1
|
Zhou Y, Wang Y, Zhao H, Guo T, Hao Y. Neonatal Fc receptor participates in endocytosis of Fc fusion protein in vivo and in vitro. Vet Immunol Immunopathol 2025; 283:110930. [PMID: 40187220 DOI: 10.1016/j.vetimm.2025.110930] [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: 11/20/2024] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
The neonatal Fc receptor (FcRn) binds to IgG CH2 and CH3 domains (the Fc segment), triggering transendocytosis. Therefore, FcRn transports biological agents across the mucosal barrier. Mucosal administration provides less stimulation to the body than other methods. However, whether FcRn is an effective carrier for antigens across bovine respiratory epithelial cells is unknown. Here, an antigen was fused with the Fc fragment and transferred through the mucosal barrier to antigen-presenting cells via active transport mediated by FcRn. We established a model of FcRn-mediated recombinant IgG Fc protein expression in bovine embryonic tracheal epithelial cells. Western blotting showed that SPA inhibited the relative transport amount of FcRn-mediated IgG Fc fusion protein. Fc fusion protein positively correlated with protein concentration and action time, with the maximum level reached at 1.4 mg/mL (protein concentration) and 18 h (action time). An FcRn-mediated transport model of the IgG Fc recombinant protein in guinea pig lungs was established, and the amount of protein transported at different time points was measured using immunohistochemistry. FcRn mediates vaccine antigen delivery through the mucosal barrier to activate immune cells in the lamina propria, laying a theoretical foundation for the clinical application of nasal mucosal immune vaccines.
Collapse
Affiliation(s)
- Yaping Zhou
- Laboratory of Microbiology and Immunology, School of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia 010018, China
| | - Yanfang Wang
- School of Basic and Forensic Medicine, Baotou Medical College, Baotou, Inner Mongolia 014040, China
| | - Hongmei Zhao
- Laboratory of Microbiology and Immunology, School of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia 010018, China
| | - Ting Guo
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, Inner Mongolia 010031, China
| | - Yongqing Hao
- Laboratory of Microbiology and Immunology, School of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia 010018, China.
| |
Collapse
|
2
|
Song M, Sun J, Lv K, Li J, Shi J, Xu Y. A comprehensive review of pathology and treatment of staphylococcus aureus osteomyelitis. Clin Exp Med 2025; 25:131. [PMID: 40299136 PMCID: PMC12040984 DOI: 10.1007/s10238-025-01595-1] [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: 12/04/2024] [Accepted: 02/10/2025] [Indexed: 04/30/2025]
Abstract
Osteomyelitis (OM) is an inflammation of the bone and bone marrow triggered by infectious pathogens which may induce progressive bone destruction. The majority of OM cases, especially the chronic OM cases, are induced by the most prevalent and devastating pathogen Staphylococcus aureus (S. aureus), partially due to its resistance mechanisms against the immune system and antibiotic therapies. Regarding the high rate of morbidity and recurrence in patients, it is pivotal to elucidate underlying mechanisms that how S. aureus enter and survive in hosts. The accumulated discoveries have identified multiple distinct strategies associated with chronicity and recurrence include biofilm development, small colony variants (SCVs), staphylococcus abscess communities (SACs), the osteocyte lacuno-canalicular network invasion (OLCN) of cortical bones, and S. aureus protein A (SpA). Unfortunately, little clinical progress has been achieved for the diagnosis and therapeutic treatment for OM patients, indicating that numerous questions remain to be solved. Therefore, we still have a long way to obtain the clear elucidation of the host-pathogen interactions which could be applied for clinical treatment of OM. In this review, we provide insights of current knowledge about how S. aureus evades immune eradication and remains persistent in hosts with recent discoveries. The common and novel treatment strategies for OM are also described. The purpose of this review is to have in-dept understanding of S. aureus OM and bring new perspectives to therapeutic fields which may be translated to the clinic.
Collapse
Affiliation(s)
- Muguo Song
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Sun
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Kehan Lv
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Junyi Li
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China
- Kunming Medical University Graduate School, Kunming, 650500, China
| | - Jian Shi
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| | - Yongqing Xu
- Department of Orthopaedics, 920 Hospital of the Joint Logistics Support Force of the PLA, Kunming, 650032, China.
| |
Collapse
|
3
|
Graham JC, Anand SS, Bercu J, Besenhofer L, de Zafra C, Feng Y, Fisher C, Hillegass J, Hutchinson R, Jolly R, Moudgal C, Nicholas T, Olszova D, Schmitz M, Semmelmann F. Safety assessment of protein A and derivation of a parenteral health-based exposure limit. Regul Toxicol Pharmacol 2024; 153:105700. [PMID: 39243930 DOI: 10.1016/j.yrtph.2024.105700] [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: 07/09/2024] [Revised: 08/18/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Protein A (PA) is a bacterial cell wall component of Staphylococcus aureus whose function is to bind to Immunoglobulin G (IgG). Given its ability to bind IgG as well as its stability and resistance to harsh acidic and basic cleaning conditions, it is commonly used in the affinity chromotography purification of biotherapeutics. This use can result in levels of PA being present in a drug product and subsequent patient exposure. Interestingly, PA was previously evaluated in clinical trials as well as supporting nonclinical studies, resulting in a database that enables the derivation of a health-based exposure limit (HBEL). Given the widespread use of PA in the pharmaceutical industry, the IQ DruSafe Impurities Safety Working Group (WG) evaluated the available information with the purpose of establishing a harmonized parenteral HBEL for PA. Based on this thorough, collaborative evaluation of nonclinical and clinical data available for PA, a parenteral HBEL of 1.2 μg/kg/dose (60 μg/dose for a 50 kg individual) is expected to be health protective for patients when it is present as an impurity in a biotherapeutic.
Collapse
Affiliation(s)
- Jessica C Graham
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | | | - Joel Bercu
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, 94404, USA
| | | | | | - Yu Feng
- Merck & Co., Inc., 126 East Lincoln Avenue, P.O. Box 2000, Rahway, NJ, 07065, USA
| | - Craig Fisher
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, MA, 02139, USA
| | - Jedd Hillegass
- Bristol Myers Squibb, 1 Squibb Drive, New Brunswick, NJ, 08901, USA
| | - Richard Hutchinson
- Johnson & Johnson Innovative Medicine, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Robert Jolly
- Eli Lilly & Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | | | | | - Daniela Olszova
- Gilead Sciences, Inc., 4010 Ocean Ranch Blvd., Oceanside, CA, 92056, USA
| | - Matthew Schmitz
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, MA, 02139, USA
| | | |
Collapse
|
4
|
Aktekin MB, Oksuz Z, Turkmenoglu B, Istifli ES, Kuzucu M, Algul O. Synthesis and evaluation of di-heterocyclic benzazole compounds as potential antibacterial and anti-biofilm agents against Staphylococcus aureus. Chem Biol Drug Des 2024; 104:e14601. [PMID: 39085984 DOI: 10.1111/cbdd.14601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/26/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Cumulative escalation in antibiotic-resistant pathogens necessitates the quest for novel antimicrobial agents, as current options continue to diminish bacterial resistance. Herein, we report the synthesis of di-heterocyclic benzazole structures (12-19) and their in vitro evaluation for some biological activities. Compounds 16 and 17 demonstrated potent antibacterial activity (MIC = 7.81 μg/mL) against Staphylococcus aureus, along with significant anti-biofilm activity. Noteworthy is the capability of Compound 17 to inhibit biofilm formation by at least 50% at sub-MIC (3.90 μg/mL) concentration. Furthermore, both compounds exhibited the potential to inhibit preformed biofilm by at least 50% at the MIC concentration (7.81 μg/mL). Additionally, Compounds 16 and 17 were examined for cytotoxic effects in HFF-1 cells, using the MTT method, and screened for binding interactions within the active site of S. aureus DNA gyrase using in silico molecular docking technique, employing AutoDock 4.2.6 and Schrödinger Glidse programs. Overall, our findings highlight Compounds 16 and 17 as promising scaffolds warranting further optimization for the development of effective antibacterial and anti-biofilm agents.
Collapse
Affiliation(s)
- Mine Buga Aktekin
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
- Department of Pharmacy Services, Vocational School of Health Services, Tarsus University, Mersin, Turkey
| | - Zehra Oksuz
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Mersin University, Mersin, Turkey
| | - Burcin Turkmenoglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, Turkey
| | - Erman Salih Istifli
- Department of Biology, Faculty of Science and Literature, Çukurova University, Adana, Turkey
| | - Mehmet Kuzucu
- Department of Biology, Faculty of Arts and Sciences, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Oztekin Algul
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mersin University, Mersin, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan, Turkey
| |
Collapse
|
5
|
Lichota A, Gwozdzinski K, Kowalczyk E, Kowalczyk M, Sienkiewicz M. Contribution of staphylococcal virulence factors in the pathogenesis of thrombosis. Microbiol Res 2024; 283:127703. [PMID: 38537329 DOI: 10.1016/j.micres.2024.127703] [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: 08/23/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Staphylococci are responsible for many infections in humans, starting with skin and soft tissue infections and finishing with invasive diseases such as endocarditis, sepsis and pneumonia, which lead to high mortality. Patients with sepsis often demonstrate activated clotting pathways, decreased levels of anticoagulants, decreased fibrinolysis, activated endothelial surfaces and activated platelets. This results in disseminated intravascular coagulation and formation of a microthrombus, which can lead to a multiorgan failure. This review describes various staphylococcal virulence factors that contribute to vascular thrombosis, including deep vein thrombosis in infected patients. The article presents mechanisms of action of different factors released by bacteria in various host defense lines, which in turn can lead to formation of blood clots in the vessels.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland.
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Lodz, Poland
| | | | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
6
|
Hajam IA, Liu GY. Linking S. aureus Immune Evasion Mechanisms to Staphylococcal Vaccine Failures. Antibiotics (Basel) 2024; 13:410. [PMID: 38786139 PMCID: PMC11117348 DOI: 10.3390/antibiotics13050410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
Vaccination arguably remains the only long-term strategy to limit the spread of S. aureus infections and its related antibiotic resistance. To date, however, all staphylococcal vaccines tested in clinical trials have failed. In this review, we propose that the failure of S. aureus vaccines is intricately linked to prior host exposure to S. aureus and the pathogen's capacity to evade adaptive immune defenses. We suggest that non-protective immune imprints created by previous exposure to S. aureus are preferentially recalled by SA vaccines, and IL-10 induced by S. aureus plays a unique role in shaping these non-protective anti-staphylococcal immune responses. We discuss how S. aureus modifies the host immune landscape, which thereby necessitates alternative approaches to develop successful staphylococcal vaccines.
Collapse
Affiliation(s)
- Irshad Ahmed Hajam
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
| | - George Y. Liu
- Department of Pediatrics, University of California San Diego, San Diego, CA 92093, USA;
- Division of Infectious Diseases, Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
7
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
8
|
Caldera JR, Tsai CM, Trieu D, Gonzalez C, Hajam IA, Du X, Lin B, Liu GY. The characteristics of pre-existing humoral imprint determine efficacy of S. aureus vaccines and support alternative vaccine approaches. Cell Rep Med 2024; 5:101360. [PMID: 38232694 PMCID: PMC10829788 DOI: 10.1016/j.xcrm.2023.101360] [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: 01/15/2023] [Revised: 08/15/2023] [Accepted: 12/10/2023] [Indexed: 01/19/2024]
Abstract
The failure of the Staphylococcus aureus (SA) IsdB vaccine trial can be explained by the recall of non-protective immune imprints from prior SA exposure. Here, we investigate natural human SA humoral imprints to understand their broader impact on SA immunizations. We show that antibody responses against SA cell-wall-associated antigens (CWAs) are non-opsonic, while antibodies against SA toxins are neutralizing. Importantly, the protective characteristics of the antibody imprints accurately predict the failure of corresponding vaccines against CWAs and support vaccination against toxins. In passive immunization platforms, natural anti-SA human antibodies reduce the efficacy of the human monoclonal antibodies suvratoxumab and tefibazumab, consistent with the results of their respective clinical trials. Strikingly, in the absence of specific humoral memory responses, active immunizations are efficacious in both naive and SA-experienced mice. Overall, our study points to a practical and predictive approach to evaluate and develop SA vaccines based on pre-existing humoral imprint characteristics.
Collapse
Affiliation(s)
- J R Caldera
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Chih-Ming Tsai
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Desmond Trieu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cesia Gonzalez
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Irshad A Hajam
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xin Du
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian Lin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Y Liu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Division of Infectious Diseases, Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
9
|
Muñoz-Hernández C, Wipf A, Ortega N, Barberá GG, Salinas J, Gonzálvez M, Martínez-Carrasco C, Candela MG. Serological and molecular survey of canine distemper virus in red foxes (Vulpes vulpes): Exploring cut-off values and the use of protein A in ELISA tests. Prev Vet Med 2023; 221:106075. [PMID: 37984159 DOI: 10.1016/j.prevetmed.2023.106075] [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: 07/27/2023] [Revised: 10/18/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
The wide distribution and ecological plasticity of the red fox (Vulpes vulpes) make it a potential reservoir for many infectious diseases shared with domestic and wild carnivores. One of such diseases is canine distemper, which is caused by an RNA virus and its main domestic reservoir is the dog. However, other carnivores can also participate in its maintenance, as shown by the recent upsurge of reported cases in wildlife in many parts of the world, and by the fact that red foxes may act as true reservoirs for canine distemper virus (CDV). The lack of validated serological tests for wildlife or other non-target species may be a handicap for monitoring this virus. In this study, serological assays were compared in 147 red fox sera using a commercial ELISA validated for its use in dogs and a non-specific modified ELISA with Protein A peroxidase conjugate to detect bound antibodies. In addition, the presence of CDV RNA in brain, spleen, lung, and liver samples from 144 foxes was investigated by a RT-qPCR. Through the comparison of the results of both ELISAs and the use of a finite mixture model of the optical density values obtained by both techniques, we adjusted the cut-off point of the commercial ELISA to obtain the seroprevalence in foxes. The overall seroprevalence detected was 53.7% (79/147) and 57.1% (84/147) by the commercial and modified ELISA, respectively, with a moderate agreement according to Cohen's Kappa statistic (κ = 0.491, z = 5.97, p < 0.0001). CDV RNA was detected in 30 out of 144 foxes, which resulted in 20.8% of CDV-infected foxes. At individual level, the results obtained by relating the serological status and the presence/absence of RNA in different organs were explained in terms of the pathogenesis of the infection. Our results highlight the convenience of adjusting the cut-off point when using an ELISA assay developed in domestic dogs for its use in foxes. Moreover, Protein A is confirmed to be a good alternative to be used in red foxes, presenting a good reactivity towards its IgG.
Collapse
Affiliation(s)
- C Muñoz-Hernández
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain; Grupo Sanidad y Biotecnología (SaBio), Instituto de Investigación en Recursos Cinegéticos, IREC (CSIC-UCLM-JCCM), 13005 Ciudad Real, Spain.
| | - A Wipf
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain.
| | - N Ortega
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain.
| | - G G Barberá
- Department of Water and Soil Conservation, CEBAS-CSIC, Campus Universitario, Espinardo 30100, Spain.
| | - J Salinas
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain.
| | - M Gonzálvez
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain; Departamento de Sanidad Animal, Grupo de Investigación en Sanidad Animal y Zoonosis (GISAZ), Universidad de Córdoba, 14014 Córdoba, Spain.
| | - C Martínez-Carrasco
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain.
| | - M G Candela
- Departamento de Sanidad Animal, Facultad de Veterinaria, Campus de Excelencia Internacional Regional "Campus Mare Nostrum", Universidad de Murcia, 30100 Murcia, Spain.
| |
Collapse
|
10
|
Bear A, Locke T, Rowland-Jones S, Pecetta S, Bagnoli F, Darton TC. The immune evasion roles of Staphylococcus aureus protein A and impact on vaccine development. Front Cell Infect Microbiol 2023; 13:1242702. [PMID: 37829608 PMCID: PMC10565657 DOI: 10.3389/fcimb.2023.1242702] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/08/2023] [Indexed: 10/14/2023] Open
Abstract
While Staphylococcus aureus (S. aureus) bacteria are part of the human commensal flora, opportunistic invasion following breach of the epithelial layers can lead to a wide array of infection syndromes at both local and distant sites. Despite ubiquitous exposure from early infancy, the life-long risk of opportunistic infection is facilitated by a broad repertoire of S. aureus virulence proteins. These proteins play a key role in inhibiting development of a long-term protective immune response by mechanisms ranging from dysregulation of the complement cascade to the disruption of leukocyte migration. In this review we describe the recent progress made in dissecting S. aureus immune evasion, focusing on the role of the superantigen, staphylococcal protein A (SpA). Evasion of the normal human immune response drives the ability of S. aureus to cause infection, often recurrently, and is also thought to be a major hindrance in the development of effective vaccination strategies. Understanding the role of S. aureus virulence protein and determining methods overcoming or subverting these mechanisms could lead to much-needed breakthroughs in vaccine and monoclonal antibody development.
Collapse
Affiliation(s)
- Alex Bear
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Thomas Locke
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | - Sarah Rowland-Jones
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| | | | | | - Thomas C. Darton
- Department of Infection, Immunity and Cardiovascular Disease, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
11
|
Sakyi ME, Kamio T, Kohyama K, Rahman MM, Shimizu K, Okada A, Inoshima Y. Assessing of the use of proteins A, G, and chimeric protein AG to detect marine mammal immunoglobulins. PLoS One 2023; 18:e0291743. [PMID: 37733771 PMCID: PMC10513184 DOI: 10.1371/journal.pone.0291743] [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: 04/20/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
In recent years, there has been an increase in infectious diseases in marine mammals, including brucellosis, infections of morbillivirus, herpesvirus, and poxvirus. Several serological diagnostic methods, including enzyme-linked immunosorbent assays, immunofluorescence assays (ELISA), and western blotting, have been used to detect antibodies against pathogens in marine mammals. However, options for commercial secondary antibodies used to detect antibodies in marine mammals are limited; therefore, the use of proteins A, G, or chimeric protein AG may provide a suitable alternative. This study aimed to assess the use of proteins A, G, and chimeric protein AG to detect marine mammal immunoglobulins. Currently, there are no comparative studies on the use of proteins A, G, and chimeric protein AG for the detection of immunoglobulins in marine mammals. In this study, we used ten pinnipeds' species (Baikal seal, California sea lion, harbor seal, northern fur seal, ringed seal, South American fur seal, South American sea lion, spotted seal, Steller sea lion, and walrus) and five cetacean species (beluga whale, bottlenose dolphin, harbor porpoise, killer whale, and Pacific white-sided dolphin) and compare binding ability to proteins A, G, or chimeric protein AG by ELISA. The results revealed that the immunoglobulins from pinniped and cetacean species reacted more strongly to protein A than protein G. In addition, the immunoglobulins of pinnipeds and cetaceans showed a strong binding ability to chimeric protein AG. These results suggest that proteins A, G, and chimeric protein AG would be used to help further develop serological assays.
Collapse
Affiliation(s)
- Michael Essien Sakyi
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Takashi Kamio
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
- Port of Nagoya Public Aquarium, Nagoya, Aichi, Japan
| | | | - Md. Matiur Rahman
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
- Faculty for Veterinary, Department of Medicine, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, Bangladesh
| | - Kaori Shimizu
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
| | - Ayaka Okada
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan
| | - Yasuo Inoshima
- Cooperative Department of Veterinary Medicine, Laboratory of Food and Environmental Hygiene, Gifu University, Gifu, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), Gifu, Japan
| |
Collapse
|
12
|
Yang J, Ostafe R, Welch CJ, Verhalen B, Budyak IL, Bruening ML. Rapid Quantitation of Various Therapeutic Monoclonal Antibodies Using Membranes with Fc-Specific Ligands. Anal Chem 2023. [PMID: 37216615 DOI: 10.1021/acs.analchem.3c00531] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Therapeutic monoclonal antibodies (mAbs) provide effective treatments for many diseases, including cancer, autoimmune disorders, and, lately, COVID-19. Monitoring the concentrations of mAbs is important during their production and subsequent processing. This work demonstrates a 5 min quantitation of most human immunoglobulin G (IgG) antibodies through capture of mAbs in membranes modified with ligands that bind to the fragment crystallizable (Fc) region. This enables binding and quantitation of most IgG mAbs. Layer-by-layer (LBL) adsorption of carboxylic acid-rich polyelectrolytes in glass-fiber membranes in 96-well plates allows functionalization of the membranes with Protein A or a peptide, oxidized Fc20 (oFc20), with high affinity for the Fc region of human IgG. mAb capture occurs in <1 min during the flow of solutions through modified membranes, and subsequent binding of a fluorophore-labeled secondary antibody enables quantitation of the captured mAbs using fluorescence. The intra- and inter-plate coefficients of variations (CV) are <10 and 15%, respectively, satisfying the acceptance criteria for many assays. The limit of detection (LOD) of 15 ng/mL is on the high end of commercial enzyme-linked immunosorbent assays (ELISAs) but certainly low enough for monitoring of manufacturing solutions. Importantly, the membrane-based method requires <5 minutes, whereas ELISAs typically take at least 90 min. Membranes functionalized with oFc20 show greater mAb binding and lower LODs than membranes with Protein A. Thus, the membrane-based 96-well-plate assay, which is effective in diluted fermentation broths and in mixtures with cell lysates, is suitable for near-real-time monitoring of the general class of human IgG mAbs during their production.
Collapse
Affiliation(s)
- Junyan Yang
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Raluca Ostafe
- Molecular Evolution, Protein Engineering and Production Facility, Purdue Institute for Inflammation, Immunology and Infection Diseases, Purdue University, West Lafayette, Indiana 47907, United States
| | - Christopher J Welch
- Indiana Consortium for Analytical Science & Engineering (ICASE), 410 W. 10th St., # 1020H, Indianapolis, Indiana 46202, United States
| | - Brandy Verhalen
- Corteva Agriscience, 8325 NW 62nd Ave, Johnston, Iowa 50131, United States
| | - Ivan L Budyak
- Biopharmaceutical Research and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Merlin L Bruening
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
13
|
Current molecular approach for diagnosis of MRSA: a meta-narrative review. Drug Target Insights 2022; 16:88-96. [PMID: 36761068 PMCID: PMC9906022 DOI: 10.33393/dti.2022.2522] [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: 11/17/2022] [Accepted: 12/31/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction: Detection and diagnosis of methicillin-resistant Staphylococcus aureus (MRSA) are important in ensuring a correct and effective treatment, further reducing its spread. A wide range of molecular approaches has been used for the diagnosis of antimicrobial resistance (AMR) in MRSA. This review aims to study and appraise widely used molecular diagnostic methods for detecting MRSA. Methods: This meta-narrative review was performed by searching PubMed using the following search terms: (molecular diagnosis) AND (antimicrobial resistance) AND (methicillin-resistant Staphylococcus aureus). Studies using molecular diagnostic techniques for the detection of MRSA were included, while non-English language, duplicates and non-article studies were excluded. After reviewing the libraries and a further manual search, 20 studies were included in this article. RAMESES publication standard for narrative reviews was used for this synthesis. Results: A total of 20 full papers were reviewed and appraised in this synthesis, consisting of PCR technique (n = 7), deoxyribonucleic acid (DNA) Microarray (n = 1), DNA sequencing (n = 2), Xpert MRSA/SA BC assay (n = 2), matrix-assisted laser desorption/ionization-time of flight (MALDI-TOF) (n = 2), MLST (n = 4), SCCmec typing (n = 1) and GENECUBE (n = 1). Discussion: Different diagnostic methods used to diagnose MRSA have been studied in this review. This study concludes that PCR has been extensively used due to its higher sensitivity and cost-effectiveness in the past five years
Collapse
|
14
|
Berry KA, Verhoef MTA, Leonard AC, Cox G. Staphylococcus aureus adhesion to the host. Ann N Y Acad Sci 2022; 1515:75-96. [PMID: 35705378 DOI: 10.1111/nyas.14807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus is a pathobiont capable of colonizing and infecting most tissues within the human body, resulting in a multitude of different clinical outcomes. Adhesion of S. aureus to the host is crucial for both host colonization and the establishment of infections. Underlying the pathogen's success is a complex and diverse arsenal of adhesins. In this review, we discuss the different classes of adhesins, including a consideration of the various adhesion sites throughout the body and the clinical outcomes of each infection type. The development of therapeutics targeting the S. aureus host-pathogen interaction is a relatively understudied area. Due to the increasing global threat of antimicrobial resistance, it is crucial that innovative and alternative approaches are considered. Neutralizing virulence factors, through the development of antivirulence agents, could reduce bacterial pathogenicity and the ever-increasing burden of S. aureus infections. This review provides insight into potentially efficacious adhesion-associated targets for the development of novel decolonizing and antivirulence strategies.
Collapse
Affiliation(s)
- Kirsten A Berry
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mackenzie T A Verhoef
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Allison C Leonard
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Georgina Cox
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
15
|
Tsai CM, Hajam IA, Caldera JR, Liu GY. Integrating complex host-pathogen immune environments into S. aureus vaccine studies. Cell Chem Biol 2022; 29:730-740. [PMID: 35594849 DOI: 10.1016/j.chembiol.2022.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/16/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022]
Abstract
Staphylococcus aureus (SA) is a leading cause of bacterial infection and antibiotic resistance globally. Therefore, development of an effective vaccine has been a major goal of the SA field for the past decades. With the wealth of understanding of pathogenesis, the failure of all SA vaccine trials has been a surprise. We argue that experimental SA vaccines have not worked because vaccines have been studied in naive laboratory animals, whereas clinical vaccine efficacy is tested in immune environments reprogrammed by SA. Here, we review the failed SA vaccines that have seemingly defied all principles of vaccinology. We describe major SA evasion strategies and suggest that they reshape the immune environment in a way that makes vaccines prone to failures. We propose that appropriate integration of concepts of host-pathogen interaction into vaccine study designs could lead to insight critical for the development of an effective SA vaccine.
Collapse
Affiliation(s)
- Chih-Ming Tsai
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Irshad A Hajam
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - J R Caldera
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - George Y Liu
- Division of Infectious Diseases, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA; Division of Infectious Diseases, Rady Children's Hospital, San Diego, CA 92123, USA.
| |
Collapse
|
16
|
Boero E, Cruz AR, Pansegrau W, Giovani C, Rooijakkers SHM, van Kessel KPM, van Strijp JAG, Bagnoli F, Manetti AGO. Natural Human Immunity Against Staphylococcal Protein A Relies on Effector Functions Triggered by IgG3. Front Immunol 2022; 13:834711. [PMID: 35359919 PMCID: PMC8963248 DOI: 10.3389/fimmu.2022.834711] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/17/2022] [Indexed: 12/02/2022] Open
Abstract
Staphylococcal protein A (SpA) is a multifunctional, highly conserved virulence factor of Staphylococcus aureus. By binding the Fc portion of all human IgG subclasses apart from IgG3, SpA interferes with antibody and complement deposition on the bacterial surface, impairing staphylococcal clearance by phagocytosis. Because of its anti-opsonic properties, SpA is not investigated as a surface antigen to mediate bacterial phagocytosis. Herein we investigate human sera for the presence of SpA-opsonizing antibodies. The screening revealed that sera containing IgG3 against SpA were able to correctly opsonize the target and drive Fcγ receptor-mediated interactions and phagocytosis. We demonstrated that IgG3 Fc is significantly more efficient in inducing phagocytosis of SpA-expressing S. aureus as compared to IgG1 Fc in an assay resembling physiological conditions. Furthermore, we show that the capacity of SpA antibodies to induce phagocytosis depends on the specific epitope recognized by the IgGs on SpA molecules. Overall, our results suggest that anti-SpA IgG3 antibodies could favor the anti-staphylococcal response in humans, paving the way towards the identification of a correlate of protection against staphylococcal infections.
Collapse
Affiliation(s)
- Elena Boero
- GSK, Siena, Italy
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ana Rita Cruz
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | | - Suzan H. M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Kok P. M. van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jos A. G. van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | | |
Collapse
|
17
|
Tahoun A, Elnafarawy HK, El-Sharkawy H, Rizk AM, Alorabi M, El-Shehawi AM, Youssef MA, Ibrahim HMM, El-Khodery S. The Prevalence and Molecular Biology of Staphylococcus aureus Isolated from Healthy and Diseased Equine Eyes in Egypt. Antibiotics (Basel) 2022; 11:antibiotics11020221. [PMID: 35203823 PMCID: PMC8868267 DOI: 10.3390/antibiotics11020221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
This work aimed to characterize S. aureus isolates from the eyes of healthy and clinically affected equines in the Kafrelsheikh Governorate, Egypt. A total of 110 animals were examined for the presence of S. aureus, which was isolated from 33 animals with ophthalmic lesions and 77 healthy animals. We also investigated the antimicrobial resistance profile, oxacillin resistance mechanism, and the major virulence factors implicated in many studies of the ocular pathology of pathogenic S. aureus. The association between S. aureus eye infections and potential risk factors was also investigated. The frequency of S. aureus isolates from clinically affected equine eyes was significantly higher than in clinically healthy equids. A significant association was found between the frequency of S. aureus isolation from clinically affected equine eyes and risk factors including age and season but not with sex or breed factors. Antimicrobial resistance to common antibiotics used to treat equine eyes was also tested. Overall, the isolates showed the highest sensitivity to sulfamethoxazole (100%) and the highest resistance to cephalosporin (90.67%) and oxacillin (90.48%). PCR was used to demonstrate that mecA was present in 100% of oxacillin- and β-lactam-resistant S. aureus strains. The virulence factor genes Spa (x region), nuc, and hlg were identified in 62.5%, 100%, and 56%, of isolates, respectively, from clinically affected equines eyes. The severity of the eye lesions increased in the presence of γ-toxin-positive S. aureus. The phylogenetic tree of the Spa (x region) gene indicated a relationship with human reference strains isolated from Egypt as well as isolates from equines in Iran and Japan. This study provides insight into the prevalence, potential risk factors, clinical pictures, zoonotic potential, antimicrobial resistance, and β-lactam resistance mechanism of S. aureus strains that cause eye infection in equines from Egypt.
Collapse
Affiliation(s)
- Amin Tahoun
- Department of Animal Medicine, Faculty of Veterinary Medicine, Kafrelshkh University, Kafrelsheikh 33511, Egypt
- Correspondence:
| | - Helmy K. Elnafarawy
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (H.K.E.); (M.A.Y.); (H.M.M.I.); (S.E.-K.)
| | - Hanem El-Sharkawy
- Department of Poultry and Rabbit Diseases, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33511, Egypt;
| | - Amira M. Rizk
- Department of Bacteriology, Mycology and Immunology, Faculty of Veterinary Medicine, Benha University, Benha 13518, Egypt;
| | - Mohammed Alorabi
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (M.A.); (A.M.E.-S.)
| | - Ahmed M. El-Shehawi
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (M.A.); (A.M.E.-S.)
| | - Mohamed A. Youssef
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (H.K.E.); (M.A.Y.); (H.M.M.I.); (S.E.-K.)
| | - Hussam M. M. Ibrahim
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (H.K.E.); (M.A.Y.); (H.M.M.I.); (S.E.-K.)
| | - Sabry El-Khodery
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt; (H.K.E.); (M.A.Y.); (H.M.M.I.); (S.E.-K.)
| |
Collapse
|
18
|
Deacy AM, Gan SKE, Derrick JP. Superantigen Recognition and Interactions: Functions, Mechanisms and Applications. Front Immunol 2021; 12:731845. [PMID: 34616400 PMCID: PMC8488440 DOI: 10.3389/fimmu.2021.731845] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.
Collapse
Affiliation(s)
- Anthony M. Deacy
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, Experimental Drug Development Centre – Bioinformatics Institute (EDDC-BII), Agency for Science Technology and Research (ASTAR), Singapore, Singapore
- James Cook University, Singapore, Singapore
| | - Jeremy P. Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Abstract
One of the defining features of Staphylococcus aureus is its ability to evade and impair the human immune response through expression of staphylococcal protein A (SpA). Herein, we describe a previously unknown mechanism by which SpA can form toxic immune complexes when in the presence of human serum, which leads to the loss of human leukocytes. Further, we demonstrate that these toxic complexes are formed specifically through SpA's interaction with intact human IgG and that, in the presence of purified IgG Fab and Fc fragments, SpA shows no such toxicity. The mechanism of action of this toxicity appears to be one mediated by necrosis and not by apoptosis, as previously hypothesized, with up to 90% of human B cells rapidly becoming necrotic following stimulation with SpA-IgG complexes. This phenomenon depends on the immunoglobulin binding capacity of SpA, as a nonbinding mutant of SpA did not induce necrosis. Importantly, immune sera raised against SpA had the capacity to significantly reduce the observed toxicity. An unprecedented toxic effect of SpA-IgG complexes on monocytes was also observed, suggesting the existence of a novel mechanism independent from the interaction of SpA with the B cell receptor. Together, these data implicate SpA in inducing indiscriminate leukocyte toxicity upon formation of complexes with IgG and highlight the requirement for vaccination strategies to inhibit this mechanism. IMPORTANCE Staphylococcus aureus is one of the largest health care threats faced by humankind, with a reported mortality rate within the United States greater than that of HIV/AIDS, tuberculosis, and viral hepatitis combined. One of the defining features of S. aureus as a human pathogen is its ability to evade and impair the human immune response through expression of staphylococcal protein A. Herein, we show that SpA induces necrosis in various immune cells by complexing with human immunoglobulins. Vaccination of mice with a nontoxigenic SpA mutant induced sera capable of inhibiting this mechanism. These observations shed new light on the toxic mechanisms of this key staphylococcal virulence factor and on protective modalities of SpA-based vaccination.
Collapse
|
20
|
Hu DL, Li S, Fang R, Ono HK. Update on molecular diversity and multipathogenicity of staphylococcal superantigen toxins. ANIMAL DISEASES 2021. [DOI: 10.1186/s44149-021-00007-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AbstractStaphylococcal superantigen (SAg) toxins are the most notable virulence factors associated with Staphylococcus aureus, which is a pathogen associated with serious community and hospital acquired infections in humans and various diseases in animals. Recently, SAg toxins have become a superfamily with 29 types, including staphylococcal enterotoxins (SEs) with emetic activity, SE-like toxins (SEls) that do not induce emesis in primate models or have yet not been tested, and toxic shock syndrome toxin-1 (TSST-1). SEs and SEls can be subdivided into classical types (SEA to SEE) and novel types (SEG to SElY, SE01, SE02, SEl26 and SEl27). The genes of SAg toxins are located in diverse accessory genetic elements and share certain structural and biological properties. SAg toxins are heat-stable proteins that exhibit pyrogenicity, superantigenicity and capacity to induce lethal hypersensitivity to endotoxin in humans and animals. They have multiple pathogenicities that can interfere with normal immune function of host, increase the chances of survival and transmission of pathogenic bacteria in host, consequently contribute to the occurrence and development of various infections, persistent infections or food poisoning. This review focuses on the following aspects of SAg toxins: (1) superfamily members of classic and novelty discovered staphylococcal SAgs; (2) diversity of gene locations and molecular structural characteristics; (3) biological characteristics and activities; (4) multi-pathogenicity of SAgs in animal and human diseases, including bovine mastitis, swine sepsis, abscesses and skin edema in pig, arthritis and septicemia in poultry, and nosocomial infections and food-borne diseases in humans.
Collapse
|
21
|
Radke EE, Li Z, Hernandez DN, El Bannoudi H, Kosakovsky Pond SL, Shopsin B, Lopez P, Fenyö D, Silverman GJ. Diversity of Functionally Distinct Clonal Sets of Human Conventional Memory B Cells That Bind Staphylococcal Protein A. Front Immunol 2021; 12:662782. [PMID: 33995388 PMCID: PMC8113617 DOI: 10.3389/fimmu.2021.662782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus, a common cause of serious and often fatal infections, is well-armed with secreted factors that disarm host immune defenses. Highly expressed in vivo during infection, Staphylococcal protein A (SpA) is reported to also contribute to nasal colonization that can be a prelude to invasive infection. Co-evolution with the host immune system has provided SpA with an Fc-antibody binding site, and a Fab-binding site responsible for non-immune superantigen interactions via germline-encoded surfaces expressed on many human BCRs. We wondered whether the recurrent exposures to S. aureus commonly experienced by adults, result in the accumulation of memory B-cell responses to other determinants on SpA. We therefore isolated SpA-specific class-switched memory B cells, and characterized their encoding VH : VL antibody genes. In SpA-reactive memory B cells, we confirmed a striking bias in usage for VH genes, which retain the surface that mediates the SpA-superantigen interaction. We postulate these interactions reflect co-evolution of the host immune system and SpA, which during infection results in immune recruitment of an extraordinarily high prevalence of B cells in the repertoire that subverts the augmentation of protective defenses. Herein, we provide the first evidence that human memory responses are supplemented by B-cell clones, and circulating-antibodies, that bind to SpA determinants independent of the non-immune Fc- and Fab-binding sites. In parallel, we demonstrate that healthy individuals, and patients recovering from S. aureus infection, both have circulating antibodies with these conventional binding specificities. These findings rationalize the potential utility of incorporating specially engineered SpA proteins into a protective vaccine.
Collapse
Affiliation(s)
- Emily E Radke
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States.,Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Zhi Li
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States.,Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, United States
| | - David N Hernandez
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Hanane El Bannoudi
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Sergei L Kosakovsky Pond
- Institute of Genomic and Evolutionary Medicine, Temple University, Philadelphia, PA, United States
| | - Bo Shopsin
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Peter Lopez
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - David Fenyö
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States.,Institute for Systems Genetics, New York University Grossman School of Medicine, New York, NY, United States
| | - Gregg J Silverman
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
22
|
Shi M, Willing SE, Kim HK, Schneewind O, Missiakas D. Peptidoglycan Contribution to the B Cell Superantigen Activity of Staphylococcal Protein A. mBio 2021; 12:e00039-21. [PMID: 33879590 PMCID: PMC8092194 DOI: 10.1128/mbio.00039-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus causes reiterative and chronic persistent infections. This can be explained by the formidable ability of this pathogen to escape immune surveillance mechanisms. Cells of S. aureus display the abundant staphylococcal protein A (SpA). SpA binds to immunoglobulin (Ig) molecules and coats the bacterial surface to prevent phagocytic uptake. SpA also binds and cross-links variable heavy 3 (VH3) idiotype (IgM) B cell receptors, promoting B cell expansion and the secretion of nonspecific VH3-IgM via a mechanism requiring CD4+ T cell help. SpA binding to antibodies is mediated by the N-terminal Ig-binding domains (IgBDs). The so-called region X, uncharacterized LysM domain, and C-terminal LPXTG sorting signal for peptidoglycan attachment complete the linear structure of the protein. Here, we report that both the LysM domain and the LPXTG motif sorting signal are required for the B cell superantigen activity of SpA in a mouse model of infection. SpA molecules purified from staphylococcal cultures are sufficient to exert B cell superantigen activity and promote immunoglobulin secretion as long as they carry intact LysM and LPXTG motif domains with bound peptidoglycan fragments. The LysM domain binds the glycan chains of peptidoglycan fragments, whereas the LPXTG motif is covalently linked to wall peptides lacking glycan. These findings emphasize the complexity of SpA interactions with B cell receptors.IMPORTANCE The LysM domain is found in all kingdoms of life. While their function in mammals is not known, LysM domains of bacteria and their phage parasites are associated with enzymes that cleave or remodel peptidoglycan. Plants recognize microbe-associated molecular patterns such as chitin via receptors endowed with LysM-containing ectodomains. In plants, such receptors play equally important roles in defense and symbiosis signaling. SpA of S. aureus carries a LysM domain that binds glycan strands of peptidoglycan to influence defined B cell responses that divert pathogen-specific adaptive immune responses.
Collapse
Affiliation(s)
- Miaomiao Shi
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
| | | | - Hwan Keun Kim
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Dominique Missiakas
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
23
|
Tam K, Lacey KA, Devlin JC, Coffre M, Sommerfield A, Chan R, O'Malley A, Koralov SB, Loke P, Torres VJ. Targeting leukocidin-mediated immune evasion protects mice from Staphylococcus aureus bacteremia. J Exp Med 2021; 217:151907. [PMID: 32602902 PMCID: PMC7478724 DOI: 10.1084/jem.20190541] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/05/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
Staphylococcus aureus is responsible for various diseases in humans, and recurrent infections are commonly observed. S. aureus produces an array of bicomponent pore-forming toxins that target and kill leukocytes, known collectively as the leukocidins. The contribution of these leukocidins to impair the development of anti–S. aureus adaptive immunity and facilitate reinfection is unclear. Using a murine model of recurrent bacteremia, we demonstrate that infection with a leukocidin mutant results in increased levels of anti–S. aureus antibodies compared with mice infected with the WT parental strain, indicating that leukocidins negatively impact the generation of anti–S. aureus antibodies in vivo. We hypothesized that neutralizing leukocidin-mediated immune subversion by vaccination may shift this host-pathogen interaction in favor of the host. Leukocidin-immunized mice produce potent leukocidin-neutralizing antibodies and robust Th1 and Th17 responses, which collectively protect against bloodstream infections. Altogether, these results demonstrate that blocking leukocidin-mediated immune evasion can promote host protection against S. aureus bloodstream infection.
Collapse
Affiliation(s)
- Kayan Tam
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Keenan A Lacey
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Joseph C Devlin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Maryaline Coffre
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Alexis Sommerfield
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Rita Chan
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Aidan O'Malley
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| | - Sergei B Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - P'ng Loke
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY.,Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
24
|
Vantucci CE, Ahn H, Fulton T, Schenker ML, Pradhan P, Wood LB, Guldberg RE, Roy K, Willett NJ. Development of systemic immune dysregulation in a rat trauma model of biomaterial-associated infection. Biomaterials 2021; 264:120405. [PMID: 33069135 PMCID: PMC8117743 DOI: 10.1016/j.biomaterials.2020.120405] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
Orthopedic biomaterial-associated infections remain a major clinical challenge, with Staphylococcus aureus being the most common pathogen. S. aureus biofilm formation enhances immune evasion and antibiotic resistance, resulting in a local, indolent infection that can persist long-term without symptoms before eventual hardware failure, bone non-union, or sepsis. Immune modulation is an emerging strategy to combat host immune evasion by S. aureus. However, most immune modulation strategies are focused on local immune responses at the site of infection, with little emphasis on understanding the infection-induced and orthopedic-related systemic immune responses of the host, and their role in local infection clearance and tissue regeneration. This study utilized a rat bone defect model to investigate how implant-associated infection affects the systemic immune response. Long-term systemic immune dysregulation was observed with a significant systemic decrease in T cells and a concomitant increase in immunosuppressive myeloid-derived suppressor cells (MDSCs) compared to non-infected controls. Further, the control group exhibited a regulated and coordinated systemic cytokine response, which was absent in the infection group. Multivariate analysis revealed high levels of MDSCs to be most correlated with the infection group, while high levels of T cells were most correlated with the control group. Locally, the infection group had attenuated macrophage infiltration and increased levels of MDSCs in the local soft tissue compared to non-infected controls. These data reveal the widespread impacts of an orthopedic infection on both the local and the systemic immune responses, uncovering promising targets for diagnostics and immunotherapies that could optimize treatment strategies and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Casey E Vantucci
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hyunhee Ahn
- The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA
| | - Travis Fulton
- The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA
| | - Mara L Schenker
- Department of Orthopaedics, Emory University, Atlanta, GA, USA; Grady Memorial Hospital, Atlanta, GA, USA
| | - Pallab Pradhan
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Levi B Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Robert E Guldberg
- Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Nick J Willett
- The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Department of Orthopaedics, Emory University, Atlanta, GA, USA.
| |
Collapse
|
25
|
Butrico CE, Cassat JE. Quorum Sensing and Toxin Production in Staphylococcus aureus Osteomyelitis: Pathogenesis and Paradox. Toxins (Basel) 2020; 12:toxins12080516. [PMID: 32806558 PMCID: PMC7471978 DOI: 10.3390/toxins12080516] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/04/2020] [Accepted: 08/10/2020] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive pathogen capable of infecting nearly every vertebrate organ. Among these tissues, invasive infection of bone (osteomyelitis) is particularly common and induces high morbidity. Treatment of osteomyelitis is notoriously difficult and often requires debridement of diseased bone in conjunction with prolonged antibiotic treatment to resolve infection. During osteomyelitis, S. aureus forms characteristic multicellular microcolonies in distinct niches within bone. Virulence and metabolic responses within these multicellular microcolonies are coordinated, in part, by quorum sensing via the accessory gene regulator (agr) locus, which allows staphylococcal populations to produce toxins and adapt in response to bacterial density. During osteomyelitis, the Agr system significantly contributes to dysregulation of skeletal homeostasis and disease severity but may also paradoxically inhibit persistence in the host. Moreover, the Agr system is subject to complex crosstalk with other S. aureus regulatory systems, including SaeRS and SrrAB, which can significantly impact the progression of osteomyelitis. The objective of this review is to highlight Agr regulation, its implications on toxin production, factors that affect Agr activation, and the potential paradoxical influences of Agr regulation on disease progression during osteomyelitis.
Collapse
Affiliation(s)
- Casey E. Butrico
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - James E. Cassat
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence: ; Tel.: +1-615-936-6494
| |
Collapse
|
26
|
Convergent Evolution of Neutralizing Antibodies to Staphylococcus aureus γ-Hemolysin C That Recognize an Immunodominant Primary Sequence-Dependent B-Cell Epitope. mBio 2020; 11:mBio.00460-20. [PMID: 32546616 PMCID: PMC7298706 DOI: 10.1128/mbio.00460-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus infection is a major public health threat in part due to the spread of antibiotic resistance and repeated failures to develop a protective vaccine. Infection is associated with production of virulence factors that include exotoxins that attack host barriers and cellular defenses, such as the leukocidin (Luk) family of bicomponent pore-forming toxins. To investigate the structural basis of antibody-mediated functional inactivation of Luk toxins, we generated a panel of murine monoclonal antibodies (MAbs) that neutralize host cell killing by the γ-hemolysin HlgCB. Staphylococcus aureus infection is a major public health threat in part due to the spread of antibiotic resistance and repeated failures to develop a protective vaccine. Infection is associated with production of virulence factors that include exotoxins that attack host barriers and cellular defenses, such as the leukocidin (Luk) family of bicomponent pore-forming toxins. To investigate the structural basis of antibody-mediated functional inactivation of Luk toxins, we generated a panel of murine monoclonal antibodies (MAbs) that neutralize host cell killing by the γ-hemolysin HlgCB. By biopanning these MAbs against a phage-display library of random Luk peptide fragments, we identified a small subregion within the rim domain of HlgC as the epitope for all the MAbs. Within the native holotoxin, this subregion folds into a conserved β-hairpin structure, with exposed key residues, His252 and Tyr253, required for antibody binding. On the basis of the phage-display results and molecular modeling, a 15-amino-acid synthetic peptide representing the minimal epitope on HlgC (HlgC241-255) was designed, and preincubation with this peptide blocked antibody-mediated HIgCB neutralization. Immunization of mice with HlgC241-255 or the homologous LukS246-260 subregion peptide elicited serum antibodies that specifically recognized the native holotoxin subunits. Furthermore, serum IgG from patients who were convalescent for invasive S. aureus infection showed neutralization of HlgCB toxin activity ex vivo, which recognized the immunodominant HlgC241-255 peptide and was dependent on His252 and Tyr253 residues. We have thus validated an efficient, rapid, and scalable experimental workflow for identification of immunodominant and immunogenic leukotoxin-neutralizing B-cell epitopes that can be exploited for new S. aureus-protective vaccines and immunotherapies.
Collapse
|
27
|
Hofstee MI, Muthukrishnan G, Atkins GJ, Riool M, Thompson K, Morgenstern M, Stoddart MJ, Richards RG, Zaat SAJ, Moriarty TF. Current Concepts of Osteomyelitis: From Pathologic Mechanisms to Advanced Research Methods. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1151-1163. [PMID: 32194053 DOI: 10.1016/j.ajpath.2020.02.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/18/2020] [Accepted: 02/27/2020] [Indexed: 01/18/2023]
Abstract
Osteomyelitis is an inflammation of the bone and bone marrow that is most commonly caused by a Staphylococcus aureus infection. Much of our understanding of the underlying pathophysiology of osteomyelitis, from the perspective of both host and pathogen, has been revised in recent years, with notable discoveries including the role played by osteocytes in the recruitment of immune cells, the invasion and persistence of S. aureus in submicron channels of cortical bone, and the diagnostic role of polymorphonuclear cells in implant-associated osteomyelitis. Advanced in vitro cell culture models, such as ex vivo culture models or organoids, have also been developed over the past decade, and have become widespread in many fields, including infectious diseases. These models better mimic the in vivo environment, allow the use of human cells, and can reduce our reliance on animals in osteomyelitis research. In this review, we provide an overview of the main pathologic concepts in osteomyelitis, with a focus on the new discoveries in recent years. Furthermore, we outline the value of modern in vitro cell culture techniques, with a focus on their current application to infectious diseases and osteomyelitis in particular.
Collapse
Affiliation(s)
- Marloes I Hofstee
- AO Research Institute Davos, Davos, Switzerland; Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research and Department of Orthopaedics, University of Rochester Medical Center, Rochester, New York
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Martijn Riool
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | | - Mario Morgenstern
- Department of Orthopedic Surgery and Traumatology, University Hospital Basel, Basel, Switzerland
| | | | | | - Sebastian A J Zaat
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, the Netherlands
| | | |
Collapse
|
28
|
Muthukrishnan G, Masters EA, Daiss JL, Schwarz EM. Mechanisms of Immune Evasion and Bone Tissue Colonization That Make Staphylococcus aureus the Primary Pathogen in Osteomyelitis. Curr Osteoporos Rep 2019; 17:395-404. [PMID: 31721069 PMCID: PMC7344867 DOI: 10.1007/s11914-019-00548-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Staphylococcus aureus is the primary pathogen responsible for osteomyelitis, which remains a major healthcare burden. To understand its dominance, here we review the unique pathogenic mechanisms utilized by S. aureus that enable it to cause incurable osteomyelitis. RECENT FINDINGS Using an arsenal of toxins and virulence proteins, S. aureus kills and usurps immune cells during infection, to produce non-neutralizing pathogenic antibodies that thwart adaptive immunity. S. aureus also has specific mechanisms for distinct biofilm formation on implants, necrotic bone tissue, bone marrow, and within the osteocyte lacuno-canicular networks (OLCN) of live bone. In vitro studies have also demonstrated potential for intracellular colonization of osteocytes, osteoblasts, and osteoclasts. S. aureus has evolved a multitude of virulence mechanisms to achieve life-long infection of the bone, most notably colonization of OLCN. Targeting S. aureus proteins involved in these pathways could provide new targets for antibiotics and immunotherapies.
Collapse
Affiliation(s)
- Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Elysia A Masters
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA
| | - John L Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.
- Department of Orthopaedics, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
29
|
Nasser A, Moradi M, Jazireian P, Safari H, Alizadeh-Sani M, Pourmand MR, Azimi T. Staphylococcus aureus versus neutrophil: Scrutiny of ancient combat. Microb Pathog 2019; 131:259-269. [PMID: 31002964 DOI: 10.1016/j.micpath.2019.04.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/11/2022]
Abstract
Staphylococcus aureus (S.aureus) is a Gram-positive bacterium that causes many infections and diseases. This pathogen can cause many types of infections such as impetigo, toxic shock syndrome toxin (TSST1), pneumonia, endocarditis, and autoimmune diseases like lupus erythematosus and can infect other healthy individuals. In the pathogenic process, colonization is a main risk factor for invasive diseases. Various factors including the cell wall-associated factors and receptors of the epithelial cells facilitate adhesion and colonization of this pathogen. S. aureus has many enzymes, toxins, and strategies to evade from the immune system either by an enzyme that lyses cellular component or by hiding from the immune system via surface antigens like protein A and second immunoglobulin-binding protein (Sbi). The strategies of this bacterium can be divided into five groups: A: Inhibit neutrophil recruitment B: Inhibit phagocytosis C: Inhibit killing by ROS, D: Neutrophil killing, and E: Resistance to antimicrobial peptide. On the other hand, innate immune system via neutrophils, the most important polymorphonuclear leukocytes, fights against bacterial cells by neutrophil extracellular trap (NET). In this review, we try to explain the role of each factor in immune evasion.
Collapse
Affiliation(s)
- Ahmad Nasser
- Microbiology Research center, Ilam University of Medical Sciences, Ilam, Iran; Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Moradi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parham Jazireian
- Department of Biology, University Campus 2,University of Guilan, Rasht, Iran
| | - Hossein Safari
- Health Promotion Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahmood Alizadeh-Sani
- Food Safety and Hygiene Division, Environmental Health Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Students Research Committee, Department of Food Sciences and Technology, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Pourmand
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Taher Azimi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Tam K, Torres VJ. Staphylococcus aureus Secreted Toxins and Extracellular Enzymes. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0039-2018. [PMID: 30873936 PMCID: PMC6422052 DOI: 10.1128/microbiolspec.gpp3-0039-2018] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus is a formidable pathogen capable of causing infections in different sites of the body in a variety of vertebrate animals, including humans and livestock. A major contribution to the success of S. aureus as a pathogen is the plethora of virulence factors that manipulate the host's innate and adaptive immune responses. Many of these immune modulating virulence factors are secreted toxins, cofactors for activating host zymogens, and exoenzymes. Secreted toxins such as pore-forming toxins and superantigens are highly inflammatory and can cause leukocyte cell death by cytolysis and clonal deletion, respectively. Coagulases and staphylokinases are cofactors that hijack the host's coagulation system. Exoenzymes, including nucleases and proteases, cleave and inactivate various immune defense and surveillance molecules, such as complement factors, antimicrobial peptides, and surface receptors that are important for leukocyte chemotaxis. Additionally, some of these secreted toxins and exoenzymes can cause disruption of endothelial and epithelial barriers through cell lysis and cleavage of junction proteins. A unique feature when examining the repertoire of S. aureus secreted virulence factors is the apparent functional redundancy exhibited by the majority of the toxins and exoenzymes. However, closer examination of each virulence factor revealed that each has unique properties that have important functional consequences. This chapter provides a brief overview of our current understanding of the major secreted virulence factors critical for S. aureus pathogenesis.
Collapse
Affiliation(s)
- Kayan Tam
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, New York, NY 10016
| |
Collapse
|
31
|
Luo Z, Li M, Wu Y, Meng Z, Martin L, Zhang L, Ogunrinde E, Zhou Z, Qin S, Wan Z, Westerink MAJ, Warth S, Liu H, Jin P, Stroncek D, Li QZ, Wang E, Wu X, Heath SL, Li Z, Alekseyenko AV, Jiang W. Systemic translocation of Staphylococcus drives autoantibody production in HIV disease. MICROBIOME 2019; 7:25. [PMID: 30764863 PMCID: PMC6376754 DOI: 10.1186/s40168-019-0646-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 02/05/2019] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increased autoreactive antibodies have been reported in HIV disease; however, the mechanism accounting for autoantibody induction in HIV remains unknown. RESULTS Herein, we show that seasonal influenza vaccination induces autoantibody production (e.g., IgG anti-nuclear antibody (ANA) and anti-double-stranded DNA antibody (anti-dsDNA)) in some viral-suppressed antiretroviral therapy (ART)-treated HIV+ subjects, but not in healthy controls. These autoantibodies were not derived from antigen-specific B cells but from activated "bystander" B cells analyzed by single-cell assay and by study of purified polyclonal ANAs from plasma. To explore the mechanism of autoantibody generation in HIV+ subjects, plasma level of microbial products, gene expression profile of B cells, and B cell receptor (BCR) repertoires were analyzed. We found that autoantibody production was associated with increased plasma level of microbial translocation; the patients with high autoantibodies had skewed B cell repertoires and upregulation of genes related to innate immune activation in response to microbial translocation. By analyzing circulating microbial 16S rDNA in plasma, the relative abundance of Staphylococcus was found to be associated with autoantibody production in HIV+ subjects. Finally, we found that injection of heat-killed Staphylococcus aureus promoted germinal center B cell responses and autoantibody production in mice, consistent with the notion that autoantibody production in HIV+ patients is triggered by microbial products. CONCLUSIONS Our results showed that translocation of Staphylococcus can promote B cell activation through enhancing germinal center response and induces autoantibody production. It uncovers a potential mechanism linking microbial translocation and autoimmunity in HIV+ disease and provides a strong rationale for targeting Staphylococcus to prevent autoantibody production.
Collapse
Affiliation(s)
- Zhenwu Luo
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Min Li
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Yongxia Wu
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zhefeng Meng
- Department of Gastroenterology, Oncology Bioinformatics Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Lisa Martin
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lumin Zhang
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Elizabeth Ogunrinde
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zejun Zhou
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Shenghui Qin
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Zhuang Wan
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Maria Anna Julia Westerink
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Stephanie Warth
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Hui Liu
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - Ping Jin
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - David Stroncek
- Cell Processing Section (CPS), Department of Transfusion Medicine, Clinical Center, NIH, Bethesda, 20892, USA
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Ena Wang
- Sidra Medical and Research Center, Doha, Qatar
| | - Xueling Wu
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY, 10016, USA
| | - Sonya L Heath
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Zihai Li
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA
| | - Alexander V Alekseyenko
- Program for Human Microbiome Research, Biomedical Informatics Center, Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Ave. BSB208D, Charleston, SC, 29425, USA.
- Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
32
|
Ulloa-Morales AJ, Goodyear CS, Silverman GJ. Essential Domain-Dependent Roles Within Soluble IgG for in vivo Superantigen Properties of Staphylococcal Protein A: Resolving the B-Cell Superantigen Paradox. Front Immunol 2018; 9:2011. [PMID: 30283436 PMCID: PMC6156153 DOI: 10.3389/fimmu.2018.02011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/15/2018] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a common commensal and frequent opportunistic pathogen that causes invasive infections that often recur. Co-evolution with the host has led to the development of toxins that affect diverse immune cell types. Recent reports have highlighted the contributions of staphylococcal protein A (SpA). This small oligomeric secreted protein contains 4–5 homologous domains with two distinct immunoglobulin-binding sites; one for IgG Fc domains, while a separate site binds an evolutionarily conserved surface on Fab encoded by VHIII clan related genes. The Fab-binding site has been implicated in in vivo supraclonal VHIII-BCR targeted B-cell depletion by an activation induced death pathway. Yet the concept of a superantigen for B lymphocytes poses a seeming paradox. Unlike TCR that are expressed only in a membrane-associated form, BCR are expressed in both a membrane BCR form and in secreted Ig forms, which permeate virtually every part of the body at high levels. We therefore asked, why circulating immunoglobulin do not block the superantigen properties of SpA? Herein, we show that soluble IgG molecules are not in vivo inhibitors of these B-cell superantigen effects but are instead essential for potentiating these properties. We also show that the Fc subclass of circulating IgG is an indirect critical determinant of the B-cell superantigen effect. In contrast, host FcγR and complement are not required for SpA mediated in vivo B-cell depletion. Unexpectedly, after VHIII-IgG2a pretreatment SpA challenge resulted in fatal anaphylactic reactions, which we speculate may have involved FcγR interactions with mast cells and basophils. Cumulatively, our findings illuminate a cunning and potent molecular strategy by which a bacterial toxin effectively confounds the contributions of host B-lymphocytes to immune defenses.
Collapse
Affiliation(s)
- Alejandro J Ulloa-Morales
- Laboratory of B-cell Immunobiology, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Gregg J Silverman
- Laboratory of B-cell Immunobiology, Department of Medicine, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
33
|
Abstract
Staphylococcus aureus is a Gram-positive opportunistic pathogen that causes superficial and invasive infections in the hospital and community. High mortality from infection emphasizes the need for improved methods for prevention and treatment. Although S. aureus possesses an arsenal of virulence factors that contribute to evasion of host defenses, few studies have examined long-term humoral and B-cell responses. Adults with acute-phase skin and soft tissue infections were recruited; blood samples were obtained; and S. aureus isolates, including methicillin-resistant strains, were subjected to genomic sequence analysis. In comparisons of acute-phase sera with convalescent-phase sera, a minority (37.5%) of patients displayed 2-fold or greater increases in antibody titers against three or more S. aureus antigens, whereas nearly half exhibited no changes, despite the presence of toxin genes in most infecting strains. Moreover, enhanced antibody responses waned over time, which could reflect a defect in B-cell memory or long-lived plasma cells. However, memory B cells reactive with a range of S. aureus antigens were prevalent at both acute-phase and convalescent-phase time points. While some memory B cells exhibited toxin-specific binding, those cross-reactive with structurally related leucocidin subunits were dominant across patients, suggesting the targeting of conserved epitopes. Memory B-cell reactivity correlated with serum antibody levels for selected S. aureus exotoxins, suggesting a relationship between the cellular and humoral compartments. Overall, although there was no global defect in the representation of anti-S. aureus memory B cells, there was evidence of restrictions in the range of epitopes recognized, which may suggest potential therapeutic approaches for augmenting host defenses. The contribution of B-cell memory and long-term antibody responses to host defenses against S. aureus exotoxins remains poorly understood. Our studies confirmed that infection did not commonly lead to enhanced long-term humoral responses. Whereas circulating memory B cells against S. aureus secreted exotoxins were prevalent, they were dominated by cross-reactivity with structurally related leucocidin subunits, consistent with recognition of conserved epitopes. These findings also provide the first evidence of a relationship between the reactivity of antistaphylococcal circulating memory B cells and serum antibody levels. In general, infection was not associated with a global defect in B-cell memory for S. aureus secreted factors, and responses were highly dominated by cross-reactivity to structurally related exotoxins, which arguably may alone be suboptimal in providing host defenses. Our studies illuminate aspects of the S. aureus-host relationship that may better inform strategies for the development of an effective protective vaccine.
Collapse
|
34
|
Kapur R, Catalina MD, Aslam R, Speck ER, Francovitch RF, Semple JW. A highly purified form of staphylococcal protein A alleviates murine immune thrombocytopenia (ITP). Br J Haematol 2017; 183:501-503. [DOI: 10.1111/bjh.14985] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Rick Kapur
- Division of Haematology and Transfusion Medicine; Lund University; Lund Sweden
| | | | - Rukhsana Aslam
- The Toronto Platelet Immunobiology Group; Toronto ON Canada
- Keenan Research Centre for Biomedical Science at St. Michael's Hospital; Toronto ON Canada
| | - Edwin R. Speck
- The Toronto Platelet Immunobiology Group; Toronto ON Canada
- Keenan Research Centre for Biomedical Science at St. Michael's Hospital; Toronto ON Canada
| | | | - John W. Semple
- Division of Haematology and Transfusion Medicine; Lund University; Lund Sweden
- The Toronto Platelet Immunobiology Group; Toronto ON Canada
- Keenan Research Centre for Biomedical Science at St. Michael's Hospital; Toronto ON Canada
- Department of Pharmacology; University of Toronto; Toronto ON Canada
- Department of Medicine; University of Toronto; Toronto ON Canada. Department of Laboratory Medicine and Pathobiology; University of Toronto; Toronto ON Canada. Canadian Blood Services; Toronto ON Canada
| |
Collapse
|