1
|
Jiang S, Matuszewska M, Chen M, Hong Y, Chen Y, Wang Z, Zhuang H, Sun L, Zhu F, Wang H, Wu X, Ji S, Holmes MA, Ba X, Chen Y, Yu Y. Emergence and spread of ST5 methicillin-resistant Staphylococcus aureus with accessory gene regulator dysfunction: genomic insights and antibiotic resistance. Microbiol Res 2025; 297:128196. [PMID: 40311457 DOI: 10.1016/j.micres.2025.128196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025]
Abstract
The globally disseminated Staphylococcus aureus ST5 clone poses a major public health threat due to its multidrug resistance and virulence. Here, we identified an agr-dysfunctional (agrA-I238K) ST5 MRSA clone that has spread across East and Southeast Asia, with recent increases in China since its emergence in the 1970s. Comparative genomic analyses identified distinct single-nucleotide polymorphisms and mobile genetic elements linked to enhanced resistance and virulence. This clone exhibits resistance to seven antimicrobial classes, including third-generation tetracyclines and fusidic acid, and shares phenotypic and genetic similarities with the vancomycin-intermediate S. aureus Mu50 strain, including reduced susceptibility to vancomycin, teicoplanin, and daptomycin. The agrA-I238K mutation attenuates hemolytic activity, increases biofilm formation, and reduces daptomycin susceptibility, suggesting a key role in the clone's success. Our results demonstrate the important role of agrA-I238K mutation in the widespread distribution of agr-dysfunctional MRSA and highlight the importance of genomic surveillance in tracking the spread of agr-dysfunctional ST5 MRSA.
Collapse
Affiliation(s)
- Shengnan Jiang
- Centre of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China; Zhejiang University School of Medicine, Hangzhou, Hangzhou, Zhejiang, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China
| | - Marta Matuszewska
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Wellcome Sanger Institute, University of Cambridge, Cambridge, UK
| | - Mengzhen Chen
- Hangzhou Center for Disease Control and Prevention, Hangzhou, China
| | - Yueqin Hong
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiyi Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhengan Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hemu Zhuang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lu Sun
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feiteng Zhu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haiping Wang
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueqing Wu
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shujuan Ji
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mark A Holmes
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Xiaoliang Ba
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| | - Yan Chen
- Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yunsong Yu
- Centre of Laboratory Medicine, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China; Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, China; Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Panigrahi M, Rajawat D, Nayak SS, Jain K, Nayak A, Rajput AS, Sharma A, Dutt T. A comprehensive review on genomic insights and advanced technologies for mastitis prevention in dairy animals. Microb Pathog 2025; 199:107233. [PMID: 39694196 DOI: 10.1016/j.micpath.2024.107233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Mastitis is a multi-etiological disease that significantly impacts milk production and reproductive efficiency. It is highly prevalent in dairy populations subjected to intensive selection for higher milk yield and where inbreeding is common. The issue is amplified by climate change and poor hygiene management, making disease control challenging. Key obstacles include antibiotic resistance, maximum residue levels, horizontal gene transfer, and limited success in breeding for resistance. Predictive genomics offers a promising solution for mastitis prevention by identifying genetic traits linked with susceptibility to mastitis. This review compiles the research and findings on genomics and its allied approaches, such as pan-genomics, epigenetics, proteomics, and transcriptomics, for diagnosing, understanding, and treating mastitis. In dairy production, artificial intelligence (AI), particularly deep learning (DL) techniques like convolutional neural networks (CNNs), has demonstrated significant potential to enhance milk production and improve farm profitability. It highlights the integration of advanced technologies like machine learning (ML), CRISPR, and pan-genomics to improve our knowledge of mastitis epidemiology, pathogen evolution, and the development of more effective diagnostic, preventive and therapeutic strategies for dairy herds. Genomic advancements provide critical insights into the complexities of mastitis, offering new avenues for understanding its dynamics. Integrating these findings with key predisposing factors can drive targeted prevention and more effective disease management.
Collapse
Affiliation(s)
- Manjit Panigrahi
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India.
| | - Divya Rajawat
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Sonali Sonejita Nayak
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Karan Jain
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Ambika Nayak
- Division of Microbiology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Atul Singh Rajput
- Livestock Production and Management Section, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Anurodh Sharma
- Division of Animal Genetics, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| | - Triveni Dutt
- Livestock Production and Management Section, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243122, UP, India
| |
Collapse
|
3
|
Varming AK, Huang Z, Hamad GM, Rasmussen KK, Ingmer H, Kilstrup M, Lo Leggio L. CI:Mor interactions in the lysogeny switches of Lactococcus lactis TP901-1 and Staphylococcus aureus φ13 bacteriophages. MICROBIOME RESEARCH REPORTS 2024; 3:15. [PMID: 38841409 PMCID: PMC11149083 DOI: 10.20517/mrr.2023.50] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 06/07/2024]
Abstract
Aim: To structurally characterize in detail the interactions between the phage repressor (CI) and the antirepressor (Mor) in the lysis-lysogeny switches of two Gram-positive bacteriophages, the lactococcal TP901-1 and staphylococcal φ13. Methods: We use crystallographic structure determination, computational structural modeling, and analysis, as well as biochemical methods, to elucidate similarities and differences in the CI:Mor interactions for the two genetic switches. Results: By comparing a newly determined and other available crystal structures for the N-terminal domain of CI (CI-NTD), we show that the CI interface involved in Mor binding undergoes structural changes upon binding in TP901-1. Most importantly, we show experimentally for the first time the direct interaction between CI and Mor for φ13, and model computationally the interaction interface. The computational modeling supports similar side chain rearrangements in TP901-1 and φ13. Conclusion: This study ascertains experimentally that, like in the TP901-1 lysogeny switch, staphylococcal φ13 CI and Mor interact with each other. The structural basis of the interaction of φ13 CI and Mor was computationally modeled and is similar to the interaction demonstrated experimentally between TP901-1 CI-NTD and Mor, likely involving similar rearrangement of residue side chains during the formation of the complex. The study identifies one CI residue, Glu69, which unusually interacts primarily through its aliphatic chain with an aromatic residue on Mor after changing its conformation compared to the un-complexed structure. This and other residues at the interface are suggested for investigation in future studies.
Collapse
Affiliation(s)
- Anders K. Varming
- Department of Chemistry, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Zhiyu Huang
- Department of Chemistry, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Ghofran M. Hamad
- Department of Chemistry, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Kim K. Rasmussen
- Department of Chemistry, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg DK-1870, Denmark
| | - Mogens Kilstrup
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby DK-2800, Denmark
| | - Leila Lo Leggio
- Department of Chemistry, University of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
4
|
Chu Yuan Kee MJ, Chen J. Phage Transduction of Staphylococcus aureus. Methods Mol Biol 2024; 2738:263-275. [PMID: 37966605 DOI: 10.1007/978-1-0716-3549-0_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Bacteriophage transduction is the major mechanism of horizontal gene transfer (HGT) among many bacteria. In Staphylococcus aureus, the phage-mediated acquisition of mobile genetic elements (MGEs) that encode virulence and antibiotic resistance genes largely contribute to its evolutionary adaptation and genetic plasticity. In molecular biology, generalized transduction is routinely used as a technique to manipulate and construct bacterial strains. Here, we describe optimized protocols for generalized transduction, applicable for the transfer of plasmid or chromosomal deoxyribonucleic acid (DNA) from donor to recipient S. aureus strains.
Collapse
Affiliation(s)
- Melissa-Jane Chu Yuan Kee
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - John Chen
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
5
|
Vinod LA, Rajendran D, Shivashankar M, Chandrasekaran N. Surface interaction of vancomycin with polystyrene microplastics and its effect on human serum albumin. Int J Biol Macromol 2024; 256:128491. [PMID: 38043666 DOI: 10.1016/j.ijbiomac.2023.128491] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Microplastics have a well-documented ability to adsorb various chemicals and contaminants found in the environment. By similar mechanisms, when medicines are stored in plastic packaging, the leaching of plastics into the contents poses the risk of possible toxicity and decreased drug efficacy. The work thus examines the presence of two categories of anthropogenic materials - microplastics (MPs) and medications - with their possible combined effects and fate in biological systems. A study on the kinetics and isotherm of the adsorption of vancomycin hydrochloride on the surface of polystyrene microspheres is performed, and the best-fitting models are obtained respectively as the pseudo-second-order model and the Temkin isotherm. Further, the interaction of each of, the drug, MPs and drug-adsorbed MPs with human serum albumin (HSA), the model protein chosen to validate the potential toxicity in humans, is determined by fluorescence spectroscopy. A thermodynamic analysis of this protein-ligand interaction shows that the process is spontaneous, endothermic and entropically favoured, and that hydrophobic forces operate between the interacting species. An unfolding of HSA is observed, disrupting its functions like the esterase activity. Competitive binding experiments with Warfarin and Ibuprofen as specific site markers on HSA reveal that all the studied ligands bind non-specifically to HSA.
Collapse
Affiliation(s)
- Lydia Ann Vinod
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Durgalakshmi Rajendran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Murugesh Shivashankar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
6
|
Jiang JH, Cameron DR, Nethercott C, Aires-de-Sousa M, Peleg AY. Virulence attributes of successful methicillin-resistant Staphylococcus aureus lineages. Clin Microbiol Rev 2023; 36:e0014822. [PMID: 37982596 PMCID: PMC10732075 DOI: 10.1128/cmr.00148-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a leading cause of severe and often fatal infections. MRSA epidemics have occurred in waves, whereby a previously successful lineage has been replaced by a more fit and better adapted lineage. Selection pressures in both hospital and community settings are not uniform across the globe, which has resulted in geographically distinct epidemiology. This review focuses on the mechanisms that trigger the establishment and maintenance of current, dominant MRSA lineages across the globe. While the important role of antibiotic resistance will be mentioned throughout, factors which influence the capacity of S. aureus to colonize and cause disease within a host will be the primary focus of this review. We show that while MRSA possesses a diverse arsenal of toxins including alpha-toxin, the success of a lineage involves more than just producing toxins that damage the host. Success is often attributed to the acquisition or loss of genetic elements involved in colonization and niche adaptation such as the arginine catabolic mobile element, as well as the activity of regulatory systems, and shift metabolism accordingly (e.g., the accessory genome regulator, agr). Understanding exactly how specific MRSA clones cause prolonged epidemics may reveal targets for therapies, whereby both core (e.g., the alpha toxin) and acquired virulence factors (e.g., the Panton-Valentine leukocidin) may be nullified using anti-virulence strategies.
Collapse
Affiliation(s)
- Jhih-Hang Jiang
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - David R Cameron
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Cara Nethercott
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Marta Aires-de-Sousa
- Laboratory of Molecular Genetics, Institutode Tecnologia Químicae Biológica António Xavier (ITQB-NOVA), Universidade Nova de Lisboa, Oeiras, Portugal
- Escola Superior de Saúde da Cruz Vermelha Portuguesa-Lisboa (ESSCVP-Lisboa), Lisbon, Portugal
| | - Anton Y Peleg
- Department of Microbiology, Infection Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
Rathi R, Kumari R, Pathak SR, Dalal V. Promising antibacterials for LLM of Staphylococcus aureus using virtual screening, molecular docking, dynamics, and MMPBSA. J Biomol Struct Dyn 2023; 41:7277-7289. [PMID: 36073371 DOI: 10.1080/07391102.2022.2119278] [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: 04/08/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
In S. aureus, lipophilic membrane (LLM) protein is a methicillin resistance factor and is an essential role in peptidoglycan metabolism. The virtual screening of antibacterial molecules against the model of LLM was performed to identify the potent antibacterial molecules. Molecular docking results of pharmacokinetic filtered molecules illustrated that five molecules had higher binding affinities than tunicamycin (TUM) and were stabled via non-covalent interactions (hydrogen bond and hydrophobic interactions) at the active site of LLM. Further, molecular dynamics results revealed that binding of identified antibacterial molecules with LLM resulted in stable LLM-inhibitor(s) complexes. Molecular Mechanics/Position-Boltzmann Surface Area (MMPBSA) analysis showed that LLM-inhibitor(s) complexes had high binding affinities in the range of -213.49 ± 2.24 to -227.42 ± 3.05 kJ/mol. The amino acid residues decomposition analysis confirmed that identified antibacterial molecules bound at the active site (Asn148, Leu149, Asp151, Asp208, His269, His271, and His272) of LLM. Noticeably, the current study found five antibacterial molecules (BDE 27575101, BDE 33638168, BDE 33672484, LAS 51502073, and BDE 25098678) were highly potent than TUM and even than earlier reported molecules. Therefore, here reported antibacterial molecules may be used directly or developed to inhibit LLM of S. aureus.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Ravi Rathi
- Amity School of Applied Sciences, Amity University Haryana, Haryana, India
| | - Reena Kumari
- Department of Mathematics and Statistics, Swami Vivekanand Subharti University, Meerut, India
| | - Seema R Pathak
- Amity School of Applied Sciences, Amity University Haryana, Haryana, India
| | - Vikram Dalal
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
8
|
Sivakumar R, Pranav PS, Annamanedi M, Chandrapriya S, Isloor S, Rajendhran J, Hegde NR. Genome sequencing and comparative genomic analysis of bovine mastitis-associated Staphylococcus aureus strains from India. BMC Genomics 2023; 24:44. [PMID: 36698060 PMCID: PMC9878985 DOI: 10.1186/s12864-022-09090-7] [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: 09/19/2022] [Accepted: 12/19/2022] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Bovine mastitis accounts for significant economic losses to the dairy industry worldwide. Staphylococcus aureus is the most common causative agent of bovine mastitis. Investigating the prevalence of virulence factors and antimicrobial resistance would provide insight into the molecular epidemiology of mastitis-associated S. aureus strains. The present study is focused on the whole genome sequencing and comparative genomic analysis of 41 mastitis-associated S. aureus strains isolated from India. RESULTS The results elucidate explicit knowledge of 15 diverse sequence types (STs) and five clonal complexes (CCs). The clonal complexes CC8 and CC97 were found to be the predominant genotypes comprising 21 and 10 isolates, respectively. The mean genome size was 2.7 Mbp with a 32.7% average GC content. The pan-genome of the Indian strains of mastitis-associated S. aureus is almost closed. The genome-wide SNP-based phylogenetic analysis differentiated 41 strains into six major clades. Sixteen different spa types were identified, and eight isolates were untypeable. The cgMLST analysis of all S. aureus genome sequences reported from India revealed that S. aureus strain MUF256, isolated from wound fluids of a diabetic patient, was the common ancestor. Further, we observed that all the Indian mastitis-associated S. aureus isolates belonging to the CC97 are mastitis-associated. We identified 17 different antimicrobial resistance (AMR) genes among these isolates, and all the isolates used in this study were susceptible to methicillin. We also identified 108 virulence-associated genes and discuss their associations with different genotypes. CONCLUSION This is the first study presenting a comprehensive whole genome analysis of bovine mastitis-associated S. aureus isolates from India. Comparative genomic analysis revealed the genome diversity, major genotypes, antimicrobial resistome, and virulome of clinical and subclinical mastitis-associated S. aureus strains.
Collapse
Affiliation(s)
- Ramamoorthy Sivakumar
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, India
| | - Parameswaran Sree Pranav
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, India
| | | | - S Chandrapriya
- Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Bengaluru, 560024, India
| | - Shrikrishna Isloor
- Department of Veterinary Microbiology, Veterinary College, Karnataka Veterinary, Animal and Fisheries Sciences University, Bengaluru, 560024, India
| | - Jeyaprakash Rajendhran
- Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, India.
| | - Nagendra R Hegde
- National Institute of Animal Biotechnology, Hyderabad, 500032, India.
| |
Collapse
|
9
|
Han X, Ortines R, Mukherjee I, Kanipakala T, Kort T, Sherchand SP, Liao G, Mednikov M, Chenine AL, Aman MJ, Nykiforuk CL, Adhikari RP. Hyperimmune Targeting Staphylococcal Toxins Effectively Protect Against USA 300 MRSA Infection in Mouse Bacteremia and Pneumonia Models. Front Immunol 2022; 13:893921. [PMID: 35655774 PMCID: PMC9152286 DOI: 10.3389/fimmu.2022.893921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Staphylococcus aureus has been acquiring multiple drug resistance and has evolved into superbugs such as Methicillin/Vancomycin-resistant S. aureus (MRSA/VRSA) and, consequently, is a major cause of nosocomial and community infections associated with high morbidity and mortality for which no FDA-approved vaccines or biotherapeutics are available. Previous efforts targeting the surface-associated antigens have failed in clinical testing. Here, we generated hyperimmune products from sera in rabbits against six major S. aureus toxins targeted by an experimental vaccine (IBT-V02) and demonstrated significant efficacy for an anti-virulence passive immunization strategy. Extensive in vitro binding and neutralizing titers were analyzed against six extracellular toxins from individual animal sera. All IBT-V02 immunized animals elicited the maximum immune response upon the first boost dose against all pore-forming vaccine components, while for superantigen (SAgs) components of the vaccine, second and third doses of a boost were needed to reach a plateau in binding and toxin neutralizing titers. Importantly, both anti-staphylococcus hyperimmune products consisting of full-length IgG (IBT-V02-IgG) purified from the pooled sera and de-speciated F(ab')2 (IBT-V02-F(ab')2) retained the binding and neutralizing titers against IBT-V02 target toxins. F(ab')2 also exhibited cross-neutralization titers against three leukotoxins (HlgAB, HlgCB, and LukED) and four SAgs (SEC1, SED, SEK, and SEQ) which were not part of IBT-V02. F(ab')2 also neutralized toxins in bacterial culture supernatant from major clinical strains of S. aureus. In vivo efficacy data generated in bacteremia and pneumonia models using USA300 S. aureus strain demonstrated dose-dependent protection by F(ab')2. These efficacy data confirmed the staphylococcal toxins as viable targets and support the further development effort of hyperimmune products as a potential adjunctive therapy for emergency uses against life-threatening S. aureus infections.
Collapse
Affiliation(s)
- Xiaobing Han
- Research and Development, Emergent BioSolutions Canada Inc., Winnipeg, MB, Canada.,Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Roger Ortines
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | - Ipsita Mukherjee
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | | | - Thomas Kort
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | | | - Grant Liao
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | - Mark Mednikov
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | - Agnes L Chenine
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | - M Javad Aman
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| | - Cory L Nykiforuk
- Research and Development, Emergent BioSolutions Canada Inc., Winnipeg, MB, Canada
| | - Rajan P Adhikari
- Integrated Biotherapeutics Inc. (IBT), Rockville, MD, United States
| |
Collapse
|
10
|
Kumari R, Rathi R, Pathak SR, Dalal V. Structural-based virtual screening and identification of novel potent antimicrobial compounds against YsxC of Staphylococcus aureus. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.132476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
11
|
Goswami C, Fox S, Holden M, Leanord A, Evans TJ. Genomic Analysis of Global Staphylococcus argenteus Strains Reveals Distinct Lineages With Differing Virulence and Antibiotic Resistance Gene Content. Front Microbiol 2021; 12:795173. [PMID: 34925305 PMCID: PMC8677677 DOI: 10.3389/fmicb.2021.795173] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Infections due to Staphylococcus argenteus have been increasingly reported worldwide and the microbe cannot be distinguished from Staphylococcus aureus by standard methods. Its complement of virulence determinants and antibiotic resistance genes remain unclear, and how far these are distinct from those produced by S. aureus remains undetermined. In order to address these uncertainties, we have collected 132 publicly available sequences from fourteen different countries, including the United Kingdom, between 2005 and 2018 to study the global genetic structure of the population. We have compared the genomes for antibiotic resistance genes, virulence determinants and mobile genetic elements such as phages, pathogenicity islands and presence of plasmid groups between different clades. 20% (n = 26) isolates were methicillin resistant harboring a mecA gene and 88% were penicillin resistant, harboring the blaZ gene. ST2250 was identified as the most frequent strain, but ST1223, which was the second largest group, contained a marginally larger number of virulence genes compared to the other STs. Novel S. argenteus pathogenicity islands were identified in our isolates harboring tsst-1, seb, sec3, ear, selk, selq toxin genes, as well as chromosomal clusters of enterotoxin and superantigen-like genes. Strain-specific type I modification systems were widespread which would limit interstrain transfer of genetic material. In addition, ST2250 possessed a CRISPR/Cas system, lacking in most other STs. S. argenteus possesses important genetic differences from S. aureus, as well as between different STs, with the potential to produce distinct clinical manifestations.
Collapse
Affiliation(s)
- Cosmika Goswami
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Stephen Fox
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Matthew Holden
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| | - Alistair Leanord
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
- Scottish Microbiology Reference Laboratories, Glasgow, United Kingdom
| | - Thomas J. Evans
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
12
|
Niedziela DA, Cormican P, Foucras G, Leonard FC, Keane OM. Bovine milk somatic cell transcriptomic response to Staphylococcus aureus is dependent on strain genotype. BMC Genomics 2021; 22:796. [PMID: 34740333 PMCID: PMC8571842 DOI: 10.1186/s12864-021-08135-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 10/28/2021] [Indexed: 01/07/2023] Open
Abstract
Background Mastitis is an economically important disease of dairy cows with Staphylococcus aureus a major cause worldwide. Challenge of Holstein-Friesian cows demonstrated that S. aureus strain MOK124, which belongs to Clonal Complex (CC)151, caused clinical mastitis, while strain MOK023, belonging to CC97, caused mild or subclinical mastitis. The aim of this study was to elucidate the molecular mechanisms of the host immune response utilising a transcriptomic approach. Milk somatic cells were collected from cows infected with either S. aureus MOK023 or MOK124 at 0, 24, 48, 72 and 168 h post-infection (hpi) and analysed for differentially expressed (DE) genes in response to each strain. Results In response to MOK023, 1278, 2278, 1986 and 1750 DE genes were found at 24, 48, 72 and 168 hpi, respectively, while 2293, 1979, 1428 and 1544 DE genes were found in response to MOK124 at those time points. Genes involved in milk production (CSN1, CSN10, CSN1S2, CSN2, a-LACTA and PRLR) were downregulated in response to both strains, with a more pronounced decrease in the MOK124 group. Immune response pathways such as NF-κB and TNF signalling were overrepresented in response to both strains at 24 hpi. These immune pathways continued to be overrepresented in the MOK023 group at 48 and 72 hpi, while the Hippo signalling, extracellular matrix interaction (ECM) and tight junction pathways were overrepresented in the MOK124 group between 48 and 168 hpi. Cellular composition analysis demonstrated that a neutrophil response was predominant in response to MOK124, while M1 macrophages were the main milk cell type post-infection in the MOK023 group. Conclusions A switch from immune response pathways to pathways involved in maintaining the integrity of the epithelial cell layer was observed in the MOK124 group from 48 hpi, which coincided with the occurrence of clinical signs in the infected animals. The higher proportion of M1 macrophages in the MOK023 group and lack of substantial neutrophil recruitment in response to MOK023 may indicate immune evasion by this strain. The results of this study highlight that the somatic cell transcriptomic response to S. aureus is dependent on the genotype of the infecting strain. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08135-7.
Collapse
Affiliation(s)
- Dagmara A Niedziela
- Animal and Bioscience Department, Teagasc, Grange, Dunsany, Co. Meath, Ireland.,School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland
| | - Paul Cormican
- Animal and Bioscience Department, Teagasc, Grange, Dunsany, Co. Meath, Ireland
| | - Gilles Foucras
- IHAP, Université de Toulouse, ENVT, INRAE, UMR1225, F-31076, Toulouse, France
| | - Finola C Leonard
- School of Veterinary Medicine, University College Dublin, Dublin 4, Ireland
| | - Orla M Keane
- Animal and Bioscience Department, Teagasc, Grange, Dunsany, Co. Meath, Ireland.
| |
Collapse
|
13
|
Achek R, El-Adawy H, Hotzel H, Hendam A, Tomaso H, Ehricht R, Neubauer H, Nabi I, Hamdi TM, Monecke S. Molecular Characterization of Staphylococcus aureus Isolated from Human and Food Samples in Northern Algeria. Pathogens 2021; 10:pathogens10101276. [PMID: 34684225 PMCID: PMC8537606 DOI: 10.3390/pathogens10101276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus is a commensal resident of the skin and nasal cavities of humans and can cause various infections. Some toxigenic strains can contaminate food matrices and cause foodborne intoxications. The present study aimed to provide relevant information (clonal complex lineages, agr types, virulence and antimicrobial resistance-associated genes) based on DNA microarray analyses as well as the origins and dissemination of several circulating clones of 60 Staphylococcus aureus isolated from food matrices (n = 24), clinical samples (n = 20), and nasal carriers (n = 16) in northern Algeria. Staphylococcus aureus were genotyped into 14 different clonal complexes. Out of 60 S. aureus, 13 and 10 isolates belonged to CC1-MSSA and CC97-MSSA, respectively. The CC 80-MRSA-IV was the predominant S. aureus strain in clinical isolates. The accessory gene regulator allele agr group III was mainly found among clinical isolates (70.4%). Panton–Valentine leukocidin genes lukF/lukS-PV were detected in 13.3% of isolates that all belonged to CC80-MRSA. The lukF/S-hlg, hlgA, and hla genes encoding for hemolysins and leucocidin components were detected in all Staphylococcusaureus isolates. Clinical and food isolates harbored more often the antibiotic resistance genes markers. Seventeen (28.3%) methicillin-resistant Staphylococcus aureus carrying the mecA gene localized on a SCCmec type IV element were identified. The penicillinase operon (blaZ/I/R) was found in 71.7% (43/60) of isolates. Food isolates belonging to CC97-MSSA carried several antibiotic resistance genes (blaZ, ermB, aphA3, sat, tetM, and tetK). The results of this study showed that all clones were found in their typical host, but interestingly, some nasal carriers had isolates assigned to CC705 thought to be absent in humans. The detection of MRSA strains among food isolates should be considered as a potential public health risk. Therefore, controlling the antibiotics prescription for a rational use in human and animal infections is mandatory.
Collapse
Affiliation(s)
- Rachid Achek
- Faculty of Nature and Life and Earth Sciences, Djilali-Bounaama University, Soufay, Khemis-Miliana 44225, Algeria;
- Laboratory of Food Hygiene and Quality Assurance System, High National Veterinary School, Oued Smar, Algiers 16059, Algeria;
| | - Hosny El-Adawy
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
- Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 35516, Egypt
- Correspondence:
| | - Helmut Hotzel
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ashraf Hendam
- Climate Change Information Center, Renewable Energy and Expert Systems (CCICREES), Agricultural Research Center, 9 Algamaa Street, Giza 12619, Egypt;
| | - Herbert Tomaso
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ralf Ehricht
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany; (R.E.); (S.M.)
- InfectoGnostics Research Campus Jena e. V., 07743 Jena, Germany
- Institute of Physical Chemistry, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Heinrich Neubauer
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, 07743 Jena, Germany; (H.H.); (H.T.); (H.N.)
| | - Ibrahim Nabi
- Faculty of Sciences, Yahia Farès University, Urban Pole, Médéa 26000, Algeria;
| | - Taha Mossadak Hamdi
- Laboratory of Food Hygiene and Quality Assurance System, High National Veterinary School, Oued Smar, Algiers 16059, Algeria;
| | - Stefan Monecke
- Leibniz Institute of Photonic Technology (IPHT), 07745 Jena, Germany; (R.E.); (S.M.)
- InfectoGnostics Research Campus Jena e. V., 07743 Jena, Germany
- Institute for Medical Microbiology and Virology, Dresden University Hospital, 01307 Dresden, Germany
| |
Collapse
|
14
|
Naorem RS, Goswami G, Gyorgy S, Fekete C. Comparative analysis of prophages carried by human and animal-associated Staphylococcus aureus strains spreading across the European regions. Sci Rep 2021; 11:18994. [PMID: 34556764 PMCID: PMC8460829 DOI: 10.1038/s41598-021-98432-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/08/2021] [Indexed: 02/08/2023] Open
Abstract
Staphylococcus aureus is a major human and animal pathogen although the animal-associated S. aureus can be a potential risk of human zoonoses. Acquisition of phage-related genomic islands determines the S. aureus species diversity. This study characterized and compared the genome architecture, distribution nature, and evolutionary relationship of 65 complete prophages carried by human and animal-associated S. aureus strains spreading across the European regions. The analyzed prophage genomes showed mosaic architecture with extensive variation in genome size. The phylogenetic analyses generated seven clades in which prophages of the animal-associated S. aureus scattered in all the clades. The S. aureus strains with the same SCCmec type, and clonal complex favored the harboring of similar prophage sequences and suggested that the frequency of phage-mediated horizontal gene transfer is higher between them. The presence of various virulence factors in prophages of animal-associated S. aureus suggested that these prophages could have more pathogenic potential than prophages of human-associated S. aureus. This study showed that the S. aureus phages are dispersed among the several S. aureus serotypes and around the European regions. Further, understanding the phage functional genomics is necessary for the phage-host interactions and could be used for tracing the S. aureus strains transmission.
Collapse
Affiliation(s)
- Romen Singh Naorem
- grid.9679.10000 0001 0663 9479Department of General and Environmental Microbiology, Institute of Biology and Sport Biology, University of Pécs, Ifjusag utja. 6, Pecs, 7624 Hungary
| | - Gunajit Goswami
- Multidisciplinary Research Unit, Jorhat Medical College and Hospital, Jorhat, Assam India
| | - Schneider Gyorgy
- grid.9679.10000 0001 0663 9479Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pecs, Hungary
| | - Csaba Fekete
- grid.9679.10000 0001 0663 9479Department of General and Environmental Microbiology, Institute of Biology and Sport Biology, University of Pécs, Ifjusag utja. 6, Pecs, 7624 Hungary
| |
Collapse
|
15
|
Rumpf C, Lange J, Schwartbeck B, Kahl BC. Staphylococcus aureus and Cystic Fibrosis-A Close Relationship. What Can We Learn from Sequencing Studies? Pathogens 2021; 10:1177. [PMID: 34578208 PMCID: PMC8466686 DOI: 10.3390/pathogens10091177] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 01/13/2023] Open
Abstract
Staphylococcus aureus is next to Pseudomonas aeruginosa the most isolated pathogen from the airways of cystic fibrosis (CF) patients, who are often infected by a dominant S. aureus clone for extended periods. To be able to persist, the pathogen has to adapt to the hostile niche of the airways to counteract host defence, antibiotic therapy and the competition with coinfecting pathogens. S. aureus is equipped with many virulence factors including adhesins, toxins that are localized on the chromosome, on plasmids or are phage-related. S. aureus is especially versatile and adaptation and evolution of the pathogen occurs by the acquisition of new genes by horizontal gene transfer (HGT), changes in nucleotides (single nucleotide variations, SNVs) that can cause a selective advantage for the bacteria and become fixed in subpopulations. Methicillin-resistant S. aureus are a special threat to CF patients due to the more severe lung disease occurring in infected patients. Today, with decreasing costs for sequencing, more and more studies using S. aureus isolates cultured from CF patients are being published, which use whole genome sequencing (WGS), multilocus sequence typing (MLST) or spa-sequence typing (spa-typing) to follow the population dynamics of S. aureus, elucidate the underlying mechanisms of phenotypic variants, newly acquired resistance or adaptation to the host response in this particular niche. In the first part of this review, an introduction to the genetic make-up and the pathogenesis of S. aureus with respect to CF is provided. The second part presents an overview of recent studies and their findings using genotypic methods such as single or multilocus sequencing and whole genome sequencing, which identify factors contributing to the adaptation of S. aureus and its evolution in the airways of individuals with CF.
Collapse
Affiliation(s)
| | | | | | - Barbara C. Kahl
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (C.R.); (J.L.); (B.S.)
| |
Collapse
|
16
|
Molecular Characterization of Staphylococcus aureus Isolated from Raw Milk Samples of Dairy Cows in Manhiça District, Southern Mozambique. Microorganisms 2021; 9:microorganisms9081684. [PMID: 34442763 PMCID: PMC8402116 DOI: 10.3390/microorganisms9081684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 08/04/2021] [Indexed: 11/18/2022] Open
Abstract
Staphylococcal infections are among the most common foodborne diseases. We performed the antibiotic susceptibility and molecular characterization of S. aureus from milk samples of dairy cows in Manhiça District. We observed a high frequency of S. aureus (41%, 58/143), in which 71% (41/58) were from commercial farms and 29% (17/58) from smallholder farms. Half of the isolates (50%, 29/58) were resistant to at least one antibiotic, with higher rates of resistance to penicillin (43%, 25/58), followed by tetracycline (16%, 9/58). Multidrug-resistant and methicillin-resistant S. aureus isolates were rare (5%, 3/58 and 3%, 2/58, respectively). The genetic diversity was low, with predominance of human-adapted strains being: ST1/CC1-t5388 (78%) and ST152-t1299 (10%), followed by ST8/CC8-t1476 (5%) and ST5/CC5-t002 (3%) and lastly, ST508/CC45-t331 and ST152-t355, with 2% each. The Panton–Valentine leukocidin (PVL) gene was detected among 14% (8/58) of the isolates, while genes encoding staphylococcal enterotoxins were scarce (3%, 2/58). Our findings revealed a high frequency of S. aureus, with high rates of resistance to the antibiotics commonly used in veterinary and human medicine. Further investigations focusing on the molecular epidemiology of S. aureus from cattle and farmers will provide detailed insights on the genetic relatedness between the strains.
Collapse
|
17
|
Genomic islands and the evolution of livestock-associated Staphylococcus aureus genomes. Biosci Rep 2021; 40:226941. [PMID: 33185245 PMCID: PMC7689654 DOI: 10.1042/bsr20202287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/23/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genomic Islands (GIs) are commonly believed to be relics of horizontal transfer and associated with specific metabolic capacities, including virulence of the strain. Horizontal gene transfer (HGT) plays a vital role in the acquisition of GIs and the evolution and adaptation of bacterial genomes. OBJECTIVE The present study was designed to predict the GIs and role of HGT in evolution of livestock-associated Staphylococcus aureus (LA-SA). METHODS GIs were predicted with two methods namely, Ensemble algorithm for Genomic Island Detection (EGID) tool, and Seq word Sniffer script. Functional characterization of GI elements was performed with clustering of orthologs. The putative donor predictions of GIs was done with the aid of the pre_GI database. RESULTS The present study predicted a pan of 46 GIs across the LA-SA genomes. Functional characterization of GI sequences revealed few unique results like the presence of metabolic operons like leuABCD and folPK genes in GIs and showed the importance of GIs in the adaptation to the host niche. The developed framework for GI donor prediction results revealed Rickettsia and Mycoplasma as the major donors of GI elements. CONCLUSIONS The role of GIs during the evolutionary race of LA-SA could be concluded from the present study. Niche adaptation of LA-SA enhanced presumably due to these GIs. Future studies could focus on the evolutionary relationships between Rickettsia and Mycoplasma sp. with S. aureus and also the evolution of Leucine/Isoleucine mosaic operon (leuABCD).
Collapse
|
18
|
Di Gregorio S, Haim MS, Vielma Vallenilla J, Cohen V, Rago L, Gulone L, Aanensen DM, Argimón S, Mollerach M. Genomic Epidemiology of CC30 Methicillin-Resistant Staphylococcus aureus Strains from Argentina Reveals Four Major Clades with Distinctive Genetic Features. mSphere 2021; 6:e01297-20. [PMID: 33692199 PMCID: PMC8546718 DOI: 10.1128/msphere.01297-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/10/2021] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus clonal complex 30 (CC30) has given rise to epidemics worldwide and is one of the most prevalent lineages in Argentina, represented by sequence type 30 methicillin-resistant S. aureus SCCmec type IV (ST30-MRSA-IV). ST30-MRSA-IV has displaced previous prevalent clones in the country and demonstrated increased virulence. Despite the burden of infections caused by ST30-MRSA-IV both in hospitals and in communities in Argentina, no detailed genome-based characterization of this clone is available to date. In this study, we used whole-genome sequencing (WGS) to evaluate the genetic diversity, population structure, and genomic characteristics of 190 CC30-MRSA strains circulating in Argentina between 2004 and 2015. Phylogenetic analysis revealed the existence of 4 major clades: ARG-1 (CC30-MRSA-IVc-spa t012), ARG-2 (ST30-MRSA-IVc-spa t021 related), ARG-3 (ST30-MRSA-IVh/j-spa t021 and related), and ARG-4 (CC30-MRSA-IVc-spa t019 and related). The clades were characterized by different distributions of antimicrobial resistance determinants, virulence genes, and mobile genetic elements (MGEs). While ARG-1 and ARG-4 were related to global epidemic MRSA-16 (EMRSA-16) and South West Pacific (SWP) clones, respectively, ARG-3 was phylogenetically distinct from previously defined CC30 epidemic clones. ARG-4, the most prevalent and geographically disseminated in the collection (N = 164), was characterized by specific MGEs and chromosomal mutations that might have contributed to its virulence and success. To our knowledge, this is the first genomic epidemiology study of CC30-MRSA in Argentina, which will serve as baseline genomic data going forward to inform public health measures for infection prevention and control.IMPORTANCE The rise in prevalence of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is of public health concern. In Argentina, several studies documented a shift in the epidemiology of CA-MRSA since 2009, with clonal complex 30 (CC30) and, in particular, sequence type 30 MRSA SCCmec type IV (ST30-MRSA-IV) replacing other clones both in communities and in hospitals and possibly displaying increased virulence. By sequencing the whole genomes of 190 CC30 MRSA isolates recovered from Argentina between 2005 and 2015, we showed that they represented a diverse population composed of 4 major clades. The predominant clade evolved from the South West Pacific clone but has acquired a distinct repertoire of mobile genetic elements, virulence genes, and chromosomal mutations that might play a role in its success. Our work is the first extensive genomic study of CC30 S. aureus in Argentina and will contribute not only to the development of genomic surveillance in the region but also to our understanding of the global epidemiology of this pathogen.
Collapse
Affiliation(s)
- Sabrina Di Gregorio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
| | - María Sol Haim
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jesús Vielma Vallenilla
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
| | - Victoria Cohen
- The Centre for Genomic Pathogen Surveillance (CGPS), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lucía Rago
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
| | - Lucía Gulone
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - David M Aanensen
- The Centre for Genomic Pathogen Surveillance (CGPS), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Silvia Argimón
- The Centre for Genomic Pathogen Surveillance (CGPS), Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Marta Mollerach
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriología y Virología Molecular (IBaViM), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
19
|
Cafiso V, Lo Verde F, Zega A, Pigola G, Rostagno R, Borrè S, Stefani S. Genomic Characterization of a New Biofilm-Forming and Adhesive ST398 Human-Adapted MSSA Lineage Causing Septic Knee Arthritis Following Surgical Reconstruction. Microorganisms 2021; 9:microorganisms9020305. [PMID: 33540689 PMCID: PMC7913009 DOI: 10.3390/microorganisms9020305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 01/04/2023] Open
Abstract
Methicillin-susceptible (MSSA) and methicillin-resistant Staphylococcus aureus (MRSA) is a pathogen commonly found in bone and joint infections, including septic arthritis. S. aureus virulence and the frailty of affected patients can cause several complications; a prompt and specific antibiotic treatment can positively affect the outcome of patients. We carried out an in-depth genomic characterization by Illumina whole genome sequencing and bioinformatics of two biofilm-producing M1 and M2 ST398 MSSA causing septic knee arthritis not-responding to antimicrobial therapy. The strains were characterized for antibiotic resistance, biofilm and adhesive properties as well as genomics, single nucleotide polymorphism phylogeny, resistomics and virulomics. Our results showed that M1 and M2 MSSA were ST398-t1451-agrI-Cap5, susceptible to cefoxitin and resistant to erythromycin and clindamycin, traits consistent with the lack of the SCCmec-locus and the presence of the sole blaZ and ermT. Furthermore, M1 and M2 were biofilm-producing and largely potentially adhesive strains, as indicated by the adhesion gene profile. Our data characterized a new human-adapted ST398 MSSA lineage, representing a "fusion" between the human-animal independent ST398 and the Livestock Associated (LA) ST398 lineages, forming biofilm and genomically predicted high adhesive, characterized by different genomic adaptation conferring a great ability to adhere to the host's extracellular matrix causing septic knee arthritis.
Collapse
Affiliation(s)
- Viviana Cafiso
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.L.V.); (A.Z.); (G.P.); (S.S.)
- Correspondence:
| | - Flavia Lo Verde
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.L.V.); (A.Z.); (G.P.); (S.S.)
| | - Alessandra Zega
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.L.V.); (A.Z.); (G.P.); (S.S.)
| | - Giuseppe Pigola
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.L.V.); (A.Z.); (G.P.); (S.S.)
| | - Roberto Rostagno
- Infectious Diseases Department of Sant’Andrea Hospital Vercelli, 13100 Vercelli, Italy; (R.R.); (S.B.)
| | - Silvio Borrè
- Infectious Diseases Department of Sant’Andrea Hospital Vercelli, 13100 Vercelli, Italy; (R.R.); (S.B.)
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.L.V.); (A.Z.); (G.P.); (S.S.)
| |
Collapse
|
20
|
Genomic analysis of European bovine Staphylococcus aureus from clinical versus subclinical mastitis. Sci Rep 2020; 10:18172. [PMID: 33097797 PMCID: PMC7584570 DOI: 10.1038/s41598-020-75179-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/09/2020] [Indexed: 12/30/2022] Open
Abstract
Intramammary infections (IMI) with Staphylococcus aureus are a common cause of bovine mastitis and can result in both clinical (CM) or subclinical mastitis (SCM). Although bacterial isolates of S. aureus differ in their virulence potential it is largely unclear which bacterial virulence factors are responsible for increased clinical severity. We performed a genome wide association study and used a generalized linear mixed model to investigate the correlation between gene carriage, lineage and clinical outcome of IMI in a collection of S. aureus isolates from cattle with CM (n = 125) and SCM (n = 151) from 11 European countries. An additional aim was to describe the genetic variation of bovine S. aureus in Europa. The dominant lineages in our collection were clonal complex (CC) 151 (81/276, 29.3%), CC97 (54/276, 19.6%), CC479 (32/276, 11.6%) and CC398 (19/276, 6.9%). Virulence and antimicrobial resistance (AMR) gene carriage was highly associated with CC. Among a selection of nine virulence and AMR genes, CC151, CC479 and CC133 carried more virulence genes than other CCs, and CC398 was associated with AMR gene carriage. Whereas CC151, CC97 were widespread in Europe, CC479, CC398 and CC8 were only found in specific countries. Compared to CC151, CC479 was associated with CM rather than SCM (OR 3.62; 95% CI 1.38-9.50) and the other CCs were not. Multiple genes were associated with CM, but due to the clustering within CC of carriage of these genes, it was not possible to differentiate between the effect of gene carriage and CC on clinical outcome of IMI. Nevertheless, this study demonstrates that characterization of S. aureus CC and virulence genes helps to predict the likelihood of the occurrence of CM following S. aureus IMI and highlights the potential benefit of diagnostics tools to identify S. aureus CC during bovine mastitis.
Collapse
|
21
|
Guimaraes AO, Cao Y, Hong K, Mayba O, Peck MC, Gutierrez J, Ruffin F, Carrasco-Triguero M, Dinoso JB, Clemenzi-Allen A, Koss CA, Maskarinec SA, Chambers HF, Fowler VG, Baruch A, Rosenberger CM. A Prognostic Model of Persistent Bacteremia and Mortality in Complicated Staphylococcus aureus Bloodstream Infection. Clin Infect Dis 2020; 68:1502-1511. [PMID: 30165412 DOI: 10.1093/cid/ciy739] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is a leading cause of bacteremia, yet there remains a significant knowledge gap in the identification of relevant biomarkers that predict clinical outcomes. Heterogeneity in the host response to invasive S. aureus infection suggests that specific biomarker signatures could be utilized to differentiate patients prone to severe disease, thereby facilitating earlier implementation of more aggressive therapies. METHODS To further elucidate the inflammatory correlates of poor clinical outcomes in patients with S. aureus bacteremia, we evaluated the association between a panel of blood proteins at initial presentation of bacteremia and disease severity outcomes using 2 cohorts of patients with S. aureus bacteremia (n = 32 and n = 124). RESULTS We identified 13 candidate proteins that were correlated with mortality and persistent bacteremia. Prognostic modeling identified interleukin (IL)-8 and CCL2 as the strongest individual predictors of mortality, with the combination of these biomarkers classifying fatal outcome with 89% sensitivity and 77% specificity (P < .0001). Baseline IL-17A levels were elevated in patients with persistent bacteremia (P < .0001), endovascular (P = .026) and metastatic tissue infections (P = .012). CONCLUSIONS These results demonstrate the potential utility of selected biomarkers to distinguish patients with the highest risk for treatment failure and bacteremia-related complications, providing a valuable tool for clinicians in the management of S. aureus bacteremia. Additionally, these biomarkers could identify patients with the greatest potential to benefit from novel therapies in clinical trials.
Collapse
Affiliation(s)
| | - Yi Cao
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Kyu Hong
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Oleg Mayba
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Melicent C Peck
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Johnny Gutierrez
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | | | - Jason B Dinoso
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | - Angelo Clemenzi-Allen
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Catherine A Koss
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | | | - Henry F Chambers
- Division of HIV, Infectious Diseases, and Global Medicine, University of California San Francisco
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University, Durham, North Carolina
| | - Amos Baruch
- Biomarker Discovery, Genentech, Inc., South San Francisco, California
| | | |
Collapse
|
22
|
Gaidar D, Jonas A, Akulenko R, Ruffing U, Herrmann M, Helms V, von Müller L. Analysis of the dynamics of Staphylococcus aureus binding to white blood cells using whole blood assay and geno-to-pheno mapping. Int J Med Microbiol 2020; 310:151411. [DOI: 10.1016/j.ijmm.2020.151411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/02/2019] [Accepted: 02/03/2020] [Indexed: 12/28/2022] Open
|
23
|
Vaiyapuri M, Joseph TC, Rao BM, Lalitha KV, Prasad MM. Methicillin-Resistant Staphylococcus aureus in Seafood: Prevalence, Laboratory Detection, Clonal Nature, and Control in Seafood Chain. J Food Sci 2019; 84:3341-3351. [PMID: 31769517 DOI: 10.1111/1750-3841.14915] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 11/30/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), a versatile pathogen bearing multiple virulence determinants, is increasingly being detected in various food-producing animals, including fish. In addition, it is a potential food poisoning agent. MRSA is not an inherent microbiota of fish; its presence is attributed to pre- or postharvest contamination through fish handlers, water, ice, and processing equipment. Several reviews have been written on MRSA in clinical as well as the food animal-producing sector, but information specific to MRSA in seafood is scant. This review puts forth insights on MRSA detection in seafood, antibiotic resistance, diversity of clones in seafood, and possible control measures in seafood production chain. Emphasis has been given on assessing the variations in the protocols employed for isolation and identification in different food matrices and lay the foundation for researchers to develop optimized procedure.
Collapse
Affiliation(s)
- Murugadas Vaiyapuri
- Microbiology, Fermentation and Biotechnology (MFB) Division, ICAR-Central Inst. of Fisheries Technology (ICAR-CIFT), Willingdon Island, Cochin, 682029, India
| | | | | | | | - Mothadaka Mukteswar Prasad
- Microbiology, Fermentation and Biotechnology (MFB) Division, ICAR-Central Inst. of Fisheries Technology (ICAR-CIFT), Willingdon Island, Cochin, 682029, India
| |
Collapse
|
24
|
Abstract
The complex regulatory role of the proteases necessitates very tight coordination and control of their expression. While this process has been well studied, a major oversight has been the consideration of proteases as a single entity rather than as 10 enzymes produced from four different promoters. As such, in this study, we comprehensively characterized the regulation of each protease promoter, discovering vast differences in the way each protease operon is controlled. Additionally, we broaden the picture of protease regulation using a global screen to identify novel loci controlling protease activity, uncovering a cadre of new effectors of protease expression. The impact of these elements on the activity of proteases and known regulators was characterized by producing a comprehensive regulatory circuit that emphasizes the complexity of protease regulation in Staphylococcus aureus. A primary function of the extracellular proteases of Staphylococcus aureus is to control the progression of infection by selectively modulating the stability of virulence factors. Consequently, a regulatory network exists to titrate protease abundance/activity to influence the accumulation, or lack thereof, of individual virulence factors. Herein, we comprehensively map this system, exploring the regulation of the four protease loci by known and novel factors. In so doing, we determined that seven major elements (SarS, SarR, Rot, MgrA, CodY, SaeR, and SarA) form the primary network of control, with the latter three being the most powerful. We note that expression of aureolysin is largely repressed by these factors, while the spl operon is subject to the strongest upregulation of any protease loci, particularly by SarR and SaeR. Furthermore, when exploring scpA expression, we find it to be profoundly influenced in opposing fashions by SarA (repressor) and SarR (activator). We also present the screening of >100 regulator mutants of S. aureus, identifying 7 additional factors (ArgR2, AtlR, MntR, Rex, XdrA, Rbf, and SarU) that form a secondary circuit of protease control. Primarily, these elements serve as activators, although we reveal XdrA as a new repressor of protease expression. With the exception or ArgR2, each of the new effectors appears to work through the primary network of regulation to influence protease production. Collectively, we present a comprehensive regulatory circuit that emphasizes the complexity of protease regulation and suggest that its existence speaks to the importance of these enzymes to S. aureus physiology and pathogenic potential. IMPORTANCE The complex regulatory role of the proteases necessitates very tight coordination and control of their expression. While this process has been well studied, a major oversight has been the consideration of proteases as a single entity rather than as 10 enzymes produced from four different promoters. As such, in this study, we comprehensively characterized the regulation of each protease promoter, discovering vast differences in the way each protease operon is controlled. Additionally, we broaden the picture of protease regulation using a global screen to identify novel loci controlling protease activity, uncovering a cadre of new effectors of protease expression. The impact of these elements on the activity of proteases and known regulators was characterized by producing a comprehensive regulatory circuit that emphasizes the complexity of protease regulation in Staphylococcus aureus.
Collapse
|
25
|
Zaatout N, Ayachi A, Kecha M, Kadlec K. Identification of staphylococci causing mastitis in dairy cattle from Algeria and characterization of Staphylococcus aureus. J Appl Microbiol 2019; 127:1305-1314. [PMID: 31356718 DOI: 10.1111/jam.14402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/29/2019] [Accepted: 07/22/2019] [Indexed: 12/26/2022]
Abstract
AIMS This study was conducted to determine the occurrence of staphylococci from cows with subclinical mastitis from independent herds in Algeria, and to characterize Staphylococcus aureus isolates. METHODS AND RESULTS Quarter milk samples were collected separately, somatic cells were counted and samples with more than 200 000 somatic cells per ml were cultured on blood agar. Staphylococci isolates were identified by routine diagnostics, and S. aureus isolates were tested for antibiotic susceptibility by disk diffusion and microdilution. Congo red agar was used to detect biofilm formation and capsule synthesis was detected on serum soft agar (SSA). The S. aureus isolates were characterized by spa typing. DNA microarray analysis was performed to detect resistance and virulence genes. Overall, 40·0% (167/418) of the cows suffered from mastitis. In 63·5% (106/167) of the cows staphylococci were identified. Nine of the 106 Staphylococcus isolates (8·5%) were S. aureus. The coagulase-negative staphylococci belonged to 14 species. All S. aureus isolates were multiresistant and biofilm forming, with 66·67% of them showing diffuse colonies on SSA and belonged to CC97-agrI-cap5. Biofilm genes (icaA/C/D), 13 genes encoding for adhesion, six genes encoding proteases, 11 genes encoding superantigen like toxins were found. Genes conferring resistance to tetracycline (tet(K)), penicillin (blaZ/I/R) and macrolide-lincosamide-streptogramin B (erm(B), erm(A)) were also detected in the S. aureus from this study. CONCLUSIONS The current investigation provides a detailed molecular and biofilm formation ability of S. aureus involved in subclinical mastitis in Algeria and shows the wide distribution of adhesion and enterotoxin(-like) genes among S. aureus responsible for causing subclinical bovine mastitis. SIGNIFICANCE AND IMPACT OF THE STUDY These findings are valuable in tracking the evolution and genomic variation of S. aureus from bovine origin.
Collapse
Affiliation(s)
- N Zaatout
- Laboratory of Applied Microbiology, Faculty of Nature and Life Sciences, University of Bejaia, Bejaia, Algeria
| | - A Ayachi
- Institute of Veterinary and Agricultural Sciences, University of Batna, Batna, Algeria
| | - M Kecha
- Laboratory of Applied Microbiology, Faculty of Nature and Life Sciences, University of Bejaia, Bejaia, Algeria
| | - K Kadlec
- Dairy Herd Consulting and Research Company (MBFG), Wunstorf, Germany
| |
Collapse
|
26
|
Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-virulence Therapy Using Natural Products. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0059-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
27
|
de Carvalho SP, de Almeida JB, de Freitas LM, Guimarães AMS, do Nascimento NC, Dos Santos AP, Campos GB, Messick JB, Timenetsky J, Marques LM. Genomic profile of Brazilian methicillin-resistant Staphylococcus aureus resembles clones dispersed worldwide. J Med Microbiol 2019; 68:693-702. [PMID: 30900970 DOI: 10.1099/jmm.0.000956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Comparative genomic analysis of strains may help us to better understand the wide diversity of their genetic profiles. The aim of this study was to analyse the genomic features of the resistome and virulome of Brazilian first methicillin-resistant Staphylococcus aureus (MRSA) isolates and their relationship to other Brazilian and international MRSA strains. METHODOLOGY The whole genomes of three MRSA strains previously isolated in Vitória da Conquista were sequenced, assembled, annotated and compared with other MRSA genomes. A phylogenetic tree was constructed and the pan-genome and accessory and core genomes were constructed. The resistomes and virulomes of all strains were identified.Results/Key findings. Phylogenetic analysis of all 49 strains indicated different clones showing high similarity. The pan-genome of the analysed strains consisted of 4484 genes, with 31 % comprising the gene portion of the core genome, 47 % comprising the accessory genome and 22 % being singletons. Most strains showed at least one gene related to virulence factors associated with immune system evasion, followed by enterotoxins. The strains showed multiresistance, with the most recurrent genes conferring resistance to beta-lactams, fluoroquinolones, aminoglycosides and macrolides. CONCLUSIONS Our comparative genomic analysis showed that there is no pattern of virulence gene distribution among the clones analysed in the different regions. The Brazilian strains showed similarity with clones from several continents.
Collapse
Affiliation(s)
- Suzi P de Carvalho
- Department of Biological Sciences, Santa Cruz State University, Ilhéus-Itabuna Road, km 16 Salobrinho, Ilhéus 45662-900, Bahia State, Brazil
| | - Jéssica B de Almeida
- Department of Biological Sciences, Santa Cruz State University, Ilhéus-Itabuna Road, km 16 Salobrinho, Ilhéus 45662-900, Bahia State, Brazil
- Multidisciplinary Institute of Health, Universidade Federal da Bahia, Rio de Contas Street, Square 17, Number 58, Candeias, Vitória da Conquista, 45029-094, Bahia State, Brazil
| | - Leandro M de Freitas
- Multidisciplinary Institute of Health, Universidade Federal da Bahia, Rio de Contas Street, Square 17, Number 58, Candeias, Vitória da Conquista, 45029-094, Bahia State, Brazil
| | - Ana Marcia S Guimarães
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, 1374 Professor Lineu Prestes Avenue, Sao Paulo, 05508-900, São Paulo State, Brazil
| | - Naíla C do Nascimento
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Andrea P Dos Santos
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Guilherme B Campos
- Multidisciplinary Institute of Health, Universidade Federal da Bahia, Rio de Contas Street, Square 17, Number 58, Candeias, Vitória da Conquista, 45029-094, Bahia State, Brazil
| | - Joanne B Messick
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA
| | - Jorge Timenetsky
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, 1374 Professor Lineu Prestes Avenue, Sao Paulo, 05508-900, São Paulo State, Brazil
| | - Lucas M Marques
- Multidisciplinary Institute of Health, Universidade Federal da Bahia, Rio de Contas Street, Square 17, Number 58, Candeias, Vitória da Conquista, 45029-094, Bahia State, Brazil
- Department of Biological Sciences, Santa Cruz State University, Ilhéus-Itabuna Road, km 16 Salobrinho, Ilhéus 45662-900, Bahia State, Brazil
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, 1374 Professor Lineu Prestes Avenue, Sao Paulo, 05508-900, São Paulo State, Brazil
| |
Collapse
|
28
|
Muthuirulandi Sethuvel DP, Devanga Ragupathi NK, Bakthavatchalam YD, Vijayakumar S, Varghese R, Shankar C, Jacob JJ, Vasudevan K, Elangovan D, Balaji V. Current strategy for local- to global-level molecular epidemiological characterisation of global antimicrobial resistance surveillance system pathogens. Indian J Med Microbiol 2019; 37:147-162. [PMID: 31745014 DOI: 10.4103/ijmm.ijmm_19_396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The prime goal of molecular epidemiology is to identify the origin and evolution of pathogens, which can potentially influence the public health worldwide. Traditional methods provide limited information which is not sufficient for outbreak investigation and studying transmission dynamics. The recent advancement of next-generation sequencing had a major impact on molecular epidemiological studies. Currently, whole-genome sequencing (WGS) has become the gold standard typing method, especially for clinically significant pathogens. Here, we aimed to describe the application of appropriate molecular typing methods for global antimicrobial resistance surveillance system pathogens based on the level of discrimination and epidemiological settings. This shows that sequence-based methods such as multi-locus sequence typing (MLST) are widely used due to cost-effectiveness and database accessibility. However, WGS is the only method of choice for studying Escherichia coli and Shigella spp. WGS is shown to have higher discrimination than other methods in typing Klebsiella pneumoniae, Acinetobacter baumannii and Salmonella spp. due to its changing accessory genome content. For Gram positives such as Streptococcus pneumoniae, WGS would be preferable to understand the evolution of the strains. Similarly, for Staphylococcus aureus, combination of MLST, staphylococcal protein A or SCCmec typing along with WGS could be the choice for epidemiological typing of hospital- and community-acquired strains. This review highlights that combinations of different typing methods should be used to get complete information since no one standalone method is sufficient to study the varying genome diversity.
Collapse
Affiliation(s)
| | | | | | - Saranya Vijayakumar
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Rosemol Varghese
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Chaitra Shankar
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Jobin John Jacob
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Karthick Vasudevan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Divyaa Elangovan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Veeraraghavan Balaji
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
29
|
Tam K, Torres VJ. Staphylococcus aureus Secreted Toxins and Extracellular Enzymes. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0039-2018. [PMID: 30873936 PMCID: PMC6422052 DOI: 10.1128/microbiolspec.gpp3-0039-2018] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus is a formidable pathogen capable of causing infections in different sites of the body in a variety of vertebrate animals, including humans and livestock. A major contribution to the success of S. aureus as a pathogen is the plethora of virulence factors that manipulate the host's innate and adaptive immune responses. Many of these immune modulating virulence factors are secreted toxins, cofactors for activating host zymogens, and exoenzymes. Secreted toxins such as pore-forming toxins and superantigens are highly inflammatory and can cause leukocyte cell death by cytolysis and clonal deletion, respectively. Coagulases and staphylokinases are cofactors that hijack the host's coagulation system. Exoenzymes, including nucleases and proteases, cleave and inactivate various immune defense and surveillance molecules, such as complement factors, antimicrobial peptides, and surface receptors that are important for leukocyte chemotaxis. Additionally, some of these secreted toxins and exoenzymes can cause disruption of endothelial and epithelial barriers through cell lysis and cleavage of junction proteins. A unique feature when examining the repertoire of S. aureus secreted virulence factors is the apparent functional redundancy exhibited by the majority of the toxins and exoenzymes. However, closer examination of each virulence factor revealed that each has unique properties that have important functional consequences. This chapter provides a brief overview of our current understanding of the major secreted virulence factors critical for S. aureus pathogenesis.
Collapse
Affiliation(s)
- Kayan Tam
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| |
Collapse
|
30
|
Anuj SA, Gajera HP, Hirpara DG, Golakiya BA. Interruption in membrane permeability of drug-resistant Staphylococcus aureus with cationic particles of nano‑silver. Eur J Pharm Sci 2019; 127:208-216. [DOI: 10.1016/j.ejps.2018.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/25/2018] [Accepted: 11/05/2018] [Indexed: 12/28/2022]
|
31
|
Blaustein RA, McFarland AG, Ben Maamar S, Lopez A, Castro-Wallace S, Hartmann EM. Pangenomic Approach To Understanding Microbial Adaptations within a Model Built Environment, the International Space Station, Relative to Human Hosts and Soil. mSystems 2019; 4:e00281-18. [PMID: 30637341 PMCID: PMC6325168 DOI: 10.1128/msystems.00281-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022] Open
Abstract
Understanding underlying mechanisms involved in microbial persistence in the built environment (BE) is essential for strategically mitigating potential health risks. To test the hypothesis that BEs impose selective pressures resulting in characteristic adaptive responses, we performed a pangenomics meta-analysis leveraging 189 genomes (accessed from GenBank) of two epidemiologically important taxa, Bacillus cereus and Staphylococcus aureus, isolated from various origins: the International Space Station (ISS; a model BE), Earth-based BEs, soil, and humans. Our objectives were to (i) identify differences in the pangenomic composition of generalist and host-associated organisms, (ii) characterize genes and functions involved in BE-associated selection, and (iii) identify genomic signatures of ISS-derived strains of potential relevance for astronaut health. The pangenome of B. cereus was more expansive than that of S. aureus, which had a dominant core component. Genomic contents of both taxa significantly correlated with isolate origin, demonstrating an importance for biogeography and potential niche adaptations. ISS/BE-enriched functions were often involved in biosynthesis, catabolism, materials transport, metabolism, and stress response. Multiple origin-enriched functions also overlapped across taxa, suggesting conserved adaptive processes. We further characterized two mobile genetic elements with local neighborhood genes encoding biosynthesis and stress response functions that distinctively associated with B. cereus from the ISS. Although antibiotic resistance genes were present in ISS/BE isolates, they were also common in counterparts elsewhere. Overall, despite differences in microbial lifestyle, some functions appear common to remaining viable in the BE, and those functions are not typically associated with direct impacts on human health. IMPORTANCE The built environment contains a variety of microorganisms, some of which pose critical human health risks (e.g., hospital-acquired infection, antibiotic resistance dissemination). We uncovered a combination of complex biological functions that may play a role in bacterial survival under the presumed selective pressures in a model built environment-the International Space Station-by using an approach to compare pangenomes of bacterial strains from two clinically relevant species (B. cereus and S. aureus) isolated from both built environments and humans. Our findings suggest that the most crucial bacterial functions involved in this potential adaptive response are specific to bacterial lifestyle and do not appear to have direct impacts on human health.
Collapse
Affiliation(s)
- Ryan A. Blaustein
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Alexander G. McFarland
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Sarah Ben Maamar
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Alberto Lopez
- Department of Microbiology-Immunology, Northwestern University, Evanston, Illinois, USA
| | - Sarah Castro-Wallace
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, Texas, USA
| | - Erica M. Hartmann
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
32
|
Loeffler A, Lloyd D. What has changed in canine pyoderma? A narrative review. Vet J 2018; 235:73-82. [DOI: 10.1016/j.tvjl.2018.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 03/23/2018] [Accepted: 04/03/2018] [Indexed: 12/28/2022]
|
33
|
Schmidt VM, Pinchbeck G, Nuttall T, Shaw S, McIntyre KM, McEwan N, Dawson S, Williams NJ. Impact of systemic antimicrobial therapy on mucosal staphylococci in a population of dogs in Northwest England. Vet Dermatol 2018; 29:192-e70. [PMID: 29664197 DOI: 10.1111/vde.12538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Antimicrobial-resistant bacteria are increasingly isolated from veterinary patients. OBJECTIVES To determine risk factors for antimicrobial resistance (AMR) among canine mucosal staphylococci following routine antimicrobial treatment with cefalexin (CFX), clavulanate-amoxicillin (AC), cefovecin (CVN), clindamycin (CD) or a fluoroquinolone (FQ). ANIMALS Mucosal swab samples (n = 463) were collected from 127 dogs pre-treatment, immediately, and at one- and three-months post-treatment. METHODS Staphylococci were identified phenotypically and biochemically as coagulase negative (CoNS) or coagulase positive (CoPS); CoPS were speciated by nuc gene PCR. Antimicrobial susceptibility was determined using disc diffusion and mecA gene carriage by PCR. Multilevel, multivariable models examined associations between risk factors and presence/absence of CoPS, meticillin resistance (MR), multidrug-resistance (MDR) and fluoroquinolone resistance (FQR). RESULTS The percentage of samples with CoNS increased and with CoPS (including S. pseudintermedius) decreased immediately post-treatment with CFX, CVN and CD (P ≤ 0.001) and one month post-treatment with CD (P = 0.003). By three months post-treatment, there was no significant difference compared to pre-treatment samples. Immediately post-treatment with FQs there was significantly increased risk of isolating MRS (P = 0.002), MDR (P = 0.002) or FQR (P = 0.013) staphylococci and of MDR following CFX treatment (P = 0.019). The percentage of samples with AMR staphylococci declined from immediately to three months post-treatment and there was no significant difference between resistance prevalence at one or three months post-treatment for most AMR traits and treatment groups. Exceptions include increased MDR following FQ (P = 0.048) or CFX (P = 0.021), at one and three months post-treatment, respectively. CONCLUSIONS AND CLINICAL IMPORTANCE Systemic antimicrobials impact on mucosal staphylococci. Immediately after therapy, the mucosa may be a reservoir for AMR staphylococci that are a source of mobile genetic elements carrying AMR genes.
Collapse
Affiliation(s)
- Vanessa M Schmidt
- Institute of Veterinary Science, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK.,Department of Epidemiology and Population Health, Institute of Infection and Global Health, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK
| | - Gina Pinchbeck
- Department of Epidemiology and Population Health, Institute of Infection and Global Health, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK
| | - Tim Nuttall
- Institute of Veterinary Science, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK.,The Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Bush Farm Road Easter Bush Campus, Roslin, Midlothian, UK
| | - Steve Shaw
- UK Vet Derm, 16 Talbot Street Whitwick, Coalville, LE67 5AW, Leicestershire, UK
| | - K Marie McIntyre
- Department of Epidemiology and Population Health, Institute of Infection and Global Health, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK.,NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, West Derby Street, Liverpool, L69 7BE, UK
| | - Neil McEwan
- Institute of Veterinary Science, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK
| | - Susan Dawson
- Institute of Veterinary Science, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK
| | - Nicola J Williams
- Department of Epidemiology and Population Health, Institute of Infection and Global Health, The University of Liverpool, Leahurst Campus Chester High Road, Neston, CH64 7TE, UK.,NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, West Derby Street, Liverpool, L69 7BE, UK
| |
Collapse
|
34
|
Stevens M, Piepers S, Supré K, De Vliegher S. Antimicrobial consumption on dairy herds and its association with antimicrobial inhibition zone diameters of non-aureus staphylococci and Staphylococcus aureus isolated from subclinical mastitis. J Dairy Sci 2018; 101:3311-3322. [DOI: 10.3168/jds.2017-13365] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/27/2017] [Indexed: 01/05/2023]
|
35
|
Development of Mycoplasma synoviae (MS) core genome multilocus sequence typing (cgMLST) scheme. Vet Microbiol 2018; 218:84-89. [PMID: 29685226 DOI: 10.1016/j.vetmic.2018.03.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/16/2018] [Accepted: 03/17/2018] [Indexed: 12/17/2022]
Abstract
Mycoplasma synoviae (MS) is a poultry pathogen with reported increased prevalence and virulence in recent years. MS strain identification is essential for prevention, control efforts and epidemiological outbreak investigations. Multiple multilocus based sequence typing schemes have been developed for MS, yet the resolution of these schemes could be limited for outbreak investigation. The cost of whole genome sequencing became close to that of sequencing the seven MLST targets; however, there is no standardized method for typing MS strains based on whole genome sequences. In this paper, we propose a core genome multilocus sequence typing (cgMLST) scheme as a standardized and reproducible method for typing MS based whole genome sequences. A diverse set of 25 MS whole genome sequences were used to identify 302 core genome genes as cgMLST targets (35.5% of MS genome) and 44 whole genome sequences of MS isolates from six countries in four continents were used for typing applying this scheme. cgMLST based phylogenetic trees displayed a high degree of agreement with core genome SNP based analysis and available epidemiological information. cgMLST allowed evaluation of two conventional MLST schemes of MS. The high discriminatory power of cgMLST allowed differentiation between samples of the same conventional MLST type. cgMLST represents a standardized, accurate, highly discriminatory, and reproducible method for differentiation between MS isolates. Like conventional MLST, it provides stable and expandable nomenclature, allowing for comparing and sharing the typing results between different laboratories worldwide.
Collapse
|
36
|
Introduction of a hydrolysis probe PCR assay for high-throughput screening of methicillin-resistant Staphylococcus aureus with the ability to include or exclude detection of Staphylococcus argenteus. PLoS One 2018; 13:e0192782. [PMID: 29425233 PMCID: PMC5806904 DOI: 10.1371/journal.pone.0192782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/30/2018] [Indexed: 01/29/2023] Open
Abstract
Many countries using sensitive screening methods for detection of carriage of methicillin-resistant Staphylococcus aureus (MRSA) have a sustained low incidence of MRSA infections. For diagnostic laboratories with high sample volumes, MRSA screening requires stability, low maintenance and high performance at a low cost. Herein we designed oligonucleotides for a new nuc targeted hydrolysis probe PCR to replace the standard in-house nuc SybrGreen PCR assay. This new, more time-efficient, PCR assay resulted in a 40% increase in daily sample capacity, with maintained high specificity and sensitivity. The assay was also able to detect Staphylococcus aureus clonal cluster 75 (CC75) lineage strains, recently re-classified as Staphylococcus argenteus, with a sensitivity considerably increased compared to our previous assay. While awaiting consensus if the CC75 lineage of S. aureus should be considered as S. argenteus, and whether methicillin-resistant S. argenteus should be included in the MRSA definition, many diagnostic laboratories need to update their MRSA assay sensitivity/specificity towards this lineage/species. The MRSA screening assay presented in this manuscript is comprised of nuc oligonucleotides separately targeting S. aureus and CC75 lineage strains/S. argenteus, thus providing high user flexibility for the detection of CC75 lineage strains/S. argenteus.
Collapse
|
37
|
Sato’o Y, Hisatsune J, Yu L, Sakuma T, Yamamoto T, Sugai M. Tailor-made gene silencing of Staphylococcus aureus clinical isolates by CRISPR interference. PLoS One 2018; 13:e0185987. [PMID: 29377933 PMCID: PMC5788344 DOI: 10.1371/journal.pone.0185987] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 09/23/2017] [Indexed: 11/30/2022] Open
Abstract
Preparing the genetically modified organisms have required much time and labor, making it the rate-limiting step but CRISPR/Cas9 technology appearance has changed this difficulty. Although reports on CRISPR/Cas9 technology such as genome editing and CRISPR interference (CRISPRi) in eukaryotes increased, those in prokaryotes especially in Staphylococci were limited. Thus, its potential in the bacteriology remains unexplored. This is attributed to ecological difference between eukaryotes and prokaryotes. Here, we constructed a novel CRISPRi plasmid vector, pBACi for Staphylococcus aureus. The transformation efficiency of S. aureus was ~104 CFU/μg DNA using a vector extracted from dcm negative, which encoded one of DNA modification genes, E. coli. Further, pBACi was introduced into various clinical isolates including that not accepting the conventional temperature-sensitive vector. dcas9 in the vector was expressed throughout the growth phases of S. aureus and this vector decreased various gene mRNA expressions based on the crRNA targeting sequences and altered the knockdown strains' phenotypes. The targeted genes included various virulence and antibiotic resistant genes. Bioinformatics suggest this vector can be introduced into wide range of low-GC Gram-positive bacteria. Because this new CRISPR/Cas9-based vector can easily prepare knockdown strains, we believe the novel vector will facilitate the characterization of the function of genes from S. aureus and other Gram-positive bacteria.
Collapse
Affiliation(s)
- Yusuke Sato’o
- Department of Bacteriology, Hiroshima University, Graduate school of Biomedical and Health Sciences, Hiroshima, Hiroshima, Japan
| | - Junzo Hisatsune
- Department of Bacteriology, Hiroshima University, Graduate school of Biomedical and Health Sciences, Hiroshima, Hiroshima, Japan
| | - Liansheng Yu
- Department of Bacteriology, Hiroshima University, Graduate school of Biomedical and Health Sciences, Hiroshima, Hiroshima, Japan
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Hiroshima University, Graduate School of Science, Hiroshima, Hiroshima, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Hiroshima University, Graduate School of Science, Hiroshima, Hiroshima, Japan
| | - Motoyuki Sugai
- Department of Bacteriology, Hiroshima University, Graduate school of Biomedical and Health Sciences, Hiroshima, Hiroshima, Japan
| |
Collapse
|
38
|
García-Pérez AN, de Jong A, Junker S, Becher D, Chlebowicz MA, Duipmans JC, Jonkman MF, van Dijl JM. From the wound to the bench: exoproteome interplay between wound-colonizing Staphylococcus aureus strains and co-existing bacteria. Virulence 2018; 9:363-378. [PMID: 29233035 PMCID: PMC5955179 DOI: 10.1080/21505594.2017.1395129] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/09/2017] [Accepted: 10/15/2017] [Indexed: 11/16/2022] Open
Abstract
Wound-colonizing microorganisms can form complex and dynamic polymicrobial communities where pathogens and commensals may co-exist, cooperate or compete with each other. The present study was aimed at identifying possible interactions between different bacteria isolated from the same chronic wound of a patient with the genetic blistering disease epidermolysis bullosa (EB). Specifically, this involved two different isolates of the human pathogen Staphylococcus aureus, and isolates of Bacillus thuringiensis and Klebsiella oxytoca. Particular focus was attributed to interactions of S. aureus with the two other species, because of the high staphylococcal prevalence among chronic wounds. Intriguingly, upon co-cultivation, none of the wound isolates inhibited each other's growth. Since the extracellular proteome of bacterial pathogens is a reservoir of virulence factors, the exoproteomes of the staphylococcal isolates in monoculture and co-culture with B. thuringiensis and K. oxytoca were characterized by Mass Spectrometry to explore the inherent relationships between these co-exisiting bacteria. This revealed a massive reduction in the number of staphylococcal exoproteins upon co-culturing with K. oxytoca or B. thuringiensis. Interestingly, this decrease was particularly evident for extracellular proteins with a predicted cytoplasmic localization, which were recently implicated in staphylococcal virulence and epidemiology. Furthermore, our exoproteome analysis uncovered potential cooperativity between the two different S. aureus isolates. Altogether, the observed exoproteome variations upon co-culturing are indicative of unprecedented adaptive mechanisms that set limits to the production of secreted staphylococcal virulence factors.
Collapse
Affiliation(s)
- Andrea N. García-Pérez
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, the Netherlands
| | - Anne de Jong
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, AG Groningen, the Netherlands
| | - Sabryna Junker
- Institute for Microbiology, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, Greifswald, Germany
| | - Dörte Becher
- Institute for Microbiology, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, Greifswald, Germany
| | - Monika A. Chlebowicz
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, the Netherlands
| | - José C. Duipmans
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, the Netherlands
| | - Marcel F. Jonkman
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, RB Groningen, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, Groningen, the Netherlands
| |
Collapse
|
39
|
Haubert L, Kroning IS, Iglesias MA, da Silva WP. First report of the Staphylococcus aureus isolate from subclinical bovine mastitis in the South of Brazil harboring resistance gene dfrG and transposon family Tn 916-1545. Microb Pathog 2017; 113:242-247. [DOI: 10.1016/j.micpath.2017.10.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 01/03/2023]
|
40
|
Singh VK, Ring RP, Aswani V, Stemper ME, Kislow J, Ye Z, Shukla SK. Phylogenetic distribution and expression of a penicillin-binding protein homologue, Ear and its significance in virulence of Staphylococcus aureus. J Med Microbiol 2017; 66:1811-1821. [PMID: 29099691 DOI: 10.1099/jmm.0.000630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Staphylococcus aureus is an opportunistic human pathogen that can cause serious infections in humans. A plethora of known and putative virulence factors are produced by staphylococci that collectively orchestrate pathogenesis. Ear protein (Escherichia coli ampicillin resistance) in S. aureus is an exoprotein in COL strain, predicted to be a superantigen, and speculated to play roles in antibiotic resistance and virulence. The goal of this study was to determine if expression of ear is modulated by single nucleotide polymorphisms in its promoter and coding sequences and whether this gene plays roles in antibiotic resistance and virulence. METHODOLOGY Promoter, coding sequences and expression of the ear gene in clinical and carriage S. aureus strains with distinct genetic backgrounds were analysed. The JE2 strain and its isogenic ear mutant were used in a systemic infection mouse model to determine the competiveness of the ear mutant.Results/Key findings. The ear gene showed a variable expression, with USA300FPR3757 showing a high-level expression compared to many of the other strains tested including some showing negligible expression. Higher expression was associated with agr type 1 but not correlated with phylogenetic relatedness of the ear gene based upon single nucleotide polymorphisms in the promoter or coding regions suggesting a complex regulation. An isogenic JE2 (USA300 background) ear mutant showed no significant difference in its growth, antibiotic susceptibility or virulence in a mouse model. CONCLUSION Our data suggests that despite being highly expressed in a USA300 genetic background, Ear is not a significant contributor to virulence in that strain.
Collapse
Affiliation(s)
- Vineet K Singh
- A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Robert P Ring
- A.T. Still University of Health Sciences, Kirksville, MO, USA
| | - Vijay Aswani
- The State University of New York, Buffalo, NY, USA
| | | | | | - Zhan Ye
- Marshfield Clinic Research Institute, Marshfield, WI, USA
| | | |
Collapse
|
41
|
Clausen M, Edslev S, Andersen P, Clemmensen K, Krogfelt K, Agner T. Staphylococcus aureus
colonization in atopic eczema and its association with filaggrin gene mutations. Br J Dermatol 2017; 177:1394-1400. [DOI: 10.1111/bjd.15470] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2017] [Indexed: 12/29/2022]
Affiliation(s)
- M.‐L. Clausen
- Department of Dermatology Bispebjerg University Hospital Copenhagen Denmark
| | - S.M. Edslev
- Department of Microbiology and Infection Control Statens Serum Institut Copenhagen Denmark
| | - P.S. Andersen
- Department of Microbiology and Infection Control Statens Serum Institut Copenhagen Denmark
- Institute of Veterinary Disease Biology University of Copenhagen Copenhagen Denmark
| | - K. Clemmensen
- Department of Dermatology Bispebjerg University Hospital Copenhagen Denmark
| | - K.A. Krogfelt
- Department of Microbiology and Infection Control Statens Serum Institut Copenhagen Denmark
| | - T. Agner
- Department of Dermatology Bispebjerg University Hospital Copenhagen Denmark
| |
Collapse
|
42
|
Harkins CP, Pettigrew KA, Oravcová K, Gardner J, Hearn RMR, Rice D, Mather AE, Parkhill J, Brown SJ, Proby CM, Holden MTG. The Microevolution and Epidemiology of Staphylococcus aureus Colonization during Atopic Eczema Disease Flare. J Invest Dermatol 2017; 138:336-343. [PMID: 28951239 PMCID: PMC5780352 DOI: 10.1016/j.jid.2017.09.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/11/2017] [Accepted: 09/12/2017] [Indexed: 01/24/2023]
Abstract
Staphylococcus aureus is an opportunistic pathogen and variable component of the human microbiota. A characteristic of atopic eczema (AE) is colonization by S. aureus, with exacerbations associated with an increased bacterial burden of the organism. Despite this, the origins and genetic diversity of S. aureus colonizing individual patients during AE disease flares is poorly understood. To examine the microevolution of S. aureus colonization, we deep sequenced S. aureus populations from nine children with moderate to severe AE and 18 non-atopic children asymptomatically carrying S. aureus nasally. Colonization by clonal S. aureus populations was observed in both AE patients and control participants, with all but one of the individuals carrying colonies belonging to a single sequence type. Phylogenetic analysis showed that disease flares were associated with the clonal expansion of the S. aureus population, occurring over a period of weeks to months. There was a significant difference in the genetic backgrounds of S. aureus colonizing AE cases versus controls (Fisher exact test, P = 0.03). Examination of intra-host genetic heterogeneity of the colonizing S. aureus populations identified evidence of within-host selection in the AE patients, with AE variants being potentially selectively advantageous for intracellular persistence and treatment resistance.
Collapse
Affiliation(s)
- Catriona P Harkins
- School of Medicine, University of St Andrews, St Andrews, UK; Department of Dermatology, Ninewells Hospital, Dundee, UK; School of Medicine, University of Dundee, Dundee, UK.
| | | | - Katarina Oravcová
- School of Medicine, University of St Andrews, St Andrews, UK; Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - June Gardner
- Department of Dermatology, Ninewells Hospital, Dundee, UK
| | - R M Ross Hearn
- Department of Dermatology, Ninewells Hospital, Dundee, UK
| | - Debbie Rice
- Scottish Children's Research Network, MACH 2 Building, Level 5, Ninewells Hospital, Dundee, UK
| | - Alison E Mather
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Julian Parkhill
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Sara J Brown
- Department of Dermatology, Ninewells Hospital, Dundee, UK; Skin Research Group, Division of Cancer Research, School of Medicine, University of Dundee, UK
| | - Charlotte M Proby
- Department of Dermatology, Ninewells Hospital, Dundee, UK; School of Medicine, University of Dundee, Dundee, UK
| | | |
Collapse
|
43
|
Sergelidis D, Angelidis AS. Methicillin-resistant Staphylococcus aureus: a controversial food-borne pathogen. Lett Appl Microbiol 2017; 64:409-418. [PMID: 28304109 DOI: 10.1111/lam.12735] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/10/2017] [Accepted: 03/11/2017] [Indexed: 12/17/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a major cause of severe healthcare-associated (HA) infections. Although during the last decade the incidence of HA invasive infections has dropped, the incidence of community-associated MRSA (CA-MRSA) infections has risen among the general population. Moreover, CA-MRSA, livestock-associated MRSA (LA-MRSA) and HA-MRSA (HA-MRSA) can be found in foods intended for human consumption. Several studies from different geographical areas have reported the presence of enterotoxin genes in several MRSA food isolates. Molecular typing studies have revealed genetic relatedness of these enterotoxigenic isolates with isolates incriminated in human infections. The contamination sources for foods, especially animal-origin foods, may be livestock as well as humans involved in animal husbandry and food-processing. Under favourable environmental conditions for growth and enterotoxin production, enterotoxigenic S. aureus isolates present in foods can cause staphylococcal food poisoning (SFP), irrespective of the contamination origin. Owing to the typically moderate clinical manifestations of SFP, the S. aureus strains responsible for SFP (cases or outbreaks) are frequently either not identified or not further characterized. Antimicrobial susceptibility testing is rarely performed, because administration of antimicrobial therapy is not required in the vast majority of cases. Staphylococcal food poisoning is the result of consumption of foods with preformed enterotoxins. Hence, similar to methicillin-sensitive enterotoxigenic S. aureus, enterotoxigenic MRSA can also act as food-borne pathogens upon favourable conditions for growth and enterotoxin production. The severity of the intoxication is not related to the antimicrobial resistance profile of the causative S. aureus strain and therefore MRSA food-borne outbreaks are not expected to be more severe. SIGNIFICANCE AND IMPACT OF THE STUDY This review evaluates the potential of methicillin-resistant Staphylococcus aureus (MRSA) as food-borne pathogens based on the current knowledge about the epidemiology of MRSA, their prevalence in livestock, foods of animal origin and humans, and their ability to produce enterotoxins.
Collapse
Affiliation(s)
- D Sergelidis
- Laboratory of Hygiene of Foods of Animal Origin, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - A S Angelidis
- Laboratory of Milk Hygiene and Technology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
44
|
Londono SC, Hartnett HE, Williams LB. Antibacterial Activity of Aluminum in Clay from the Colombian Amazon. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:2401-2408. [PMID: 28121138 DOI: 10.1021/acs.est.6b04670] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The problems of antibiotic overuse compel us to seek alternative antibacterial agents. Some clays have been shown to kill antibiotic-resistant human pathogens and may provide an alternative to known antibiotics. Here we show that Al toxicity plays a central role in the antibacterial action of a kaolin-rich clay from the Colombian Amazon (AMZ). Antibacterial susceptibility testing shows minimum inhibitory concentrations of 80 mg/mL against a model Escherichia coli (ATCC 25922). The clay buffered the media pH to ∼4.6 and Eh values to +360 mV. Chemical analysis of AMZ and bacteria showed that Al, P, and transition metals (Fe, Cu, Mn, and Zn) were exchanged during incubation at 37 °C. Only Al derived from the clay exceeded the minimum inhibitory concentrations for E. coli under acidic conditions. Ion imaging showed elevated Al levels in the bacterial membrane, and high intracellular Fe levels, relative to those of untreated controls. Phosphorus depletion in E. coli after reaction with AMZ, together with evidence of membrane permeabilization, suggests that Al reacts with membrane phospholipids, enhancing intracellular transport of metals. These results highlight the importance of dissolved Al for amplifying the toxicity of transition metals to human pathogens.
Collapse
Affiliation(s)
- S Carolina Londono
- School of Earth & Space Exploration, Arizona State University , Tempe, Arizona 85287-1404, United States
| | - Hilairy E Hartnett
- School of Earth & Space Exploration, Arizona State University , Tempe, Arizona 85287-1404, United States
- School of Molecular Sciences, Arizona State University , Tempe, Arizona 85281, United States
| | - Lynda B Williams
- School of Earth & Space Exploration, Arizona State University , Tempe, Arizona 85287-1404, United States
| |
Collapse
|
45
|
Hayden MK, Lolans K, Haffenreffer K, Avery TR, Kleinman K, Li H, Kaganov RE, Lankiewicz J, Moody J, Septimus E, Weinstein RA, Hickok J, Jernigan J, Perlin JB, Platt R, Huang SS. Chlorhexidine and Mupirocin Susceptibility of Methicillin-Resistant Staphylococcus aureus Isolates in the REDUCE-MRSA Trial. J Clin Microbiol 2016; 54:2735-2742. [PMID: 27558180 PMCID: PMC5078551 DOI: 10.1128/jcm.01444-16] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/18/2016] [Indexed: 12/23/2022] Open
Abstract
Whether targeted or universal decolonization strategies for the control of methicillin-resistant Staphylococcus aureus (MRSA) select for resistance to decolonizing agents is unresolved. The REDUCE-MRSA trial (ClinicalTrials registration no. NCT00980980) provided an opportunity to investigate this question. REDUCE-MRSA was a 3-arm, cluster-randomized trial of either screening and isolation without decolonization, targeted decolonization with chlorhexidine and mupirocin, or universal decolonization without screening to prevent MRSA infection in intensive-care unit (ICU) patients. Isolates from the baseline and intervention periods were collected and tested for susceptibility to chlorhexidine gluconate (CHG) by microtiter dilution; mupirocin susceptibility was tested by Etest. The presence of the qacA or qacB gene was determined by PCR and DNA sequence analysis. A total of 3,173 isolates were analyzed; 2 were nonsusceptible to CHG (MICs, 8 μg/ml), and 5/814 (0.6%) carried qacA or qacB At baseline, 7.1% of MRSA isolates expressed low-level mupirocin resistance, and 7.5% expressed high-level mupirocin resistance. In a mixed-effects generalized logistic regression model, the odds of mupirocin resistance among clinical MRSA isolates or MRSA isolates acquired in an ICU in intervention versus baseline periods did not differ across arms, although estimates were imprecise due to small numbers. Reduced susceptibility to chlorhexidine and carriage of qacA or qacB were rare among MRSA isolates in the REDUCE-MRSA trial. The odds of mupirocin resistance were no different in the intervention versus baseline periods across arms, but the confidence limits were broad, and the results should be interpreted with caution.
Collapse
Affiliation(s)
- Mary K Hayden
- Department of Medicine (Infectious Diseases), Rush University Medical Center, Chicago, Illinois, USA
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Karen Lolans
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Katherine Haffenreffer
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Taliser R Avery
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Ken Kleinman
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Haiying Li
- Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Rebecca E Kaganov
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Julie Lankiewicz
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Julia Moody
- Hospital Corporation of America, Nashville, Tennessee, USA
| | - Edward Septimus
- Division of Internal Medicine, Texas A&M Health Science Center College of Medicine, Houston, Texas, USA
| | - Robert A Weinstein
- Department of Medicine (Infectious Diseases), Rush University Medical Center, Chicago, Illinois, USA
- Cook County Health and Hospitals System, Chicago, Illinois, USA
| | - Jason Hickok
- Hospital Corporation of America, Nashville, Tennessee, USA
| | - John Jernigan
- Office of HAI Prevention Research and Evaluation, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Richard Platt
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
| | - Susan S Huang
- Division of Infectious Diseases, University of California-Irvine School of Medicine, Orange, California, USA
| |
Collapse
|
46
|
Planet PJ, Narechania A, Chen L, Mathema B, Boundy S, Archer G, Kreiswirth B. Architecture of a Species: Phylogenomics of Staphylococcus aureus. Trends Microbiol 2016; 25:153-166. [PMID: 27751626 DOI: 10.1016/j.tim.2016.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/07/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022]
Abstract
A deluge of whole-genome sequencing has begun to give insights into the patterns and processes of microbial evolution, but genome sequences have accrued in a haphazard manner, with biased sampling of natural variation that is driven largely by medical and epidemiological priorities. For instance, there is a strong bias for sequencing epidemic lineages of methicillin-resistant Staphylococcus aureus (MRSA) over sensitive isolates (methicillin-sensitive S. aureus: MSSA). As more diverse genomes are sequenced the emerging picture is of a highly subdivided species with a handful of relatively clonal groups (complexes) that, at any given moment, dominate in particular geographical regions. The establishment of hegemony of particular clones appears to be a dynamic process of successive waves of replacement of the previously dominant clone. Here we review the phylogenomic structure of a diverse range of S. aureus, including both MRSA and MSSA. We consider the utility of the concept of the 'core' genome and the impact of recombination and horizontal transfer. We argue that whole-genome surveillance of S. aureus populations could lead to better forecasting of antibiotic resistance and virulence of emerging clones, and a better understanding of the elusive biological factors that determine repeated strain replacement.
Collapse
Affiliation(s)
- Paul J Planet
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA; Department of Pediatrics, Division of Pediatric Infectious Diseases, Children's Hospital of Philadelphia & University of Pennsylvania, Philadelphia, PA, USA.
| | - Apurva Narechania
- Sackler Institute for Comparative Genomics, American Museum of Natural History, New York, NY, USA
| | - Liang Chen
- Public Health Research Institute Center, New Jersey Medical School, Rutgers, Newark, NJ, USA
| | - Barun Mathema
- Public Health Research Institute Center, New Jersey Medical School, Rutgers, Newark, NJ, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Sam Boundy
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Gordon Archer
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Barry Kreiswirth
- Public Health Research Institute Center, New Jersey Medical School, Rutgers, Newark, NJ, USA
| |
Collapse
|
47
|
Molecular Epidemiology of Staphylococcus aureus in the General Population in Northeast Germany: Results of the Study of Health in Pomerania (SHIP-TREND-0). J Clin Microbiol 2016; 54:2774-2785. [PMID: 27605711 PMCID: PMC5078557 DOI: 10.1128/jcm.00312-16] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 08/24/2016] [Indexed: 01/05/2023] Open
Abstract
Population-based studies on Staphylococcus aureus nasal colonization are scarce. We examined the prevalence, resistance, and molecular diversity of S. aureus in the general population in Northeast Germany. Nasal swabs were obtained from 3,891 adults in the large-scale population-based Study of Health in Pomerania (SHIP-TREND). Isolates were characterized using spa genotyping, as well as antibiotic resistance and virulence gene profiling. We observed an S. aureus prevalence of 27.2%. Nasal S. aureus carriage was associated with male sex and inversely correlated with age. Methicillin-resistant S. aureus (MRSA) accounted for 0.95% of the colonizing S. aureus strains. MRSA carriage was associated with frequent visits to hospitals, nursing homes, or retirement homes within the previous 24 months. All MRSA strains were resistant to multiple antibiotics. Most MRSA isolates belonged to the pandemic European hospital-acquired MRSA sequence type 22 (HA-MRSA-ST22) lineage. We also detected one livestock-associated MRSA ST398 (LA-MRSA-ST398) isolate, as well as six livestock-associated methicillin-susceptible S. aureus (LA-MSSA) isolates (clonal complex 1 [CC1], CC97, and CC398). spa typing revealed a diverse but also highly clonal S. aureus population structure. We identified a total of 357 spa types, which were grouped into 30 CCs or sequence types. The major seven CCs (CC30, CC45, CC15, CC8, CC7, CC22, and CC25) included 75% of all isolates. Virulence gene patterns were strongly linked to the clonal background. In conclusion, MSSA and MRSA prevalences and the molecular diversity of S. aureus in Northeast Germany are consistent with those of other European countries. The detection of HA-MRSA and LA-MRSA within the general population indicates possible transmission from hospitals and livestock, respectively, and should be closely monitored.
Collapse
|
48
|
The Plasmidome of Firmicutes: Impact on the Emergence and the Spread of Resistance to Antimicrobials. Microbiol Spectr 2016; 3:PLAS-0039-2014. [PMID: 26104702 DOI: 10.1128/microbiolspec.plas-0039-2014] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The phylum Firmicutes is one of the most abundant groups of prokaryotes in the microbiota of humans and animals and includes genera of outstanding relevance in biomedicine, health care, and industry. Antimicrobial drug resistance is now considered a global health security challenge of the 21st century, and this heterogeneous group of microorganisms represents a significant part of this public health issue.The presence of the same resistant genes in unrelated bacterial genera indicates a complex history of genetic interactions. Plasmids have largely contributed to the spread of resistance genes among Staphylococcus, Enterococcus, and Streptococcus species, also influencing the selection and ecological variation of specific populations. However, this information is fragmented and often omits species outside these genera. To date, the antimicrobial resistance problem has been analyzed under a "single centric" perspective ("gene tracking" or "vehicle centric" in "single host-single pathogen" systems) that has greatly delayed the understanding of gene and plasmid dynamics and their role in the evolution of bacterial communities.This work analyzes the dynamics of antimicrobial resistance genes using gene exchange networks; the role of plasmids in the emergence, dissemination, and maintenance of genes encoding resistance to antimicrobials (antibiotics, heavy metals, and biocides); and their influence on the genomic diversity of the main Gram-positive opportunistic pathogens under the light of evolutionary ecology. A revision of the approaches to categorize plasmids in this group of microorganisms is given using the 1,326 fully sequenced plasmids of Gram-positive bacteria available in the GenBank database at the time the article was written.
Collapse
|
49
|
Comparative genomics of toxigenic and non-toxigenic Staphylococcus hyicus. Vet Microbiol 2016; 185:34-40. [DOI: 10.1016/j.vetmic.2016.01.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 01/21/2016] [Accepted: 01/23/2016] [Indexed: 11/18/2022]
|
50
|
Goldberg GW, Marraffini LA. Resistance and tolerance to foreign elements by prokaryotic immune systems - curating the genome. Nat Rev Immunol 2016; 15:717-24. [PMID: 26494050 DOI: 10.1038/nri3910] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
To engage in adaptive symbioses or genetic exchange, organisms must interact with foreign, non-self elements despite the risks of predation and parasitism. By surveying the interface between self and non-self, immune systems can help ensure the benevolence of these interactions without isolating their hosts altogether. In this Essay, we examine prokaryotic restriction-modification and CRISPR-Cas (clustered, regularly interspaced palindromic repeat-CRISPR-associated proteins) activities and discuss their analogy to mammalian immune pathways. We further explain how their capacities for resistance and tolerance are optimized to reduce parasitism and immunopathology during encounters with non-self.
Collapse
Affiliation(s)
- Gregory W Goldberg
- Laboratory of Bacteriology, The Rockefeller University, 1230 York Avenue, New York City, New York 10065, USA
| | - Luciano A Marraffini
- Laboratory of Bacteriology, The Rockefeller University, 1230 York Avenue, New York City, New York 10065, USA
| |
Collapse
|