1
|
Martins Y, Cerqueira e Costa MO, Palumbo MC, F. Do Porto D, Custódio FL, Trevizani R, Nicolás MF. PAPreC: A Pipeline for Antigenicity Prediction Comparison Methods across Bacteria. ACS OMEGA 2025; 10:5415-5429. [PMID: 39989760 PMCID: PMC11840615 DOI: 10.1021/acsomega.4c07147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Antigenicity prediction plays a crucial role in vaccine development, antibody-based therapies, and diagnostic assays, as this predictive approach helps assess the potential of molecular structures to induce and recruit immune cells and drive antibody production. Several existing prediction methods, which target complete proteins and epitopes identified through reverse vaccinology, face limitations regarding input data constraints, feature extraction strategies, and insufficient flexibility for model evaluation and interpretation. This work presents PAPreC (Pipeline for Antigenicity Prediction Comparison), an open-source, versatile workflow (available at https://github.com/YasCoMa/paprec_nx_workflow) designed to address these challenges. PAPreC systematically examines three key factors: the selection of training data sets, feature extraction methods (including physicochemical descriptors and ESM-2 encoder-derived embeddings), and diverse classifiers. It provides automated model evaluation, interpretability through SHapley Additive exPlanations (SHAP) analysis, and applicability domain assessments, enabling researchers to identify optimal configurations for their specific data sets. Applying PAPreC to IEDB data as a reference, we demonstrate its effectiveness across the ESKAPE pathogen group. A case study involving Pseudomonas aeruginosa and Staphylococcus aureus shows that specific feature configurations are more suitable for different sequence types, and that ESM-2 embeddings enhance model performance. Moreover, our results indicate that separate models for Gram-positive and Gram-negative bacteria are not required. PAPreC offers a comprehensive, adaptable, and robust framework to streamline and improve antigenicity prediction for diverse bacterial data sets.
Collapse
Affiliation(s)
- Yasmmin
C. Martins
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Maiana O. Cerqueira e Costa
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| | - Miranda C. Palumbo
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Dario F. Do Porto
- Department
of Biological Chemistry, Faculty of Exact and Natural Sciences, University of Buenos Aires - UBA, Av. Int. Cantilo, C1428 Buenos Aires, Argentina
| | - Fábio L. Custódio
- Department
of Computational Mechanics, National Laboratory
for Scientific Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| | - Raphael Trevizani
- Biotechnology, Oswaldo Cruz Foundation
- Fiocruz, Street São
José S/N, 61760-000 Eusébio, Brazil
| | - Marisa Fabiana Nicolás
- Bioinformatics
Laboratory, National Laboratory for Scientific
Computing, Av. Getúlio Vargas 333, 25651-075 Petrópolis, Brazil
| |
Collapse
|
2
|
Coll F, Blane B, Bellis KL, Matuszewska M, Wonfor T, Jamrozy D, Toleman MS, Geoghegan JA, Parkhill J, Massey RC, Peacock SJ, Harrison EM. The mutational landscape of Staphylococcus aureus during colonisation. Nat Commun 2025; 16:302. [PMID: 39805814 PMCID: PMC11730646 DOI: 10.1038/s41467-024-55186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Staphylococcus aureus is an important human pathogen and a commensal of the human nose and skin. Survival and persistence during colonisation are likely major drivers of S. aureus evolution. Here we applied a genome-wide mutation enrichment approach to a genomic dataset of 3060 S. aureus colonization isolates from 791 individuals. Despite limited within-host genetic diversity, we observed an excess of protein-altering mutations in metabolic genes, in regulators of quorum-sensing (agrA and agrC) and in known antibiotic targets (fusA, pbp2, dfrA and ileS). We demonstrated the phenotypic effect of multiple adaptive mutations in vitro, including changes in haemolytic activity, antibiotic susceptibility, and metabolite utilisation. Nitrogen metabolism showed the strongest evidence of adaptation, with the assimilatory nitrite reductase (nasD) and urease (ureG) showing the highest mutational enrichment. We identified a nasD natural mutant with enhanced growth under urea as the sole nitrogen source. Inclusion of 4090 additional isolate genomes from 731 individuals revealed eight more genes including sasA/sraP, darA/pstA, and rsbU with signals of adaptive variation that warrant further characterisation. Our study provides a comprehensive picture of the heterogeneity of S. aureus adaptive changes during colonisation, and a robust methodological approach applicable to study in host adaptive evolution in other bacterial pathogens.
Collapse
Affiliation(s)
- Francesc Coll
- Applied Microbial Genomics Unit, Department of Molecular Basis of Disease, Institute of Biomedicine of Valencia (IBV-CSIC), Valencia, Spain.
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
- Parasites & Microbes Programme, Wellcome Sanger Institute, Hinxton, UK.
| | - Beth Blane
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Katherine L Bellis
- Parasites & Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marta Matuszewska
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Toska Wonfor
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
- Department of Microbes, Infection & Microbiomes, College of Medicine & Health, University of Birmingham, Birmingham, UK
| | - Dorota Jamrozy
- Parasites & Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | | | - Joan A Geoghegan
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
- Department of Microbes, Infection & Microbiomes, College of Medicine & Health, University of Birmingham, Birmingham, UK
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Julian Parkhill
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ruth C Massey
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Ewan M Harrison
- Parasites & Microbes Programme, Wellcome Sanger Institute, Hinxton, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Ke S, Kil H, Roggy C, Shields T, Quinn Z, Quinn AP, Small JM, Towne FD, Brooks AE, Brooks BD. Potential Therapeutic Targets for Combination Antibody Therapy Against Staphylococcus aureus Infections. Antibiotics (Basel) 2024; 13:1046. [PMID: 39596740 PMCID: PMC11591076 DOI: 10.3390/antibiotics13111046] [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: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Despite the significant advances in antibiotic treatments and therapeutics, Staphylococcus aureus (S. aureus) remains a formidable pathogen, primarily due to its rapid acquisition of antibiotic resistance. Known for its array of virulence factors, including surface proteins that promote adhesion to host tissues, enzymes that break down host barriers, and toxins that contribute to immune evasion and tissue destruction, S. aureus poses a serious health threat. Both the Centers for Disease Control and Prevention (CDC) and the World Health Organization (WHO) classify S. aureus as an ESKAPE pathogen, recognizing it as a critical threat to global health. The increasing prevalence of drug-resistant S. aureus underscores the need for new therapeutic strategies. This review discusses a promising approach that combines monoclonal antibodies targeting multiple S. aureus epitopes, offering synergistic efficacy in treating infections. Such strategies aim to reduce the capacity of the pathogen to develop resistance, presenting a potent adjunct or alternative to conventional antibiotic treatments.
Collapse
Affiliation(s)
- Sharon Ke
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Hyein Kil
- Department of Surgery, Virtua Health, Camden, NJ 08103, USA
| | - Conner Roggy
- Department of Orthopaedic Surgery, Community Memorial Healthcare, Ventura, CA 93003, USA
| | - Ty Shields
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Zachary Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Alyssa P. Quinn
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - James M. Small
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Francina D. Towne
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Amanda E. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| | - Benjamin D. Brooks
- Department of Biomedical Sciences, College of Osteopathic Medicine, Rocky Vista University, Parker, CO 80134, USA
| |
Collapse
|
4
|
Vozza EG, Kelly AM, Daly CM, O'Rourke SA, Carlile SR, Morris B, Dunne A, McLoughlin RM. Type 1 interferons promote Staphylococcus aureus nasal colonization by inducing phagocyte apoptosis. Cell Death Discov 2024; 10:403. [PMID: 39271670 PMCID: PMC11399434 DOI: 10.1038/s41420-024-02173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/23/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Staphylococcus aureus is an important human commensal which persistently colonizes up to 30% of the human population, predominantly within the nasal cavity. The commensal lifestyle of S. aureus is complex, and the mechanisms underpinning colonization are not fully understood. S. aureus can induce an immunosuppressive environment in the nasal tissue (NT) by driving IL-10 and IL-27 to facilitate nasal colonization, indicating that S. aureus has the capacity to modulate the local immune environment for its commensal habitation. Mounting evidence suggests commensal bacteria drive type 1 interferons (IFN-I) to establish an immunosuppressive environment and whilst S. aureus can induce IFN-I during infection, its role in colonization has not yet been examined. Here, we show that S. aureus preferentially induces IFN signaling in macrophages. This IFN-I in turn upregulates expression of proapoptotic genes within macrophages culminating in caspase-3 cleavage. Importantly, S. aureus was found to drive phagocytic cell apoptosis in the nasal tissue during nasal colonization in an IFN-I dependent manner with colonization significantly reduced under caspase-3 inhibition. Overall, loss of IFN-I signaling significantly diminished S. aureus nasal colonization implicating a pivotal role for IFN-I in controlling S. aureus persistence during colonization through its ability to induce phagocyte apoptosis. Together, this study reveals a novel strategy utilized by S. aureus to circumvent host immunity in the nasal mucosa to facilitate nasal colonization.
Collapse
Affiliation(s)
- Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alanna M Kelly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Clíodhna M Daly
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Sinead A O'Rourke
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Simon R Carlile
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Brenda Morris
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling Dunne
- Molecular Immunology Group, School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
5
|
Bhattacharya M, Horswill AR. The role of human extracellular matrix proteins in defining Staphylococcus aureus biofilm infections. FEMS Microbiol Rev 2024; 48:fuae002. [PMID: 38337187 PMCID: PMC10873506 DOI: 10.1093/femsre/fuae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 01/26/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024] Open
Abstract
Twenty to forty one percent of the world's population is either transiently or permanently colonized by the Gram-positive bacterium, Staphylococcus aureus. In 2017, the CDC designated methicillin-resistant S. aureus (MRSA) as a serious threat, reporting ∼300 000 cases of MRSA-associated hospitalizations annually, resulting in over 19 000 deaths, surpassing that of HIV in the USA. S. aureus is a proficient biofilm-forming organism that rapidly acquires resistance to antibiotics, most commonly methicillin (MRSA). This review focuses on a large group of (>30) S. aureus adhesins, either surface-associated or secreted that are designed to specifically bind to 15 or more of the proteins that form key components of the human extracellular matrix (hECM). Importantly, this includes hECM proteins that are pivotal to the homeostasis of almost every tissue environment [collagen (skin), proteoglycans (lung), hemoglobin (blood), elastin, laminin, fibrinogen, fibronectin, and fibrin (multiple organs)]. These adhesins offer S. aureus the potential to establish an infection in every sterile tissue niche. These infections often endure repeated immune onslaught, developing into chronic, biofilm-associated conditions that are tolerant to ∼1000 times the clinically prescribed dose of antibiotics. Depending on the infection and the immune response, this allows S. aureus to seamlessly transition from colonizer to pathogen by subtly manipulating the host against itself while providing the time and stealth that it requires to establish and persist as a biofilm. This is a comprehensive discussion of the interaction between S. aureus biofilms and the hECM. We provide particular focus on the role of these interactions in pathogenesis and, consequently, the clinical implications for the prevention and treatment of S. aureus biofilm infections.
Collapse
Affiliation(s)
- Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, United States
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, CO 80045, United States
| |
Collapse
|
6
|
Narongpun P, Chanchaithong P, Yamagishi J, Thapa J, Nakajima C, Suzuki Y. Whole-Genome Investigation of Zoonotic Transmission of Livestock-Associated Methicillin-Resistant Staphylococcus aureus Clonal Complex 398 Isolated from Pigs and Humans in Thailand. Antibiotics (Basel) 2023; 12:1745. [PMID: 38136779 PMCID: PMC10741195 DOI: 10.3390/antibiotics12121745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) has been widespread globally in pigs and humans for decades. Nasal colonization of LA-MRSA is regarded as an occupational hazard to people who are regularly involved in livestock production. Our previous study suggested pig-to-human transmission caused by LA-MRSA clonal complex (CC) 398, using traditional molecular typing methods. Instead, this study aimed to investigate the zoonotic transmission of LA-MRSA CC398 using whole genome sequencing (WGS) technologies. A total of 63 LA-MRSA isolates were identified and characterized in Thailand. Further, the 16 representatives of LA-MRSA CC9 and CC398, including porcine and worker isolates, were subjected to WGS on the Illumina Miseq platform. Core-genome single nucleotide polymorphism (SNP)-based analyses verify the zoonotic transmission caused by LA-MRSA CC398 in two farms. WGS-based characterization suggests the emergence of a novel staphylococcal cassette chromosome (SCC) mec type, consisting of multiple cassette chromosome recombinase (ccr) gene complexes via genetic recombination. Additionally, the WGS analyses revealed putative multi-resistant plasmids and several cross-resistance genes, conferring resistance against drugs of last resort used in humans such as quinupristin/dalfopristin and linezolid. Significantly, LA-MRSA isolates, in this study, harbored multiple virulence genes that may become a serious threat to an immunosuppressive population, particularly for persons who are in close contact with LA-MRSA carriers.
Collapse
Affiliation(s)
- Pawarut Narongpun
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (P.N.); (J.T.)
| | - Pattrarat Chanchaithong
- Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Junya Yamagishi
- Division of Collaboration and Education, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan;
| | - Jeewan Thapa
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (P.N.); (J.T.)
| | - Chie Nakajima
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (P.N.); (J.T.)
- International Collaboration Unit, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
| | - Yasuhiko Suzuki
- Division of Bioresources, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan; (P.N.); (J.T.)
- International Collaboration Unit, Hokkaido University International Institute for Zoonosis Control, Sapporo 001-0020, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo 001-0020, Japan
| |
Collapse
|
7
|
Francis D, Veeramanickathadathil Hari G, Koonthanmala Subash A, Bhairaddy A, Joy A. The biofilm proteome of Staphylococcus aureus and its implications for therapeutic interventions to biofilm-associated infections. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 138:327-400. [PMID: 38220430 DOI: 10.1016/bs.apcsb.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
Staphylococcus aureus is a major healthcare concern due to its ability to inflict life-threatening infections and evolve antibiotic resistance at an alarming pace. It is frequently associated with hospital-acquired infections, especially device-associated infections. Systemic infections due to S. aureus are difficult to treat and are associated with significant mortality and morbidity. The situation is worsened by the ability of S. aureus to form social associations called biofilms. Biofilms embed a community of cells with the ability to communicate with each other and share resources within a polysaccharide or protein matrix. S. aureus establish biofilms on tissues and conditioned abiotic surfaces. Biofilms are hyper-tolerant to antibiotics and help evade host immune responses. Biofilms exacerbate the severity and recalcitrance of device-associated infections. The development of a biofilm involves various biomolecules, such as polysaccharides, proteins and nucleic acids, contributing to different structural and functional roles. Interconnected signaling pathways and regulatory molecules modulate the expression of these molecules. A comprehensive understanding of the molecular biology of biofilm development would help to devise effective anti-biofilm therapeutics. Although bactericidal agents, antimicrobial peptides, bacteriophages and nano-conjugated anti-biofilm agents have been employed with varying levels of success, there is still a requirement for effective and clinically viable anti-biofilm therapeutics. Proteins that are expressed and utilized during biofilm formation, constituting the biofilm proteome, are a particularly attractive target for anti-biofilm strategies. The proteome can be explored to identify potential anti-biofilm drug targets and utilized for rational drug discovery. With the aim of uncovering the biofilm proteome, this chapter explores the mechanism of biofilm formation and its regulation. Furthermore, it explores the antibiofilm therapeutics targeted against the biofilm proteome.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India.
| | | | | | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College (Autonomous), Bengaluru, India
| |
Collapse
|
8
|
Schwermann N, Winstel V. Functional diversity of staphylococcal surface proteins at the host-microbe interface. Front Microbiol 2023; 14:1196957. [PMID: 37275142 PMCID: PMC10232760 DOI: 10.3389/fmicb.2023.1196957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/21/2023] [Indexed: 06/07/2023] Open
Abstract
Surface proteins of Gram-positive pathogens are key determinants of virulence that substantially shape host-microbe interactions. Specifically, these proteins mediate host invasion and pathogen transmission, drive the acquisition of heme-iron from hemoproteins, and subvert innate and adaptive immune cell responses to push bacterial survival and pathogenesis in a hostile environment. Herein, we briefly review and highlight the multi-facetted roles of cell wall-anchored proteins of multidrug-resistant Staphylococcus aureus, a common etiological agent of purulent skin and soft tissue infections as well as severe systemic diseases in humans. In particular, we focus on the functional diversity of staphylococcal surface proteins and discuss their impact on the variety of clinical manifestations of S. aureus infections. We also describe mechanistic and underlying principles of staphylococcal surface protein-mediated immune evasion and coupled strategies S. aureus utilizes to paralyze patrolling neutrophils, macrophages, and other immune cells. Ultimately, we provide a systematic overview of novel therapeutic concepts and anti-infective strategies that aim at neutralizing S. aureus surface proteins or sortases, the molecular catalysts of protein anchoring in Gram-positive bacteria.
Collapse
Affiliation(s)
- Nicoletta Schwermann
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Volker Winstel
- Research Group Pathogenesis of Bacterial Infections, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
9
|
Elahimanesh M, Najafi M. Cross talk between bacterial and human gene networks enriched using ncRNAs in IBD disease. Sci Rep 2023; 13:7704. [PMID: 37169818 PMCID: PMC10175251 DOI: 10.1038/s41598-023-34780-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a long-term inflammatory immune-mediated gut illness with several extra-intestinal complications. The aims of this study were to identify a novel network-based meta-analysis approach on the basis of the combinations of the differentially expressed genes (DEGs) from microarray data, to enrich the functional modules from human protein-protein interaction (PPI) and gene ontology (GO) data, and to profile the ncRNAs on the genes involved in IBD. The gene expression profiles of GSE126124, GSE87473, GSE75214, and GSE95095 are obtained from the Gene Expression Omnibus (GEO) database based on the study criteria between 2017 and 2022. The DEGs were screened by the R software. DEGs were then used to examine gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. The ncRNAs including the miRNAs and ceRNAs were predicted on the PPIs visualized using Cytoscape. Enrichment analysis of genes with differential expression (n = 342) using KEGG and GO showed that the signaling pathways related with staphylococcus aureus and pertussis bacterial infections may stimulate the immune system and exacerbate IBD via the interaction with human proteins including Fibrinogen gamma chain (FGG), Keratin 10 (KRT10), and Toll like receptor 4 (TLR4). By building a ceRNA network, lncRNA XIST and NEAT1 were determined by affecting common miRNAs, hsa-miR-6875-5p, hsa-miR-1908-5p, hsa-miR-186-5p, hsa-miR-6763-5p, hsa-miR-4436a, and hsa-miR-520a-5p. Additionally, the chromosome regions including NM_001039703 and NM_006267, which produce the most potent circRNAs play a significant role in the ceRNA network of IBD. Also, we predicted the siRNAs that would be most effective against the bacterial genes in staphylococcus aureus and pertussis infections. These findings suggested that three genes (FGG, KRT10, and TLR4), six miRNAs (hsa-miR-6875-5p, hsa-miR-1908-5p, hsa-miR-186-5p, hsa-miR-4436a, hsa-miR-520a-5p, and hsa-miR-6763-5p), two lncRNAs (XIST and NEAT1), and chromosomal regions including NM_001039703 and NM_006267 with the production of the most effective circRNAs are involved in the ncRNA-associated ceRNA network of IBD. These ncRNA profiles are related to the described gene functions and may play therapeutic targets in controlling inflammatory bowel disease.
Collapse
Affiliation(s)
- Mohammad Elahimanesh
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Clinical Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
- Microbial Biotechnology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Berry KA, Verhoef MTA, Leonard AC, Cox G. Staphylococcus aureus adhesion to the host. Ann N Y Acad Sci 2022; 1515:75-96. [PMID: 35705378 DOI: 10.1111/nyas.14807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus is a pathobiont capable of colonizing and infecting most tissues within the human body, resulting in a multitude of different clinical outcomes. Adhesion of S. aureus to the host is crucial for both host colonization and the establishment of infections. Underlying the pathogen's success is a complex and diverse arsenal of adhesins. In this review, we discuss the different classes of adhesins, including a consideration of the various adhesion sites throughout the body and the clinical outcomes of each infection type. The development of therapeutics targeting the S. aureus host-pathogen interaction is a relatively understudied area. Due to the increasing global threat of antimicrobial resistance, it is crucial that innovative and alternative approaches are considered. Neutralizing virulence factors, through the development of antivirulence agents, could reduce bacterial pathogenicity and the ever-increasing burden of S. aureus infections. This review provides insight into potentially efficacious adhesion-associated targets for the development of novel decolonizing and antivirulence strategies.
Collapse
Affiliation(s)
- Kirsten A Berry
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mackenzie T A Verhoef
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Allison C Leonard
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Georgina Cox
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
11
|
Dollery SJ, Harro JM, Wiggins TJ, Wille BP, Kim PC, Tobin JK, Bushnell RV, Tasker NJPER, MacLeod DA, Tobin GJ. Select Whole-Cell Biofilm-Based Immunogens Protect against a Virulent Staphylococcus Isolate in a Stringent Implant Model of Infection. Vaccines (Basel) 2022; 10:833. [PMID: 35746441 PMCID: PMC9231243 DOI: 10.3390/vaccines10060833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Many microbes of concern to human health remain without vaccines. We have developed a whole-microbe inactivation technology that enables us to rapidly inactivate large quantities of a pathogen while retaining epitopes that were destroyed by previous inactivation methods. The method that we call UVC-MDP inactivation can be used to make whole-cell vaccines with increased potency. We and others are exploring the possibility of using improved irradiation-inactivation technologies to develop whole-cell vaccines for numerous antibiotic-resistant microbes. Here, we apply UVC-MDP to produce candidate MRSA vaccines which we test in a stringent tibia implant model of infection challenged with a virulent MSRA strain. We report high levels of clearance in the model and observe a pattern of protection that correlates with the immunogen protein profile used for vaccination.
Collapse
Affiliation(s)
- Stephen J. Dollery
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Janette M. Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Taralyn J. Wiggins
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Brendan P. Wille
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Peter C. Kim
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - John K. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Ruth V. Bushnell
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Naomi J. P. E. R. Tasker
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - David A. MacLeod
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Gregory J. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| |
Collapse
|
12
|
Alfaiz FA. Molecular studies of immunological enzyme clumping factor B for the inhibition of Staphylococcus aureus with essential oils of Nigella sativa. J Mol Recognit 2021; 34:e2941. [PMID: 34626016 DOI: 10.1002/jmr.2941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/14/2021] [Accepted: 09/26/2021] [Indexed: 11/09/2022]
Abstract
Essential oils from black cumin seeds (Nigella sativa) have largely been used in the manufacturing of nutraceuticals and functional food products due to the presence of a wide variety of bioactive compounds. However, their applications in the pharmaceutical sector have recently attracted interest and started blooming. The present research elucidates the in silico and in vitro efficacies of active leads from essential oil of N sativa against the human pathogenic bacterium Staphylococcus aureus. Biofilm development has become an inevitable situation in the health care sector. Lowering the efficacies of antimicrobial drugs is one of the vital ramifications that resulted in the emergence of multidrug resistance. Clumping factor B (clfB) of S aureus plays a key role in the human immune functions during pathogenesis. Through STRING analysis, the interacting protein partners of clfB were found to regulate biofilm pathway. Therefore, eight ligands from essential oil are docked with the critical clfB protein, which revealed p-cymene, thymoquinone and carvacrol as the robust ligands with highest binding affinity. Therefore, antibiofilm potential of N sativa essential oil at in vitro states was evaluated against S aureus. Further, real time PCR analysis showed that the expression of clfB and intercellular adhesion gene (icaA and icaD) was significantly altered upon treatment with essential oil. Altogether, the findings confirmed the antibiofilm efficacy of N sativa essential oil against S aureus. Hence, the essential oil from N. sativa was envisaged to be promising candidate to treat S aureus biofilm mediated infection.
Collapse
Affiliation(s)
- Faiz Abdulaziz Alfaiz
- Department of Biology, College of Science in Zulfi, Majmaah University, Majmaah, Saudi Arabia
| |
Collapse
|
13
|
Millar EV, Bennett JW, Barin B, Carey PM, Law NN, English CE, Schwartz MM, Cochrane T, Ellis MW, Tribble DR, Timothy Cooke M, Hennessey JP. Safety, immunogenicity, and efficacy of NDV-3A against Staphylococcus aureus colonization: A phase 2 vaccine trial among US Army Infantry trainees. Vaccine 2021; 39:3179-3188. [PMID: 33962841 PMCID: PMC10430023 DOI: 10.1016/j.vaccine.2021.04.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Military trainees are at increased risk for Staphylococcus aureus colonization and infection. Disease prevention strategies are needed, but a S. aureus vaccine does not currently exist. METHODS We enrolled US Army Infantry trainees (Fort Benning, GA) in a phase 2, randomized, double-blind, placebo-controlled trial of NDV-3A, a vaccine containing a recombinant adhesin/invasion protein of Candida albicans that has structural similarity to the S. aureus protein clumping factor A. Study participants received one intramuscular dose of NDV-3A or placebo (adjuvant alone) within 72 h of arrival on base. Longitudinal nasal and oral (throat) swabs were collected throughout the 14-week Infantry training cycle. Safety, immunogenicity, and efficacy of NDV-3A against S. aureus nasal / oral acquisition were the endpoints. RESULTS The NDV-3A candidate had minimal reactogenicity and elicited robust antigen-specific B- and T-cell responses. During the 56-day post-vaccination period, there was no difference in the incidence of S. aureus nasal acquisition between those who were randomized to receive NDV-3A vs. placebo (25.6% vs. 29.1%; vaccine efficacy [VE]: 12.1%; p = 0.31). In time-to-event analysis, there was no difference between study groups with respect to the S. aureus colonization-free interval (VE: 13%; p = 0.29). Similarly, the efficacy of NDV-3A against S. aureus oral acquisition was poor (VE: 2.4%; p = 0.52). CONCLUSIONS A single dose of NDV-3A did not prevent nasal nor oral acquisition of S. aureus in a population of military trainees at high risk for colonization.
Collapse
Affiliation(s)
- Eugene V Millar
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States.
| | - Jason W Bennett
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Burc Barin
- The Emmes Company, Rockville, MD, United States
| | - Patrick M Carey
- Benning Martin Army Community Hospital, Fort Benning, GA, United States
| | - Natasha N Law
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States; Benning Martin Army Community Hospital, Fort Benning, GA, United States
| | - Caroline E English
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | | | | | - Michael W Ellis
- University of Toledo Medical Center, Toledo, OH, United States
| | - David R Tribble
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | | | |
Collapse
|
14
|
Monoclonal Antibodies Targeting Surface-Exposed and Secreted Proteins from Staphylococci. Vaccines (Basel) 2021; 9:vaccines9050459. [PMID: 34064471 PMCID: PMC8147999 DOI: 10.3390/vaccines9050459] [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: 03/19/2021] [Revised: 04/14/2021] [Accepted: 04/30/2021] [Indexed: 02/01/2023] Open
Abstract
Staphylococci (specifically Staphylococcus aureus and Staphylococcus epidermidis) are the causative agents of diseases ranging from superficial skin and soft tissue infections to severe conditions such as fatal pneumonia, bacteremia, sepsis and endocarditis. The widespread and indiscriminate use of antibiotics has led to serious problems of resistance to staphylococcal disease and has generated a renewed interest in alternative therapeutic agents such as vaccines and antibodies. Staphylococci express a large repertoire of surface and secreted virulence factors, which provide mechanisms (adhesion, invasion and biofilm development among others) for both bacterial survival in the host and evasion from innate and adaptive immunity. Consequently, the development of antibodies that target specific antigens would provide an effective protective strategy against staphylococcal infections. In this review, we report an update on efforts to develop anti-staphylococci monoclonal antibodies (and their derivatives: minibodies, antibody–antibiotic conjugates) and the mechanism by which such antibodies can help fight infections. We also provide an overview of mAbs used in clinical trials and highlight their therapeutic potential in various infectious contexts.
Collapse
|
15
|
Kasela M, Grzegorczyk A, Nowakowicz-Dębek B, Malm A. The Prevalence of Virulence Determinants and Antibiotic Resistance Patterns in Methicillin-Resistant Staphylococcus aureus in a Nursing Home in Poland. Pathogens 2021; 10:pathogens10040427. [PMID: 33916758 PMCID: PMC8065860 DOI: 10.3390/pathogens10040427] [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: 02/24/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 11/16/2022] Open
Abstract
Nursing homes (NH) contribute to the regional spread of methicillin-resistant Staphylococcus aureus (MRSA). Moreover, residents are vulnerable to the colonization and subsequent infection of MRSA etiology. We aimed at investigating the molecular and phenotypic characteristics of 21 MRSA collected from the residents and personnel in an NH (Lublin, Poland) during 2018. All MRSA were screened for 20 genes encoding virulence determinants (sea-see, eta, etb, tst, lukS-F-PV, eno, cna, ebpS, fib, bbp, fnbA, fnbB, icaADBC) and for resistance to 18 antimicrobials. To establish the relatedness and clonal complexes of MRSA in NH we applied multiple-locus variable-number tandem-repeat fingerprinting (MLVF), pulse field gel electrophoresis (PFGE), multilocus sequence typing (MLST) and staphylococcal cassette chromosome mec (SCCmec) typing. We identified four sequence types (ST) among two clonal complexes (CC): ST (CC22) known as EMRSA-15 as well as three novel STs—ST6295 (CC8), ST6293 (CC8) and ST6294. All tested MRSA were negative for sec, eta, etb, lukS-F-PV, bbp and ebpS genes. The most prevalent gene encoding toxin was sed (52.4%; n = 11/21), and adhesins were eno and fnbA (100%). Only 9.5% (n = 2/21) of MRSA were classified as multidrug-resistant. The emergence of novel MRSA with a unique virulence and the presence of epidemic clone EMRSA-15 creates challenges for controlling the spread of MRSA in NH.
Collapse
Affiliation(s)
- Martyna Kasela
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland; (A.G.); (A.M.)
- Correspondence:
| | - Agnieszka Grzegorczyk
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland; (A.G.); (A.M.)
| | - Bożena Nowakowicz-Dębek
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Anna Malm
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland; (A.G.); (A.M.)
| |
Collapse
|
16
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
17
|
Clow F, Peterken K, Pearson V, Proft T, Radcliff FJ. PilVax, a novel Lactococcus lactis-based mucosal vaccine platform, stimulates systemic and mucosal immune responses to Staphylococcus aureus. Immunol Cell Biol 2020; 98:369-381. [PMID: 32150301 DOI: 10.1111/imcb.12325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 03/07/2020] [Indexed: 12/28/2022]
Abstract
Most pathogens initiate infection via the mucosa, therefore delivery of vaccines directly to the mucosa is likely to be advantageous for stimulating protective immunity at the site of entry. PilVax is a novel mucosal vaccine platform that harnesses Lactococcus lactis bacteria engineered to stably express multiple copies of vaccine peptide antigens within pili, hair-like structures which extend from the cell wall. This strategy elicited systemic and mucosal antibody responses to a model antigen after intranasal immunization, but has not been tested for its capacity to stimulate protective mucosal immunity. A well-characterized linear B-cell epitope, D3(22-33) , from the fibronectin-binding protein A of Staphylococcus aureus was successfully introduced into PilVax and delivered intranasally to mice. Specific antipeptide immunoglobulin (Ig) G and IgA antibodies were detected in the serum and respiratory mucosa of vaccinated mice. Responses to the major pilus backbone protein Spy0128 were also assessed; robust antibody responses to this antigen were generated both systemically and in the respiratory and intestinal mucosa. Mice were challenged intranasally with the mouse-adapted S. aureus JSNZ strain and the S. aureus load quantified 7 days after challenge. Unexpectedly, exposure to PilVax, irrespective of the presence of the peptide, resulted in a significant reduction in S. aureus load in both the intestine and nasal mucosa (both P < 0.05) when compared with unvaccinated control mice. The mechanism(s) of protection are unclear, but merit further investigation to determine whether PilVax is a suitable platform for delivery of vaccine candidate antigens to the mucosa.
Collapse
Affiliation(s)
- Fiona Clow
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kelly Peterken
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Victoria Pearson
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Fiona J Radcliff
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
Abstract
Staphylococcus aureus is an important pathogen responsible for nosocomial and community-acquired infections in humans, and methicillin-resistant S. aureus (MRSA) infections have continued to increase despite widespread preventative measures. S. aureus can colonize the female vaginal tract, and reports have suggested an increase in MRSA infections in pregnant and postpartum women as well as outbreaks in newborn nurseries. Currently, little is known about specific factors that promote MRSA vaginal colonization and subsequent infection. To study S. aureus colonization of the female reproductive tract in a mammalian system, we developed a mouse model of S. aureus vaginal carriage and demonstrated that both hospital-associated and community-associated MRSA isolates can colonize the murine vaginal tract. Immunohistochemical analysis revealed an increase in neutrophils in the vaginal lumen during MRSA colonization. Additionally, we observed that a mutant lacking fibrinogen binding adhesins exhibited decreased persistence within the mouse vagina. To further identify novel factors that promote vaginal colonization, we performed RNA sequencing to determine the transcriptome of MRSA growing in vivo during vaginal carriage at 5 h, 1 day, and 3 days postinoculation. Over 25% of the bacterial genes were differentially regulated at all time points during colonization compared to laboratory cultures. The most highly induced genes were those involved in iron acquisition, including the Isd system and siderophore transport systems. Mutants deficient in these pathways did not persist as well during in vivo colonization. These results reveal that fibrinogen binding and the capacity to overcome host nutritional limitation are important determinants of MRSA vaginal colonization.IMPORTANCE Staphylococcus aureus is an opportunistic pathogen able to cause a wide variety of infections in humans. Recent reports have suggested an increasing prevalence of MRSA in pregnant and postpartum women, coinciding with the increased incidence of MRSA infections in neonatal intensive care units (NICUs) and newborn nurseries. Vertical transmission from mothers to infants at delivery is a likely route of MRSA acquisition by the newborn; however, essentially nothing is known about host and bacterial factors that influence MRSA carriage in the vagina. Here, we established a mouse model of vaginal colonization and observed that multiple MRSA strains can persist in the vaginal tract. Additionally, we determined that MRSA interactions with fibrinogen and iron uptake can promote vaginal persistence. This study is the first to identify molecular mechanisms which govern vaginal colonization by MRSA, the critical initial step preceding infection and neonatal transmission.
Collapse
|
19
|
Leonard AC, Petrie LE, Cox G. Bacterial Anti-adhesives: Inhibition of Staphylococcus aureus Nasal Colonization. ACS Infect Dis 2019; 5:1668-1681. [PMID: 31374164 DOI: 10.1021/acsinfecdis.9b00193] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bacterial adhesion to the skin and mucosa is often a fundamental and early step in host colonization, the establishment of bacterial infections, and pathology. This process is facilitated by adhesins on the surface of the bacterial cell that recognize host cell molecules. Interfering with bacterial host cell adhesion, so-called anti-adhesive therapeutics, offers promise for the development of novel approaches to control bacterial infections. In this review, we focus on the discovery of anti-adhesives targeting the high priority pathogen Staphylococcus aureus. This organism remains a major clinical burden, and S. aureus nasal colonization is associated with poor clinical outcomes. We describe the molecular basis of nasal colonization and highlight potentially efficacious targets for the development of novel nasal decolonization strategies.
Collapse
Affiliation(s)
- Allison C. Leonard
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Laurenne E. Petrie
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| | - Georgina Cox
- College of Biological Sciences, Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Rd E, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
20
|
The MSCRAMM Family of Cell-Wall-Anchored Surface Proteins of Gram-Positive Cocci. Trends Microbiol 2019; 27:927-941. [PMID: 31375310 DOI: 10.1016/j.tim.2019.06.007] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/10/2019] [Accepted: 06/19/2019] [Indexed: 01/21/2023]
Abstract
The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.
Collapse
|
21
|
Clumping factor B is an important virulence factor during Staphylococcus aureus skin infection and a promising vaccine target. PLoS Pathog 2019; 15:e1007713. [PMID: 31009507 PMCID: PMC6497315 DOI: 10.1371/journal.ppat.1007713] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 05/02/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus expresses a number of cell wall-anchored proteins that mediate adhesion and invasion of host cells and tissues and promote immune evasion, consequently contributing to the virulence of this organism. The cell wall-anchored protein clumping factor B (ClfB) has previously been shown to facilitate S. aureus nasal colonization through high affinity interactions with the cornified envelope in the anterior nares. However, the role of ClfB during skin and soft tissue infection (SSTI) has never been investigated. This study reveals a novel role for ClfB during SSTIs. ClfB is crucial in determining the abscess structure and bacterial burden early in infection and this is dependent upon a specific interaction with the ligand loricrin which is expressed within the abscess tissue. Targeting ClfB using a model vaccine that induced both protective humoral and cellular responses, leads to protection during S. aureus skin infection. This study therefore identifies ClfB as an important antigen for future SSTI vaccines. Staphylococcus aureus is the leading cause of skin and soft tissue infections (SSTIs), the treatment of which is becoming increasingly difficult due to antibiotic resistance. An anti-S. aureus vaccine offers a potential solution, but a better understanding of how S. aureus causes pathology during SSTI is required to identify effective vaccine targets. Here, we identify an important virulence determinant during S. aureus SSTI. Clumping factor B (ClfB), a surface protein expressed by S. aureus is shown to promote skin abscess formation by binding to the host protein loricrin. Targeting ClfB using a model vaccine conferred significant protection during S. aureus SSTI. In this study, we uncover an entirely novel mechanism by which S. aureus forms abscesses during skin infection, identifying an important therapeutic target for treating S. aureus SSTI.
Collapse
|
22
|
Laux C, Peschel A, Krismer B. Staphylococcus aureus Colonization of the Human Nose and Interaction with Other Microbiome Members. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0029-2018. [PMID: 31004422 PMCID: PMC11590430 DOI: 10.1128/microbiolspec.gpp3-0029-2018] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Indexed: 12/11/2022] Open
Abstract
Staphylococcus aureus is usually regarded as a bacterial pathogen due to its ability to cause multiple types of invasive infections. Nevertheless, S. aureus colonizes about 30% of the human population asymptomatically in the nares, either transiently or persistently, and can therefore be regarded a human commensal as well, although carriage increases the risk of infection. Whereas many facets of the infection processes have been studied intensively, little is known about the commensal lifestyle of S. aureus. Recent studies highlight the major role of the composition of the highly variable nasal microbiota in promoting or inhibiting S. aureus colonization. Competition for limited nutrients, trace elements, and epithelial attachment sites, different susceptibilities to host defense molecules and the production of antimicrobial molecules by bacterial competitors may determine whether nasal bacteria outcompete each other. This chapter summarizes our knowledge about mechanisms that are used by S. aureus for efficient nasal colonization and strategies used by other nasal bacteria to interfere with its colonization. An improved understanding of naturally evolved mechanisms might enable us to develop new strategies for pathogen eradication.
Collapse
Affiliation(s)
- Claudia Laux
- University of Tübingen, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Infection Biology Unit, 72076 Tübingen, Germany
| | - Andreas Peschel
- University of Tübingen, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Infection Biology Unit, 72076 Tübingen, Germany
| | - Bernhard Krismer
- University of Tübingen, Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Infection Biology Unit, 72076 Tübingen, Germany
| |
Collapse
|
23
|
Casadei E, Salinas I. Comparative models for human nasal infections and immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:212-222. [PMID: 30513304 PMCID: PMC7102639 DOI: 10.1016/j.dci.2018.11.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/30/2018] [Accepted: 11/30/2018] [Indexed: 05/09/2023]
Abstract
The human olfactory system is a mucosal surface and a major portal of entry for respiratory and neurotropic pathogens into the body. Understanding how the human nasopharynx-associated lymphoid tissue (NALT) halts the progression of pathogens into the lower respiratory tract or the central nervous system is key for developing effective cures. Although traditionally mice have been used as the gold-standard model for the study of human nasal diseases, mouse models present important caveats due to major anatomical and functional differences of the human and murine olfactory system and NALT. We summarize the NALT anatomy of different animal groups that have thus far been used to study host-pathogen interactions at the olfactory mucosa and to test nasal vaccines. The goal of this review is to highlight the strengths and limitations of each animal model of nasal immunity and to identify the areas of research that require further investigation to advance human health.
Collapse
Affiliation(s)
- Elisa Casadei
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA.
| | - Irene Salinas
- University of New Mexico, Department of Biology, Center for Evolutionary and Theoretical Immunology (CETI), Albuquerque, NM, USA
| |
Collapse
|
24
|
Majelan PA, Mahdavi M, Yazdi MH, Salimi E, Pourmand MR. Recombinant Staphylococcal Antigen-F (r-ScaF), a novel vaccine candidate against methicillin resistant Staphylococcus aureus infection: Potency and efficacy studies. Microb Pathog 2019; 127:159-165. [DOI: 10.1016/j.micpath.2018.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/24/2018] [Accepted: 11/26/2018] [Indexed: 01/06/2023]
|
25
|
Schneewind O, Missiakas D. Sortases, Surface Proteins, and Their Roles in Staphylococcus aureus Disease and Vaccine Development. Microbiol Spectr 2019; 7:10.1128/microbiolspec.psib-0004-2018. [PMID: 30737913 PMCID: PMC6386163 DOI: 10.1128/microbiolspec.psib-0004-2018] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Indexed: 12/27/2022] Open
Abstract
Sortases cleave short peptide motif sequences at the C-terminal end of secreted surface protein precursors and either attach these polypeptides to the peptidoglycan of Gram-positive bacteria or promote their assembly into pilus structures that are also attached to peptidoglycan. Sortase A, the enzyme first identified in the human pathogen Staphylococcus aureus, binds LPXTG motif sorting signals, cleaves between threonine (T) and glycine (G) residues, and forms an acyl enzyme between its active-site cysteine thiol and the carboxyl group of threonine (T). Sortase A acyl enzyme is relieved by the nucleophilic attack of the cross bridge amino group within lipid II, thereby generating surface protein linked to peptidoglycan precursor. Such products are subsequently incorporated into the cell wall envelope by enzymes of the peptidoglycan synthesis pathway. Surface proteins linked to peptidoglycan may be released from the bacterial envelope to diffuse into host tissues and fulfill specific biological functions. S. aureus sortase A is essential for host colonization and for the pathogenesis of invasive diseases. Staphylococcal sortase-anchored surface proteins fulfill key functions during the infectious process, and vaccine-induced antibodies targeting surface proteins may provide protection against S. aureus. Alternatively, small-molecule inhibitors of sortase may be useful agents for the prevention of S. aureus colonization and invasive disease.
Collapse
Affiliation(s)
- Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, IL 60637
| | | |
Collapse
|
26
|
Sakr A, Brégeon F, Mège JL, Rolain JM, Blin O. Staphylococcus aureus Nasal Colonization: An Update on Mechanisms, Epidemiology, Risk Factors, and Subsequent Infections. Front Microbiol 2018; 9:2419. [PMID: 30349525 PMCID: PMC6186810 DOI: 10.3389/fmicb.2018.02419] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/21/2018] [Indexed: 02/02/2023] Open
Abstract
Up to 30% of the human population are asymptomatically and permanently colonized with nasal Staphylococcus aureus. To successfully colonize human nares, S. aureus needs to establish solid interactions with human nasal epithelial cells and overcome host defense mechanisms. However, some factors like bacterial interactions in the human nose can influence S. aureus colonization and sometimes prevent colonization. On the other hand, certain host characteristics and environmental factors can predispose to colonization. Nasal colonization can cause opportunistic and sometimes life-threatening infections such as surgical site infections or other infections in non-surgical patients that increase morbidity, mortality as well as healthcare costs.
Collapse
Affiliation(s)
- Adèle Sakr
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix-Marseille Université, Marseille, France.,Service de Pharmacologie Clinique et Pharmacovigilance, AP-HM, Pharmacologie Intégrée et Interface Clinique et Industriel, Institut des Neurosciences Timone - UMR AMU-INSERM 1106, Aix-Marseille Université, Marseille, France
| | - Fabienne Brégeon
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix-Marseille Université, Marseille, France
| | - Jean-Louis Mège
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix-Marseille Université, Marseille, France
| | - Jean-Marc Rolain
- Faculté de Médecine et de Pharmacie, IRD, APHM, MEPHI, IHU Méditerranée Infection, Aix-Marseille Université, Marseille, France
| | - Olivier Blin
- Service de Pharmacologie Clinique et Pharmacovigilance, AP-HM, Pharmacologie Intégrée et Interface Clinique et Industriel, Institut des Neurosciences Timone - UMR AMU-INSERM 1106, Aix-Marseille Université, Marseille, France
| |
Collapse
|
27
|
Abstract
Vascular graft infection is a devastating complication of vascular reconstructive surgery. The infection can occur early in the postoperative period and is largely due to intraoperative contamination or by contiguous extension from a nearby infection. It can also occur years after implantation. Staphylococci remain the most common organisms and biofilm production makes eradication difficult. Factors commonly reported to predispose to vascular graft infection are periodontal disease, nasal colonization with Staphylococcus aureus, bacteremia, certain graft characteristics, diabetes mellitus, postoperative hyperglycemia, location of the incision, wound infection, and emergency procedure. Management consists of antibiotic and surgical therapy. Preventive methods are described.
Collapse
Affiliation(s)
- Amal Gharamti
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut, Cairo Street, Riad El Solh, Beirut 1107 2020, Lebanon
| | - Zeina A Kanafani
- Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut, Cairo Street, Riad El Solh, Beirut 1107 2020, Lebanon; Division of Infectious Diseases, Department of Internal Medicine, American University of Beirut Medical Center, Cairo Street, PO Box 11-0236/11D, Riad El Solh, Beirut 1107 2020, Lebanon.
| |
Collapse
|
28
|
Staphylococcal Protein A Contributes to Persistent Colonization of Mice with Staphylococcus aureus. J Bacteriol 2018; 200:JB.00735-17. [PMID: 29440258 DOI: 10.1128/jb.00735-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus persistently colonizes the nasopharynx in humans, which increases the risk for invasive diseases, such as skin infection and bacteremia. Nasal colonization triggers IgG responses against staphylococcal surface antigens; however, these antibodies cannot prevent subsequent colonization or disease. Here, we describe S. aureus WU1, a multilocus sequence type 88 (ST88) isolate that persistently colonizes the nasopharynx in mice. We report that staphylococcal protein A (SpA) is required for persistence of S. aureus WU1 in the nasopharynx. Compared to animals colonized by wild-type S. aureus, mice colonized with the Δspa variant mount increased IgG responses against staphylococcal colonization determinants. Immunization of mice with a nontoxigenic SpA variant, which cannot cross-link B cell receptors and divert antibody responses, elicits protein A-neutralizing antibodies that promote IgG responses against colonizing S. aureus and diminish pathogen persistence.IMPORTANCE Staphylococcus aureus persistently colonizes the nasopharynx in about one-third of the human population, thereby promoting community- and hospital-acquired infections. Antibiotics are currently used for decolonization of individuals at increased risk of infection. However, the efficacy of antibiotics is limited by recolonization and selection for drug-resistant strains. Here, we propose a model of how staphylococcal protein A (SpA), a B cell superantigen, modifies host immune responses during colonization to support continued persistence of S. aureus in the nasopharynx. We show that this mechanism can be thwarted by vaccine-induced anti-SpA antibodies that promote IgG responses against staphylococcal antigens and diminish colonization.
Collapse
|
29
|
Speziale P, Rindi S, Pietrocola G. Antibody-Based Agents in the Management of Antibiotic-Resistant Staphylococcus aureus Diseases. Microorganisms 2018. [PMID: 29533985 PMCID: PMC5874639 DOI: 10.3390/microorganisms6010025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is a human pathogen that can cause a wide spectrum of diseases, including sepsis, pneumonia, arthritis, and endocarditis. Ineffective treatment of a number of staphylococcal infections with antibiotics is due to the development and spread of antibiotic-resistant strains following decades of antibiotic usage. This has generated renewed interest within the scientific community in alternative therapeutic agents, such as anti-S. aureus antibodies. Although the role of antibodies in the management of S. aureus diseases is controversial, the success of this pathogen in neutralizing humoral immunity clearly indicates that antibodies offer the host extensive protection. In this review, we report an update on efforts to develop antibody-based agents, particularly monoclonal antibodies, and their therapeutic potential in the passive immunization approach to the treatment and prevention of S. aureus infections.
Collapse
Affiliation(s)
- Pietro Speziale
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
- Department of Industrial and Information Engineering, University of Pavia, 27100 Pavia, Italy.
| | - Simonetta Rindi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy.
| | | |
Collapse
|
30
|
Humoral immune consequences of Staphylococcus aureus ST239-associated bacteremia. Eur J Clin Microbiol Infect Dis 2017; 37:255-263. [PMID: 29103153 DOI: 10.1007/s10096-017-3124-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 01/21/2023]
Abstract
The humoral immune responses against 46 different staphylococcal antigens in 27 bacteremia patients infected by clonally related methicillin-resistant Staphylococcus aureus (MRSA) strains of a single sequence type (ST) 239 were investigated. A group of non-infected patients (n = 31) hospitalized for different reasons served as controls. All strains were confirmed as ST 239 by S. aureus and mecA-specific PCR, spa, and multi-locus sequence typing (MLST). In each bacteremia patient, a unique pattern of S. aureus antigen-specific immune responses after infection was observed. Antibody levels among bacteremia patients were significantly higher than controls for HlgB (P = 0.001), LukD (P = 0.009), LukF (P = 0.0001), SEA (P = 0.0001), SEB (P = 0.011), SEC (P = 0.010), SEQ (P = 0.049), IsaA (P = 0.043), IsdA (P = 0.038), IsdH (P = 0.01), SdrD (P = 0.001), SdrE (P = 0.046), EsxA (P = 0.0001), and SA0104 (P = 0.0001). On the other hand, the antibody levels were significantly higher among controls for SSL3 (P = 0.009), SSL9 (P = 0.002), and SSL10 (P = 0.007) when the IgG level on the day of infection was compared with that measured on the day of admission. Diversity was observed in the immune response against the antigens. However, a set of antigens (IsaA, IsdA, IsdH, SdrD, and HlgB) triggered a similar type of immune response in different individuals. We suggest that these antigens could be considered when developing a multi-component (passive) vaccine. SEA and/or its specific antibodies seem to play a critical role during ST239 MRSA bacteremia and SEA-targeted therapy may be a strategy to be considered.
Collapse
|
31
|
Krismer B, Weidenmaier C, Zipperer A, Peschel A. The commensal lifestyle of Staphylococcus aureus and its interactions with the nasal microbiota. Nat Rev Microbiol 2017; 15:675-687. [PMID: 29021598 DOI: 10.1038/nrmicro.2017.104] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Although human colonization by facultative bacterial pathogens, such as Staphylococcus aureus, represents a major risk factor for invasive infections, the commensal lifestyle of such pathogens has remained a neglected area of research. S. aureus colonizes the nares of approximately 30% of the human population and recent studies suggest that the composition of highly variable nasal microbiota has a major role in promoting or inhibiting S. aureus colonization. Competition for epithelial attachment sites or limited nutrients, different susceptibilities to host defence molecules and the production of antimicrobial molecules may determine whether nasal bacteria outcompete each other. In this Review, we discuss recent insights into mechanisms that are used by S. aureus to prevail in the human nose and the counter-strategies that are used by other nasal bacteria to interfere with its colonization. Understanding such mechanisms will be crucial for the development of new strategies for the eradication of endogenous facultative pathogens.
Collapse
Affiliation(s)
- Bernhard Krismer
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, 72076 Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Christopher Weidenmaier
- German Centre for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany.,Interfaculty Institute of Microbiology and Infection Medicine, Medical Microbiology and Hygiene, University of Tübingen, 72076 Tübingen, Germany
| | - Alexander Zipperer
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, 72076 Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology, University of Tübingen, 72076 Tübingen, Germany.,German Centre for Infection Research (DZIF), Partner Site Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
32
|
Flaxman A, van Diemen PM, Yamaguchi Y, Allen E, Lindemann C, Rollier CS, Milicic A, Wyllie DH. Development of persistent gastrointestinal S. aureus carriage in mice. Sci Rep 2017; 7:12415. [PMID: 28963555 PMCID: PMC5622074 DOI: 10.1038/s41598-017-12576-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 09/01/2017] [Indexed: 01/09/2023] Open
Abstract
One fifth to one quarter of the human population is asymptomatically, naturally and persistently colonised by Staphylococcus aureus. Observational human studies indicate that although the whole population is intermittently exposed, some individuals lose S. aureus rapidly. Others become persistent carriers, as assessed by nasal cultures, with many individuals colonised for decades. Current animal models of S. aureus colonisation are expensive and normally require antibiotics. Importantly, these animal models have not yet contributed to our poor understanding of the dichotomy in human colonisation status. Here, we identify a single strain of S. aureus found to be persistently colonising the gastrointestinal tract of BALB/c mice. Phylogenetic analyses suggest it diverged from a human ST15 lineage in the recent past. We show that murine carriage of this organism occurs in the bowel and nares, is acquired early in life, and can persist for months. Importantly, we observe the development of persistent and non-persistent gastrointestinal carriage states in genetically identical mice. We developed a needle- and antibiotic-free model in which we readily induced S. aureus colonisation of the gastrointestinal tract experimentally by environmental exposure. Using our experimental model, impact of adaptive immunity on S. aureus colonisation could be assessed. Vaccine efficacy to eliminate colonisation could also be investigated using this model.
Collapse
Affiliation(s)
- Amy Flaxman
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Pauline M van Diemen
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Yuko Yamaguchi
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Elizabeth Allen
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Claudia Lindemann
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, CCVTM, Oxford, UK.,The NIHR Oxford Biomedical Research Centre, Oxford University Hospitals, Oxford, UK
| | | | - David H Wyllie
- Jenner Institute, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Oxford, UK.
| |
Collapse
|
33
|
Combining Two Methods of Global Sensitivity Analysis to Investigate MRSA Nasal Carriage Model. Bull Math Biol 2017; 79:2258-2272. [PMID: 28752384 DOI: 10.1007/s11538-017-0329-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 07/20/2017] [Indexed: 10/19/2022]
Abstract
We apply two different sensitivity techniques to a model of bacterial colonization of the anterior nares to better understand the dynamics of Staphylococcus aureus nasal carriage. Specifically, we use partial rank correlation coefficients to investigate sensitivity as a function of time and identify a reduced model with fewer than half of the parameters of the full model. The reduced model is used for the calculation of Sobol' indices to identify interacting parameters by their additional effects indices. Additionally, we found that the model captures an interesting characteristic of the biological phenomenon related to the initial population size of the infection; only two parameters had any significant additional effects, and these parameters have biological evidence suggesting they are connected but not yet completely understood. Sensitivity is often applied to elucidate model robustness, but we show that combining sensitivity measures can lead to synergistic insight into both model and biological structures.
Collapse
|
34
|
Fernandez S, Ledo C, Lattar S, Noto Llana M, Bertelli AM, Di Gregorio S, Sordelli DO, Gómez MI, Mollerach ME. High virulence of methicillin resistant Staphylococcus aureus ST30-SCC mec IVc- spa t019, the dominant community-associated clone in Argentina. Int J Med Microbiol 2017; 307:191-199. [DOI: 10.1016/j.ijmm.2017.05.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 05/11/2017] [Accepted: 05/14/2017] [Indexed: 10/19/2022] Open
|
35
|
Yang Y, Yu R, Yang X, Liu S, Fang T, Song X, Hou L, Yu C, Xu J, Fu L, Yi S, Chen W. Protection against Staphylococcus aureus and tetanus infections by a combined vaccine containing SasA and TeNT‑Hc in mice. Mol Med Rep 2017; 15:2369-2373. [PMID: 28259925 DOI: 10.3892/mmr.2017.6227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/19/2016] [Indexed: 11/06/2022] Open
Abstract
In developing countries, trauma patients and neonates are vulnerable to Staphylococcus aureus (S. aureus) and Clostridium tetani infections. It has been suggested that a combined vaccine against the two infections may be a reliable and cost‑effective strategy. Previous studies have indicated that the S. aureus surface protein A (SasA) and the C fragment of tetanus neurotoxin (TeNT‑Hc) may be suitable candidates for a vaccine against S. aureus and tetanus infections, respectively. In the present study, mice were immunized with a combined vaccine containing SasA and TeNT‑Hc, which induced a robust immune response to both antigens, and mutual interference between SasA and TeNT‑Hc was not observed. In the S.aureus challenge model, the combined vaccine fully protected BALB/c mice against lethal intraperitoneal challenges with 3x109 colony‑forming units of a methicillin‑resistant S. aureus USA300 strain. In the TeNT challenge model, the combined vaccine conferred complete protection against a lethal dose of (2x103) xLD50 tetanus toxin. These results implied that SasA and TeNT‑Hc promising components for a combined vaccine against S. aureus and tetanus infections.
Collapse
Affiliation(s)
- Yilong Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Rui Yu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Xiuxu Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Shuling Liu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Ting Fang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Xiaohong Song
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Lihua Hou
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Changming Yu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Ling Fu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Shaoqiong Yi
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| |
Collapse
|
36
|
Liu B, Park S, Thompson CD, Li X, Lee JC. Antibodies to Staphylococcus aureus capsular polysaccharides 5 and 8 perform similarly in vitro but are functionally distinct in vivo. Virulence 2016; 8:859-874. [PMID: 27936346 DOI: 10.1080/21505594.2016.1270494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The capsular polysaccharide (CP) produced by Staphylococcus aureus is a virulence factor that allows the organism to evade uptake and killing by host neutrophils. Polyclonal antibodies to the serotype 5 (CP5) and type 8 (CP8) capsular polysaccharides are opsonic and protect mice against experimental bacteremia provoked by encapsulated staphylococci. Thus, passive immunotherapy using CP antibodies has been considered for the prevention or treatment of invasive antibiotic-resistant S. aureus infections. In this report, we generated monoclonal antibodies (mAbs) against S. aureus CP5 or CP8. Backbone specific mAbs reacted with native and O-deacetylated CPs, whereas O-acetyl specific mAbs reacted only with native CPs. Reference strains of S. aureus and a selection of clinical isolates reacted by colony immunoblot with the CP5 and CP8 mAbs in a serotype-specific manner. The mAbs mediated in vitro CP type-specific opsonophagocytic killing of S. aureus strains, and mice passively immunized with CP5 mAbs were protected against S. aureus bacteremia. Neither CP8-specific mAbs or polyclonal antibodies protected mice against bacteremia provoked by serotype 8 S. aureus clinical isolates, although these same antibodies did protect against a serotype 5 S. aureus strain genetically engineered to produce CP8. We detected soluble CP8 in culture supernatants of serotype 8 clinical isolates and in the plasma of infected animals. Serotype 5 S. aureus released significantly less soluble CP5 in vitro and in vivo. The release of soluble CP8 by S. aureus may contribute to the inability of CP8 vaccines or antibodies to protect against serotype 8 staphylococcal infections.
Collapse
Affiliation(s)
- Bo Liu
- a Division of Infectious Diseases , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| | - Saeyoung Park
- a Division of Infectious Diseases , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| | - Christopher D Thompson
- a Division of Infectious Diseases , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| | - Xue Li
- a Division of Infectious Diseases , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA.,b Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Jean C Lee
- a Division of Infectious Diseases , Brigham and Women's Hospital and Harvard Medical School , Boston , MA , USA
| |
Collapse
|
37
|
Crosby HA, Kwiecinski J, Horswill AR. Staphylococcus aureus Aggregation and Coagulation Mechanisms, and Their Function in Host-Pathogen Interactions. ADVANCES IN APPLIED MICROBIOLOGY 2016; 96:1-41. [PMID: 27565579 DOI: 10.1016/bs.aambs.2016.07.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The human commensal bacterium Staphylococcus aureus can cause a wide range of infections ranging from skin and soft tissue infections to invasive diseases like septicemia, endocarditis, and pneumonia. Muticellular organization almost certainly contributes to S. aureus pathogenesis mechanisms. While there has been considerable focus on biofilm formation and its role in colonizing prosthetic joints and indwelling devices, less attention has been paid to nonsurface-attached group behavior like aggregation and clumping. S. aureus is unique in its ability to coagulate blood, and it also produces multiple fibrinogen-binding proteins that facilitate clumping. Formation of clumps, which are large, tightly packed groups of cells held together by fibrin(ogen), has been demonstrated to be important for S. aureus virulence and immune evasion. Clumps of cells are able to avoid detection by the host's immune system due to a fibrin(ogen) coat that acts as a shield, and the size of the clumps facilitates evasion of phagocytosis. In addition, clumping could be an important early step in establishing infections that involve tight clusters of cells embedded in host matrix proteins, such as soft tissue abscesses and endocarditis. In this review, we discuss clumping mechanisms and regulation, as well as what is known about how clumping contributes to immune evasion.
Collapse
Affiliation(s)
- H A Crosby
- University of Iowa, Iowa City, IA, United States
| | - J Kwiecinski
- University of Iowa, Iowa City, IA, United States
| | - A R Horswill
- University of Iowa, Iowa City, IA, United States
| |
Collapse
|
38
|
Host-Bacterial Crosstalk Determines Staphylococcus aureus Nasal Colonization. Trends Microbiol 2016; 24:872-886. [PMID: 27474529 DOI: 10.1016/j.tim.2016.06.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/13/2016] [Accepted: 06/29/2016] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus persistently colonizes the anterior nares of approximately one fifth of the population and nasal carriage is a significant risk factor for infection. Recent advances have significantly refined our understanding of S. aureus-host communication during nasal colonization. Novel bacterial adherence mechanisms in the nasal epithelium have been identified, and novel roles for both the innate and the adaptive immune response in controlling S. aureus nasal colonization have been defined, through the use of both human and rodent models. It is clear that S. aureus maintains a unique, complex relationship with the host immune system and that S. aureus nasal colonization is overall a multifactorial process which is as yet incompletely understood.
Collapse
|
39
|
Dayan GH, Mohamed N, Scully IL, Cooper D, Begier E, Eiden J, Jansen KU, Gurtman A, Anderson AS. Staphylococcus aureus: the current state of disease, pathophysiology and strategies for prevention. Expert Rev Vaccines 2016; 15:1373-1392. [PMID: 27118628 DOI: 10.1080/14760584.2016.1179583] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus is both a commensal organism and also an important opportunistic human pathogen, causing a variety of community and hospital-associated pathologies, such as bacteremia-sepsis, endocarditis, pneumonia, osteomyelitis, arthritis and skin diseases. The resurgence of S. aureus during the last decade in many settings has been facilitated not only by bacterial antibiotic resistance mechanisms but also by the emergence of new S. aureus clonal types with increased expression of virulence factors and the capacity to neutralize the host immune response. Prevention of the spread of S. aureus infection relies on the use of contact precautions and adequate procedures for infection control that so far have not been fully effective. Prevention using a prophylactic vaccine would complement these processes, having the potential to bring additional, significant progress toward decreasing invasive disease due to S. aureus.
Collapse
Affiliation(s)
- Gustavo H Dayan
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Naglaa Mohamed
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Ingrid L Scully
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - David Cooper
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Elizabeth Begier
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Joseph Eiden
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | - Kathrin U Jansen
- a Pfizer Vaccine Research and Development , Pearl River , NY , USA
| | | | | |
Collapse
|
40
|
Becker SC, Roach DR, Chauhan VS, Shen Y, Foster-Frey J, Powell AM, Bauchan G, Lease RA, Mohammadi H, Harty WJ, Simmons C, Schmelcher M, Camp M, Dong S, Baker JR, Sheen TR, Doran KS, Pritchard DG, Almeida RA, Nelson DC, Marriott I, Lee JC, Donovan DM. Triple-acting Lytic Enzyme Treatment of Drug-Resistant and Intracellular Staphylococcus aureus. Sci Rep 2016; 6:25063. [PMID: 27121552 PMCID: PMC4848530 DOI: 10.1038/srep25063] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/11/2016] [Indexed: 12/15/2022] Open
Abstract
Multi-drug resistant bacteria are a persistent problem in modern health care, food safety and animal health. There is a need for new antimicrobials to replace over used conventional antibiotics. Here we describe engineered triple-acting staphylolytic peptidoglycan hydrolases wherein three unique antimicrobial activities from two parental proteins are combined into a single fusion protein. This effectively reduces the incidence of resistant strain development. The fusion protein reduced colonization by Staphylococcus aureus in a rat nasal colonization model, surpassing the efficacy of either parental protein. Modification of a triple-acting lytic construct with a protein transduction domain significantly enhanced both biofilm eradication and the ability to kill intracellular S. aureus as demonstrated in cultured mammary epithelial cells and in a mouse model of staphylococcal mastitis. Interestingly, the protein transduction domain was not necessary for reducing the intracellular pathogens in cultured osteoblasts or in two mouse models of osteomyelitis, highlighting the vagaries of exactly how protein transduction domains facilitate protein uptake. Bacterial cell wall degrading enzyme antimicrobials can be engineered to enhance their value as potent therapeutics.
Collapse
Affiliation(s)
| | | | | | - Yang Shen
- Institute for Bioscience and Biotechnology Research, University of MD, Rockville, MD, USA.,Department of Veterinary Medicine, University of MD, College Park, MD, USA
| | | | | | - Gary Bauchan
- ARS, USDA, 10300 Baltimore Ave, Beltsville, MD, USA
| | | | | | | | - Chad Simmons
- ARS, USDA, 10300 Baltimore Ave, Beltsville, MD, USA
| | | | - Mary Camp
- ARS, USDA, 10300 Baltimore Ave, Beltsville, MD, USA
| | - Shengli Dong
- Biochemistry, Univ. Alabama, Birmingham, Birmingham, AL, USA
| | - John R Baker
- Biochemistry, Univ. Alabama, Birmingham, Birmingham, AL, USA
| | | | - Kelly S Doran
- Biology, San Diego State University, San Diego, CA, USA
| | | | | | - Daniel C Nelson
- Institute for Bioscience and Biotechnology Research, University of MD, Rockville, MD, USA.,Department of Veterinary Medicine, University of MD, College Park, MD, USA
| | - Ian Marriott
- Biology, Univ. North Carolina, Charlotte, Charlotte, NC, USA
| | - Jean C Lee
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
41
|
Abtahi H, Farhangnia L, Ghaznavi-Rad E. In Vitro and in Vivo Antistaphylococcal Activity Determination of the New Recombinant Lysostaphin Protein. Jundishapur J Microbiol 2016; 9:e28489. [PMID: 27217919 PMCID: PMC4870841 DOI: 10.5812/jjm.28489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Revised: 05/31/2015] [Accepted: 06/14/2015] [Indexed: 11/28/2022] Open
Abstract
Background: Bacterial infection by antibiotic-resistant Staphylococcus aureus strains is a worldwide concern and the development of novel antistaphylococcal agents is acutely needed. Lysostaphin, an example of such novel agents, is a bacteriocin secreted by S. simulans to kill S. aureus through proteolysis of the Staphylococcus cell wall. Objectives: The aim of this study was to evaluate the in vitro and in vivo antistaphylococcal activity of recombinant lysostaphin. Materials and Methods: The in vitro study of the recombinant lysostaphin activity against S. aureus was determined by turbidimetric assay. For in vivo investigation, two groups of rats were inoculated with 1.4 × 109 CFU S. aureus. Five days after the nasal instillation of S. aureus, treatment in one of the groups was performed with a single dose (200 μg/dose) of recombinant lysostaphin formulated in Eucerin-based cream. Results: Recombinant lysostaphin at 100 μg/mL concentration showed a significant decrease of the optical density compared to the control samples. The in vivo study demonstrated that a single dose (200 μg/dose) of recombinant lysostaphin cream significantly reduced nasal colonization in all the treated animals compared to the untreated ones. Conclusions: These results demonstrated that the recombinant lysostaphin produced in this study was able to kill nasal S. aureus in rats. It can be recommended for human clinical trial studies.
Collapse
Affiliation(s)
- Hamid Abtahi
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, IR Iran
| | - Leila Farhangnia
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, IR Iran
| | - Ehsanollah Ghaznavi-Rad
- Department of Microbiology and Immunology, Molecular and Medicine Research Center, Faculty of Medicine, Arak University of Medical Sciences, Arak, IR Iran
- Corresponding author: Ehsanollah Ghaznavi-Rad, Department of Microbiology and Immunology, Molecular and Medicine Research Center, Faculty of Medicine, Arak University of Medical Sciences, Arak, IR Iran. Tel/Fax: +98-8634173526, E-mail:
| |
Collapse
|
42
|
Askarian F, Ajayi C, Hanssen AM, van Sorge NM, Pettersen I, Diep DB, Sollid JUE, Johannessen M. The interaction between Staphylococcus aureus SdrD and desmoglein 1 is important for adhesion to host cells. Sci Rep 2016; 6:22134. [PMID: 26924733 PMCID: PMC4770587 DOI: 10.1038/srep22134] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/03/2016] [Indexed: 12/27/2022] Open
Abstract
Staphylococcus aureus is known as a frequent colonizer of the skin and mucosa. Among bacterial factors involved in colonization are adhesins such as the microbial surface components recognizing adhesive matrix molecules (MSCRAMMs). Serine aspartate repeat containing protein D (SdrD) is involved in adhesion to human squamous cells isolated from the nose. Here, we identify Desmoglein 1 (Dsg1) as a novel interaction partner for SdrD. Genetic deletion of sdrD in S. aureus NCTC8325-4 through allelic replacement resulted in decreased bacterial adherence to Dsg1- expressing HaCaT cells in vitro. Complementary gain-of-function was demonstrated by heterologous expression of SdrD in Lactococcus lactis, which increased adherence to HaCaT cells. Also ectopic expression of Dsg1 in HEK293 cells resulted in increased adherence of S. aureus NCTC8325-4 in vitro. Increased adherence of NCTC8325-4, compared to NCTC8325-4ΔsdrD, to the recombinant immobilized Dsg1 demonstrated direct interaction between SdrD and Dsg1. Specificity of SdrD interaction with Dsg1 was further verified using flow cytometry and confirmed binding of recombinant SdrD to HaCaT cells expressing Dsg1 on their surface. These data demonstrate that Dsg1 is a host ligand for SdrD.
Collapse
Affiliation(s)
- Fatemeh Askarian
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Clement Ajayi
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Anne-Merethe Hanssen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Nina M van Sorge
- Medical Microbiology, University Medical Center Utrecht, Utrecht 3584CX, The Netherlands
| | - Ingvild Pettersen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Dzung B Diep
- Department of Chemistry, Biotechnology and Food Science, Norwegian University of Life Science, Ås, Norway
| | - Johanna U E Sollid
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| | - Mona Johannessen
- Research group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT-The Artic University of Norway, Norway
| |
Collapse
|
43
|
A new class of phenylhydrazinylidene derivatives as inhibitors of Staphylococcus aureus biofilm formation. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1535-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
44
|
Lacey KA, Geoghegan JA, McLoughlin RM. The Role of Staphylococcus aureus Virulence Factors in Skin Infection and Their Potential as Vaccine Antigens. Pathogens 2016; 5:pathogens5010022. [PMID: 26901227 PMCID: PMC4810143 DOI: 10.3390/pathogens5010022] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 01/27/2016] [Accepted: 02/03/2016] [Indexed: 01/13/2023] Open
Abstract
Staphylococcus aureus (S. aureus) causes the vast majority of skin and soft tissue infections (SSTIs) in humans. S. aureus has become increasingly resistant to antibiotics and there is an urgent need for new strategies to tackle S. aureus infections. Vaccines offer a potential solution to this epidemic of antimicrobial resistance. However, the development of next generation efficacious anti-S. aureus vaccines necessitates a greater understanding of the protective immune response against S. aureus infection. In particular, it will be important to ascertain if distinct immune mechanisms are required to confer protection at distinct anatomical sites. Recent discoveries have highlighted that interleukin-17-producing T cells play a particularly important role in the immune response to S. aureus skin infection and suggest that vaccine strategies to specifically target these types of T cells may be beneficial in the treatment of S. aureus SSTIs. S. aureus expresses a large number of cell wall-anchored (CWA) proteins, which are covalently attached to the cell wall peptidoglycan. The virulence potential of many CWA proteins has been demonstrated in infection models; however, there is a paucity of information regarding their roles during SSTIs. In this review, we highlight potential candidate antigens for vaccines targeted at protection against SSTIs.
Collapse
Affiliation(s)
- Keenan A Lacey
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Joan A Geoghegan
- Microbiology Department, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Rachel M McLoughlin
- Host Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
45
|
Parker D, Ahn D, Cohen T, Prince A. Innate Immune Signaling Activated by MDR Bacteria in the Airway. Physiol Rev 2016; 96:19-53. [PMID: 26582515 DOI: 10.1152/physrev.00009.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Health care-associated bacterial pneumonias due to multiple-drug resistant (MDR) pathogens are an important public health problem and are major causes of morbidity and mortality worldwide. In addition to antimicrobial resistance, these organisms have adapted to the milieu of the human airway and have acquired resistance to the innate immune clearance mechanisms that normally prevent pneumonia. Given the limited efficacy of antibiotics, bacterial clearance from the airway requires an effective immune response. Understanding how specific airway pathogens initiate and regulate innate immune signaling, and whether this response is excessive, leading to host-induced pathology may guide future immunomodulatory therapy. We will focus on three of the most important causes of health care-associated pneumonia, Staphylococcus aureus, Pseudomonas aeruginosa, and Klebsiella pneumoniae, and review the mechanisms through which an inappropriate or damaging innate immune response is stimulated, as well as describe how airway pathogens cause persistent infection by evading immune activation.
Collapse
Affiliation(s)
- Dane Parker
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Danielle Ahn
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Taylor Cohen
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| | - Alice Prince
- Departments of Pediatrics and Pharmacology, Columbia University, New York, New York
| |
Collapse
|
46
|
Reyes-Robles T, Lubkin A, Alonzo F, Lacy DB, Torres VJ. Exploiting dominant-negative toxins to combat Staphylococcus aureus pathogenesis. EMBO Rep 2016; 17:428-40. [PMID: 26882549 DOI: 10.15252/embr.201540994] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus (S. aureus) is a human pathogen that relies on the subversion of host phagocytes to support its pathogenic lifestyle. S. aureus strains can produce up to five beta-barrel, bi-component, pore-forming leukocidins that target and kill host phagocytes. Thus, preventing immune cell killing by these toxins is likely to boost host immunity. Here, we describe the identification of glycine-rich motifs within the membrane-penetrating stem domains of the leukocidin subunits that are critical for killing primary human neutrophils. Remarkably, leukocidins lacking these glycine-rich motifs exhibit dominant-negative inhibitory effects toward their wild-type toxin counterparts as well as other leukocidins. Biochemical and cellular assays revealed that these dominant-negative toxins work by forming mixed complexes that are impaired in pore formation. The dominant-negative leukocidins inhibited S. aureus cytotoxicity toward primary human neutrophils, protected mice from lethal challenge by wild-type leukocidin, and reduced bacterial burden in a murine model of bloodstream infection. Thus, we describe the first example of staphylococcal bi-component dominant-negative toxins and their potential as novel therapeutics to combat S. aureus infection.
Collapse
Affiliation(s)
- Tamara Reyes-Robles
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - D Borden Lacy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
47
|
Kiedrowski MR, Paharik AE, Ackermann LW, Shelton AU, Singh SB, Starner TD, Horswill AR. Development of an in vitro colonization model to investigate Staphylococcus aureus interactions with airway epithelia. Cell Microbiol 2016; 18:720-32. [PMID: 26566259 DOI: 10.1111/cmi.12543] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 11/03/2015] [Accepted: 11/04/2015] [Indexed: 12/27/2022]
Abstract
Staphylococcus aureus is a bacterial pathogen responsible for a wide range of diseases and is also a human commensal colonizing the upper respiratory tract. Strains belonging to the clonal complex group CC30 are associated with colonization, although the colonization state itself is not clearly defined. In this work, we developed a co-culture model with S. aureus colonizing the apical surface of polarized human airway epithelial cells. The S. aureus are grown at the air-liquid interface to allow an in-depth evaluation of a simulated colonization state. Exposure to wild-type, S. aureus bacteria or conditioned media killed airway epithelial cells within 1 day, while mutant S. aureus strains lacking alpha-toxin (hla) persisted on viable cells for at least 2 days. Recent S. aureus CC30 isolates are natural hla mutants, and we observed that these strains displayed reduced toxicity toward airway epithelial cells. Quantitative real-time polymerase chain reaction of known virulence factors showed the expression profile of S. aureus grown in co-culture correlates with results from previous human colonization studies. Microarray analysis indicated significant shifts in S. aureus physiology in the co-culture model toward lipid and amino acid metabolism. The development of the in vitro colonization model will enable further study of specific S. aureus interactions with the host epithelia.
Collapse
Affiliation(s)
- Megan R Kiedrowski
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexandra E Paharik
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Laynez W Ackermann
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Annie U Shelton
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sachinkumar B Singh
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Timothy D Starner
- Department of Pediatrics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexander R Horswill
- Department of Microbiology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
48
|
Cascioferro S, Raffa D, Maggio B, Raimondi MV, Schillaci D, Daidone G. Sortase A Inhibitors: Recent Advances and Future Perspectives. J Med Chem 2015; 58:9108-23. [DOI: 10.1021/acs.jmedchem.5b00779] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Stella Cascioferro
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
- IEMEST, Istituto Euromediterraneo di Scienza e Tecnologia, Via Emerico Amari, 123, 90139 Palermo, Italy
| | - Demetrio Raffa
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Benedetta Maggio
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Maria Valeria Raimondi
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Domenico Schillaci
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Giuseppe Daidone
- Dipartimento
di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione
di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
49
|
Misawa Y, Kelley KA, Wang X, Wang L, Park WB, Birtel J, Saslowsky D, Lee JC. Staphylococcus aureus Colonization of the Mouse Gastrointestinal Tract Is Modulated by Wall Teichoic Acid, Capsule, and Surface Proteins. PLoS Pathog 2015. [PMID: 26201029 PMCID: PMC4511793 DOI: 10.1371/journal.ppat.1005061] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Staphylococcus aureus colonizes the nose, throat, skin, and gastrointestinal (GI) tract of humans. GI carriage of S. aureus is difficult to eradicate and has been shown to facilitate the transmission of the bacterium among individuals. Although staphylococcal colonization of the GI tract is asymptomatic, it increases the likelihood of infection, particularly skin and soft tissue infections caused by USA300 isolates. We established a mouse model of persistent S. aureus GI colonization and characterized the impact of selected surface antigens on colonization. In competition experiments, an acapsular mutant colonized better than the parental strain Newman, whereas mutants defective in sortase A and clumping factor A showed impaired ability to colonize the GI tract. Mutants lacking protein A, clumping factor B, poly-N-acetyl glucosamine, or SdrCDE showed no defect in colonization. An S. aureus wall teichoic acid (WTA) mutant (ΔtagO) failed to colonize the mouse nose or GI tract, and the tagO and clfA mutants showed reduced adherence in vitro to intestinal epithelial cells. The tagO mutant was recovered in lower numbers than the wild type strain in the murine stomach and duodenum 1 h after inoculation. This reduced fitness correlated with the in vitro susceptibility of the tagO mutant to bile salts, proteases, and a gut-associated defensin. Newman ΔtagO showed enhanced susceptibility to autolysis, and an autolysin (atl) tagO double mutant abrogated this phenotype. However, the atl tagO mutant did not survive better in the mouse GI tract than the tagO mutant. Our results indicate that the failure of the tagO mutant to colonize the GI tract correlates with its poor adherence and susceptibility to bactericidal factors within the mouse gut, but not to enhanced activity of its major autolysin. Staphylococcus aureus persistently colonizes ~20% of the human population, and 40–60% of humans are intermittently colonized by this bacterium. The most common reservoir for S. aureus is the anterior nares, and the incidence of staphylococcal disease in higher in individuals who are colonized. Rectal colonization by S. aureus isolates, reflecting gastrointestinal (GI) carriage, has recently been recognized as an important reservoir from which person to person transmission occurs. We developed a murine model of S. aureus GI colonization to investigate bacterial factors that promote staphylococcal colonization of the gut. We identified several surface-associated S. aureus antigens that modulate colonization of the GI tract and identified a surface glycopolymer (cell wall teichoic acid) as critical for the early steps in colonization. The failure of the teichoic acid mutant to colonize the GI tract can be attributed to its defects in bacterial adherence and to its enhanced susceptibility to mammalian host defenses unique to the gastrointestinal tract. Efforts to develop antimicrobials that target WTA may lead to an overall reduction in asymptomatic colonization by antibiotic-resistant S. aureus and may impact the incidence of invasive disease.
Collapse
Affiliation(s)
- Yoshiki Misawa
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kathryn A. Kelley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Xiaogang Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Linhui Wang
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wan Beom Park
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Johannes Birtel
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David Saslowsky
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jean C. Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
50
|
Which are important targets in development of S. aureus mastitis vaccine? Res Vet Sci 2015; 100:88-99. [DOI: 10.1016/j.rvsc.2015.03.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/04/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
|