1
|
Won HK, Lee J, Park KE, Choi JP, Song WJ. Exploration of IgE Specific to Staphylococcal Serine Protease-Like Protein A as a Phenotypic Marker in Late-Onset Asthma. Lung 2025; 203:56. [PMID: 40249510 DOI: 10.1007/s00408-025-00810-w] [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: 02/26/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE Staphylococcus aureus (SA) secretes pro-allergic molecules, including staphylococcal enterotoxins (SEs) and serine protease-like proteins (Spls). While IgE sensitization to SE has been relatively well documented in relation to severe eosinophilic late-onset asthma, the clinical implications of IgE sensitization to Spls remain unclear. We explored the clinical relevance of Spl-IgE in late-onset asthmatics. METHODS Adults with late-onset asthma (onset age ≥ 40 years) were prospectively enrolled. Demographic and clinical characteristics were assessed, and serum levels of total IgE, SE-IgE, and SplA-IgE were measured. Nasal swabs were obtained to assess SA colonization. RESULTS Among 109 participants, SplA-IgE levels were significantly associated with blood eosinophilia, total IgE, SE-IgE sensitization, and male sex, but not with SA colonization, asthma severity, or lung function. CONCLUSION Sensitization to SplA-IgE may indicate a type 2 inflammatory phenotype, but its role in asthma warrants further investigation.
Collapse
Affiliation(s)
- Ha-Kyeong Won
- Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Jiwon Lee
- Institute of Allergy and Clinical Immunology, Asan Medical Research Center, Seoul, Korea
| | | | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Korea.
| |
Collapse
|
2
|
Zheng Y, Jiang Y, Niu J, Deng W, Ban S, Xu Y, Wu Q, Shi Y. Temperature and wind speed help fermentation-sourced microbiota reconstruct the airborne microbiota in the Chinese baijiu fermentation region. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 378:124769. [PMID: 40037255 DOI: 10.1016/j.jenvman.2025.124769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Microorganisms are vital components of airborne particles and are closely linked to human health and industrial processes. As industrialization advances, factories are exacerbating their impact on environmental microbial communities, an area where our understanding remains limited. In this study, we investigated air microorganisms surrounding Chinese baijiu fermentation facilities, aiming to explore the dispersal mechanisms of fermentation-sourced microbiota and their potential impacts on local airborne microbial communities. The results revealed a significant overlap between microbial communities from fermentation processes and the surrounding air, with 14 genera commonly found in both habitats, suggesting widespread and substantial dispersal of factory microorganisms. Due to this dispersal, fermentation-sourced Saccharopolyspora and Streptomyces have become central nodes in the airborne microbial network. Environmental factors played a pivotal role in the microbiota dispersal process. Higher temperatures may facilitate microbial proliferation and increase the dispersal of fermentation microorganisms (r = 0.30, P < 0.01), while higher wind speeds may lead to a dilution effect and limit the uniform dispersal of microorganisms (r = -0.22, P < 0.05). Furthermore, air-resident microorganisms restricted the proliferation of fermentation microorganisms (r = -0.60, P < 0.001). These results confirm the modifying effect of fermentation facilities on environmental microbial communities and establish a theoretical foundation for optimizing factory site selection.
Collapse
Affiliation(s)
- Yifu Zheng
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Yingli Jiang
- Sichuan Langjiu Group Co., Ltd., 610213, Luzhou, China
| | - Jiao Niu
- Sichuan Langjiu Group Co., Ltd., 610213, Luzhou, China
| | - Wanyu Deng
- Sichuan Langjiu Group Co., Ltd., 610213, Luzhou, China
| | - Shibo Ban
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Yan Xu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China
| | - Qun Wu
- Key Laboratory of Industrial Biotechnology of Ministry of Education, State Key Laboratory of Food Science and Technology, School of Biotechnology, Jiangnan University, 214122, Wuxi, China.
| | - Yu Shi
- State Key Laboratory of Crop Stress Adaptation and Improvement, School of Life Sciences, Henan University, 475004, Kaifeng, China.
| |
Collapse
|
3
|
Rasquel-Oliveira FS, Ribeiro JM, Martelossi-Cebinelli G, Costa FB, Nakazato G, Casagrande R, Verri WA. Staphylococcus aureus in Inflammation and Pain: Update on Pathologic Mechanisms. Pathogens 2025; 14:185. [PMID: 40005560 PMCID: PMC11858194 DOI: 10.3390/pathogens14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/23/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Staphylococcus aureus (S. aureus) is a Gram-positive bacterium of significant clinical importance, known for its versatility and ability to cause a wide array of infections, such as osteoarticular, pulmonary, cardiovascular, device-related, and hospital-acquired infections. This review describes the most recent evidence of the pathogenic potential of S. aureus, which is commonly part of the human microbiota but can lead to severe infections. The prevalence of pathogenic S. aureus in hospital and community settings contributes to substantial morbidity and mortality, particularly in individuals with compromised immune systems. The immunopathogenesis of S. aureus infections involves intricate interactions with the host immune and non-immune cells, characterized by various virulence factors that facilitate adherence, invasion, and evasion of the host's defenses. This review highlights the complexity of S. aureus infections, ranging from mild to life-threatening conditions, and underscores the growing public health concern posed by multidrug-resistant strains, including methicillin-resistant S. aureus (MRSA). This article aims to provide an updated perspective on S. aureus-related infections, highlighting the main diseases linked to this pathogen, how the different cell types, virulence factors, and signaling molecules are involved in the immunopathogenesis, and the future perspectives to overcome the current challenges to treat the affected individuals.
Collapse
Affiliation(s)
- Fernanda S. Rasquel-Oliveira
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Jhonatan Macedo Ribeiro
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Geovana Martelossi-Cebinelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Fernanda Barbosa Costa
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| | - Gerson Nakazato
- Department of Microbiology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil (G.N.)
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil; (F.S.R.-O.)
| |
Collapse
|
4
|
Chhiba KD, Patel GB, Peters AT. Anti-IgE therapy in chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2025; 155:24-30. [PMID: 39551440 DOI: 10.1016/j.jaci.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a chronic inflammatory condition characterized by type 2 (T2) immune responses with significant impacts on quality of life and health care costs. Local IgE production in nasal polyp tissue plays a key role in the T2 inflammatory cascade. Omalizumab, an anti-IgE monoclonal antibody, is an effective treatment for some patients with CRSwNP regardless of the patient's allergic status. Clinical trials, including the pivotal POLYP 1 and POLYP 2 studies, demonstrated omalizumab's efficacy in reducing nasal polyp size, improving symptom scores, and enhancing quality of life, particularly in patients with comorbid asthma and aspirin-exacerbated respiratory disease. As we summarize in this review, omalizumab's effect appears to involve the reduction in local IgE and T2 inflammation; however, this remains poorly understood. Notably, omalizumab's effectiveness appears to be partially sustained after long-term therapy, though symptoms and inflammation begin to return at discontinuation. Ongoing research is needed to determine the optimal duration of therapy and potential for biologics to modify the disease course. Additionally, further studies are needed to identify biomarkers to predict treatment response and to compare omalizumab with other biologics such as dupilumab in head-to-head trials. Omalizumab is one of the key T2-targeted therapeutic options for CRSwNP, with sustained effectiveness and strong safety profile.
Collapse
Affiliation(s)
- Krishan D Chhiba
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Gayatri B Patel
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Anju T Peters
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
5
|
Scherr F, Darisipudi MN, Börner FR, Austermeier S, Hoffmann F, Eberhardt M, Abdurrahman G, Saade C, von Eggeling F, Kasper L, Holtfreter S, Bröker BM, Kiehntopf M. Alpha-1-antitrypsin as novel substrate for S. aureus' Spl proteases - implications for virulence. Front Immunol 2024; 15:1481181. [PMID: 39628483 PMCID: PMC11611844 DOI: 10.3389/fimmu.2024.1481181] [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: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 12/06/2024] Open
Abstract
Background The serine protease like (Spl) proteases of Staphylococcus aureus are a family of six proteases whose function and impact on virulence are poorly understood. Here we propose alpha-1-antitrypsin (AAT), an important immunomodulatory serine protease inhibitor as target of SplD, E and F. AAT is an acute phase protein, interacting with many proteases and crucial for prevention of excess tissue damage by neutrophil elastase during the innate immune response to infections. Methods We used MALDI-TOF-MS to identify the cleavage site of Spl proteases within AAT's reactive center loop (RCL) and LC-MS/MS to quantify the resulting peptide cleavage product in in vitro digestions of AAT and heterologous expressed proteases or culture supernatants from different S. aureus strains. We further confirmed proteolytic cleavage and formation of a covalent complex with Western Blots, investigated AAT's inhibitory potential against Spls and examined the NETosis inhibitory activity of AAT-Spl-digestions. Results SplD, E and F, but not A or B, cleave AAT in its RCL, resulting in the release of a peptide consisting of AAT's C-terminal 36 amino acids (C36). Synthetic C36, as well as AAT-SplD/E/F-digestions exhibit NETosis inhibition. Only SplE, but not D or F, was partly inhibited by AAT, forming a covalent complex. Conclusion We unraveled a new virulence trait of S. aureus, where SplD/E/F cleave and inactivate AAT while the cleavage product C36 inhibits NETosis.
Collapse
Affiliation(s)
- Franziska Scherr
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | | | - Friedemann R. Börner
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Sophie Austermeier
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Martin Eberhardt
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Christopher Saade
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Matrix-assisted Laser Desorption/Ionization (MALDI) Imaging and Clinical Biophotonics, Jena University Hospital, Jena, Germany
| | - Lydia Kasper
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology -Hans Knoell Institute Jena (HKI), Jena, Germany
- Institute of Novel and Emerging Infectious Diseases (INNT), Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| |
Collapse
|
6
|
Chen X, Missiakas D. Novel Antibody-Based Protection/Therapeutics in Staphylococcus aureus. Annu Rev Microbiol 2024; 78:425-446. [PMID: 39146354 DOI: 10.1146/annurev-micro-041222-024605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Staphylococcus aureus is a commensal of the skin and nares of humans as well as the causative agent of infections associated with significant mortality. The acquisition of antibiotic resistance traits complicates the treatment of such infections and has prompted the development of monoclonal antibodies. The selection of protective antigens is typically guided by studying the natural antibody responses to a pathogen. What happens when the pathogen masks these antigens and subverts adaptive responses, or when the pathogen inhibits or alters the effector functions of antibodies? S. aureus is constantly exposed to its human host and has evolved all these strategies. Here, we review how anti-S. aureus targets have been selected and how antibodies have been engineered to overcome the formidable immune evasive activities of this pathogen. We discuss the prospects of antibody-based therapeutics in the context of disease severity, immune competence, and history of past infections.
Collapse
Affiliation(s)
- Xinhai Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, China
| | - Dominique Missiakas
- Department of Microbiology, The University of Chicago, Chicago, Illinois, USA;
| |
Collapse
|
7
|
Kline SN, Saito Y, Archer NK. Staphylococcus aureus Proteases: Orchestrators of Skin Inflammation. DNA Cell Biol 2024; 43:483-491. [PMID: 38957987 PMCID: PMC11535466 DOI: 10.1089/dna.2024.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Skin homeostasis relies on a delicate balance between host proteases and protease inhibitors along with those secreted from microbial communities, as disruption to this harmony contributes to the pathogenesis of inflammatory skin disorders, including atopic dermatitis and Netherton's syndrome. In addition to being a prominent cause of skin and soft tissue infections, the gram-positive bacterium Staphylococcus aureus is a key player in inflammatory skin conditions due to its array of 10 secreted proteases. Herein we review how S. aureus proteases augment the development of inflammation in skin disorders. These mechanisms include degradation of skin barrier integrity, immune dysregulation and pruritis, and impairment of host defenses. Delineating the diverse roles of S. aureus proteases has the potential to reveal novel therapeutic strategies, such as inhibitors of proteases or their cognate target, as well as neutralizing vaccines to alleviate the burden of inflammatory skin disorders in patients.
Collapse
Affiliation(s)
- Sabrina N. Kline
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yoshine Saito
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Xie H, Li L, Guo Y, Zhou L, Ma L, He A, Lai H, He Y, Liu Y, Chen H, Luo L, Huang Y, Sha X, Zhang H, Yan J, Zhang Q, Tao A. Pseudomonas aeruginosa exotoxin A as a novel allergen induced Non-T H2 inflammation in a murine model of steroid-insensitive asthma. Heliyon 2024; 10:e37512. [PMID: 39315215 PMCID: PMC11417555 DOI: 10.1016/j.heliyon.2024.e37512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Background Despite the immediate in vivo occurrence of anaphylactic and allergic reactions following treatment with Pseudomonas aeruginosa exotoxin A (PEA)-based immunotoxins, the immunological role of PEA in asthma pathogenesis remains unclear. Objective This study investigated the allergenic potential of PEA and the specific type of asthma induced. Methods Recombinant PEA (rPEA) lacking domain Ia (to eliminate non-specific cytotoxicity) was expressed, purified, and employed to detect serum PEA-specific IgE levels in asthmatic patients. Competitive ELISA assays were used to assess rPEA's IgE binding capacity and allergenicity. Additionally, rPEA-challenged C57BL/6 mice were subjected to inflammatory endotyping and therapeutic assays to characterize the allergic nature of PEA. Results PEA-specific IgE was identified in 17 (14.2 %) of 120 asthma patients. The rPEA-sensitized and challenged mice had increased PEA-specific immunoglobulins (such as IgE, IgG1 and IgG2a) and developed asthma-like phenotypes with airway hyperresponsiveness, severe airway inflammation, and airway remodeling. Lungs from these mice displayed significant increases in neutrophils and IL-17A+ cells. Innate lymphoid cells (ILCs) produced type 2 cytokines (IL-4, IL-5, and IL-13), whereas Th cells did not. Nonetheless, airway inflammation, rather than hyperresponsiveness, was elicited in non-sensitized mice upon challenge with rPEA. Importantly, rPEA-induced asthmatic mice were unresponsive to dexamethasone treatment. Conclusion PEA is a novel allergen that sensitizes asthmatic patients. Furthermore, mice developed steroid-resistant asthma, characterized by an atypical cytokine profile associated with non-TH2 inflammation, only after being sensitized and challenged with rPEA. These findings suggest a potentially significant role for PEA in asthma development, warranting consideration in clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Huancheng Xie
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Linmei Li
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Yuhe Guo
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Linghui Zhou
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Linyi Ma
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Andong He
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - He Lai
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Ying He
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Yongping Liu
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Huifang Chen
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Liping Luo
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Yuyi Huang
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Xiangyin Sha
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Huanping Zhang
- , Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Jie Yan
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| | - Qingling Zhang
- , Guangdong Provincial Key Laboratory of Allergy & Immunology, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ailin Tao
- , The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, 250 Changgang Road East, Guangzhou, 510260, China
| |
Collapse
|
9
|
Odunitan TT, Apanisile BT, Akinboade MW, Abdulazeez WO, Oyaronbi AO, Ajayi TM, Oyekola SA, Ibrahim NO, Nafiu T, Afolabi HO, Olayiwola DM, David OT, Adeyemo SF, Ayodeji OD, Akinade EM, Saibu OA. Microbial mysteries: Staphylococcus aureus and the enigma of carcinogenesis. Microb Pathog 2024; 194:106831. [PMID: 39089512 DOI: 10.1016/j.micpath.2024.106831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Staphylococcus aureus, a common human pathogen, has long been the focus of scientific investigation due to its association with various infections. However, recent research has unveiled a tantalizing enigma surrounding this bacterium and its potential involvement in carcinogenesis. Chronic S. aureus infections have been linked to an elevated risk of certain cancers, including skin cancer and oral cancer. This review explores the current state of knowledge regarding this connection, examining epidemiological evidence, pathogenic mechanisms, and biological interactions that suggest a correlation. Although initial studies point to a possible link, the precise mechanisms through which S. aureus may contribute to cancer development remain elusive. Emerging evidence suggests that the chronic inflammation induced by persistent S. aureus infections may create a tumor-promoting environment. This inflammation can lead to DNA damage, disrupt cellular signaling pathways, and generate an immunosuppressive microenvironment conducive to cancer progression. Additionally, S. aureus produces a variety of toxins and metabolites that can directly interact with host cells, potentially inducing oncogenic transformations. Despite these insights, significant gaps remain in our understanding of the exact biological processes involved. This review emphasizes the urgent need for more comprehensive research to clarify these microbiological mysteries. Understanding the role of S. aureus in cancer development could lead to novel strategies for cancer prevention and treatment, potentially transforming therapeutic approaches.
Collapse
Affiliation(s)
- Tope T Odunitan
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria; Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria; Ehigie's Biochemistry and Biocomputational Laboratory, Ogbomosho, Oyo State, Nigeria.
| | - Boluwatife T Apanisile
- Department of Nutrition and Dietetics, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Modinat W Akinboade
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Waliu O Abdulazeez
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adegboye O Oyaronbi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Temitope M Ajayi
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Samuel A Oyekola
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Najahtulahi O Ibrahim
- Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Tawakalitu Nafiu
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Hezekiah O Afolabi
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Dolapo M Olayiwola
- Department of Medical Laboratory Science, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oladunni T David
- Microbiology Unit, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Stephen F Adeyemo
- Department of Biological Sciences, First Technical University, Ibadan, Oyo State, Nigeria; Division of Medical Artificial Intelligence, Helix Biogen Institute, Ogbomosho, Oyo State, Nigeria
| | - Oluwatobi D Ayodeji
- Department of Nursing, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Esther M Akinade
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Oluwatosin A Saibu
- Department of Chemistry and Biochemistry, New Mexico State University, USA
| |
Collapse
|
10
|
De Donato DP, Effner R, Nordengrün M, Lechner A, Darisipudi MN, Volz T, Hagl B, Bröker BM, Renner ED. Staphylococcus aureus Serine protease-like protein A (SplA) induces IL-8 by keratinocytes and synergizes with IL-17A. Cytokine 2024; 180:156634. [PMID: 38810500 DOI: 10.1016/j.cyto.2024.156634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Serine protease-like (Spl) proteins produced by Staphylococcus (S.) aureus have been associated with allergic inflammation. However, effects of Spls on the epidermal immune response have not been investigated. OBJECTIVES To assess the epidermal immune response to SplA, SplD and SplE dependent on differentiation of keratinocytes and a Th2 or Th17 cytokine milieu. METHODS Human keratinocytes of healthy controls and a STAT3-hyper-IgE syndrome (STAT3-HIES) patient were cultured in different calcium concentrations in the presence of Spls and Th2 or Th17 cytokines. Keratinocyte-specific IL-8 production and concomitant migration of neutrophils were assessed. RESULTS SplE and more significantly SplA, induced IL-8 in keratinocytes. Suprabasal-like keratinocytes showed a higher Spl-mediated IL-8 production and neutrophil migration compared to basal-like keratinocytes. Th17 cytokines amplified Spl-mediated IL-8 production, which correlated with neutrophil recruitment. Neutrophil recruitment by keratinocytes of the STAT3-HIES patient was similar to healthy control cells. CONCLUSION S. aureus-specific Spl proteases synergized with IL-17A on human keratinocytes with respect to IL-8 release and neutrophil migration, highlighting the importance of keratinocytes and Th17 immunity in barrier function.
Collapse
Affiliation(s)
- D P De Donato
- Translational Immunology in Environmental Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany; Vascular Surgery, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - R Effner
- Translational Immunology in Environmental Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany; Translational Immunology, Faculty of Medicine, University of Augsburg, Germany
| | - M Nordengrün
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - A Lechner
- Translational Immunology in Environmental Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany; Translational Immunology, Faculty of Medicine, University of Augsburg, Germany
| | - M N Darisipudi
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - T Volz
- Department of Dermatology and Allergology, School of Medicine, Technical University of Munich, Munich, Germany
| | - B Hagl
- Translational Immunology in Environmental Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany; Translational Immunology, Faculty of Medicine, University of Augsburg, Germany
| | - B M Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - E D Renner
- Translational Immunology in Environmental Medicine, School of Medicine and Health, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Munich, German Research Center for Environmental Health, Neuherberg, Germany; Translational Immunology, Faculty of Medicine, University of Augsburg, Germany; Department of Pediatrics, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany.
| |
Collapse
|
11
|
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
|
12
|
Cullum E, Perez-Betancourt Y, Shi M, Gkika E, Schneewind O, Missiakas D, Golovkina T. Deficiency in non-classical major histocompatibility class II-like molecule, H2-O confers protection against Staphylococcus aureus in mice. PLoS Pathog 2024; 20:e1012306. [PMID: 38843309 PMCID: PMC11185455 DOI: 10.1371/journal.ppat.1012306] [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: 01/23/2024] [Revised: 06/18/2024] [Accepted: 05/29/2024] [Indexed: 06/19/2024] Open
Abstract
Staphylococcus aureus is a human-adapted pathogen that replicates by asymptomatically colonizing its host. S. aureus is also the causative agent of purulent skin and soft tissue infections as well as bloodstream infections that result in the metastatic seeding of abscess lesions in all organ tissues. Prolonged colonization, infection, disease relapse, and recurrence point to the versatile capacity of S. aureus to bypass innate and adaptive immune defenses as well as the notion that some hosts fail to generate protective immune responses. Here, we find a genetic trait that provides protection against this pathogen. Mice lacking functional H2-O, the equivalent of human HLA-DO, inoculated with a mouse-adapted strain of S. aureus, efficiently decolonize the pathogen. Further, these decolonized animals resist subsequent bloodstream challenge with methicillin-resistant S. aureus. A genetic approach demonstrates that T-cell dependent B cell responses are required to control S. aureus colonization and infection in H2-O-deficient mice. Reduced bacterial burdens in these animals correlate with increased titers and enhanced phagocytic activity of S. aureus-specific antibodies. H2-O negatively regulates the loading of high affinity peptides on major histocompatibility class II (MHC-II) molecules. Thus, we hypothesize that immune responses against S. aureus are derepressed in mice lacking H2-O because more high affinity peptides are presented by MHC-II. We speculate that loss-of-function HLA-DO alleles may similarly control S. aureus replication in humans.
Collapse
Affiliation(s)
- Emily Cullum
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
| | - Yunys Perez-Betancourt
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Miaomiao Shi
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Eirinaios Gkika
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Dominique Missiakas
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Howard T. Ricketts Laboratory, University of Chicago, Chicago, Illinois, United States of America
| | - Tatyana Golovkina
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Immunology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Microbiology, University of Chicago, Chicago, Illinois, United States of America
- Committee on Genetics, Genomics and System Biology, University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
13
|
van Dalen R, Elsherbini AMA, Harms M, Alber S, Stemmler R, Peschel A. Secretory IgA impacts the microbiota density in the human nose. MICROBIOME 2023; 11:233. [PMID: 37865781 PMCID: PMC10589987 DOI: 10.1186/s40168-023-01675-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/24/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Respiratory mucosal host defense relies on the production of secretory IgA (sIgA) antibodies, but we currently lack a fundamental understanding of how sIgA is induced by contact with microbes and how such immune responses may vary between humans. Defense of the nasal mucosal barrier through sIgA is critical to protect from infection and to maintain homeostasis of the microbiome, which influences respiratory disorders and hosts opportunistic pathogens. METHODS We applied IgA-seq analysis to nasal microbiota samples from male and female healthy volunteers, to identify which bacterial genera and species are targeted by sIgA on the level of the individual host. Furthermore, we used nasal sIgA from the same individuals in sIgA deposition experiments to validate the IgA-seq outcomes. CONCLUSIONS We observed that the amount of sIgA secreted into the nasal mucosa by the host varied substantially and was negatively correlated with the bacterial density, suggesting that nasal sIgA limits the overall bacterial capacity to colonize. The interaction between mucosal sIgA antibodies and the nasal microbiota was highly individual with no obvious differences between potentially invasive and non-invasive bacterial species. Importantly, we could show that for the clinically relevant opportunistic pathogen and frequent nasal resident Staphylococcus aureus, sIgA reactivity was in part the result of epitope-independent interaction of sIgA with the antibody-binding protein SpA through binding of sIgA Fab regions. This study thereby offers a first comprehensive insight into the targeting of the nasal microbiota by sIgA antibodies. It thereby helps to better understand the shaping and homeostasis of the nasal microbiome by the host and may guide the development of effective mucosal vaccines against bacterial pathogens. Video Abstract.
Collapse
Affiliation(s)
- Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- Present Address: Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ahmed M A Elsherbini
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Mareike Harms
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Svenja Alber
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Regine Stemmler
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
14
|
Bonzano L, Borgia F, Casella R, Miniello A, Nettis E, Gangemi S. Microbiota and IL-33/31 Axis Linkage: Implications and Therapeutic Perspectives in Atopic Dermatitis and Psoriasis. Biomolecules 2023; 13:1100. [PMID: 37509136 PMCID: PMC10377073 DOI: 10.3390/biom13071100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/24/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Microbiome dysbiosis and cytokine alternations are key features of atopic dermatitis (AD) and psoriasis (PsO), two of the most prevalent and burdensome pruritic skin conditions worldwide. Interleukin (IL)-33 and IL-31 have been recognized to be major players who act synergistically in the pathogenesis and maintenance of different chronic inflammatory conditions and pruritic skin disorders, including AD and PsO, and their potential role as therapeutic targets is being thoroughly investigated. The bidirectional interplay between dysbiosis and immunological changes has been extensively studied, but there is still debate regarding which of these two factors is the actual causative culprit behind the aetiopathological process that ultimately leads to AD and PsO. We conducted a literature review on the Pubmed database assessing articles of immunology, dermatology, microbiology and allergology with the aim to strengthen the hypothesis that dysbiosis is at the origin of the IL-33/IL-31 dysregulation that contributes to the pathogenesis of AD and PsO. Finally, we discussed the therapeutic options currently in development for the treatment of these skin conditions targeting IL-31, IL-33 and/or the microbiome.
Collapse
Affiliation(s)
- Laura Bonzano
- Dermatology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98122 Messina, Italy
| | - Rossella Casella
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70120 Bari, Italy
| | - Andrea Miniello
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70120 Bari, Italy
| | - Eustachio Nettis
- Department of Emergency and Organ Transplantation, School of Allergology and Clinical Immunology, University of Bari Aldo Moro, Policlinico di Bari, 70120 Bari, Italy
| | - Sebastiano Gangemi
- School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| |
Collapse
|
15
|
Xu Z, Yan J, Wen W, Zhang N, Bachert C. Pathophysiology and management of Staphylococcus aureus in nasal polyp disease. Expert Rev Clin Immunol 2023; 19:981-992. [PMID: 37409375 DOI: 10.1080/1744666x.2023.2233700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is a common pathogen that frequently colonizes the sinonasal cavity. Recent studies demonstrated the essential role of Staphylococcus aureus in the pathophysiology of uncontrolled severe chronic rhinosinusitis with nasal polyps (NP) by initiating an immune response to the germ and its products, resulting in type 2 inflammation. AREAS COVERED This review aims to summarize the evidence for the role of S. aureus in the development of NP disease including S. aureus-related virulence factors, the pathophysiologic mechanisms used by S. aureus, and the synergistic effects of S. aureus and other pathogens. It also describes the current management of S. aureus associated with NPs as well as potential therapeutic strategies that are used in clinical practice. EXPERT OPINION S. aureus is able to damage the nasal mucosal epithelial barrier, impair the clearance of the host immune system, and trigger adaptive and innate immune reactions which lead to the formation of inflammation and nasal polyp growth. Further studies should focus on the development of novel therapeutic strategies, such as biologics, bacteriophages, probiotics, and nanomedicine, which could be used to treat S. aureus and its immunological consequences in the future.
Collapse
Affiliation(s)
- Zhaofeng Xu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Jieying Yan
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Weiping Wen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Nan Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
- Division of ENT Diseases, Stockholm, Sweden
- Clinic for ENT Diseases and Head and Neck Surgery, University Clinic Münster, Münster, Germany
| |
Collapse
|
16
|
Won HK, Yoo Y, Lee J, Kang N, Lee JH, Choi JP, Kim TB, Cho SH, Song WJ. Clinical Relevance of Staphylococcus aureus Nasal Colonization and Staphylococcal Enterotoxin-Specific IgE Sensitization in Late-Onset Asthma. Lung 2023:10.1007/s00408-023-00624-8. [PMID: 37253986 DOI: 10.1007/s00408-023-00624-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/24/2023] [Indexed: 06/01/2023]
Abstract
This prospective observational study examined whether Staphylococcus aureus (SA) nasal colonization and staphylococcal enterotoxin (SE)-specific IgE sensitization synergistically affect clinical outcomes of adults with late-onset asthma (onset age ≥ 40 years). Nasal swabs were taken to evaluate SA colonization. Serum SE-IgE level was measured. Subjects were classified into 4 groups according to SA colonization and SE-IgE sensitization positivity. Among 181 patients with late-onset asthma recruited, the proportions of SA/SE (‒/‒), SA/SE (+ /‒), SA/SE (‒/ +), and SA/SE (+ / +) were 33.7%, 15.5%, 28.2%, and 22.6%, respectively. Severe asthma was more frequent in the SA/SE (+ / +) group than in the SA/SE (‒/‒) group (41.5% vs. 13.1%). The relationship of SA/SE (+ / +) with severe asthma was significant in multivariate logistic regression (vs. SA/SE (‒/‒); adjusted odds ratio: 4.36; 95% confidence intervals: 1.50‒12.73; p = 0.007), whereas SA/SE (+ /‒) or SA/SE (‒/ +) was not. In conclusion, SA nasal colonization and SE-IgE sensitization may synergistically affect disease severity in late-onset asthmatics.
Collapse
Affiliation(s)
- Ha-Kyeong Won
- Division of Pulmonology and Allergy, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Youngsang Yoo
- Department of Pulmonary, Allergy, and Critical Care Medicine, Gangneung Asan Hospital, Gangneung, Korea
| | - Jiwon Lee
- Institute of Allergy and Clinical Immunology, Asan Medical Research Center, Seoul, Korea
| | - Noeul Kang
- Division of Allergy, Department of Medicine, Samsung Medical Center, Seoul, Korea
| | - Ji-Hyang Lee
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
17
|
Al-Trad EI, Che Hamzah AM, Puah SM, Chua KH, Hanifah MZ, Ayub Q, Palittapongarnpim P, Kwong SM, Chew CH, Yeo CC. Complete Genome Sequence and Analysis of a ST573 Multidrug-Resistant Methicillin-Resistant Staphylococcus aureus SauR3 Clinical Isolate from Terengganu, Malaysia. Pathogens 2023; 12:pathogens12030502. [PMID: 36986424 PMCID: PMC10053073 DOI: 10.3390/pathogens12030502] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/17/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a World Health Organization-listed priority pathogen. Scarce genomic data are available for MRSA isolates from Malaysia. Here, we present the complete genome sequence of a multidrug-resistant MRSA strain SauR3, isolated from the blood of a 6-year-old patient hospitalized in Terengganu, Malaysia, in 2016. S. aureus SauR3 was resistant to five antimicrobial classes comprising nine antibiotics. The genome was sequenced on the Illumina and Oxford Nanopore platforms and hybrid assembly was performed to obtain its complete genome sequence. The SauR3 genome consists of a circular chromosome of 2,800,017 bp and three plasmids designated pSauR3-1 (42,928 bp), pSauR3-2 (3011 bp), and pSauR3-3 (2473 bp). SauR3 belongs to sequence type 573 (ST573), a rarely reported sequence type of the staphylococcal clonal complex 1 (CC1) lineage, and harbors a variant of the staphylococcal cassette chromosome mec (SCCmec) type V (5C2&5) element which also contains the aac(6')-aph(2″) aminoglycoside-resistance genes. pSauR3-1 harbors several antibiotic resistance genes in a 14,095 bp genomic island (GI), previously reported in the chromosome of other staphylococci. pSauR3-2 is cryptic, whereas pSauR3-3 encodes the ermC gene that mediates inducible resistance to macrolide-lincosamide-streptogramin B (iMLSB). The SauR3 genome can potentially be used as a reference genome for other ST573 isolates.
Collapse
Affiliation(s)
- Esra'a I Al-Trad
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| | | | - Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Muhamad Zarul Hanifah
- Monash University Malaysia Genomics Facility, School of Science, Monash University, Bandar Sunway 47500, Malaysia
| | - Qasim Ayub
- Monash University Malaysia Genomics Facility, School of Science, Monash University, Bandar Sunway 47500, Malaysia
| | - Prasit Palittapongarnpim
- Pornchai Matangkasombut Center for Microbial Genomics (CENMIG), Mahidol University, Bangkok 10400, Thailand
| | - Stephen M Kwong
- Infectious Diseases & Microbiology, School of Medicine, Western Sydney University, Campbelltown 2560, Australia
| | - Ching Hoong Chew
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Kuala Nerus 21300, Malaysia
| | - Chew Chieng Yeo
- Centre for Research in Infectious Diseases and Biotechnology (CeRIDB), Faculty of Medicine, Universiti Sultan Zainal Abidin, Kuala Terengganu 20400, Malaysia
| |
Collapse
|
18
|
Badolati I, van der Heiden M, Brodin D, Zuurveld M, Szilágyi S, Björkander S, Sverremark-Ekström E. Staphylococcus aureus-derived factors promote human Th9 cell polarization and enhance a transcriptional program associated with allergic inflammation. Eur J Immunol 2023; 53:e2250083. [PMID: 36550071 DOI: 10.1002/eji.202250083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/09/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
T helper (Th) 9 cells, characterized by robust secretion of IL-9, have been increasingly associated with allergic diseases. However, whether and how Th9 cells are modulated by environmental stimuli remains poorly understood. In this study, we show that in vitro exposure of human PBMCs or isolated CD4 T-cells to Staphylococcus (S.) aureus-derived factors, including its toxins, potently enhances Th9 cell frequency and IL-9 secretion. Furthermore, as revealed by RNA sequencing analysis, S. aureus increases the expression of Th9-promoting factors at the transcriptional level, such as FOXO1, miR-155, and TNFRSF4. The addition of retinoic acid (RA) dampens the Th9 responses promoted by S. aureus and substantially changes the transcriptional program induced by this bacterium, while also altering the expression of genes associated with allergic inflammation. Together, our results demonstrate a strong influence of microbial and dietary factors on Th9 cell polarization, which may be important in the context of allergy development and treatment.
Collapse
Affiliation(s)
- Isabella Badolati
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marieke van der Heiden
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - David Brodin
- Bioinformatics and Expression Analysis Core Facility, Karolinska Institutet, Huddinge, Sweden
| | - Marit Zuurveld
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Szilvia Szilágyi
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sophia Björkander
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
19
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
20
|
Valverde-Molina J, García-Marcos L. Microbiome and Asthma: Microbial Dysbiosis and the Origins, Phenotypes, Persistence, and Severity of Asthma. Nutrients 2023; 15:nu15030486. [PMID: 36771193 PMCID: PMC9921812 DOI: 10.3390/nu15030486] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
The importance of the microbiome, and of the gut-lung axis in the origin and persistence of asthma, is an ongoing field of investigation. The process of microbial colonisation in the first three years of life is fundamental for health, with the first hundred days of life being critical. Different factors are associated with early microbial dysbiosis, such as caesarean delivery, artificial lactation and antibiotic therapy, among others. Longitudinal cohort studies on gut and airway microbiome in children have found an association between microbial dysbiosis and asthma at later ages of life. A low α-diversity and relative abundance of certain commensal gut bacterial genera in the first year of life are associated with the development of asthma. Gut microbial dysbiosis, with a lower abundance of Phylum Firmicutes, could be related with increased risk of asthma. Upper airway microbial dysbiosis, especially early colonisation by Moraxella spp., is associated with recurrent viral infections and the development of asthma. Moreover, the bacteria in the respiratory system produce metabolites that may modify the inception of asthma and is progression. The role of the lung microbiome in asthma development has yet to be fully elucidated. Nevertheless, the most consistent finding in studies on lung microbiome is the increased bacterial load and the predominance of proteobacteria, especially Haemophilus spp. and Moraxella catarrhalis. In this review we shall update the knowledge on the association between microbial dysbiosis and the origins of asthma, as well as its persistence, phenotypes, and severity.
Collapse
Affiliation(s)
- José Valverde-Molina
- Department of Paediatrics, Santa Lucía General University Hospital, 30202 Cartagena, Spain
| | - Luis García-Marcos
- Paediatric Allergy and Pulmonology Units, Virgen de la Arrixaca University Children’s Hospital, University of Murcia and IMIB Biomedical Research Institute, 20120 Murcia, Spain
- Correspondence:
| |
Collapse
|
21
|
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
|
22
|
In Silico Genome-Scale Analysis of Molecular Mechanisms Contributing to the Development of a Persistent Infection with Methicillin-Resistant Staphylococcus aureus (MRSA) ST239. Int J Mol Sci 2022; 23:ijms232416086. [PMID: 36555727 PMCID: PMC9781258 DOI: 10.3390/ijms232416086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
The increasing frequency of isolation of methicillin-resistant Staphylococcus aureus (MRSA) limits the chances for the effective antibacterial therapy of staphylococcal diseases and results in the development of persistent infection such as bacteremia and osteomyelitis. The aim of this study was to identify features of the MRSAST239 0943-1505-2016 (SA943) genome that contribute to the formation of both acute and chronic musculoskeletal infections. The analysis was performed using comparative genomics data of the dominant epidemic S. aureus lineages, namely ST1, ST8, ST30, ST36, and ST239. The SA943 genome encodes proteins that provide resistance to the host's immune system, suppress immunological memory, and form biofilms. The molecular mechanisms of adaptation responsible for the development of persistent infection were as follows: amino acid substitution in PBP2 and PBP2a, providing resistance to ceftaroline; loss of a large part of prophage DNA and restoration of the nucleotide sequence of beta-hemolysin, that greatly facilitates the escape of phagocytosed bacteria from the phagosome and formation of biofilms; dysfunction of the AgrA system due to the presence of psm-mec and several amino acid substitutions in the AgrC; partial deletion of the nucleotide sequence in genomic island vSAβ resulting in the loss of two proteases of Spl-operon; and deletion of SD repeats in the SdrE amino acid sequence.
Collapse
|
23
|
Shin SH, Ye MK, Park J, Geum SY. Immunopathologic Role of Eosinophils in Eosinophilic Chronic Rhinosinusitis. Int J Mol Sci 2022; 23:ijms232113313. [PMID: 36362100 PMCID: PMC9658199 DOI: 10.3390/ijms232113313] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a diverse chronic inflammatory disease of the sinonasal mucosa. CRS manifests itself in a variety of clinical and immunologic patterns. The histological hallmark of eosinophilic CRS (ECRS) is eosinophil infiltration. ECRS is associated with severe disease severity, increased comorbidity, and a higher recurrence rate, as well as thick mucus production. Eosinophils play an important role in these ECRS clinical characteristics. Eosinophils are multipotential effector cells that contribute to host defense against nonphagocytable pathogens, as well as allergic and nonallergic inflammatory diseases. Eosinophils interact with Staphylococcus aureus, Staphylococcal enterotoxin B, and fungi, all of which were found in the tissue of CRS patients. These interactions activate Th2 immune responses in the sinonasal mucosa and exacerbate local inflammation. Activated eosinophils were discovered not only in the tissue but also in the sinonasal cavity secretion. Eosinophil extracellular traps (EETs) are extracellular microbes trapping and killing structures found in the secretions of CRS patients with intact granule protein and filamentous chromatic structures. At the same time, EET has a negative effect by causing an epithelial barrier defect. Eosinophils also influence the local tissue microenvironment by exchanging signals with other immune cells and structural cells. As a result, eosinophils are multifaceted leukocytes that contribute to various physiologic and pathologic processes of the upper respiratory mucosal immune system. The goal of this review is to summarize recent research on the immunopathologic properties and immunologic role of eosinophils in CRS.
Collapse
|
24
|
The Changes in Bacterial Microbiome Associated with Immune Disorder in Allergic Respiratory Disease. Microorganisms 2022; 10:microorganisms10102066. [PMID: 36296340 PMCID: PMC9610723 DOI: 10.3390/microorganisms10102066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/05/2022] [Accepted: 10/16/2022] [Indexed: 12/02/2022] Open
Abstract
Allergic respiratory disease is a worldwide and increasingly prevalent health problem. Many researchers have identified complex changes in the microbiota of the respiratory and intestinal tracts in patients with allergic respiratory diseases. These affect immune response and influence the progression of disease. However, the diversity of bacterial changes in such cases make it difficult to identify a specific microorganism to target for adjustment. Recent research evidence suggests that common bacterial variations present in allergic respiratory disease are associated with immune disorders. This finding could lead to the discovery of potential therapeutic targets in cases of allergic respiratory disease. In this review, we summarize current knowledge of bacteria changes in cases of allergic respiratory disease, to identify changes commonly associated with immune disorders, and thus provide a theoretical basis for targeting therapies of allergic respiratory disease through effective modulation of key bacteria.
Collapse
|
25
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
26
|
Shaghayegh G, Cooksley C, Ramezanpour M, Wormald PJ, Psaltis AJ, Vreugde S. Chronic Rhinosinusitis, S. aureus Biofilm and Secreted Products, Inflammatory Responses, and Disease Severity. Biomedicines 2022; 10:1362. [PMID: 35740385 PMCID: PMC9220248 DOI: 10.3390/biomedicines10061362] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a persistent inflammation of the nasal cavity and paranasal sinuses associated with tissue remodelling, dysfunction of the sinuses' natural defence mechanisms, and induction of different inflammatory clusters. The etiopathogenesis of CRS remains elusive, and both environmental factors, such as bacterial biofilms and the host's general condition, are thought to play a role. Bacterial biofilms have significant clinical relevance due to their potential to cause resistance to antimicrobial therapy and host defenses. Despite substantial medical advances, some CRS patients suffer from recalcitrant disease that is unresponsive to medical and surgical treatments. Those patients often have nasal polyps with tissue eosinophilia, S. aureus-dominant mucosal biofilm, comorbid asthma, and a severely compromised quality of life. This review aims to summarise the contemporary knowledge of inflammatory cells/pathways in CRS, the role of bacterial biofilm, and their impact on the severity of the disease. Here, an emphasis is placed on S. aureus biofilm and its secreted products. A better understanding of these factors might offer important diagnostic and therapeutic perceptions for recalcitrant disease.
Collapse
Affiliation(s)
- Gohar Shaghayegh
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Clare Cooksley
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Mahnaz Ramezanpour
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Peter-John Wormald
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Alkis James Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Sarah Vreugde
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| |
Collapse
|
27
|
Ma PY, Chong CW, Than LTL, Sulong AB, Ho KL, Neela VK, Sekawi Z, Liew YK. Impact of IsaA Gene Disruption: Decreasing Staphylococcal Biofilm and Alteration of Transcriptomic and Proteomic Profiles. Microorganisms 2022; 10:microorganisms10061119. [PMID: 35744637 PMCID: PMC9229027 DOI: 10.3390/microorganisms10061119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
Staphylococcus aureus expresses diverse proteins at different stages of growth. The immunodominant staphylococcal antigen A (IsaA) is one of the proteins that is constitutively produced by S. aureus during colonisation and infection. SACOL2584 (or isaA) is the gene that encodes this protein. It has been suggested that IsaA can hydrolyse cell walls, and there is still need to study isaA gene disruption to analyse its impact on staphylococcal phenotypes and on alteration to its transcription and protein profiles. In the present study, the growth curve in RPMI medium (which mimics human plasma), autolytic activity, cell wall morphology, fibronectin and fibrinogen adhesion and biofilm formation of S. aureus SH1000 (wildtype) was compared to that of S. aureus MS001 (isaA mutant). RNA sequencing and liquid chromatography–tandem mass spectrometry were carried out on samples of both S. aureus strains taken during the exponential growth phase, followed by bioinformatics analysis. Disruption of isaA had no obvious effect on the growth curve and autolysis ability or thickness of cell walls, but this study revealed significant strength of fibronectin adherence in S. aureus MS001. In particular, the isaA mutant formed less biofilm than S. aureus SH1000. In addition, proteomics and transcriptomics showed that the adhesin/biofilm-related genes and hemolysin genes, such as sasF, sarX and hlgC, were consistently downregulated with isaA gene disruption. The majority of the upregulated genes or proteins in S. aureus MS001 were pur genes. Taken together, this study provides insight into how isaA disruption changes the expression of other genes and has implications regarding biofilm formation and biological processes.
Collapse
Affiliation(s)
- Pei Yee Ma
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Anita Binti Sulong
- Department of Medical Microbiology and Immunology, Pusat Perubatan UKM, Kuala Lumpur 56000, Malaysia;
| | - Ket Li Ho
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia;
| | - Vasantha Kumari Neela
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Zamberi Sekawi
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43400, Malaysia; (L.T.L.T.); (V.K.N.); (Z.S.)
| | - Yun Khoon Liew
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur 57000, Malaysia;
- Correspondence:
| |
Collapse
|
28
|
Calabrese C, Seccia V, Pelaia C, Spinelli F, Morini P, Rizzi A, Detoraki A. S. aureus and IgE-mediated diseases: pilot or copilot? A narrative review. Expert Rev Clin Immunol 2022; 18:639-647. [PMID: 35507006 DOI: 10.1080/1744666x.2022.2074402] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION S. aureus is a major opportunistic pathogen that has been implicated in the pathogenesis of several chronic inflammatory diseases including bronchial asthma, chronic rhinosinusitis with nasal polyps (CRSwNP), chronic spontaneous urticaria (CSU), and atopic dermatitis. S. aureus can induce the production of both polyclonal and specific IgE that can elicit an inflammatory cascade. AREAS COVERED The link between the sensitization to S. aureus enterotoxins and the severity of several chronic inflammatory diseases is reviewed in detail, as well as its therapeutic implications. EXPERT OPINION An anti-IgE strategy to inhibit S. aureus enterotoxins would be a valid approach to treat several endotypes of severe asthma, CRSwNP and CSU in which IgE against S. aureus enterotoxins should represent, not only a marker of severity of the diseases but also a target of a treatment.
Collapse
Affiliation(s)
- Cecilia Calabrese
- Department of Translational Medical Sciences, Institute of Respiratory Diseases, University of Campania "L. Vanvitelli", Naples, Italy
| | - Veronica Seccia
- Otolaryngology Audiology, and Phoniatric Operative Unit, Department of Surgical, Medical, Molecular Pathology, and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Corrado Pelaia
- Department of Health Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | | | | | | | - Aikaterini Detoraki
- Division of Internal Medicine and Clinical Immunology, Department of Internal Medicine, Clinical Immunology, Clinical Pathology and Infectious Diseases, Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| |
Collapse
|
29
|
Farag AK, Roesner LM, Wieschowski S, Heratizadeh A, Eiz‐Vesper B, Kwok WW, Valenta R, Werfel T. Specific T cells targeting Staphylococcus aureus fibronectin-binding protein 1 induce a type 2/type 1 inflammatory response in sensitized atopic dermatitis patients. Allergy 2022; 77:1245-1253. [PMID: 34601735 PMCID: PMC11497299 DOI: 10.1111/all.15120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 08/13/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is one of the most common inflammatory skin diseases worldwide and Staphylococcus aureus colonization and secondary infections occur in the majority of AD patients. Allergic sensitizations against microbial antigens have been discussed as possible trigger factors of AD. Recently, we reported IgE sensitization against fibronectin-binding protein 1 (FBP1), an essential virulence component in S. aureus, in a subgroup of patients suffering from AD. To expand these findings by investigating delayed-type immune reactions, the objective of this study was to detect and phenotypically characterize FBP1-specific T cells as possible trigger factors in AD. METHODS Immunodominant T-cell epitopes were mapped by proliferation testing of patient-derived FBP1-specific T-cell lines after stimulation with single 15mer peptides, which were derived from different functional domains of the FBP1 sequence. Major histocompatibility complex class II tetramers carrying immunodominant epitopes successfully stained T helper cells in 8 out of 8 HLA-matched, IgE-sensitized AD patients. RESULTS Cytokine profiling of multimer-sorted cells revealed that predominantly the type 2 cytokines IL-13 and IL-4 were secreted by these cells. In contrast, IL-17, the marker cytokine for response to extracellular pathogens, was scarcely detectable. CONCLUSIONS We demonstrate that FBP1 contains immunodominant peptides that induce a specific pro-inflammatory T helper cell response with increased Th2 levels that can drive an allergic inflammation in sensitized AD patients.
Collapse
Affiliation(s)
- Ahmed K. Farag
- Division of Dermatology and AllergyDepartment of Immunodermatology and Allergy ResearchHannover Medical SchoolHannoverGermany
| | - Lennart M. Roesner
- Division of Dermatology and AllergyDepartment of Immunodermatology and Allergy ResearchHannover Medical SchoolHannoverGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical SchoolHannoverGermany
| | - Susanne Wieschowski
- Division of Dermatology and AllergyDepartment of Immunodermatology and Allergy ResearchHannover Medical SchoolHannoverGermany
| | - Annice Heratizadeh
- Division of Dermatology and AllergyDepartment of Immunodermatology and Allergy ResearchHannover Medical SchoolHannoverGermany
| | - Britta Eiz‐Vesper
- Institute of Transfusion Medicine and Transplant EngineeringHannover Medical SchoolHannoverGermany
| | - William W. Kwok
- Benaroya Research Institute at Virginia MasonSeattleWashingtonUSA
- Department of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Rudolf Valenta
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- NRC Institute of Immunology FMBA of RussiaMoscowRussia
- Department of Clinical Immunology and AllergySechenov First Moscow State Medical UniversityMoscowRussia
- Karl Landsteiner University of Health SciencesKremsAustria
| | - Thomas Werfel
- Division of Dermatology and AllergyDepartment of Immunodermatology and Allergy ResearchHannover Medical SchoolHannoverGermany
- Cluster of Excellence RESIST (EXC 2155)Hannover Medical SchoolHannoverGermany
| |
Collapse
|
30
|
Krueger A, Zaugg J, Chisholm S, Linedale R, Lachner N, Teoh SM, Tuong ZK, Lukowski SW, Morrison M, Soyer HP, Hugenholtz P, Hill MM, Frazer IH. Secreted Toxins From Staphylococcus aureus Strains Isolated From Keratinocyte Skin Cancers Mediate Pro-tumorigenic Inflammatory Responses in the Skin. Front Microbiol 2022; 12:789042. [PMID: 35145494 PMCID: PMC8822148 DOI: 10.3389/fmicb.2021.789042] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/08/2021] [Indexed: 12/27/2022] Open
Abstract
Squamous cell carcinoma (SCC) is a common type of skin cancer that typically arises from premalignant precursor lesions named actinic keratoses (AK). Chronic inflammation is a well-known promoter of skin cancer progression. AK and SCC have been associated with an overabundance of the bacterium Staphylococcus aureus (S. aureus). Certain secreted products from S. aureus are known to promote cutaneous pro-inflammatory responses; however, not all S. aureus strains produce these. As inflammation plays a key role in SCC development, we investigated the pro-inflammatory potential and toxin secretion profiles of skin-cancer associated S. aureus. Sterile culture supernatants (“secretomes”) of S. aureus clinical strains isolated from AK and SCC were applied to human keratinocytes in vitro. Some S. aureus secretomes induced keratinocytes to overexpress inflammatory mediators that have been linked to skin carcinogenesis, including IL-6, IL-8, and TNFα. A large phenotypic variation between the tested clinical strains was observed. Strains that are highly pro-inflammatory in vitro also caused more pronounced skin inflammation in mice. Proteomic characterization of S. aureus secretomes using mass spectrometry established that specific S. aureus enzymes and cytolytic toxins, including hemolysins, phenol-soluble modulins, and serine proteases, as well as currently uncharacterized proteins, correlate with the pro-inflammatory S. aureus phenotype. This study is the first to describe the toxin secretion profiles of AK and SCC-associated S. aureus, and their potential to induce a pro-inflammatory environment in the skin. Further studies are needed to establish whether these S. aureus products promote SCC development by mediating chronic inflammation.
Collapse
Affiliation(s)
- Annika Krueger
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Julian Zaugg
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Sarah Chisholm
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Richard Linedale
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Nancy Lachner
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Siok Min Teoh
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Zewen K. Tuong
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Samuel W. Lukowski
- The Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Mark Morrison
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - H. Peter Soyer
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Philip Hugenholtz
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD, Australia
| | - Michelle M. Hill
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Ian H. Frazer
- Faculty of Medicine, The University of Queensland Diamantina Institute, Translational Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- *Correspondence: Ian H. Frazer,
| |
Collapse
|
31
|
Naeem M, Manzoor S, Abid MUH, Tareen MBK, Asad M, Mushtaq S, Ehsan N, Amna D, Xu B, Hazafa A. Fungal Proteases as Emerging Biocatalysts to Meet the Current Challenges and Recent Developments in Biomedical Therapies: An Updated Review. J Fungi (Basel) 2022; 8:109. [PMID: 35205863 PMCID: PMC8875690 DOI: 10.3390/jof8020109] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 02/07/2023] Open
Abstract
With the increasing world population, demand for industrialization has also increased to fulfill humans' living standards. Fungi are considered a source of essential constituents to produce the biocatalytic enzymes, including amylases, proteases, lipases, and cellulases that contain broad-spectrum industrial and emerging applications. The present review discussed the origin, nature, mechanism of action, emerging aspects of genetic engineering for designing novel proteases, genome editing of fungal strains through CRISPR technology, present challenges and future recommendations of fungal proteases. The emerging evidence revealed that fungal proteases show a protective role to many environmental exposures and discovered that an imbalance of protease inhibitors and proteases in the epithelial barriers leads to the protection of chronic eosinophilic airway inflammation. Moreover, mitoproteases recently were found to execute intense proteolytic processes that are crucial for mitochondrial integrity and homeostasis function, including mitochondrial biogenesis, protein synthesis, and apoptosis. The emerging evidence revealed that CRISPR/Cas9 technology had been successfully developed in various filamentous fungi and higher fungi for editing of specific genes. In addition to medical importance, fungal proteases are extensively used in different industries such as foods to prepare butter, fruits, juices, and cheese, and to increase their shelf life. It is concluded that hydrolysis of proteins in industries is one of the most significant applications of fungal enzymes that led to massive usage of proteomics.
Collapse
Affiliation(s)
- Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang 050025, China;
| | - Saba Manzoor
- Department of Zoology, University of Sialkot, Sialkot 51310, Pakistan;
| | | | | | - Mirza Asad
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| | - Sajida Mushtaq
- Department of Zoology, Government College Women University, Sialkot 51040, Pakistan;
| | - Nazia Ehsan
- Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| | - Dua Amna
- Institute of Food Science & Nutrition, Bahauddin Zakariya University, Multan 60800, Pakistan;
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University (BNU-HKBU) United International College, Zhuhai 519087, China
| | - Abu Hazafa
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38040, Pakistan;
| |
Collapse
|
32
|
Adamovich SN, Ushakov IA, Oborina EN, Vashchenko AV. Silatrane-sulfonamide hybrids: Synthesis, characterization, and evaluation of biological activity. J Organomet Chem 2022. [DOI: 10.1016/j.jorganchem.2021.122150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
33
|
Jeong J, Lee HK. The Role of CD4 + T Cells and Microbiota in the Pathogenesis of Asthma. Int J Mol Sci 2021; 22:11822. [PMID: 34769255 PMCID: PMC8584410 DOI: 10.3390/ijms222111822] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
Asthma, a chronic respiratory disease involving variable airflow limitations, exhibits two phenotypes: eosinophilic and neutrophilic. The asthma phenotype must be considered because the prognosis and drug responsiveness of eosinophilic and neutrophilic asthma differ. CD4+ T cells are the main determinant of asthma phenotype. Th2, Th9 and Tfh cells mediate the development of eosinophilic asthma, whereas Th1 and Th17 cells mediate the development of neutrophilic asthma. Elucidating the biological roles of CD4+ T cells is thus essential for developing effective asthma treatments and predicting a patient's prognosis. Commensal bacteria also play a key role in the pathogenesis of asthma. Beneficial bacteria within the host act to suppress asthma, whereas harmful bacteria exacerbate asthma. Recent literature indicates that imbalances between beneficial and harmful bacteria affect the differentiation of CD4+ T cells, leading to the development of asthma. Correcting bacterial imbalances using probiotics reportedly improves asthma symptoms. In this review, we investigate the effects of crosstalk between the microbiota and CD4+ T cells on the development of asthma.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
34
|
The protease SplB of Staphylococcus aureus targets host complement components and inhibits complement-mediated bacterial opsonophagocytosis. J Bacteriol 2021; 204:e0018421. [PMID: 34633872 PMCID: PMC8765433 DOI: 10.1128/jb.00184-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that can cause life-threatening infections, particularly in immunocompromised individuals. The high-level virulence of S. aureus largely relies on its diverse and variable collection of virulence factors and immune evasion proteins, including the six serine protease-like proteins SplA to SplF. Spl proteins are expressed by most clinical isolates of S. aureus, but little is known about the molecular mechanisms by which these proteins modify the host’s immune response for the benefit of the bacteria. Here, we identify SplB as a protease that inactivates central human complement proteins, i.e., C3, C4, and the activation fragments C3b and C4b, by preferentially cleaving their α-chains. SplB maintained its proteolytic activity in human serum, degrading C3 and C4. SplB further cleaved the components of the terminal complement pathway, C5, C6, C7, C8, and C9. In contrast, the important soluble human complement regulators factor H and C4b-binding protein (C4BP), as well as C1q, were left intact. Thereby, SplB reduced C3b-mediated opsonophagocytosis by human neutrophils as well as C5b-9 deposition on the bacterial surface. In conclusion, we identified the first physiological substrates of the S. aureus extracellular protease SplB. This enzyme inhibits all three complement pathways and blocks opsonophagocytosis. Thus, SplB can be considered a novel staphylococcal complement evasion protein. IMPORTANCE The success of bacterial pathogens in immunocompetent humans depends on the control and inactivation of host immunity. S. aureus, like many other pathogens, efficiently blocks host complement attack early in infection. Aiming to understand the role of the S. aureus-encoded orphan proteases of the Spl operon, we asked whether these proteins play a role in immune escape. We found that SplB inhibits all three complement activation pathways as well as the lytic terminal complement pathway. This blocks the opsonophagocytosis of the bacteria by neutrophils. We also clarified the molecular mechanisms: SplB cleaves the human complement proteins C3, C4, C5, C6, C7, C8, and C9 as well as factor B but not the complement inhibitors factor H and C4BP. Thus, we identify the first physiological substrates of the extracellular protease SplB of S. aureus and characterize SplB as a novel staphylococcal complement evasion protein.
Collapse
|
35
|
Abstract
The human skin is our outermost layer and serves as a protective barrier against external insults. Advances in next generation sequencing have enabled the discoveries of a rich and diverse community of microbes - bacteria, fungi and viruses that are residents of this surface. The genomes of these microbes also revealed the presence of many secretory enzymes. In particular, proteases which are hydrolytic enzymes capable of protein cleavage and degradation are of special interest in the skin environment which is enriched in proteins and lipids. In this minireview, we will focus on the roles of these skin-relevant microbial secreted proteases, both in terms of their widely studied roles as pathogenic agents in tissue invasion and host immune inactivation, and their recently discovered roles in inter-microbial interactions and modulation of virulence factors. From these studies, it has become apparent that while microbial proteases are capable of a wide range of functions, their expression is tightly regulated and highly responsive to the environments the microbes are in. With the introduction of new biochemical and bioinformatics tools to study protease functions, it will be important to understand the roles played by skin microbial secretory proteases in cutaneous health, especially the less studied commensal microbes with an emphasis on contextual relevance.
Collapse
|
36
|
Patel N, Nair M. The small RNA RsaF regulates the expression of secreted virulence factors in Staphylococcus aureus Newman. J Microbiol 2021; 59:920-930. [PMID: 34554453 DOI: 10.1007/s12275-021-1205-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/26/2022]
Abstract
The pathogenesis of Staphylococcus aureus, from local infections to systemic dissemination, is mediated by a battery of virulence factors that are regulated by intricate mechanisms, which include regulatory proteins and small RNAs (sRNAs) as key regulatory molecules. We have investigated the involvement of sRNA RsaF, in the regulation of pathogenicity genes hyaluronate lyase (hysA) and serine proteaselike protein D (splD), by employing S. aureus strains with disruption and overexpression of rsaF. Staphylococcus aureus strain with disruption of rsaF exhibited marked down-regulation of hysA transcripts by 0.2 to 0.0002 fold, and hyaluronate lyase activity by 0.2-0.1 fold, as well as increased biofilm formation, during growth from log phase to stationery phase. These mutants also displayed down-regulation of splD transcripts by 0.8 to 0.005 fold, and reduced activity of multiple proteases by zymography. Conversely, overexpression of rsaF resulted in a 2- to 4- fold increase in hysA mRNA levels and hyaluronidase activity. Both hysA and splD mRNAs demonstrated an increased stability in RsaF+ strains. In silico RNA-RNA interaction indicated a direct base pairing of RsaF with hysA and splD mRNAs, which was established in electrophoretic mobility shift assays. The findings demonstrate a positive regulatory role for small RNA RsaF in the expression of the virulence factors, HysA and SplD.
Collapse
Affiliation(s)
- Niralee Patel
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - Mrinalini Nair
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
37
|
Renz H, Bachert C, Berek C, Hamelmann E, Levi‐Schaffer F, Raap U, Simon H, Ploetz S, Taube C, Valent P, Voehringer D, Werfel T, Zhang N, Ring J. Physiology and pathology of eosinophils: Recent developments: Summary of the Focus Workshop Organized by DGAKI. Scand J Immunol 2021; 93:e13032. [PMID: 33624312 PMCID: PMC11475402 DOI: 10.1111/sji.13032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/10/2021] [Accepted: 02/21/2021] [Indexed: 12/15/2022]
Abstract
Over the last century, eosinophils have been regarded ambiguously either as 'friends' or 'foes'. Recent developments have greatly enhanced our understanding of the role and function of eosinophils in health and disease. Pathogenic eosinophilic inflammation can lead to severe diseases in various organs, such as the gastrointestinal tract, airways, heart and skin. In a 2-day focus workshop of the German Society for Allergology and Clinical Immunology (DGAKI), the state of the art was discussed and practical recommendations for diagnosis and treatment of eosinophilic diseases, with a particular focus on new biologics, such as anti-interleukin 5 and anti-interleukin 5R, were derived.
Collapse
Affiliation(s)
- Harald Renz
- Institute of Laboratory MedicineUniversities of Giessen and Marburg Lung Center (UGMLC)German Center for Lung Research (DZL)Philipps Universität MarburgMarburgGermany
| | - Claus Bachert
- Upper Airways Research Laboratory and Department of Oto‐Rhino‐LaryngologyGhent University and Ghent University HospitalGhentBelgium
- Division of ENT DiseasesCLINTECKarolinska InstituteUniversity of StockholmStockholmSweden
| | - Claudia Berek
- Deutsches Rheuma ForschungszentrumEin Institut der LeibnizgemeinschaftBerlinGermany
| | - Eckard Hamelmann
- Klinik für Kinder‐ und JugendmedizinEvangelisches Klinikum BethelBielefeldGermany
- Allergy Center of the Ruhr UniversityBochumGermany
| | - Francesca Levi‐Schaffer
- School of PharmacyFaculty of MedicineThe Institute for Drug ResearchThe Hebrew University of JerusalemIsrael
| | - Ulrike Raap
- Clinics of Dermatology and AllergyFaculty of Medical Health and SciencesUniversity of OldenburgGermany
| | - Hans‐Uwe Simon
- Institute of PharmacologyUniversity of BernBernSwitzerland
| | | | - Christian Taube
- Department of Pulmonary MedicineUniversity Hospital Essen—RuhrlandklinikEssenGermany
| | - Peter Valent
- Department of Internal Medicine IDivision of Hematology and Hemostaseology, and Ludwig Boltzmann Institute for Hematology & OncologyMedical University of ViennaViennaAustria
| | - David Voehringer
- Department of Infection BiologyUniversity Hospital Erlangen and Friedrich‐Alexander University Erlangen‐NurembergErlangenGermany
| | - Thomas Werfel
- Klinik für DermatologieAllergologie und VenerologieMedizinische Hochschule HannoverHannoverGermany
| | - Nan Zhang
- Upper Airways Research Laboratory and Department of Oto‐Rhino‐LaryngologyGhent University and Ghent University HospitalGhentBelgium
| | - Johannes Ring
- Deptment of Dermatology and Allergology BiedersteinTechnical University Munich (TUM)MunichGermany
| |
Collapse
|
38
|
Th2 inflammatory responses in the development of nasal polyps and chronic rhinosinusitis. Curr Opin Allergy Clin Immunol 2021; 20:1-8. [PMID: 31567293 DOI: 10.1097/aci.0000000000000588] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
PURPOSE OF REVIEW Pathogenesis of nasal polyp has been largely studied based on innate and adaptive immunity of sinonasal mucosa. So far, various factors have been identified that trigger an inflammatory response in the pathogenesis of nasal polyps. In this review, we summarized recently updated information in the understanding of mechanisms in the development of chronic rhinosinusitis with nasal polyp (CRSwNP) focusing on Th2 inflammation. RECENT FINDINGS Endotype of CRSwNP presented mainly Th2-skewed inflammation, and it has been associated with refractoriness and comorbidities. Staphylococcus aureus can drive Th2 inflammation by producing enterotoxins and serine protease-like protein. Moreover, S. aureus directly affected mucosal barrier function and enhanced Th2 cytokine production by fast induction of epithelial-derived innate cytokines. Epithelial-derived innate cytokines, including TSLP, IL-25, and IL-33, promote Th2 responses via the development of innate lymphoid cells. Mast cell expresses IL-5, IL-13, and periostin, and it plays a role in the pathogenesis of nasal polyps through orchestrating eosinophil infiltration. Formation of eosinophil extracellular traps and Charcot-Leyden crystals is strongly associated with disease severity and viscous mucus plug production. Therefore, it needs to be investigated mechanistically. The role of neutrophils in Th2 inflammation has been poorly understood but appears to enhance Th2 inflammation and make it more resistant to steroid therapy. SUMMARY There is growing evidence of the role of S. aureus in innate and adaptive immunity, which contribute to Th2 inflammation in CRSwNP. Innate immunity, including epithelial-derived cytokines, plays a crucial role in the development of CRSwNP by inducing various pathways and need to be investigated more as Th2-targeted biomarkers. Recently, the role of neutrophilic inflammation in Th2 inflammation has started to be studied but still remains unclear.
Collapse
|
39
|
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
|
40
|
Frey AM, Chaput D, Shaw LN. Insight into the human pathodegradome of the V8 protease from Staphylococcus aureus. Cell Rep 2021; 35:108930. [PMID: 33826899 PMCID: PMC8054439 DOI: 10.1016/j.celrep.2021.108930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/03/2020] [Accepted: 03/11/2021] [Indexed: 12/03/2022] Open
Abstract
Staphylococcus aureus possesses ten extracellular proteases with mostly unknown targets in the human proteome. To assist with bacterial protease target discovery, we have applied and compared two N-terminomics methods to investigate cleavage of human serum proteins by S. aureus V8 protease, discovering 85 host-protein targets. Among these are virulence-relevant complement, iron sequestration, clotting cascade, and host protease inhibitor proteins. Protein cleavage sites have been identified, providing insight into the disruption of host protein function by V8. Complement proteins are cleaved within peptidase and sushi domains, and host protease inhibitors are cleaved outside their protease-trapping motifs. Our data highlight the potential for further application of N-terminomics in discovery of bacterial protease substrates in other host niches and provide omics-scale insight into the role of the V8 protease in S. aureus pathogenesis. S. aureus-secreted proteases are central to disease causation, but the discovery of their host substrates has been limited. Frey et al. use N-terminomic approaches to uncover human serum targets of the V8 protease that are from virulence-relevant processes such as the host inflammatory network and nutrient sequestration.
Collapse
Affiliation(s)
- Andrew Michael Frey
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA
| | - Lindsey Neil Shaw
- Department of Cell Biology, Microbiology & Molecular Biology, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
41
|
Nordengrün M, Abdurrahman G, Treffon J, Wächter H, Kahl BC, Bröker BM. Allergic Reactions to Serine Protease-Like Proteins of Staphylococcus aureus. Front Immunol 2021; 12:651060. [PMID: 33833764 PMCID: PMC8021911 DOI: 10.3389/fimmu.2021.651060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/08/2021] [Indexed: 11/13/2022] Open
Abstract
In cystic fibrosis (CF) infectious and allergic airway inflammation cause pulmonary exacerbations that destroy the lungs. Staphylococcus aureus is a common long-term colonizer and cause of recurrent airway infections in CF. The pathogen is also associated with respiratory allergy; especially the staphylococcal serine protease-like proteins (Spls) can induce type 2 immune responses in humans and mice. We measured the serum IgE levels specific to 7 proteases of S. aureus by ELISA, targeting 5 Spls (76 CF patients and 46 controls) and the staphopains A and B (16 CF patients and 46 controls). Then we compared cytokine release and phenotype of T cells that had been stimulated with Spls between 5 CF patients and 5 controls. CF patients had strongly increased serum IgE binding to all Spls but not to the staphopains. Compared to healthy controls, their Spl-stimulated T cells released more type 2 cytokines (IL-4, IL-5, IL-13) and more IL-6 with no difference in the secretion of type 1- or type 3 cytokines (IFNγ, IL-17A, IL-17F). IL-10 production was low in CF T cells. The phenotype of the Spl-exposed T cells shifted towards a Th2 or Th17 profile in CF but to a Th1 profile in controls. Sensitization to S. aureus Spls is common in CF. This discovery could explain episodes of allergic inflammation of hitherto unknown causation in CF and extend the diagnostic and therapeutic portfolio.
Collapse
Affiliation(s)
- Maria Nordengrün
- Department of Immunology, Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Goran Abdurrahman
- Department of Immunology, Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Janina Treffon
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany.,Institute of Hygiene, University Hospital Münster, Münster, Germany
| | - Hannah Wächter
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Barbara C Kahl
- Institute of Medical Microbiology, University Hospital Münster, Münster, Germany
| | - Barbara M Bröker
- Department of Immunology, Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
42
|
Orlandi RR, Kingdom TT, Smith TL, Bleier B, DeConde A, Luong AU, Poetker DM, Soler Z, Welch KC, Wise SK, Adappa N, Alt JA, Anselmo-Lima WT, Bachert C, Baroody FM, Batra PS, Bernal-Sprekelsen M, Beswick D, Bhattacharyya N, Chandra RK, Chang EH, Chiu A, Chowdhury N, Citardi MJ, Cohen NA, Conley DB, DelGaudio J, Desrosiers M, Douglas R, Eloy JA, Fokkens WJ, Gray ST, Gudis DA, Hamilos DL, Han JK, Harvey R, Hellings P, Holbrook EH, Hopkins C, Hwang P, Javer AR, Jiang RS, Kennedy D, Kern R, Laidlaw T, Lal D, Lane A, Lee HM, Lee JT, Levy JM, Lin SY, Lund V, McMains KC, Metson R, Mullol J, Naclerio R, Oakley G, Otori N, Palmer JN, Parikh SR, Passali D, Patel Z, Peters A, Philpott C, Psaltis AJ, Ramakrishnan VR, Ramanathan M, Roh HJ, Rudmik L, Sacks R, Schlosser RJ, Sedaghat AR, Senior BA, Sindwani R, Smith K, Snidvongs K, Stewart M, Suh JD, Tan BK, Turner JH, van Drunen CM, Voegels R, Wang DY, Woodworth BA, Wormald PJ, Wright ED, Yan C, Zhang L, Zhou B. International consensus statement on allergy and rhinology: rhinosinusitis 2021. Int Forum Allergy Rhinol 2021; 11:213-739. [PMID: 33236525 DOI: 10.1002/alr.22741] [Citation(s) in RCA: 514] [Impact Index Per Article: 128.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
I. EXECUTIVE SUMMARY BACKGROUND: The 5 years since the publication of the first International Consensus Statement on Allergy and Rhinology: Rhinosinusitis (ICAR-RS) has witnessed foundational progress in our understanding and treatment of rhinologic disease. These advances are reflected within the more than 40 new topics covered within the ICAR-RS-2021 as well as updates to the original 140 topics. This executive summary consolidates the evidence-based findings of the document. METHODS ICAR-RS presents over 180 topics in the forms of evidence-based reviews with recommendations (EBRRs), evidence-based reviews, and literature reviews. The highest grade structured recommendations of the EBRR sections are summarized in this executive summary. RESULTS ICAR-RS-2021 covers 22 topics regarding the medical management of RS, which are grade A/B and are presented in the executive summary. Additionally, 4 topics regarding the surgical management of RS are grade A/B and are presented in the executive summary. Finally, a comprehensive evidence-based management algorithm is provided. CONCLUSION This ICAR-RS-2021 executive summary provides a compilation of the evidence-based recommendations for medical and surgical treatment of the most common forms of RS.
Collapse
Affiliation(s)
| | | | | | | | | | - Amber U Luong
- University of Texas Medical School at Houston, Houston, TX
| | | | - Zachary Soler
- Medical University of South Carolina, Charleston, SC
| | - Kevin C Welch
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | | | | | | | - Claus Bachert
- Ghent University, Ghent, Belgium.,Karolinska Institute, Stockholm, Sweden.,Sun Yatsen University, Gangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David A Gudis
- Columbia University Irving Medical Center, New York, NY
| | - Daniel L Hamilos
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | - Richard Harvey
- University of New South Wales and Macquarie University, Sydney, New South Wales, Australia
| | | | | | | | | | - Amin R Javer
- University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | - Valerie Lund
- Royal National Throat Nose and Ear Hospital, UCLH, London, UK
| | - Kevin C McMains
- Uniformed Services University of Health Sciences, San Antonio, TX
| | | | - Joaquim Mullol
- IDIBAPS Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | - Alkis J Psaltis
- University of Adelaide, Adelaide, South Australia, Australia
| | | | | | | | - Luke Rudmik
- University of Calgary, Calgary, Alberta, Canada
| | - Raymond Sacks
- University of New South Wales, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | - De Yun Wang
- National University of Singapore, Singapore, Singapore
| | | | | | | | - Carol Yan
- University of California San Diego, La Jolla, CA
| | - Luo Zhang
- Capital Medical University, Beijing, China
| | - Bing Zhou
- Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Structural Determinants of Substrate Specificity of SplF Protease from Staphylococcus aureus. Int J Mol Sci 2021; 22:ijms22042220. [PMID: 33672341 PMCID: PMC7926377 DOI: 10.3390/ijms22042220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/16/2021] [Accepted: 02/19/2021] [Indexed: 11/17/2022] Open
Abstract
Accumulating evidence suggests that six proteases encoded in the spl operon of a dangerous human pathogen, Staphylococcus aureus, may play a role in virulence. Interestingly, SplA, B, D, and E have complementary substrate specificities while SplF remains to be characterized in this regard. Here, we describe the prerequisites of a heterologous expression system for active SplF protease and characterize the enzyme in terms of substrate specificity and its structural determinants. Substrate specificity of SplF is comprehensively profiled using combinatorial libraries of peptide substrates demonstrating strict preference for long aliphatic sidechains at the P1 subsite and significant selectivity for aromatic residues at P3. The crystal structure of SplF was provided at 1.7 Å resolution to define the structural basis of substrate specificity of SplF. The obtained results were compared and contrasted with the characteristics of other Spl proteases determined to date to conclude that the spl operon encodes a unique extracellular proteolytic system.
Collapse
|
44
|
Cristinziano L, Poto R, Criscuolo G, Ferrara AL, Galdiero MR, Modestino L, Loffredo S, de Paulis A, Marone G, Spadaro G, Varricchi G. IL-33 and Superantigenic Activation of Human Lung Mast Cells Induce the Release of Angiogenic and Lymphangiogenic Factors. Cells 2021; 10:cells10010145. [PMID: 33445787 PMCID: PMC7828291 DOI: 10.3390/cells10010145] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
Human lung mast cells (HLMCs) express the high-affinity receptor FcεRI for IgE and are strategically located in different compartments of human lung, where they play a role in several inflammatory disorders and cancer. Immunoglobulin superantigens (e.g., protein A of Staphylococcus aureus and protein L of Peptostreptococcus magnus) bind to the variable regions of either the heavy (VH3) or light chain (κ) of IgE. IL-33 is a cytokine expressed by epithelial cells that exerts pleiotropic functions in the lung. The present study investigated whether immunoglobulin superantigens protein A and protein L and IL-33 caused the release of inflammatory (histamine), angiogenic (VEGF-A) and lymphangiogenic (VEGF-C) factors from HLMCs. The results show that protein A and protein L induced the rapid (30 min) release of preformed histamine from HLMCs. By contrast, IL-33 did not induce the release of histamine from lung mast cells. Prolonged incubation (12 h) of HLMCs with superantigens and IL-33 induced the release of VEGF-A and VEGF-C. Preincubation with IL-33 potentiated the superantigenic release of histamine, angiogenic and lymphangiogenic factors from HLMCs. Our results suggest that IL-33 might enhance the inflammatory, angiogenic and lymphangiogenic activities of lung mast cells in pulmonary disorders.
Collapse
Affiliation(s)
- Leonardo Cristinziano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
45
|
Lyly A, Laulajainen-Hongisto A, Gevaert P, Kauppi P, Toppila-Salmi S. Monoclonal Antibodies and Airway Diseases. Int J Mol Sci 2020; 21:E9477. [PMID: 33322143 PMCID: PMC7763928 DOI: 10.3390/ijms21249477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies, biologics, are a relatively new treatment option for severe chronic airway diseases, asthma, allergic rhinitis, and chronic rhinosinusitis (CRS). In this review, we focus on the physiological and pathomechanisms of monoclonal antibodies, and we present recent study results regarding their use as a therapeutic option against severe airway diseases. Airway mucosa acts as a relative barrier, modulating antigenic stimulation and responding to environmental pathogen exposure with a specific, self-limited response. In severe asthma and/or CRS, genome-environmental interactions lead to dysbiosis, aggravated inflammation, and disease. In healthy conditions, single or combined type 1, 2, and 3 immunological response pathways are invoked, generating cytokine, chemokine, innate cellular and T helper (Th) responses to eliminate viruses, helminths, and extracellular bacteria/fungi, correspondingly. Although the pathomechanisms are not fully known, the majority of severe airway diseases are related to type 2 high inflammation. Type 2 cytokines interleukins (IL) 4, 5, and 13, are orchestrated by innate lymphoid cell (ILC) and Th subsets leading to eosinophilia, immunoglobulin E (IgE) responses, and permanently impaired airway damage. Monoclonal antibodies can bind or block key parts of these inflammatory pathways, resulting in less inflammation and improved disease control.
Collapse
Affiliation(s)
- Annina Lyly
- Inflammation Centre, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, P.O. Box 160, 00029 HUS Helsinki, Finland;
- Department of Otorhinolaryngology—Head and Neck Surgery, Helsinki University Hospital, University of Helsinki, 00029 HUS Helsinki, Finland;
| | - Anu Laulajainen-Hongisto
- Department of Otorhinolaryngology—Head and Neck Surgery, Helsinki University Hospital, University of Helsinki, 00029 HUS Helsinki, Finland;
| | - Philippe Gevaert
- Department of Otorhinolaryngology, Upper Airway Research Laboratory, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Paula Kauppi
- Heart and Lung Center, Pulmonary Department, University of Helsinki and Helsinki University Hospital, 00029 HUS Helsinki, Finland;
| | - Sanna Toppila-Salmi
- Inflammation Centre, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, P.O. Box 160, 00029 HUS Helsinki, Finland;
- Medicum, Haartman Institute, University of Helsinki, 00029 HUS Helsinki, Finland
| |
Collapse
|
46
|
Castagnoli R, Licari A, Brambilla I, Tosca M, Ciprandi G, Marseglia GL. An update on the role of chronic rhinosinusitis with nasal polyps as a co-morbidity in severe asthma. Expert Rev Respir Med 2020; 14:1197-1205. [PMID: 32875924 DOI: 10.1080/17476348.2020.1812388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Chronic rhinosinusitis and asthma are heterogeneous diseases with complex pathogenesis. The presence of chronic rhinosinusitis with nasal polyps has been associated with increased asthma exacerbation frequency and may represent a predictor of future exacerbations in severe asthma. AREAS COVERED This review provides the clinician with an overview of the prevalence and clinical impact of the chronic rhinosinusitis with nasal polyps in severe asthma and summarizes recommended therapeutic approaches, including innovative biologic therapies. To select relevant literature for inclusion in this review, we conducted a literature search using the PubMed and ClinicalTrials.gov databases, using terms 'chronic rhinosinusitis with nasal polyps' AND 'asthma' OR 'severe asthma.' The literature review was performed for publication years 2010-2020, restricting the articles to humans and English language publications. EXPERT OPINION Biological therapies have opened new perspectives in the treatment of upper and lower airway allergic diseases. Care pathways in severe asthma are almost consolidated, while they still rely on phenotypic rather than endotypic features in chronic rhinosinusitis with nasal polyps. Unveiling the correlation between clinical phenotypes and molecular endotypes will allow better stratification of patients with chronic rhinosinusitis with nasal polyps to identify candidates who benefit most from biological therapy.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia , Pavia, Italy
| | - Amelia Licari
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia , Pavia, Italy
| | - Ilaria Brambilla
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia , Pavia, Italy
| | - Mariangela Tosca
- Pediatric Allergy Center, Istituto Giannina Gaslini , Genoa, Italy
| | | | - Gian Luigi Marseglia
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, University of Pavia , Pavia, Italy
| |
Collapse
|
47
|
Bachert C, Desrosiers MY, Hellings PW, Laidlaw TM. The Role of Biologics in Chronic Rhinosinusitis with Nasal Polyps. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 9:1099-1106. [PMID: 33227522 DOI: 10.1016/j.jaip.2020.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022]
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a common and heterogeneous inflammatory disease of the upper respiratory tract. This article provides expert opinion and points of view from both allergists and rhinologists who specialize in CRSwNP. Despite the potential value of biomarker-based endotyping to provide guidance regarding optimal care and treatment choices for patients with CRSwNP, current practice is largely not biomarker-based. In general, there is agreement that for patients with symptomatic CRSwNP who have failed a trial of a course of at least 3 months of intranasal steroids and a short course of oral corticosteroids, a surgical intervention will often be the next treatment of choice. Biologics may be considered before an initial surgery in patients with comorbid severe asthma and in those for whom surgery is less available, refused by the patient, or likely to be associated with a higher-than-average complication rate. Biologic use immediately following surgery may be considered in patients who have a history of nasal polyp recurrence within 12 months of a prior surgery. For many patients with recalcitrant disease, a combination of sinus surgery and use of a biologic that is targeted to their precise endotype may be the optimal treatment strategy, though which surgical approach and which biologics are best for each patient are debates that remain ongoing.
Collapse
Affiliation(s)
- Claus Bachert
- Upper Airways Research Laboratory and Department of Oto-Rhino-Laryngology, Ghent University, Gent, Belgium; First Affiliated Hospital, Sun Yat-sen University, International Airway Research Center, Guangzhou, China; Division of ENT Diseases, CLINTEC, Karolinska Institute, University of Stockholm, Stockholm, Sweden
| | - Martin Y Desrosiers
- Department of Otolaryngology, Centre de Recherche du Centre Hospitalier de L'Universite de Montreal, Montreal, Quebec, Canada
| | - Peter W Hellings
- Upper Airways Research Laboratory and Department of Oto-Rhino-Laryngology, Ghent University, Gent, Belgium; Clinical Division of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Otorhinolaryngology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Tanya M Laidlaw
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass.
| |
Collapse
|
48
|
Bachert C, Marple B, Schlosser RJ, Hopkins C, Schleimer RP, Lambrecht BN, Bröker BM, Laidlaw T, Song WJ. Adult chronic rhinosinusitis. Nat Rev Dis Primers 2020; 6:86. [PMID: 33122665 DOI: 10.1038/s41572-020-00218-1] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Chronic rhinosinusitis (CRS) occurs in >10% of the adult population in Europe and the USA and can be differentiated into CRS without nasal polyps and CRS with nasal polyps (CRSwNP). Both phenotypes are characterized by a high disease burden and an overlapping spectrum of symptoms, with facial pain and loss of smell being the most differentiating. Great progress has been made in the understanding of CRS pathophysiology: from the epithelium and epithelial-mesenchymal transition to innate and adaptive immunity pathways and, finally, on the role of eosinophils and Staphylococcus aureus in the persistence of disease. Although clinical manifestations and diagnostic tools (including nasal endoscopy and imaging) have undergone major changes over the past few years, management (including pharmacotherapy, surgery and biologics) has experienced enormous progress based on the growing knowledge of key mediators in severe CRSwNP. The introduction of endotyping has led to a differentiation of 'tailored' surgical approaches, focusing on the mucosal concept in those with severe CRSwNP and on the identification of patients eligible for extended surgery and possibly biologics in the future.
Collapse
Affiliation(s)
- Claus Bachert
- Sun Yat-sen University, International Airway Research Center, First Affiliated Hospital, Guangzhou, China.
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium.
- Division of ENT diseases, CLINTEC, Karolinska Institute, University of Stockholm, Stockholm, Sweden.
| | - Bradley Marple
- University of Texas, Southwestern Medical Center, Department of Otolaryngology - Head and Neck Surgery, Dallas, TX, USA
| | - Rodney J Schlosser
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC, USA
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB-UGhent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, Netherlands
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Tanya Laidlaw
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
49
|
Poddighe D, Vangelista L. Staphylococcus aureus Infection and Persistence in Chronic Rhinosinusitis: Focus on Leukocidin ED. Toxins (Basel) 2020; 12:678. [PMID: 33126405 PMCID: PMC7692112 DOI: 10.3390/toxins12110678] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is thought to be a multifactorial disease that includes a direct involvement of bacteria that trigger inflammation and contribute to CRS pathogenesis. Staphylococcus aureus infection and persistence is associated with chronic rhinosinusitis (CRS), and it may be particularly relevant in the form with nasal polyps (CRSwNP). The large array of exotoxins deployed by S. aureus is instrumental for the bacterium to warrant its infection and dissemination in different human body districts. Here, we analyze the common Th2 environment in CRSwNP and prospect a possible dynamic role played by S. aureus leukocidins in promoting this chronic inflammation, considering leukocidin ED (LukED) as a strong prototype candidate worth of therapeutic investigation. CCR5 is an essential target for LukED to exert its cytotoxicity towards T cells, macrophages and dendritic cells. Therefore, CCR5 blockade might be an interesting therapeutic option for CRS and, more specifically, persistent and relapsing CRSwNP. In this perspective, the arsenal of CCR5 antagonists being developed to inhibit HIV-1 entry (CCR5 being the major HIV-1 co-receptor) could be easily repurposed for CRS therapeutic investigation. Finally, direct targeting of LukED by neutralizing antibodies could represent an important additional solution to S. aureus infection.
Collapse
Affiliation(s)
- Dimitri Poddighe
- Department of Medicine, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan;
- Department of Pediatrics, National Research Center for Maternal and Child Health, Nur-Sultan 010000, Kazakhstan
| | - Luca Vangelista
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Nur-Sultan 010000, Kazakhstan
| |
Collapse
|
50
|
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
|