1
|
Unterfrauner I, Bragatto-Hess N, Studhalter T, Farshad M, Uçkay I. General skin and nasal decolonization with octenisan® set before and after elective orthopedic surgery in selected patients at elevated risk for revision surgery and surgical site infections-a single-center, unblinded, superiority, randomized controlled trial (BALGDEC trial). Trials 2024; 25:461. [PMID: 38978089 PMCID: PMC11229206 DOI: 10.1186/s13063-024-08173-y] [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: 03/23/2023] [Accepted: 06/05/2023] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND The preoperative body surface and nasal decolonization may reduce the risk of surgical site infections (SSI) but yields conflicting results in the current orthopedic literature. METHODS We perform a single-center, randomized-controlled, superiority trial in favor of the preoperative decolonization using a commercial product (octenidine® set). We will randomize a total number of 1000 adult elective orthopedic patients with a high risk for SSI and/or wound complications (age ≥ 80 years, chronic immune-suppression, American Society of Anesthesiologists score 3-4 points) between a decolonization (octenisan® wash lotion 1 × per day and octenisan® md nasal gel 2-3 × per day; during 5 days) and no decolonization. Decolonized patients will additionally fill a questionnaire regarding the practical difficulties, the completeness, and the adverse events of decolonization. The primary outcomes are SSI and revision surgeries for postoperative wound problems until 6 weeks postoperatively (or 1 year for surgeries with implants or bone). Secondary outcomes are unplanned revision surgeries for non-infectious problems and all adverse events. With 95% event-free surgeries in the decolonization arm versus 90% in the control arm, we formally need 2 × 474 elective orthopedic surgeries included during 2 years. DISCUSSION In selected adult orthopedic patients with a high risk for SSI, the presurgical decolonization may reduce postoperative wound problems, including SSI. TRIAL REGISTRATION ClinicalTrial.gov NCT05647252. Registered on 9 December 2022. PROTOCOL VERSION 2 (5 December 2022).
Collapse
Affiliation(s)
- Ines Unterfrauner
- Orthopedic Surgery, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Nadja Bragatto-Hess
- Infection Control, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Thorsten Studhalter
- Infection Control, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
| | - Mazda Farshad
- Orthopedic Surgery, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland
- Medical Direction, Balgrist University Hospital, University of Zurich, Forchstrasse 340, Zurich, 8008, Switzerland
| | - Ilker Uçkay
- Infection Control, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland.
- Unit for Clinical and Applied Research, Balgrist University Hospital, University of Zurich, Forchstrasse 340, 8008, Zurich, Switzerland.
| |
Collapse
|
2
|
Abdurrahman G, Pospich R, Steil L, Gesell Salazar M, Izquierdo González JJ, Normann N, Mrochen D, Scharf C, Völker U, Werfel T, Bröker BM, Roesner LM, Gómez-Gascón L. The extracellular serine protease from Staphylococcus epidermidis elicits a type 2-biased immune response in atopic dermatitis patients. Front Immunol 2024; 15:1352704. [PMID: 38895118 PMCID: PMC11183529 DOI: 10.3389/fimmu.2024.1352704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/02/2024] [Indexed: 06/21/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with skin barrier defects and a misdirected type 2 immune response against harmless antigens. The skin microbiome in AD is characterized by a reduction in microbial diversity with a dominance of staphylococci, including Staphylococcus epidermidis (S. epidermidis). Objective To assess whether S. epidermidis antigens play a role in AD, we screened for candidate allergens and studied the T cell and humoral immune response against the extracellular serine protease (Esp). Methods To identify candidate allergens, we analyzed the binding of human serum IgG4, as a surrogate of IgE, to S. epidermidis extracellular proteins using 2-dimensional immunoblotting and mass spectrometry. We then measured serum IgE and IgG1 binding to recombinant Esp by ELISA in healthy and AD individuals. We also stimulated T cells from AD patients and control subjects with Esp and measured the secreted cytokines. Finally, we analyzed the proteolytic activity of Esp against IL-33 and determined the cleavage sites by mass spectrometry. Results We identified Esp as the dominant candidate allergen of S. epidermidis. Esp-specific IgE was present in human serum; AD patients had higher concentrations than controls. T cells reacting to Esp were detectable in both AD patients and healthy controls. The T cell response in healthy adults was characterized by IL-17, IL-22, IFN-γ, and IL-10, whereas the AD patients' T cells lacked IL-17 production and released only low amounts of IL-22, IFN-γ, and IL-10. In contrast, Th2 cytokine release was higher in T cells from AD patients than from healthy controls. Mature Esp cleaved and activated the alarmin IL-33. Conclusion The extracellular serine protease Esp of S. epidermidis can activate IL-33. As an antigen, Esp elicits a type 2-biased antibody and T cell response in AD patients. This suggests that S. epidermidis can aggravate AD through the allergenic properties of Esp.
Collapse
Affiliation(s)
- Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Rebecca Pospich
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Leif Steil
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | | | - Nicole Normann
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Daniel Mrochen
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Scharf
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Lennart M. Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Lidia Gómez-Gascón
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
3
|
Zhu Z, Hu Z, Li S, Fang R, Ono HK, Hu DL. Molecular Characteristics and Pathogenicity of Staphylococcus aureus Exotoxins. Int J Mol Sci 2023; 25:395. [PMID: 38203566 PMCID: PMC10778951 DOI: 10.3390/ijms25010395] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Staphylococcus aureus stands as one of the most pervasive pathogens given its morbidity and mortality worldwide due to its roles as an infectious agent that causes a wide variety of diseases ranging from moderately severe skin infections to fatal pneumonia and sepsis. S. aureus produces a variety of exotoxins that serve as important virulence factors in S. aureus-related infectious diseases and food poisoning in both humans and animals. For example, staphylococcal enterotoxins (SEs) produced by S. aureus induce staphylococcal foodborne poisoning; toxic shock syndrome toxin-1 (TSST-1), as a typical superantigen, induces toxic shock syndrome; hemolysins induce cell damage in erythrocytes and leukocytes; and exfoliative toxin induces staphylococcal skin scalded syndrome. Recently, Panton-Valentine leucocidin, a cytotoxin produced by community-associated methicillin-resistant S. aureus (CA-MRSA), has been reported, and new types of SEs and staphylococcal enterotoxin-like toxins (SEls) were discovered and reported successively. This review addresses the progress of and novel insights into the molecular structure, biological activities, and pathogenicity of both the classic and the newly identified exotoxins produced by S. aureus.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Shaowen Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| |
Collapse
|
4
|
Sirichoat A, Kaewseekhao B, Nithichanon A, Roytrakul S, Faksri K. Proteomic Profiles and Protein Network Analysis of Primary Human Leukocytes Revealed Possible Clearance Biomarkers for Staphylococcus aureus Infection. Curr Microbiol 2023; 80:335. [PMID: 37665379 DOI: 10.1007/s00284-023-03450-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/18/2023] [Indexed: 09/05/2023]
Abstract
Staphylococcus aureus is a serious pathogen that can survive within host cells after a typical course of treatment completion, leading to chronic infection. Knowledge of host proteomic patterns after clearance of this pathogen from cells is limited. Here, we looked for S. aureus clearance biomarkers produced by in vitro-infected leukocytes. Extracellular proteins from primary human leukocytes infected with S. aureus ATCC 25923 were investigated as possible treatment-monitoring clearance biomarkers by applying a proteomics approach combining liquid chromatography with tandem mass spectrometry (LC-MS/MS) and protein interaction network analysis. It was found that the expression patterns of proteins secreted by S. aureus-infected leukocytes differed among stages of infection. Proteomic profiles showed that an ATPase, aminophospholipid transporter-like, Class I, type 8A, member 2 (ATP8A2) was expressed in the clearance stage and was not detected at any earlier stage or in uninfected controls. Protein network analysis showed that TERF2 (telomeric repeat-binding factor 2), ZNF440 (zinc finger protein 440), and PPP1R14A (phosphatase 1 regulatory subunit 14A) were up-regulated, while GLE1, an essential RNA-export mediator, was suppressed in both infection and clearance stages, suggesting their potential roles in S. aureus infection and clearance. These findings are the first to report that the ATP8A2 has potential as a clearance biomarker for S. aureus infection.
Collapse
Affiliation(s)
- Auttawit Sirichoat
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Benjawan Kaewseekhao
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Arnone Nithichanon
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand
| | - Sittiruk Roytrakul
- Genome Institute, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Kiatichai Faksri
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
- Research and Diagnostic Center for Emerging Infectious Diseases (RCEID), Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
5
|
Thiriard A, Meyer B, Eberhardt CS, Loevy N, Grazioli S, Adouan W, Fontannaz P, Marechal F, L’Huillier AG, Siegrist CA, Georges D, Putignano A, Marchant A, Didierlaurent AM, Blanchard-Rohner G. Antibody response in children with multisystem inflammatory syndrome related to COVID-19 (MIS-C) compared to children with uncomplicated COVID-19. Front Immunol 2023; 14:1107156. [PMID: 37006315 PMCID: PMC10050384 DOI: 10.3389/fimmu.2023.1107156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
ObjectivesTo comprehensively analyze the quality of the antibody response between children with Multisystem inflammatory syndrome (MIS-C) and age-matched controls at one month after SARS-CoV-2 exposure, and infected in the same time-period.MethodsSerum from 20 MIS-C children at admission, and 14 control children were analyzed. Antigen specific antibody isotypes and subclasses directed against various antigens of SARS-CoV-2 as well as against human common coronavirus (HCoVs) and commensal or pathogenic microorganisms were assessed by a bead-based multiplexed serological assay and by ELISA. The functionality of these antibodies was also assessed using a plaque reduction neutralization test, a RBD-specific avidity assay, a complement deposition assay and an antibody-dependent neutrophil phagocytosis (ADNP) assay.ResultsChildren with MIS-C developed a stronger IgA antibody response in comparison to children with uncomplicated COVID-19, while IgG and IgM responses are largely similar in both groups. We found a typical class-switched antibody profile with high level of IgG and IgA titers and a measurable low IgM due to relatively recent SARS-CoV-2 infection (one month). SARS-CoV-2-specific IgG antibodies of MIS-C children had higher functional properties (higher neutralization activity, avidity and complement binding) as compared to children with uncomplicated COVID-19. There was no difference in the response to common endemic coronaviruses between both groups. However, MIS-C children had a moderate increase against mucosal commensal and pathogenic strains, reflecting a potential association between a disruption of the mucosal barrier with the disease.ConclusionEven if it is still unclear why some children develop a MIS-C, we show here that MIS-C children produce higher titers of IgA antibodies, and IgG antibodies with higher functionality, which could reflect the local gastro-intestinal mucosal inflammation potentially induced by a sustained SARS-CoV-2 gut infection leading to continuous release of SARS-CoV-2 antigens.
Collapse
Affiliation(s)
- Anaïs Thiriard
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Benjamin Meyer
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Christiane S. Eberhardt
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Natasha Loevy
- Pediatric Platform for Clinical Research, Department of Woman, Child and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Serge Grazioli
- Division of Neonatal and Pediatric Intensive Care, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Wafae Adouan
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Paola Fontannaz
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Fabienne Marechal
- Pediatric Platform for Clinical Research, Department of Woman, Child and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arnaud G. L’Huillier
- Pediatric Infectious Diseases Unit, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Daphnée Georges
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Enzymology and Protein Folding, Centre for Protein Engineering, InBioS, University of Liège, Liège, Belgium
| | - Antonella Putignano
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Arnaud M. Didierlaurent
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Geraldine Blanchard-Rohner
- Centre for Vaccinology, Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Pediatric Immunology and Vaccinology Unit, Children’s Hospital of Geneva, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
- *Correspondence: Geraldine Blanchard-Rohner,
| |
Collapse
|
6
|
Jorde I, Schreiber J, Stegemann-Koniszewski S. The Role of Staphylococcus aureus and Its Toxins in the Pathogenesis of Allergic Asthma. Int J Mol Sci 2022; 24:ijms24010654. [PMID: 36614093 PMCID: PMC9820472 DOI: 10.3390/ijms24010654] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 01/01/2023] Open
Abstract
Bronchial asthma is one of the most common chronic diseases worldwide and affects more than 300 million patients. Allergic asthma affects the majority of asthmatic children as well as approximately 50% of adult asthmatics. It is characterized by a Th2-mediated immune response against aeroallergens. Many aspects of the overall pathophysiology are known, while the underlying mechanisms and predisposing factors remain largely elusive today. Over the last decade, respiratory colonization with Staphylococcus aureus (S. aureus), a Gram-positive facultative bacterial pathogen, came into focus as a risk factor for the development of atopic respiratory diseases. More than 30% of the world’s population is constantly colonized with S. aureus in their nasopharynx. This colonization is mostly asymptomatic, but in immunocompromised patients, it can lead to serious complications including pneumonia, sepsis, or even death. S. aureus is known for its ability to produce a wide range of proteins including toxins, serine-protease-like proteins, and protein A. In this review, we provide an overview of the current knowledge about the pathophysiology of allergic asthma and to what extent it can be affected by different toxins produced by S. aureus. Intensifying this knowledge might lead to new preventive strategies for atopic respiratory diseases.
Collapse
|
7
|
Feng J, Sun D, Wang L, Li X, Guan J, Wei L, Yue D, Wang X, Zhao Y, Yang H, Song W, Wang B. Biochanin A as an α-hemolysin inhibitor for combating methicillin-resistant Staphylococcus aureus infection. World J Microbiol Biotechnol 2021; 38:6. [PMID: 34837116 DOI: 10.1007/s11274-021-03182-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a multidrug-resistant pathogen that poses a significant risk to global health today. In S. aureus, α-hemolysin is an important virulence factor as it contributes to the capacity of the bacteria to infect the host. Here, we showed that biochanin A (bioA), an isoflavone present in red clover, cabbage and alfalfa, effectively inhibited hemolytic activity at a dose as low as 32 μg/mL. Further, western blot and RT-qPCR data showed that bioA reduced the production and expression of MRSA hemolysin in a dose-dependent manner. In addition, when different concentrations of bioA were added to a coculture system of A549 cells and S. aureus, it could significantly decrease cell injury. Importantly, the in vivo study showed that bioA could protect mice from pneumonia caused by a lethal dose of MRSA, as evidenced by improving their survival and reducing the number of bacterial colonies in lung tissues, the secretion of hemolysin into alveolar lavage fluid and the degree of pulmonary edema. In conclusion, biochanin A protected the host from MRSA infection by inhibiting the expression of the hemolysin of MRSA, which may provide experimental evidence for its development to a potential anti-MRSA drug.
Collapse
Affiliation(s)
- Jiaxuan Feng
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Dazhong Sun
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Li Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xueting Li
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Lin Wei
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Donghui Yue
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Xingye Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yicheng Zhao
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Haimiao Yang
- Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Bingmei Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
8
|
Raineri EJM, Altulea D, van Dijl JM. Staphylococcal trafficking and infection - from 'nose to gut' and back. FEMS Microbiol Rev 2021; 46:6321165. [PMID: 34259843 PMCID: PMC8767451 DOI: 10.1093/femsre/fuab041] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/11/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus is an opportunistic human pathogen, which is a leading cause of infections worldwide. The challenge in treating S. aureus infection is linked to the development of multidrug-resistant strains and the mechanisms employed by this pathogen to evade the human immune defenses. In addition, S. aureus can hide asymptomatically in particular ‘protective’ niches of the human body for prolonged periods of time. In the present review, we highlight recently gained insights in the role of the human gut as an endogenous S. aureus reservoir next to the nasopharynx and oral cavity. In addition, we address the contribution of these ecological niches to staphylococcal transmission, including the roles of particular triggers as modulators of the bacterial dissemination. In this context, we present recent advances concerning the interactions between S. aureus and immune cells to understand their possible roles as vehicles of dissemination from the gut to other body sites. Lastly, we discuss the factors that contribute to the switch from colonization to infection. Altogether, we conclude that an important key to uncovering the pathogenesis of S. aureus infection lies hidden in the endogenous staphylococcal reservoirs, the trafficking of this bacterium through the human body and the subsequent immune responses.
Collapse
Affiliation(s)
- Elisa J M Raineri
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dania Altulea
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
9
|
Fernandez J, Sanders H, Henn J, Wilson JM, Malone D, Buoninfante A, Willms M, Chan R, DuMont AL, McLahan C, Grubb K, Romanello A, van den Dobbelsteen G, Torres VJ, Poolman JT. Vaccination with Detoxified Leukocidin AB Reduces Bacterial Load in a Staphylococcus aureus Minipig Deep Surgical Wound Infection Model. J Infect Dis 2021; 225:1460-1470. [PMID: 33895843 PMCID: PMC9016470 DOI: 10.1093/infdis/jiab219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Vaccines against Staphylococcus aureus have eluded researchers for >3 decades while the burden of staphylococcal diseases has increased. Early vaccine attempts mainly used rodents to characterize preclinical efficacy, and all subsequently failed in human clinical efficacy trials. More recently, leukocidin AB (LukAB) has gained interest as a vaccine antigen. We developed a minipig deep surgical wound infection model offering 3 independent efficacy readouts: bacterial load at the superficial and at the deep-seated surgical site, and dissemination of bacteria. Due to similarities with humans, minipigs are an attractive option to study novel vaccine candidates. With this model, we characterized the efficacy of a LukAB toxoid as vaccine candidate. Compared to control animals, a 3-log reduction of bacteria at the deep-seated surgical site was observed in LukAB-treated minipigs and dissemination of bacteria was dramatically reduced. Therefore, LukAB toxoids may be a useful addition to S. aureus vaccines and warrant further study.
Collapse
Affiliation(s)
| | - H Sanders
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - J Henn
- Bacterial Vaccines, Spring House, PA, USA
| | | | - D Malone
- Bacterial Vaccines, Spring House, PA, USA
| | - A Buoninfante
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - M Willms
- Bacterial Vaccines, Spring House, PA, USA
| | - R Chan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - A L DuMont
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - C McLahan
- In Vivo Sciences, Spring House, PA, USA
| | - K Grubb
- Bacterial Vaccines, Spring House, PA, USA
| | | | | | - V J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY, USA
| | - J T Poolman
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| |
Collapse
|
10
|
Mrochen DM, Trübe P, Jorde I, Domanska G, van den Brandt C, Bröker BM. Immune Polarization Potential of the S. aureus Virulence Factors SplB and GlpQ and Modulation by Adjuvants. Front Immunol 2021; 12:642802. [PMID: 33936060 PMCID: PMC8081891 DOI: 10.3389/fimmu.2021.642802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/30/2021] [Indexed: 11/16/2022] Open
Abstract
Protection against Staphylococcus aureus is determined by the polarization of the anti-bacterial immune effector mechanisms. Virulence factors of S. aureus can modulate these and induce differently polarized immune responses in a single individual. We proposed that this may be due to intrinsic properties of the bacterial proteins. To test this idea, we selected two virulence factors, the serine protease-like protein B (SplB) and the glycerophosphoryl diester phosphodiesterase (GlpQ). In humans naturally exposed to S. aureus, SplB induces a type 2-biased adaptive immune response, whereas GlpQ elicits type 1/type 3 immunity. We injected the recombinant bacterial antigens into the peritoneum of S. aureus-naïve C57BL/6N mice and analyzed the immune response. This was skewed by SplB toward a Th2 profile including specific IgE, whereas GlpQ was weakly immunogenic. To elucidate the influence of adjuvants on the proteins’ polarization potential, we studied Montanide ISA 71 VG and Imject™Alum, which promote a Th1 and Th2 response, respectively. Alum strongly increased antibody production to the Th2-polarizing protein SplB, but did not affect the response to GlpQ. Montanide enhanced the antibody production to both S. aureus virulence factors. Montanide also augmented the inflammation in general, whereas Alum had little effect on the cellular immune response. The adjuvants did not override the polarization potential of the S. aureus proteins on the adaptive immune response.
Collapse
Affiliation(s)
- Daniel M Mrochen
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Patricia Trübe
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Ilka Jorde
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Grazyna Domanska
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
11
|
Meyer TC, Michalik S, Holtfreter S, Weiss S, Friedrich N, Völzke H, Kocher T, Kohler C, Schmidt F, Bröker BM, Völker U. A Comprehensive View on the Human Antibody Repertoire Against Staphylococcus aureus Antigens in the General Population. Front Immunol 2021; 12:651619. [PMID: 33777051 PMCID: PMC7987813 DOI: 10.3389/fimmu.2021.651619] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Our goal was to provide a comprehensive overview of the antibody response to Staphylococcus aureus antigens in the general population as a basis for defining disease-specific profiles and diagnostic signatures. We tested the specific IgG and IgA responses to 79 staphylococcal antigens in 996 individuals from the population-based Study of Health in Pomerania. Using a dilution-based multiplex suspension array, we extended the dynamic range of specific antibody detection to seven orders of magnitude, allowing the precise quantification of high and low abundant antibody specificities in the same sample. The observed IgG and IgA antibody responses were highly heterogeneous with differences between individuals as well as between bacterial antigens that spanned several orders of magnitude. Some antigens elicited significantly more IgG than IgA and vice versa. We confirmed a strong influence of colonization on the antibody response and quantified the influence of sex, smoking, age, body mass index, and serum glucose on anti-staphylococcal IgG and IgA. However, all host parameters tested explain only a small part of the extensive variability in individual response to the different antigens of S. aureus.
Collapse
Affiliation(s)
- Tanja C Meyer
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Weiss
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Unit of Periodontology, University Medicine Greifswald, Greifswald, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Frank Schmidt
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,Proteomics Core, Weill Cornell Medicine-Qatar, Qatar Foundation-Education City, Doha, Qatar
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Department Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Jorde I, Hildebrand CB, Kershaw O, Lücke E, Stegemann-Koniszewski S, Schreiber J. Modulation of Allergic Sensitization and Allergic Inflammation by Staphylococcus aureus Enterotoxin B in an Ovalbumin Mouse Model. Front Immunol 2020; 11:592186. [PMID: 33193436 PMCID: PMC7649385 DOI: 10.3389/fimmu.2020.592186] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/05/2020] [Indexed: 01/02/2023] Open
Abstract
The superantigen Staphylococcus aureus (S. aureus) enterotoxin B (SEB) has been proposed a central player in the associations between S. aureus nasal colonization and the development of allergic asthma. Previously, SEB has been shown to aggravate allergic sensitization and allergic airway inflammation (AAI) in experimental mouse models. Aiming at understanding the underlying immunological mechanisms, we tested the hypothesis that intranasal (i.n.) SEB-treatment divergently modulates AAI depending on the timing and intensity of the SEB-encounter. In an ovalbumin-mediated mouse model of AAI, we treated mice i.n. with 50 ng or 500 ng SEB either together with the allergic challenge or prior to the peripheral sensitization. We observed SEB to affect different hallmark parameters of AAI depending on the timing and the dose of treatment. SEB administered i.n. together with the allergic challenge significantly modulated respiratory leukocyte accumulation, intensified lymphocyte activation and, at the higher dose, induced a strong type-1 and pro-inflammatory cytokine response and alleviated airway hyperreactivity in AAI. SEB administered i.n. prior to the allergic sensitization at the lower dose significantly boosted the specific IgE response while administration of the higher dose led to a significantly reduced recruitment of immune cells, including eosinophils, to the respiratory tract and to a significantly dampened Th-2 cytokine response without inducing a Th-1 or pro-inflammatory response. We show a remarkably versatile potential for SEB to either aggravate or alleviate different parameters of allergic sensitization and AAI. Our study thereby not only highlights the complexity of the associations between S. aureus and allergic asthma but possibly even points at prophylactic and therapeutic pathways.
Collapse
Affiliation(s)
- Ilka Jorde
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Christina B Hildebrand
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Olivia Kershaw
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Eva Lücke
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Sabine Stegemann-Koniszewski
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens Schreiber
- Experimental Pneumology, Department of Pneumology, University Hospital Magdeburg/Medical Faculty, Health Campus Immunology, Infectiology and Inflammation (GC-I³), Otto-von-Guericke-University, Magdeburg, Germany
| |
Collapse
|
13
|
Israelsson E, Chaussabel D, Fischer RSB, Moore HC, Robinson DA, Dunkle JW, Essigmann HT, Record S, Brown EL. Characterization of peripheral blood mononuclear cells gene expression profiles of pediatric Staphylococcus aureus persistent and non-carriers using a targeted assay. Microbes Infect 2020; 22:540-549. [PMID: 32758644 DOI: 10.1016/j.micinf.2020.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/26/2020] [Accepted: 07/25/2020] [Indexed: 11/28/2022]
Abstract
Defects in innate immunity affect many different physiologic systems and several studies of patients with primary immunodeficiency disorders demonstrated the importance of innate immune system components in disease prevention or colonization of bacterial pathogens. To assess the role of the innate immune system on nasal colonization with Staphylococcus aureus, innate immune responses in pediatric S. aureus nasal persistent carriers (n = 14) and non-carriers (n = 15) were profiled by analyzing co-clustered gene sets (modules). We stimulated previously frozen peripheral blood mononuclear cells (PBMCs) from these subjects with i) a panel of TLR ligands, ii) live S. aureus (either a mixture of strains or stimulation with respective carriage isolates), or iii) heat-killed S. aureus. We found no difference in responses between carriers and non-carriers when PBMCs were stimulated with a panel of TLR ligands. However, PBMC gene expression profiles differed between persistent and non-S. aureus carriers following stimulation with either live or dead S. aureus. These observations suggest that individuals susceptible to persistent carriage with S. aureus may possess differences in their live/dead bacteria recognition pathway and that innate pathway signaling is different between persistent and non-carriers of S. aureus.
Collapse
Affiliation(s)
- Elisabeth Israelsson
- Department of Systems Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Damien Chaussabel
- Systems Biology Department, Sidra Medical and Research Center, Doha, Qatar
| | - Rebecca S B Fischer
- Texas A&M Health Science Center School of Public Health, Department of Epidemiology and Biostatistics, College Station, TX, USA
| | - Heather C Moore
- Baylor College of Medicine, Complex Care Clinic, Texas Children's Hospital, Houston, TX, USA
| | - D Ashley Robinson
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jesse W Dunkle
- Icahn School of Medicine, Mount Sinai Hospital, Institute for Advanced Medicine, New York, NY, USA
| | - Heather T Essigmann
- Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center, Houston, TX, USA
| | - Sharron Record
- Texas Children's Hospital, Department of Pediatrics, TX, USA
| | - Eric L Brown
- Division of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
14
|
Dreisbach A, Wang M, van der Kooi-Pol MM, Reilman E, Koedijk DGAM, Mars RAT, Duipmans J, Jonkman M, Benschop JJ, Bonarius HPJ, Groen H, Hecker M, Otto A, Bäsell K, Bernhardt J, Back JW, Becher D, Buist G, van Dijl JM. Tryptic Shaving of Staphylococcus aureus Unveils Immunodominant Epitopes on the Bacterial Cell Surface. J Proteome Res 2020; 19:2997-3010. [DOI: 10.1021/acs.jproteome.0c00043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Annette Dreisbach
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Magdalena M. van der Kooi-Pol
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Ewoud Reilman
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Dennis G. A. M. Koedijk
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Ruben A. T. Mars
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - José Duipmans
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Marcel Jonkman
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Joris J. Benschop
- Pepscan Therapeutics BV, P. O. Box 2098, 8203 AB Lelystad, the Netherlands
| | | | - Herman Groen
- IQ Therapeutics, Rozenburglaan 13a, 9727 DL Groningen, the Netherlands
| | - Michael Hecker
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Andreas Otto
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Katrin Bäsell
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Jörg Bernhardt
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Jaap Willem Back
- Pepscan Therapeutics BV, P. O. Box 2098, 8203 AB Lelystad, the Netherlands
| | - Dörte Becher
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
15
|
Early-Stage Staphylococcus aureus Bloodstream Infection Causes Changes in the Concentrations of Lipoproteins and Acute-Phase Proteins and Is Associated with Low Antibody Titers against Bacterial Virulence Factors. mSystems 2020; 5:5/1/e00632-19. [PMID: 31964768 PMCID: PMC6977072 DOI: 10.1128/msystems.00632-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
S. aureus sepsis has a high complication and mortality rate. Given the limited therapeutic possibilities, effective prevention strategies, e.g., a vaccine, or the early identification of high-risk patients would be important but are not available. Our study showed an acute-phase response in patients with S. aureus bloodstream infection and evidence that lipoproteins are downregulated in plasma. Using immunoproteomics, stratification of patients appears to be achievable, since at the early stages of systemic S. aureus infection patients had low preexisting anti-S. aureus antibody levels. This strengthens the notion that a robust immune memory for S. aureus protects against infections with the pathogen. Systemic and quantitative investigations of human plasma proteins (proteomics) and Staphylococcus aureus-specific antibodies (immunoproteomics) provide complementary information and hold promise for the discovery of biomarkers in Staphylococcus aureus bloodstream infection (SABSI). Usually, data-dependent acquisition (DDA) is used for proteome analysis of serum or plasma, but data-independent acquisition (DIA) is more comprehensive and reproducible. In this prospective cohort study, we aimed to identify biomarkers associated with the early stages of SABSI using a serum DIA proteomic and immunoproteomic approach. Sera from 49 SABSI patients and 43 noninfected controls were analyzed. In total, 608 human serum proteins were identified with DIA. A total of 386 proteins could be quantified, of which 9 proteins, mainly belonging to acute-phase proteins, were significantly increased, while 7 high-density lipoproteins were lower in SABSI. In SABSI, total anti-S. aureus serum IgG was reduced compared with controls as shown by immunoproteomic quantification of IgG binding to 143 S. aureus antigens. IgG binding to 48 of these anti-S. aureus proteins was significantly lower in SABSI, while anti-Ecb IgG was the only one increased in SABSI. Serum IgG binding to autoinducing peptide MsrB, FadB, EsxA, Pbp2, FadB, SspB, or SodA was very low in SABSI. This marker panel discriminated early SABSI from controls with 95% sensitivity and 100% specificity according to random forest prediction. This holds promise for patient stratification according to their risk of S. aureus infection, underlines the protective function of the adaptive immune system, and encourages further efforts in the development of a vaccine against S. aureus. IMPORTANCES. aureus sepsis has a high complication and mortality rate. Given the limited therapeutic possibilities, effective prevention strategies, e.g., a vaccine, or the early identification of high-risk patients would be important but are not available. Our study showed an acute-phase response in patients with S. aureus bloodstream infection and evidence that lipoproteins are downregulated in plasma. Using immunoproteomics, stratification of patients appears to be achievable, since at the early stages of systemic S. aureus infection patients had low preexisting anti-S. aureus antibody levels. This strengthens the notion that a robust immune memory for S. aureus protects against infections with the pathogen.
Collapse
|
16
|
Flaxman A, Yamaguchi Y, van Diemen PM, Rollier C, Allen E, Elshina E, Wyllie DH. Heterogeneous early immune responses to the S. aureus EapH2 antigen induced by gastrointestinal tract colonisation impact the response to subsequent vaccination. Vaccine 2019; 37:494-501. [PMID: 30503080 DOI: 10.1016/j.vaccine.2018.11.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 11/19/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022]
Abstract
INTRODUCTION S. aureus is a pathogen to which individuals are exposed shortly after birth, with immune responses to S. aureus increasing during childhood. There is marked heterogeneity between the anti- S. aureus immune responses of different humans, the basis of which is not fully understood. METHODS To investigate development of anti-S. aureus immune responses, we studied S. aureus colonised mice under controlled conditions. Mice were either acquired colonised from breeding colonies, or experimentally colonised by exposure to a cage environment which had been sprayed with a S. aureus suspension. Colonisation was monitored by sequential stool sampling, and immunoglobulin levels against both whole fixed S. aureus and individual S. aureus antigens quantified. The immunological impact of colonisation on subsequent vaccination was investigated. RESULTS Colonised BALB/c and BL/6 mice develop serum anti- S. aureus cell surface IgG1 antibodies. Responses were proportional to the cumulative S. aureus bioburden in the mice, and were higher in BALB/c mice, which have higher colonisation levels, than in C57BL/6 animals. We observed marked variation in the induction of anti-cell surface antibodies, even in genetically identical mice experimentally colonised with the same S. aureus clone. Heterogeneity was also evident when monitoring immune responses to the secreted S. aureus protein EapH2. Approximately 50% of colonised mice developed anti-EapH2 responses (responders); in other mice, responses were not significantly different to those in uncolonised mice (non-responders). Following vaccination with a replication deficient adenovirus expressing EapH2, less anti-EapH2 antibody was generated in non-responder than responder animals. CONCLUSIONS In genetically identical mice, S. aureus colonisation results in all-or-nothing antibody responses against some antigens, including EapH2. For antigens involved in colonisation success by microbes, apparently stochastic early immune responses may impact both vaccine responses and the establishment of an animal-specific microbiome.
Collapse
Affiliation(s)
- Amy Flaxman
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Yuko Yamaguchi
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Pauline M van Diemen
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Christine Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Biomedical Research Centre, CCVTM, Churchill Drive, UK
| | - Elizabeth Allen
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - Elizaveta Elshina
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK
| | - David H Wyllie
- Jenner Institute, University of Oxford, Centre for Cellular and Molecular Physiology, Oxford, UK.
| |
Collapse
|
17
|
Buchan KD, Foster SJ, Renshaw SA. Staphylococcus aureus: setting its sights on the human innate immune system. MICROBIOLOGY-SGM 2019; 165:367-385. [PMID: 30625113 DOI: 10.1099/mic.0.000759] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Staphylococcus aureus has colonized humans for at least 10 000 years, and today inhabits roughly a third of the population. In addition, S. aureus is a major pathogen that is responsible for a significant disease burden, ranging in severity from mild skin and soft-tissue infections to life-threatening endocarditis and necrotizing pneumonia, with treatment often hampered by resistance to commonly available antibiotics. Underpinning its versatility as a pathogen is its ability to evade the innate immune system. S. aureus specifically targets innate immunity to establish and sustain infection, utilizing a large repertoire of virulence factors to do so. Using these factors, S. aureus can resist phagosomal killing, impair complement activity, disrupt cytokine signalling and target phagocytes directly using proteolytic enzymes and cytolytic toxins. Although most of these virulence factors are well characterized, their importance during infection is less clear, as many display species-specific activity against humans or against animal hosts, including cows, horses and chickens. Several staphylococcal virulence factors display species specificity for components of the human innate immune system, with as few as two amino acid changes reducing binding affinity by as much as 100-fold. This represents a major issue for studying their roles during infection, which cannot be examined without the use of humanized infection models. This review summarizes the major factors S. aureus uses to impair the innate immune system, and provides an in-depth look into the host specificity of S. aureus and how this problem is being approached.
Collapse
Affiliation(s)
- Kyle D Buchan
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Simon J Foster
- 2Department of Molecular Biology and Biotechnology, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| | - Stephen A Renshaw
- 1The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Western Bank, Sheffield S10 2TN, UK
| |
Collapse
|
18
|
Immunogenicity and protective efficacy of recombinant alkaline shock protein 23 from Staphylococcus aureus in a murine model. Cent Eur J Immunol 2018; 43:371-377. [PMID: 30799984 PMCID: PMC6384426 DOI: 10.5114/ceji.2018.81348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/08/2016] [Indexed: 11/29/2022] Open
Abstract
The diversity and severity of infections caused and the rapid emergence of antibiotic resistance necessitates the development of a vaccine against Staphylococcus aureus. None of the antigens tried as vaccine candidates so far has been translated into a clinically viable vaccine. Recent research data suggest that antigens with the potential to activate cell mediated immunity along with humoral immunity would be the key to the development of a vaccine. Alkaline shock protein 23, a membrane-anchored protein involved in the stress response, has been identified as a CD4+ T cell antigen from S. aureus. In the present study, we report the evaluation of immunogenicity and protective efficacy of a recombinant alkaline shock protein 23 from S. aureus in mouse models. The gene coding for the protein was cloned and expressed in Escherichia coli, purified using immobilized metal iron affinity chromatography, sequence-confirmed using mass spectrometry and intraperitoneally administered to BALB/c mice. Serum titers of IgG, IgG1, and IgG2a in response to the protein were measured on post-immunization days 21, 35 and 42 using indirect ELISA and compared to control mice injected with PBS. Our results showed that the protein induced significantly higher (p < 0.01) antibody responses in immunized mice compared to the control mice. The mean serum antibody titers of IgG, IgG1 and IgG2a three weeks after the last immunization were found to be 25600, 25600 and 12800 respectively. Moreover, we found that immunization with Asp23 protected mice from a lethal dose of S. aureus strain USA300.
Collapse
|
19
|
Jorge AM, Schneider J, Unsleber S, Xia G, Mayer C, Peschel A. Staphylococcus aureus counters phosphate limitation by scavenging wall teichoic acids from other staphylococci via the teichoicase GlpQ. J Biol Chem 2018; 293:14916-14924. [PMID: 30068554 DOI: 10.1074/jbc.ra118.004584] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/26/2018] [Indexed: 11/06/2022] Open
Abstract
Staphylococcus aureus is part of the human nasal and skin microbiomes along with other bacterial commensals and opportunistic pathogens. Nutrients are scarce in these habitats, demanding effective nutrient acquisition and competition strategies. How S. aureus copes with phosphate limitation is still unknown. Wall teichoic acid (WTA), a polyol-phosphate polymer, could serve as a phosphate source, but whether S. aureus can utilize it during phosphate starvation remains unknown. S. aureus secretes a glycerophosphodiesterase, GlpQ, that cleaves a broad variety of glycerol-3-phosphate (GroP) headgroups of deacylated phospholipids, providing this bacterium with GroP as a carbon and phosphate source. Here we demonstrate that GlpQ can also use glycerophosphoglycerol derived from GroP WTA from coagulase-negative Staphylococcus lugdunensis, Staphylococcus capitis, and Staphylococcus epidermidis, which share the nasal and skin habitats with S. aureus Therefore, S. aureus GlpQ is the first reported WTA-hydrolyzing enzyme, or teichoicase, from Staphylococcus Activity assays revealed that unmodified WTA is the preferred GlpQ substrate, and the results from MS analysis suggested that GlpQ uses an exolytic cleavage mechanism. Importantly, GlpQ did not hydrolyze the ribitol-5-phosphate WTA polymers of S. aureus, underscoring its role in interspecies competition rather than in S. aureus cell wall homeostasis or WTA recycling. glpQ expression was strongly up-regulated under phosphate limitation, and GlpQ allowed S. aureus to grow in the presence of GroP WTA as the sole phosphate source. Our study reveals a novel and unprecedented strategy of S. aureus for acquiring phosphate from bacterial competitors under the phosphate-limiting conditions in the nasal or skin environments.
Collapse
Affiliation(s)
- Ana Maria Jorge
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany, .,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Jonathan Schneider
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Sandra Unsleber
- the Microbiology/Biotechnology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
| | - Guoqing Xia
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| | - Christoph Mayer
- the Microbiology/Biotechnology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany
| | - Andreas Peschel
- From the Infection Biology Department, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, 72076 Tübingen, Germany.,the German Center for Infection Research, Partner Site Tübingen, University of Tübingen, 72076 Tübingen, Germany, and
| |
Collapse
|
20
|
Staphylococcus aureus Toxins and Their Molecular Activity in Infectious Diseases. Toxins (Basel) 2018; 10:toxins10060252. [PMID: 29921792 PMCID: PMC6024779 DOI: 10.3390/toxins10060252] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/14/2018] [Accepted: 06/15/2018] [Indexed: 12/04/2022] Open
Abstract
Staphylococcus aureus is a microorganism resident in the skin and nasal membranes with a dreadful pathogenic potential to cause a variety of community and hospital-acquired infections. The frequency of these infections is increasing and their treatment is becoming more difficult. The ability of S. aureus to form biofilms and the emergence of multidrug-resistant strains are the main reasons determining the challenge in dealing with these infections. S. aureus' infectious capacity and its success as a pathogen is related to the expression of virulence factors, among which the production of a wide variety of toxins is highlighted. For this reason, a better understanding of S. aureus toxins is needed to enable the development of new strategies to reduce their production and consequently improve therapeutic approaches. This review focuses on understanding the toxin-based pathogenesis of S. aureus and their role on infectious diseases.
Collapse
|
21
|
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes superficial and invasive infections in the hospital and community. High mortality from infection emphasizes the need for improved methods for prevention and treatment. Although S. aureus possesses an arsenal of virulence factors that contribute to evasion of host defenses, few studies have examined long-term humoral and B-cell responses. Adults with acute-phase skin and soft tissue infections were recruited; blood samples were obtained; and S. aureus isolates, including methicillin-resistant strains, were subjected to genomic sequence analysis. In comparisons of acute-phase sera with convalescent-phase sera, a minority (37.5%) of patients displayed 2-fold or greater increases in antibody titers against three or more S. aureus antigens, whereas nearly half exhibited no changes, despite the presence of toxin genes in most infecting strains. Moreover, enhanced antibody responses waned over time, which could reflect a defect in B-cell memory or long-lived plasma cells. However, memory B cells reactive with a range of S. aureus antigens were prevalent at both acute-phase and convalescent-phase time points. While some memory B cells exhibited toxin-specific binding, those cross-reactive with structurally related leucocidin subunits were dominant across patients, suggesting the targeting of conserved epitopes. Memory B-cell reactivity correlated with serum antibody levels for selected S. aureus exotoxins, suggesting a relationship between the cellular and humoral compartments. Overall, although there was no global defect in the representation of anti-S. aureus memory B cells, there was evidence of restrictions in the range of epitopes recognized, which may suggest potential therapeutic approaches for augmenting host defenses. The contribution of B-cell memory and long-term antibody responses to host defenses against S. aureus exotoxins remains poorly understood. Our studies confirmed that infection did not commonly lead to enhanced long-term humoral responses. Whereas circulating memory B cells against S. aureus secreted exotoxins were prevalent, they were dominated by cross-reactivity with structurally related leucocidin subunits, consistent with recognition of conserved epitopes. These findings also provide the first evidence of a relationship between the reactivity of antistaphylococcal circulating memory B cells and serum antibody levels. In general, infection was not associated with a global defect in B-cell memory for S. aureus secreted factors, and responses were highly dominated by cross-reactivity to structurally related exotoxins, which arguably may alone be suboptimal in providing host defenses. Our studies illuminate aspects of the S. aureus-host relationship that may better inform strategies for the development of an effective protective vaccine.
Collapse
|
22
|
Human Immunoglobulin G Cannot Inhibit Fibrinogen Binding by the Genetically Diverse A Domain of Staphylococcus aureus Fibronectin-Binding Protein A. mSphere 2018; 3:mSphere00590-17. [PMID: 29564394 PMCID: PMC5853482 DOI: 10.1128/msphere.00590-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/14/2018] [Indexed: 11/20/2022] Open
Abstract
The fibronectin-binding protein A (FnBPA) is a cell surface-associated protein of Staphylococcus aureus which mediates adherence to the host extracellular matrix and is important for bacterial virulence. Previously, substantial sequence diversity was found among strains in the fibrinogen-binding A domain of this protein, and 7 different isotypes were described. The effect of this sequence diversity on the human antibody response, in terms of both antibody production and antibody function, remains unclear. In this study, we identify five different FnBPA A domain isotypes based on the sequence results of 22 clinical S. aureus isolates, obtained from the same number of patients suffering from bacteremia. Using a bead-based Luminex technique, we measure the patients' total immunoglobulin G (IgG) against the 7 FnBPA isotypes at the onset and during the time course of bacteremia (median of 10 serum samples per patient over a median of 35 days). A significant increase in IgG against the FnBPA A domain, including the isotype carried by the infecting strain, is observed in only three out of 22 patients (14%) after the onset of bacteremia. Using a Luminex-based FnBPA-fibrinogen-binding assay, we find that preincubation of recombinant FnBPA isotypes with IgG from diverse patients does not interfere with binding to fibrinogen. This observation is confirmed using an alternative Luminex-based assay and enzyme-linked immunosorbent assay (ELISA). IMPORTANCE Despite the many in vitro and murine in vivo studies involving FnBPA, the actual presence of this virulence factor during human infection is less well established. Furthermore, it is currently unknown to what extent sequence variation in such a virulence factor affects the human antibody response and the ability of antibodies to interfere with FnBPA function. This study sheds new light on these issues. First, the uniform presence of a patient's IgG against FnBPA indicates the presence and importance of this virulence factor during S. aureus pathogenesis. Second, the absence of an increase in antibody production in most patients following bacteremia indicates the complexity of S. aureus-host interactions, possibly involving immune evasion or lack of expression of FnBPA during invasive infection. Finally, we provide new insights into the inability of human antibodies to interfere with FnBPA-fibrinogen binding. These observations should be taken into account during the development of novel vaccination approaches.
Collapse
|
23
|
Romero Pastrana F, Neef J, Koedijk DGAM, de Graaf D, Duipmans J, Jonkman MF, Engelmann S, van Dijl JM, Buist G. Human antibody responses against non-covalently cell wall-bound Staphylococcus aureus proteins. Sci Rep 2018; 8:3234. [PMID: 29459694 PMCID: PMC5818649 DOI: 10.1038/s41598-018-21724-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 02/06/2018] [Indexed: 12/28/2022] Open
Abstract
Human antibody responses to pathogens, like Staphylococcus aureus, are important indicators for in vivo expression and immunogenicity of particular bacterial components. Accordingly, comparing the antibody responses to S. aureus components may serve to predict their potential applicability as antigens for vaccination. The present study was aimed at assessing immunoglobulin G (IgG) responses elicited by non-covalently cell surface-bound proteins of S. aureus, which thus far received relatively little attention. To this end, we applied plasma samples from patients with the genetic blistering disease epidermolysis bullosa (EB) and healthy S. aureus carriers. Of note, wounds of EB patients are highly colonized with S. aureus and accordingly these patients are more seriously exposed to staphylococcal antigens than healthy individuals. Ten non-covalently cell surface-bound proteins of S. aureus, namely Atl, Eap, Efb, EMP, IsaA, LukG, LukH, SA0710, Sle1 and SsaA2, were selected by bioinformatics and biochemical approaches. These antigens were recombinantly expressed, purified and tested for specific IgG responses using human plasma. We show that high exposure of EB patients to S. aureus is mirrored by elevated IgG levels against all tested non-covalently cell wall-bound staphylococcal antigens. This implies that these S. aureus cell surface proteins are prime targets for the human immune system.
Collapse
Affiliation(s)
- Francisco Romero Pastrana
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - Jolanda Neef
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - Dennis G A M Koedijk
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - Douwe de Graaf
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - José Duipmans
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - Marcel F Jonkman
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| | - Susanne Engelmann
- Institute of Microbiology, Technical University Braunschweig, Inhoffenstrasse 7, D-38124, Braunschweig, Germany.,Helmholtz Institute for Infection Research, Microbial Proteomics, Inhoffenstrasse 7, D-38124, Braunschweig, Germany
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands.
| | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30001, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
24
|
Sampedro GR, Bubeck Wardenburg J. Staphylococcus aureus in the Intensive Care Unit: Are These Golden Grapes Ripe for a New Approach? J Infect Dis 2017; 215:S64-S70. [PMID: 28003353 DOI: 10.1093/infdis/jiw581] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Staphylococcus aureus is the leading cause of infection in the setting of critical illness and injury. This pathogen causes life-threatening infection in otherwise healthy individuals and also complicates the clinical course of patients requiring intensive care as a result of their primary medical or surgical disease processes. S. aureus infection in the intensive care unit (ICU) most commonly manifests as sepsis, ventilator-associated pneumonia, and infection of surgical sites and indwelling medical devices. With the epidemic spread of methicillin-resistant S. aureus, many cases of staphylococcal infection in the ICU are now classified as drug resistant, prompting hospital-based screening for methicillin-resistant S. aureus and implementation of both isolation practices and decolonization strategies in ICU patients. The genetic adaptability of S. aureus, heterogeneity of disease presentation, clinical course, and outcome between individual S. aureus-infected ICU patients remains enigmatic, suggesting a need to define disease classification subtypes that inform disease progression and therapy. We propose that S. aureus infection in the ICU now presents a unique opportunity for individualized risk stratification coupled with the investigation of novel approaches to mitigate disease. Given our increasing knowledge of the molecular pathogenesis of S. aureus disease, we suggest that the application of molecular pathological epidemiology to S. aureus infection can usher in a new era of highly focused personalized therapy that may be particularly beneficial in the setting of critical illness and injury.
Collapse
Affiliation(s)
- Georgia R Sampedro
- Departments of 1 Microbiology and.,Pediatrics, University of Chicago, Illinois
| | | |
Collapse
|
25
|
Bouvet C, Gjoni S, Zenelaj B, Lipsky BA, Hakko E, Uçkay I. Staphylococcus aureus soft tissue infection may increase the risk of subsequent staphylococcal soft tissue infections. Int J Infect Dis 2017; 60:44-48. [PMID: 28487239 DOI: 10.1016/j.ijid.2017.05.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/03/2017] [Accepted: 05/02/2017] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Staphylococcus aureus is the most common cause of soft tissue infections. It is unknown, however, if a patient who has had such an infection is at greater risk for future soft tissue infections with S. aureus. METHODS We conducted an epidemiological survey of adult patients hospitalized in the only public hospital in Geneva for treatment (usually combined surgical and medical) of a soft tissue infection caused by S. aureus. By reviewing nursing and medical records from the emergency department and hospital wards, we assessed whether or not they developed any other soft tissue infections (excluding a recurrence) after or before the index one. RESULTS Among 1023 index episodes of soft tissue infections, 670 (65%) were caused by S. aureus, of which 47 were caused by methicillin-resistant strains (30 healthcare-associated and 17 community-acquired). The patients' median age was 51 years and 334 (34%) were immune-compromised. The median time span between the patient's first and last consultation (for any reason) in our hospital was 21.4 years (interquartile range, 10-30 years). In addition to their index infection, 124 patients (12%) developed a new nosocomial or community-acquired soft tissue infection. Among the index cases with an S. aureus infection, 92 (14%) had another soft tissue infection, compared to 32 (9%) who had a non-staphylococcal index infection (Pearson-χ2-test; p=0.03). Similarly, patients with an index S. aureus infection, compared to those with a non-S. aureus infection, had a higher rate of another soft tissue infection caused by S. aureus (χ2-test; p<0.01). In multivariate analysis, an index infection due to S. aureus shows a high association to further S. aureus soft tissue infections (logistic regression; odds ratio 2.5, 95% confidence interval 1.4-4.6). CONCLUSION Among adult patients hospitalised for a soft tissue infection, those infected with S. aureus (compared with other pathogens) may be at higher risk of a subsequent soft tissue infection, particularly with S. aureus.
Collapse
Affiliation(s)
- Cindy Bouvet
- Orthopaedic Surgery Service, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland
| | - Shpresa Gjoni
- Division of General Medical Rehabilitation, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland
| | - Besa Zenelaj
- Orthopaedic Surgery Service, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland
| | - Benjamin A Lipsky
- Service of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland; Division of Medical Sciences, Green Templeton College, University of Oxford, Oxford, United Kingdom
| | - Elif Hakko
- Anadolu Sağlık Hospital, Istanbul, Turkey
| | - Ilker Uçkay
- Orthopaedic Surgery Service, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland; Service of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland; Infection Control Program, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Switzerland.
| |
Collapse
|
26
|
Partial Immunoblotting of 2D-Gels: A Novel Method to Identify Post-Translationally Modified Proteins Exemplified for the Myelin Acetylome. Proteomes 2017; 5:proteomes5010003. [PMID: 28248254 PMCID: PMC5372224 DOI: 10.3390/proteomes5010003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 11/17/2022] Open
Abstract
Post-translational modifications (PTMs) play a key role in regulating protein function, yet their identification is technically demanding. Here, we present a straightforward workflow to systematically identify post-translationally modified proteins based on two-dimensional gel electrophoresis. Upon colloidal Coomassie staining the proteins are partially transferred, and the investigated PTMs are immunodetected. This strategy allows tracking back the immunopositive antigens to the corresponding spots on the original gel, from which they are excised and mass spectrometrically identified. Candidate proteins are validated on the same membrane by immunodetection using a second fluorescence channel. We exemplify the power of partial immunoblotting with the identification of lysine-acetylated proteins in myelin, the oligodendroglial membrane that insulates neuronal axons. The excellent consistency of the detected fluorescence signals at all levels allows the differential comparison of PTMs across multiple conditions. Beyond PTM screening, our multi-level workflow can be readily adapted to clinical applications such as identifying auto-immune antigens or host-pathogen interactions.
Collapse
|
27
|
Jorge AM, Schneider J, Unsleber S, Göhring N, Mayer C, Peschel A. Utilization of glycerophosphodiesters by Staphylococcus aureus. Mol Microbiol 2016; 103:229-241. [PMID: 27726204 DOI: 10.1111/mmi.13552] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2016] [Indexed: 01/02/2023]
Abstract
The facultative pathogen Staphylococcus aureus colonizes the human anterior nares and causes infections of various organ systems. Which carbon, energy, and phosphate sources can be utilized by S. aureus in nutrient-poor habitats has remained largely unknown. We describe that S. aureus secretes a glycerophosphodiesterase (glycerophosphodiester phosphodiesterase, EC 3.1.4.46), GlpQ, degrading the glycerophosphodiester (GPD) head groups of phospholipids such as human phosphatidylcholine (GroPC). Deletion of glpQ completely abolished the GroPC-degrading activity in S. aureus culture supernatants. GroPC has been detected in human tissues and body fluids probably as a result of phospholipid remodelling and degradation. Notably, GroPC promoted S. aureus growth under carbon- and phosphate-limiting conditions in a GlpQ-dependent manner indicating that GlpQ permits S. aureus to utilize GPD-derived glycerol-3-phosphate as a carbon and phosphate sources. Thus, S. aureus can use a broader spectrum of nutrients than previously thought which underscores its capacity to adapt to the highly variable and nutrient-poor surroundings.
Collapse
Affiliation(s)
- Ana Maria Jorge
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Jonathan Schneider
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Sandra Unsleber
- Interfaculty Institute of Microbiology and Infection Medicine, Microbiology/Biotechnology Department, University of Tübingen, Germany
| | - Nadine Göhring
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| | - Christoph Mayer
- Interfaculty Institute of Microbiology and Infection Medicine, Microbiology/Biotechnology Department, University of Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Infection Biology Department, University of Tübingen, Germany
| |
Collapse
|
28
|
Stentzel S, Gläser R, Bröker BM. Elucidating the anti-Staphylococcus aureusantibody response by immunoproteomics. Proteomics Clin Appl 2016; 10:1011-1019. [DOI: 10.1002/prca.201600050] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/20/2016] [Accepted: 08/08/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| | - Regine Gläser
- Department of Dermatology; University Hospital Schleswig-Holstein; Kiel Germany
| | - Barbara M. Bröker
- Department of Immunology; University Medicine Greifswald; Greifswald Germany
| |
Collapse
|
29
|
Vu CH, Kolata J, Stentzel S, Beyer A, Gesell Salazar M, Steil L, Pané-Farré J, Rühmling V, Engelmann S, Götz F, van Dijl JM, Hecker M, Mäder U, Schmidt F, Völker U, Bröker BM. Adaptive immune response to lipoproteins of Staphylococcus aureus in healthy subjects. Proteomics 2016; 16:2667-2677. [PMID: 27324828 PMCID: PMC5096053 DOI: 10.1002/pmic.201600151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/31/2016] [Accepted: 06/16/2016] [Indexed: 01/03/2023]
Abstract
Staphylococcus aureus is a frequent commensal but also a dangerous pathogen, causing many forms of infection ranging from mild to life‐threatening conditions. Among its virulence factors are lipoproteins, which are anchored in the bacterial cell membrane. Lipoproteins perform various functions in colonization, immune evasion, and immunomodulation. These proteins are potent activators of innate immune receptors termed Toll‐like receptors 2 and 6. This study addressed the specific B‐cell and T‐cell responses directed to lipoproteins in human S. aureus carriers and non‐carriers. 2D immune proteomics and ELISA approaches revealed that titers of antibodies (IgG) binding to S. aureus lipoproteins were very low. Proliferation assays and cytokine profiling data showed only subtle responses of T cells; some lipoproteins did not elicit proliferation. Hence, the robust activation of the innate immune system by S. aureus lipoproteins does not translate into a strong adaptive immune response. Reasons for this may include inaccessibility of lipoproteins for B cells as well as ineffective processing and presentation of the antigens to T cells.
Collapse
Affiliation(s)
- Chi Hai Vu
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Julia Kolata
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.,Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sebastian Stentzel
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Anica Beyer
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Leif Steil
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Jan Pané-Farré
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Vanessa Rühmling
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Susanne Engelmann
- Institute of Microbiology, University of Greifswald, Greifswald, Germany.,Helmholtz Center for Infection Research, Microbial Proteomics, Braunschweig, Germany.,Institute for Microbiology, University of Braunschweig, Braunschweig, Germany
| | - Friedrich Götz
- Department of Microbial Genetics, University of Tübingen, Tübingen, Germany
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Michael Hecker
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Ulrike Mäder
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Frank Schmidt
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
30
|
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
|
31
|
Stentzel S, Teufelberger A, Nordengrün M, Kolata J, Schmidt F, van Crombruggen K, Michalik S, Kumpfmüller J, Tischer S, Schweder T, Hecker M, Engelmann S, Völker U, Krysko O, Bachert C, Bröker BM. Staphylococcal serine protease-like proteins are pacemakers of allergic airway reactions to Staphylococcus aureus. J Allergy Clin Immunol 2016; 139:492-500.e8. [PMID: 27315768 DOI: 10.1016/j.jaci.2016.03.045] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 02/15/2016] [Accepted: 03/22/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND A substantial subgroup of asthmatic patients have "nonallergic" or idiopathic asthma, which often takes a severe course and is difficult to treat. The cause might be allergic reactions to the gram-positive pathogen Staphylococcus aureus, a frequent colonizer of the upper airways. However, the driving allergens of S aureus have remained elusive. OBJECTIVE We sought to search for potentially allergenic S aureus proteins and characterize the immune response directed against them. METHODS S aureus extracellular proteins targeted by human serum IgG4 were identified by means of immunoblotting to screen for potential bacterial allergens. Candidate antigens were expressed as recombinant proteins and used to analyze the established cellular and humoral immune responses in healthy adults and asthmatic patients. The ability to induce a type 2 immune response in vivo was tested in a mouse asthma model. RESULTS We identified staphylococcal serine protease-like proteins (Spls) as dominant IgG4-binding S aureus proteins. SplA through SplF are extracellular proteases of unknown function expressed by S aureus in vivo. Spls elicited IgE antibody responses in most asthmatic patients. In healthy S aureus carriers and noncarriers, peripheral blood T cells elaborated TH2 cytokines after stimulation with Spls, as is typical for allergens. In contrast, TH1/TH17 cytokines, which dominated the response to S aureus α-hemolysin, were of low concentration or absent. In mice inhalation of SplD without adjuvant induced lung inflammation characterized by TH2 cytokines and eosinophil infiltration. CONCLUSION We identify Spls as triggering allergens released by S aureus, opening prospects for diagnosis and causal therapy of asthma.
Collapse
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Maria Nordengrün
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Julia Kolata
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany; Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Frank Schmidt
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | | | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany; Junior Group Applied Proteomics, ZIK FunGene, University Medicine Greifswald, Greifswald, Germany
| | - Jana Kumpfmüller
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Department of Biomolecular Chemistry, Leibniz Institute for Natural Product Research and Infection Biology, HKI, Jena, Germany
| | - Sebastian Tischer
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Schweder
- Department of Pharmaceutical Biotechnology, Institute of Pharmacy, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Michael Hecker
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Susanne Engelmann
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany; Institute for Microbiology, University of Braunschweig, Braunschweig, Germany; Helmholtz Center for Infection Research, Microbial Proteomics, Braunschweig, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Olga Krysko
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium; Division of Ear, Nose, and Throat Diseases, Clintec, Karolinska Institute, Stockholm, Sweden
| | - Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
32
|
Selle M, Hertlein T, Oesterreich B, Klemm T, Kloppot P, Müller E, Ehricht R, Stentzel S, Bröker BM, Engelmann S, Ohlsen K. Global antibody response to Staphylococcus aureus live-cell vaccination. Sci Rep 2016; 6:24754. [PMID: 27103319 PMCID: PMC4840433 DOI: 10.1038/srep24754] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/05/2016] [Indexed: 02/06/2023] Open
Abstract
The pathogen Staphylococcus aureus causes a broad range of severe diseases and is feared for its ability to rapidly develop resistance to antibiotic substances. The increasing number of highly resistant S. aureus infections has accelerated the search for alternative treatment options to close the widening gap in anti-S. aureus therapy. This study analyses the humoral immune response to vaccination of Balb/c mice with sublethal doses of live S. aureus. The elicited antibody pattern in the sera of intravenously and intramuscularly vaccinated mice was determined using of a recently developed protein array. We observed a specific antibody response against a broad set of S. aureus antigens which was stronger following i.v. than i.m. vaccination. Intravenous but not intramuscular vaccination protected mice against an intramuscular challenge infection with a high bacterial dose. Vaccine protection was correlated with the strength of the anti-S. aureus antibody response. This study identified novel vaccine candidates by using protein microarrays as an effective tool and showed that successful vaccination against S. aureus relies on the optimal route of administration.
Collapse
Affiliation(s)
- Martina Selle
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Tobias Hertlein
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Babett Oesterreich
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Theresa Klemm
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Peggy Kloppot
- University Greifswald, Institute for Microbiology, Greifswald, Germany
| | - Elke Müller
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Ralf Ehricht
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Sebastian Stentzel
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Barbara M Bröker
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Susanne Engelmann
- Technical University Braunschweig, Institute for Microbiology, Braunschweig, Germany.,Helmholtz-Zentrum für Infektionsforschung, Mikrobielle Proteomik, Braunschweig, Germany
| | - Knut Ohlsen
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| |
Collapse
|
33
|
Bröker BM, Mrochen D, Péton V. The T Cell Response to Staphylococcus aureus. Pathogens 2016; 5:pathogens5010031. [PMID: 26999219 PMCID: PMC4810152 DOI: 10.3390/pathogens5010031] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/04/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a dangerous pathogen and a leading cause of both nosocomial and community acquired bacterial infection worldwide. However, on the other hand, we are all exposed to this bacterium, often within the first hours of life, and usually manage to establish equilibrium and coexist with it. What does the adaptive immune system contribute toward lifelong control of S. aureus? Will it become possible to raise or enhance protective immune memory by vaccination? While in the past the S. aureus-specific antibody response has dominated this discussion, the research community is now coming to appreciate the role that the cellular arm of adaptive immunity, the T cells, plays. There are numerous T cell subsets, each with differing functions, which together have the ability to orchestrate the immune response to S. aureus and hence to tip the balance between protection and pathology. This review summarizes the state of the art in this dynamic field of research.
Collapse
Affiliation(s)
- Barbara M Bröker
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Daniel Mrochen
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| | - Vincent Péton
- Department of Immunology, University Medicine Greifswald, Sauerbruchstraße DZ7, 17475 Greifswald, Germany.
| |
Collapse
|
34
|
Omics Approaches for the Study of Adaptive Immunity to Staphylococcus aureus and the Selection of Vaccine Candidates. Proteomes 2016; 4:proteomes4010011. [PMID: 28248221 PMCID: PMC5217363 DOI: 10.3390/proteomes4010011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/05/2016] [Accepted: 03/01/2016] [Indexed: 01/20/2023] Open
Abstract
Staphylococcus aureus is a dangerous pathogen both in hospitals and in the community. Due to the crisis of antibiotic resistance, there is an urgent need for new strategies to combat S. aureus infections, such as vaccination. Increasing our knowledge about the mechanisms of protection will be key for the successful prevention or treatment of S. aureus invasion. Omics technologies generate a comprehensive picture of the physiological and pathophysiological processes within cells, tissues, organs, organisms and even populations. This review provides an overview of the contribution of genomics, transcriptomics, proteomics, metabolomics and immunoproteomics to the current understanding of S. aureus‑host interaction, with a focus on the adaptive immune response to the microorganism. While antibody responses during colonization and infection have been analyzed in detail using immunoproteomics, the full potential of omics technologies has not been tapped yet in terms of T-cells. Omics technologies promise to speed up vaccine development by enabling reverse vaccinology approaches. In consequence, omics technologies are powerful tools for deepening our understanding of the “superbug” S. aureus and for improving its control.
Collapse
|
35
|
Mehraj J, Witte W, Akmatov MK, Layer F, Werner G, Krause G. Epidemiology of Staphylococcus aureus Nasal Carriage Patterns in the Community. Curr Top Microbiol Immunol 2016; 398:55-87. [PMID: 27370344 DOI: 10.1007/82_2016_497] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive opportunistic pathogen that colonizes frequently and asymptomatically the anterior nares of humans and animals. It can cause different kinds of infections and is considered to be an important nosocomial pathogen. Nasal carriage of S. aureus can be permanent or intermittent and may build the reservoir for autogenous infections and cross-transmission to other individuals. Most of the studies on the epidemiology of S. aureus performed in the past were focused on the emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) in healthcare settings. There are, however, a number of more recent epidemiological studies have aimed at analysing carriage patterns over time in the community settings providing new insights on risk factors for colonization and important data for the development of strategies to prevent infections. This chapter aims to give a review of current epidemiological studies on S. aureus carriage patterns in the general community and put them into perspective with recent, yet unpublished, investigations on the S. aureus epidemiology in the general population in northern Germany.
Collapse
Affiliation(s)
- Jaishri Mehraj
- Department of Epidemiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany.,Hannover Medical School, Hannover, Germany
| | - Wolfgang Witte
- The Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Manas K Akmatov
- Department of Epidemiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, Hannover, Germany
| | - Franziska Layer
- The Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Guido Werner
- The Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Gérard Krause
- Department of Epidemiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany. .,Hannover Medical School, Hannover, Germany.
| |
Collapse
|
36
|
Brown AF, Murphy AG, Lalor SJ, Leech JM, O’Keeffe KM, Mac Aogáin M, O’Halloran DP, Lacey KA, Tavakol M, Hearnden CH, Fitzgerald-Hughes D, Humphreys H, Fennell JP, van Wamel WJ, Foster TJ, Geoghegan JA, Lavelle EC, Rogers TR, McLoughlin RM. Memory Th1 Cells Are Protective in Invasive Staphylococcus aureus Infection. PLoS Pathog 2015; 11:e1005226. [PMID: 26539822 PMCID: PMC4634925 DOI: 10.1371/journal.ppat.1005226] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/23/2015] [Indexed: 12/15/2022] Open
Abstract
Mechanisms of protective immunity to Staphylococcus aureus infection in humans remain elusive. While the importance of cellular immunity has been shown in mice, T cell responses in humans have not been characterised. Using a murine model of recurrent S. aureus peritonitis, we demonstrated that prior exposure to S. aureus enhanced IFNγ responses upon subsequent infection, while adoptive transfer of S. aureus antigen-specific Th1 cells was protective in naïve mice. Translating these findings, we found that S. aureus antigen-specific Th1 cells were also significantly expanded during human S. aureus bloodstream infection (BSI). These Th1 cells were CD45RO+, indicative of a memory phenotype. Thus, exposure to S. aureus induces memory Th1 cells in mice and humans, identifying Th1 cells as potential S. aureus vaccine targets. Consequently, we developed a model vaccine comprising staphylococcal clumping factor A, which we demonstrate to be an effective human T cell antigen, combined with the Th1-driving adjuvant CpG. This novel Th1-inducing vaccine conferred significant protection during S. aureus infection in mice. This study notably advances our understanding of S. aureus cellular immunity, and demonstrates for the first time that a correlate of S. aureus protective immunity identified in mice may be relevant in humans. Staphylococcus aureus is a leading cause of skin, soft tissue and bone infections and, most seriously, bloodstream infection. When S. aureus does get into the bloodstream, it is more likely to kill than any other bacterial infection, despite all the treatments modern medicine has to offer. It has thus far developed resistance to all antibiotics licensed to treat it. Thus, there is an urgent need to develop a vaccine against S. aureus. However, such a vaccine remains elusive. This is largely due to the fact that we have a very limited understanding of how our immune system fights this infection. Here, we examine how certain T cells of the mouse immune system effectively recognise and respond to S. aureus, and show that transferring these cells to other mice improves their ability to clear infection. We then demonstrate that a vaccine which drives these specific T cells also improves clearance of infection. Until recently, it was not known if human T cells could recognise and respond to S. aureus. Here we show, for the first time, that these cells are expanded in patients with S. aureus bloodstream infection, and suggest that they should be targeted in anti-S. aureus vaccines.
Collapse
Affiliation(s)
- Aisling F. Brown
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Alison G. Murphy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Stephen J. Lalor
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - John M. Leech
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate M. O’Keeffe
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Micheál Mac Aogáin
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Dara P. O’Halloran
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Keenan A. Lacey
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mehri Tavakol
- Department of Medical Microbiology & Infectious Diseases, Erasmus Medical Center, Rotterdam, Netherlands
| | - Claire H. Hearnden
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | - Hilary Humphreys
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Microbiology, Beaumont Hospital, Dublin, Ireland
| | - Jérôme P. Fennell
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Microbiology, Adelaide Meath & National Children’s Hospital, Dublin, Ireland
| | - Willem J. van Wamel
- Department of Medical Microbiology & Infectious Diseases, Erasmus Medical Center, Rotterdam, Netherlands
| | - Timothy J. Foster
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Ed C. Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, Dublin, Ireland
| | - Thomas R. Rogers
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Microbiology, St. James's Hospital, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
37
|
Specific serum IgG at diagnosis of Staphylococcus aureus bloodstream invasion is correlated with disease progression. J Proteomics 2015; 128:1-7. [DOI: 10.1016/j.jprot.2015.06.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/11/2015] [Accepted: 06/30/2015] [Indexed: 11/16/2022]
|
38
|
Prevalence of enterotoxin genes in Staphylococcus aureus colonising food handlers: does nasal carriage status matter? Eur J Clin Microbiol Infect Dis 2015; 34:2177-81. [PMID: 26306787 DOI: 10.1007/s10096-015-2465-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
This study investigated the association between the presence of staphylococcal enterotoxin (SE) genes and nasal carriage status, and determined temporal changes in the prevalence of these genes in Staphylococcus aureus strains isolated from healthy carriers between 2002 and 2011. Three large samples of food handlers recruited in 2002, 2003 and 2011 were nasally sampled on two occasions to determine S. aureus colonisation status. Those carrying the same spa type on both occasions were defined as persistent carriers. Genes for SEs SEA-SEU were amplified and associations between carriage status and presence of SE genes were investigated. Although 80 % of nasal isolates harboured at least one SE gene over the sampling period, persistent carriers were significantly more likely to harbour enterotoxigenic S. aureus than transiently colonised subjects [odds ratio (OR) 2.52-3.06]. Strains from persistent carriers more commonly harboured sea, seb and sem. The prevalence of classical SE genes and sej, sem, sen, seo, seq and ses was stable over time, but seh, sel, sep, ser, set and selu increased significantly. Increased toxigenicity of isolates from persistent carriers is consistent with the elevated antibody levels to classical SEs previously reported in persistent carriers, supporting the hypothesis that superantigen production in the nasal cavity may enhance colonisation.
Collapse
|
39
|
van den Berg S, de Vogel CP, van Belkum A, Bakker-Woudenberg IAJM. Mild Staphylococcus aureus Skin Infection Improves the Course of Subsequent Endogenous S. aureus Bacteremia in Mice. PLoS One 2015; 10:e0129150. [PMID: 26060995 PMCID: PMC4464736 DOI: 10.1371/journal.pone.0129150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 05/05/2015] [Indexed: 01/02/2023] Open
Abstract
Staphylococcus aureus carriers with S. aureus bacteremia may have a reduced mortality risk compared to non-carriers. A role for the immune system is suggested. Here, we study in mice the effect of mild S. aureus skin infection prior to endogenous or exogenous S. aureus bacteremia, and evaluate protection in relation to anti-staphylococcal antibody levels. Skin infections once or twice by a clinical S. aureus isolate (isolate P) or S. aureus strain 8325-4 were induced in mice free of S. aureus and anti-staphylococcal antibodies. Five weeks later, immunoglobulin G (IgG) levels in blood against 25 S. aureus antigens were determined, and LD50 or LD100 bacteremia caused by S. aureus isolate P was induced. S. aureus skin infections led to elevated levels of anti-staphylococcal IgG in blood. One skin infection improved the course of subsequent severe endogenous bacteremia only. A second skin infection further improved animal survival rate, which was associated with increased pre-bacteremia IgG levels against Efb, IsaA, LukD, LukE, Nuc, PrsA and WTA. In conclusion, S. aureus isolate P skin infection in mice reduces the severity of subsequent endogenous S. aureus bacteremia only. Although cellular immune effects cannot be rules out, anti-staphylococcal IgG against specified antigens may contribute to this effect.
Collapse
Affiliation(s)
- Sanne van den Berg
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
- * E-mail:
| | - Corné P. de Vogel
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alex van Belkum
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
- bioMérieux, Microbiology R&D, La Balme les Grottes, France
| | | |
Collapse
|
40
|
Superantigens Modulate Bacterial Density during Staphylococcus aureus Nasal Colonization. Toxins (Basel) 2015; 7:1821-36. [PMID: 26008236 PMCID: PMC4448176 DOI: 10.3390/toxins7051821] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/15/2015] [Indexed: 02/06/2023] Open
Abstract
Superantigens (SAgs) are potent microbial toxins that function to activate large numbers of T cells in a T cell receptor (TCR) Vβ-specific manner, resulting in excessive immune system activation. Staphylococcus aureus possesses a large repertoire of distinct SAgs, and in the context of host-pathogen interactions, staphylococcal SAg research has focused primarily on the role of these toxins in severe and invasive diseases. However, the contribution of SAgs to colonization by S. aureus remains unclear. We developed a two-week nasal colonization model using SAg-sensitive transgenic mice expressing HLA-DR4, and evaluated the role of SAgs using two well-studied stains of S. aureus. S. aureus Newman produces relatively low levels of staphylococcal enterotoxin A (SEA), and although we did not detect significant TCR-Vβ specific changes during wild-type S. aureus Newman colonization, S. aureus Newman Δsea established transiently higher bacterial loads in the nose. S. aureus COL produces relatively high levels of staphylococcal enterotoxin B (SEB), and colonization with wild-type S. aureus COL resulted in clear Vβ8-specific T cell skewing responses. S. aureus COL Δseb established consistently higher bacterial loads in the nose. These data suggest that staphylococcal SAgs may be involved in regulating bacterial densities during nasal colonization.
Collapse
|
41
|
Kloppot P, Selle M, Kohler C, Stentzel S, Fuchs S, Liebscher V, Müller E, Kale D, Ohlsen K, Bröker BM, Zipfel PF, Kahl BC, Ehricht R, Hecker M, Engelmann S. Microarray-based identification of human antibodies against Staphylococcus aureus antigens. Proteomics Clin Appl 2015; 9:1003-11. [PMID: 25676254 DOI: 10.1002/prca.201400123] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 01/06/2015] [Accepted: 02/05/2015] [Indexed: 02/03/2023]
Abstract
PURPOSE The mortality rate of patients with Staphylococcus aureus infections is alarming and urgently demands new strategies to attenuate the course of these infections or to detect them at earlier stages. EXPERIMENTAL DESIGN To study the adaptive immune response to S. aureus antigens in healthy human volunteers, a protein microarray containing 44 S. aureus proteins was developed using the ArrayStrip platform technology. RESULTS Testing plasma samples from 15 S. aureus carriers and 15 noncarriers 21 immunogenic S. aureus antigens have been identified. Seven antigens were recognized by antibodies present in at least 60% of the samples, representing the core S. aureus immunome of healthy individuals. S. aureus-specific serum immunoglobulin G (IgG) levels were significantly lower in noncarriers than in carriers specifically anti-IsaA, anti-SACOL0479, and anti-SACOL0480 IgGs were found at lower frequencies and quantities. Twenty-two antigens present on the microarray were encoded by all S. aureus carrier isolates. Nevertheless, the immune system of the carriers was responsive to only eight of them and with different intensities. CONCLUSION AND CLINICAL RELEVANCE The established protein microarray allows a broad profiling of the S. aureus-specific antibody response and can be used to identify S. aureus antigens that might serve as vaccines or diagnostic markers.
Collapse
Affiliation(s)
- Peggy Kloppot
- Institut für Mikrobiologie, Universität Greifswald, Greifswald, Germany
| | - Martina Selle
- Institut für Molekulare Infektionsbiologie, Universität Würzburg, Würzburg, Germany
| | - Christian Kohler
- Institut für Mikrobiologie, Universität Greifswald, Greifswald, Germany
| | - Sebastian Stentzel
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Stephan Fuchs
- Institut für Mikrobiologie, Universität Greifswald, Greifswald, Germany
| | - Volkmar Liebscher
- Institut für Mathematik und Informatik, Universität Greifswald, Greifswald, Germany
| | | | - Devika Kale
- Institut für Medizinische Mikrobiologie, Universitätsklinikum Münster, Münster, Germany
| | - Knut Ohlsen
- Institut für Molekulare Infektionsbiologie, Universität Würzburg, Würzburg, Germany
| | - Barbara M Bröker
- Institut für Immunologie und Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Peter F Zipfel
- Infektionsbiologie, Hans-Knöll-Institut Jena, Münster, Germany.,Friedrich Schiller Universität, Jena, Germany
| | - Barbara C Kahl
- Institut für Medizinische Mikrobiologie, Universitätsklinikum Münster, Münster, Germany
| | | | - Michael Hecker
- Institut für Mikrobiologie, Universität Greifswald, Greifswald, Germany
| | - Susanne Engelmann
- Institut für Mikrobiologie, Universität Greifswald, Greifswald, Germany.,Institut für Mikrobiologie, TU Braunschweig, Braunschweig, Germany.,Mikrobielle Proteomik, Helmholtzzentrum für Infektionsforschung, Braunschweig, Germany
| |
Collapse
|
42
|
Comparative Exoproteomics and Host Inflammatory Response in Staphylococcus aureus Skin and Soft Tissue Infections, Bacteremia, and Subclinical Colonization. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:593-603. [PMID: 25809633 DOI: 10.1128/cvi.00493-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 03/21/2015] [Indexed: 11/20/2022]
Abstract
The exoproteome of Staphylococcus aureus contains enzymes and virulence factors that are important for host adaptation. We investigated the exoprotein profiles and cytokine/chemokine responses obtained in three different S. aureus-host interaction scenarios by using two-dimensional gel electrophoresis (2-DGE) and two-dimensional immunoblotting (2D-IB) combined with tandem mass spectrometry (MS/MS) and cytometric bead array techniques. The scenarios included S. aureus bacteremia, skin and soft tissue infections (SSTIs), and healthy carriage. By the 2-DGE approach, 12 exoproteins (the chaperone protein DnaK, a phosphoglycerate kinase [Pgk], the chaperone GroEL, a multisensor hybrid histidine kinase, a 3-methyl-2-oxobutanoate hydroxymethyltransferase [PanB], cysteine synthase A, an N-acetyltransferase, four isoforms of elongation factor Tu [EF-Tu], and one signature protein spot that could not be reliably identified by MS/MS) were found to be consistently present in more than 50% of the bacteremia isolates, while none of the SSTI or healthy-carrier isolates showed any of these proteins. By the 2D-IB approach, we also identified five antigens (methionine aminopeptidase [MetAPs], exotoxin 15 [Set15], a peptidoglycan hydrolase [LytM], an alkyl hydroperoxide reductase [AhpC], and a haptoglobin-binding heme uptake protein [HarA]) specific for SSTI cases. Cytokine and chemokine production varied during the course of different infection types and carriage. Monokine induced by gamma interferon (MIG) was more highly stimulated in bacteremia patients than in SSTI patients and healthy carriers, especially during the acute phase of infection. MIG could therefore be further explored as a potential biomarker of bacteremia. In conclusion, 12 exoproteins from bacteremia isolates, MIG production, and five antigenic proteins identified during SSTIs should be further investigated for potential use as diagnostic markers.
Collapse
|
43
|
Kolata JB, Kühbandner I, Link C, Normann N, Vu CH, Steil L, Weidenmaier C, Bröker BM. The Fall of a Dogma? Unexpected High T-Cell Memory Response to Staphylococcus aureus in Humans. J Infect Dis 2015; 212:830-8. [PMID: 25737563 DOI: 10.1093/infdis/jiv128] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 02/20/2015] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Though Staphylococcus aureus is a major pathogen, vaccine trials have failed. In contrast, class-switched antibodies specific to S. aureus are common, implying immune memory formation and suggesting a large pool of S. aureus-reactive helper T-cells. OBJECTIVE To elucidate the cellular arm of S. aureus-specific immune memory, the T-cell response in humans was characterized. METHODS The proliferative response of human peripheral blood mononuclear cells (PBMCs) to S. aureus antigens and the frequency of S. aureus-specific T-cells were quantified by (3)H-thymidine incorporation; cytokine release was measured by flow cytometry. RESULTS Staphylococcus aureus particles and extracellular proteins elicited pronounced proliferation in PBMCs of healthy adults. This reflected a memory response with high frequencies of T-cells being activated by single S. aureus antigens. The whole S. aureus-specific T-cell pool was estimated to comprise 3.6% of T-cells with 35-fold differences between individuals (range, 0.2%-5.7%). When exposed to S. aureus antigens, the T-cells released predominantly but not solely T helper (Th)1/Th17 cytokines. CONCLUSIONS The large number of S. aureus antigen-reactive memory T-lymphocytes is likely to influence the course of S. aureus infection. To enable rational vaccine design, the naturally acquired human T-cell memory needs to be explored at high priority.
Collapse
Affiliation(s)
- Julia B Kolata
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| | - Iris Kühbandner
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| | - Christopher Link
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| | - Nicole Normann
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| | - Chi Hai Vu
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| | - Leif Steil
- Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald
| | - Christopher Weidenmaier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Eberhard-Karls-University, Germany
| | - Barbara M Bröker
- Institute of Immunology and Transfusion Medicine, Department of Immunology, University Medicine Greifswald
| |
Collapse
|
44
|
Rozemeijer W, Fink P, Rojas E, Jones CH, Pavliakova D, Giardina P, Murphy E, Liberator P, Jiang Q, Girgenti D, Peters RPH, Savelkoul PHM, Jansen KU, Anderson AS, Kluytmans J. Evaluation of approaches to monitor Staphylococcus aureus virulence factor expression during human disease. PLoS One 2015; 10:e0116945. [PMID: 25719409 PMCID: PMC4342157 DOI: 10.1371/journal.pone.0116945] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/17/2014] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus is a versatile pathogen of medical significance, using multiple virulence factors to cause disease. A prophylactic S. aureus 4-antigen (SA4Ag) vaccine comprising capsular polysaccharide (types 5 and 8) conjugates, clumping factor A (ClfA) and manganese transporter C (MntC) is under development. This study was designed to characterize S. aureus isolates recovered from infected patients and also to investigate approaches for examining expression of S. aureus vaccine candidates and the host response during human infection. Confirmation of antigen expression in different disease states is important to support the inclusion of these antigens in a prophylactic vaccine. Hospitalized patients with diagnosed S. aureus wound (27) or bloodstream (24) infections were enrolled. Invasive and nasal carriage S. aureus isolates were recovered and characterized for genotypic diversity. S. aureus antigen expression was evaluated directly by real-time, quantitative, reverse-transcriptase PCR (qRT-PCR) analysis and indirectly by serology using a competitive Luminex immunoassay. Study isolates were genotypically diverse and all had the genes encoding the antigens present in the SA4Ag vaccine. S. aureus nasal carriage was detected in 55% of patients, and in those subjects 64% of the carriage isolates matched the invasive strain. In swab samples with detectable S. aureus triosephosphate isomerase housekeeping gene expression, RNA transcripts encoding the S. aureus virulence factors ClfA, MntC, and capsule polysaccharide were detected by qRT-PCR. Antigen expression was indirectly confirmed by increases in antibody titer during the course of infection from acute to convalescent phase. Demonstration of bacterial transcript expression together with immunological response to the SA4Ag antigens in a clinically relevant patient population provides support for inclusion of these antigens in a prophylactic vaccine.
Collapse
Affiliation(s)
| | - Pamela Fink
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Eduardo Rojas
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - C. Hal Jones
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Danka Pavliakova
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Peter Giardina
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Ellen Murphy
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Paul Liberator
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Qin Jiang
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Douglas Girgenti
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | | | | | - Kathrin U. Jansen
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
| | - Annaliesa S. Anderson
- Pfizer Vaccine Research and Development, Pearl River, New York, United States of America
- * E-mail:
| | - Jan Kluytmans
- VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
45
|
van den Berg S, Koedijk DGAM, Back JW, Neef J, Dreisbach A, van Dijl JM, Bakker-Woudenberg IAJM, Buist G. Active immunization with an octa-valent Staphylococcus aureus antigen mixture in models of S. aureus bacteremia and skin infection in mice. PLoS One 2015; 10:e0116847. [PMID: 25710376 PMCID: PMC4339199 DOI: 10.1371/journal.pone.0116847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/15/2014] [Indexed: 11/18/2022] Open
Abstract
Proteomic studies with different Staphylococcus aureus isolates have shown that the cell surface-exposed and secreted proteins IsaA, LytM, Nuc, the propeptide of Atl (pro-Atl) and four phenol-soluble modulins α (PSMα) are invariantly produced by this pathogen. Therefore the present study was aimed at investigating whether these proteins can be used for active immunization against S. aureus infection in mouse models of bacteremia and skin infection. To this end, recombinant His-tagged fusions of IsaA, LytM, Nuc and pro-Atl were isolated from Lactococcus lactis or Escherichia coli, while the PSMα1-4 peptides were chemically synthesized. Importantly, patients colonized by S. aureus showed significant immunoglobulin G (IgG) responses against all eight antigens. BALB/cBYJ mice were immunized subcutaneously with a mixture of the antigens at day one (5 μg each), and boosted twice (25 μg of each antigen) with 28 days interval. This resulted in high IgG responses against all antigens although the response against pro-Atl was around one log lower compared to the other antigens. Compared to placebo-immunized mice, immunization with the octa-valent antigen mixture did not reduce the S. aureus isolate P load in blood, lungs, spleen, liver, and kidneys in a bacteremia model in which the animals were challenged for 14 days with a primary load of 3 × 105 CFU. Discomfort scores and animal survival rates over 14 days did not differ between immunized mice and placebo-immunized mice upon bacteremia with S. aureus USA300 (6 × 105 CFU). In addition, this immunization did not reduce the S. aureus isolate P load in mice with skin infection. These results show that the target antigens are immunogenic in both humans and mice, but in the used animal models do not result in protection against S. aureus infection.
Collapse
Affiliation(s)
- Sanne van den Berg
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
- * E-mail:
| | - Dennis G. A. M. Koedijk
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Jolanda Neef
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Annette Dreisbach
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
46
|
Nissen M, Marshall H, Richmond P, Shakib S, Jiang Q, Cooper D, Rill D, Baber J, Eiden J, Gruber W, Jansen KU, Emini EA, Anderson AS, Zito ET, Girgenti D. A randomized phase I study of the safety and immunogenicity of three ascending dose levels of a 3-antigen Staphylococcus aureus vaccine (SA3Ag) in healthy adults. Vaccine 2015; 33:1846-54. [PMID: 25707693 DOI: 10.1016/j.vaccine.2015.02.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 10/24/2022]
Abstract
BACKGROUND Staphylococcus aureus is a common cause of healthcare-acquired morbidity and mortality and increased healthcare resource utilization. A prophylactic vaccine is being developed that may reduce this disease burden. METHODS Volunteers in good general health aged 50-85 (n=312) and 18-24 (n=96) years were randomized to receive a single intramuscular dose of one of three dose levels of a non-adjuvanted, 3-antigen S. aureus vaccine (SA3Ag) or placebo. SA3Ag antigens included capsular polysaccharides 5 and 8 (CP5 and CP8), each conjugated to cross-reactive material 197 (CRM197), and recombinant clumping factor A (ClfA). Safety, tolerability, and immunogenicity were evaluated. RESULTS At day 29 post-vaccination, robust immune responses were observed in both age cohorts at all three SA3Ag dose levels. In the primary analysis population, the 50- to 85-year age stratum, geometric mean-fold-rises in competitive Luminex(®) immunoassay antibody titers from baseline ranged from 29.2 to 83.7 (CP5), 14.1 to 31.0 (CP8), and 37.1 to 42.9 (ClfA), all (P<0.001) exceeding the pre-defined two-fold rise criteria. Similar rises in opsonophagocytic activity assay titers demonstrated functionality of the immune response. Most injection-site reactions were mild in severity and there were no substantial differences (SA3Ag vs. placebo) with regard to systemic or adverse events. CONCLUSIONS In this study of healthy adults aged 50-85 and 18-24 years, SA3Ag elicited a rapid and robust immune response and was well tolerated, with no notable safety concerns.
Collapse
Affiliation(s)
- Michael Nissen
- Queensland Paediatric Infectious Diseases Clinical Trials Centre, Royal Children's Hospital and Children's Health Queensland, Brisbane, QLD, Australia.
| | - Helen Marshall
- Vaccinology and Immunology Research Trials Unit, Women's and Children's Hospital and Robinson Research Institute and School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, SA, Australia.
| | - Peter Richmond
- University of Western Australia School of Paediatrics and Child Health & Telethon Kids Institute, Perth, WA, Australia
| | | | | | | | | | - James Baber
- Pfizer Australia Pty Ltd, Sydney, NSW, Australia
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Low anti-staphylococcal IgG responses in granulomatosis with polyangiitis patients despite long-term Staphylococcus aureus exposure. Sci Rep 2015; 5:8188. [PMID: 25641235 PMCID: PMC5389034 DOI: 10.1038/srep08188] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/07/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic nasal carriage of the bacterium Staphylococcus aureus in patients with the autoimmune disease granulomatosis with polyangiitis (GPA) is a risk factor for disease relapse. To date, it was neither known whether GPA patients show similar humoral immune responses to S. aureus as healthy carriers, nor whether specific S. aureus types are associated with GPA. Therefore, this study was aimed at assessing humoral immune responses of GPA patients against S. aureus antigens in relation to the genetic diversity of their nasal S. aureus isolates. A retrospective cohort study was conducted, including 85 GPA patients and 18 healthy controls (HC). Humoral immune responses against S. aureus were investigated by determining serum IgG levels against 59 S. aureus antigens. Unexpectedly, patient sera contained lower anti-staphylococcal IgG levels than sera from HC, regardless of the patients' treatment, while total IgG levels were similar or higher. Furthermore, 210 S. aureus isolates obtained from GPA patients were characterized by different typing approaches. This showed that the S. aureus population of GPA patients is highly diverse and mirrors the general S. aureus population. Our combined findings imply that GPA patients are less capable of mounting a potentially protective antibody response to S. aureus than healthy individuals.
Collapse
|
48
|
Rongpharpi SR, Duggal S, Kalita H, Duggal AK. Staphylococcus aureus bacteremia: targeting the source. Postgrad Med 2014; 126:167-75. [PMID: 25295661 DOI: 10.3810/pgm.2014.09.2811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bacteremia due to Staphylococcus aureus is one of the major causes of morbidity and mortality in India, but studies targeting the source of Staphylococcus aureus bacteremia are lacking. S. aureus has a vivid armamentarium consisting of toxins, adhesins, and other virulence factors by virtue of which it can cause varied types of infections, sometimes of a serious nature. This review highlights the possible causes of S. aureus bacteremia, and discusses the necessity of tracing its source and eliminating it with proper antibiotic therapy to avoid recurrences or relapses.
Collapse
Affiliation(s)
- Sharon Rainy Rongpharpi
- Senior Resident, Department of Microbiology, Dr. Baba Saheb Ambedkar Hospital, New Delhi, India
| | | | | | | |
Collapse
|
49
|
van Kessel KPM, Bestebroer J, van Strijp JAG. Neutrophil-Mediated Phagocytosis of Staphylococcus aureus. Front Immunol 2014; 5:467. [PMID: 25309547 PMCID: PMC4176147 DOI: 10.3389/fimmu.2014.00467] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/12/2014] [Indexed: 01/13/2023] Open
Abstract
Initial elimination of invading Staphylococcus aureus from the body is mediated by professional phagocytes. The neutrophil is the major phagocyte of the innate immunity and plays a key role in the host defense against staphylococcal infections. Opsonization of the bacteria with immunoglobulins and complement factors enables efficient recognition by the neutrophil that subsequently leads to intracellular compartmentalization and killing. Here, we provide a review of the key processes evolved in neutrophil-mediated phagocytosis of S. aureus and briefly describe killing. As S. aureus is not helpless against the professional phagocytes, we will also highlight its immune evasion arsenal related to phagocytosis.
Collapse
Affiliation(s)
- Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Jovanka Bestebroer
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht , Utrecht , Netherlands
| |
Collapse
|
50
|
Stentzel S, Vu HC, Weyrich AM, Jehmlich N, Schmidt F, Salazar MG, Steil L, Völker U, Bröker BM. Altered immune proteome ofStaphylococcus aureusunder iron-restricted growth conditions. Proteomics 2014; 14:1857-67. [DOI: 10.1002/pmic.201300512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/16/2014] [Accepted: 05/22/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Sebastian Stentzel
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Hai Chi Vu
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Anna Maria Weyrich
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| | - Nico Jehmlich
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Frank Schmidt
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
- ZIK-FunGene Junior Research Group “Applied Proteomics”; University Medicine Greifswald; Greifswald Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Leif Steil
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics; University Medicine Greifswald; Greifswald Germany
| | - Barbara M. Bröker
- Department of Immunology; Institute of Immunology and Transfusion Medicine; University Medicine Greifswald; Greifswald Germany
| |
Collapse
|