Sequences, Nucleic Acid
- Ribotyping
- Rifampin/pharmacology
- Selection, Genetic
- Sequence Analysis, DNA
- Spectinomycin/pharmacology
- Sputum/microbiology
- beta-Lactamases/analysis
Collapse
Affiliation(s)
- Michael E Watson
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri-Columbia, Columbia, MO 65212, USA
- Seattle Biomedical Research Institute, 307 Westlake Ave N, Suite 500, Seattle, WA 98109, USA
| | - Jane L Burns
- Division of Infectious Diseases, Children's Hospital and Regional Medical Center, 4800 Sand Point Way, Seattle, WA 98105, USA
| | - Arnold L Smith
- Seattle Biomedical Research Institute, 307 Westlake Ave N, Suite 500, Seattle, WA 98109, USA
| |
Collapse
|