1
|
Tan Z, Yang W, O'Brien NA, Pan X, Ramadan S, Marsh T, Hammer N, Cywes-Bentley C, Vinacur M, Pier GB, Gildersleeve JC, Huang X. A comprehensive synthetic library of poly-N-acetyl glucosamines enabled vaccine against lethal challenges of Staphylococcus aureus. Nat Commun 2024; 15:3420. [PMID: 38658531 PMCID: PMC11043332 DOI: 10.1038/s41467-024-47457-4] [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: 10/19/2023] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
Poly-β-(1-6)-N-acetylglucosamine (PNAG) is an important vaccine target, expressed on many pathogens. A critical hurdle in developing PNAG based vaccine is that the impacts of the number and the position of free amine vs N-acetylation on its antigenicity are not well understood. In this work, a divergent strategy is developed to synthesize a comprehensive library of 32 PNAG pentasaccharides. This library enables the identification of PNAG sequences with specific patterns of free amines as epitopes for vaccines against Staphylococcus aureus (S. aureus), an important human pathogen. Active vaccination with the conjugate of discovered PNAG epitope with mutant bacteriophage Qβ as a vaccine carrier as well as passive vaccination with diluted rabbit antisera provides mice with near complete protection against infections by S. aureus including methicillin-resistant S. aureus (MRSA). Thus, the comprehensive PNAG pentasaccharide library is an exciting tool to empower the design of next generation vaccines.
Collapse
Affiliation(s)
- Zibin Tan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Center for Cancer Immunology, Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, 518000, China
| | - Weizhun Yang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
| | - Nicholas A O'Brien
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Xingling Pan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Sherif Ramadan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
- Chemistry Department, Faculty of Science, Benha University, Benha, Qaliobiya, 13518, Egypt
| | - Terence Marsh
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, 48824, USA
| | - Neal Hammer
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, 48824, USA
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mariana Vinacur
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeffrey C Gildersleeve
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
2
|
Bei J, Wu J, Liu J. Re-N-acetylation of group B Streptococcus type Ia capsular polysaccharide improves the immunogenicity of glycoconjugate vaccines. Carbohydr Polym 2024; 330:121848. [PMID: 38368118 DOI: 10.1016/j.carbpol.2024.121848] [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: 11/16/2023] [Revised: 12/28/2023] [Accepted: 01/19/2024] [Indexed: 02/19/2024]
Abstract
The capsular polysaccharides (CPS) of Group B Streptococcus play a crucial role as virulence determinants and are potential candidates for antigenic components in vaccine formulations. Alkaline treatments are commonly used to extract polysaccharides owing to their efficiency and cost-effectiveness; however, they may induce the removal of N-acetyl groups from CPS. This study involved re-N-acetylation of CPS Ia to improve its biological functionality. The structural modifications and enhanced antigenicity of CPS Ia were observed after re-N-acetylation. The tetanus toxoid (TT) was conjugated with either partially de-N-acetylated or fully re-N-acetylated CPS. As a result, the conjugate containing re-N-acetylated CPS (IaReN-TT) enhanced the induction of IgG antibody levels and functional antibodies in mice. Both passive and active protection assays substantiated the superior protective efficacy of IaReN-TT, suggesting that the re-N-acetylation of CPS Ia could be a critical step in refining the immunogenic profile of glycoconjugate vaccines.
Collapse
Affiliation(s)
- Jiaming Bei
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education; School of Biotechnology, Jiangnan University, Wuxi 214122, China; Suzhou Juwei Biotech Co., Ltd, Suzhou 215000, China
| | - Jianrong Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education; School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Jia Liu
- Suzhou Juwei Biotech Co., Ltd, Suzhou 215000, China
| |
Collapse
|
3
|
Li D, Wang J, Wang X, Qiao Z, Wang L, Wang P, Song N, Li M. β-Glycosylations with 2-Deoxy-2-(2,4-dinitrobenzenesulfonyl)-amino-glucosyl/galactosyl Selenoglycosides: Assembly of Partially N-Acetylated β-(1 → 6)-Oligoglucosaminosides. J Org Chem 2023; 88:9004-9025. [PMID: 37306475 DOI: 10.1021/acs.joc.3c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
An efficient protocol has been established for β-glycosylations with 2-deoxy-2-(2,4-dinitrobenzenesulfonyl)amino (2dDNsNH)-glucopyranosyl/galactopyranosyl selenoglycosides using PhSeCl/AgOTf as an activating system. The reaction features highly β-selective glycosylation with a wide range of alcohol acceptors that are either sterically hindered or poorly nucleophilic. Thioglycoside- and selenoglycoside-based alcohols prove to be viable nucleophiles, opening up new opportunities for one-pot construction of oligosaccharides. The power of this approach is highlighted by the efficient assembly of tri-, hexa-, and nonasaccharides composed of β-(1 → 6)-glucosaminosyl residues based on one-pot preparation of a triglucosaminosyl thioglycoside with DNs, phthaloyl, and 2,2,2-trichloroethoxycarbonyl as the protecting groups of amino groups. These glycans are potential antigens for developing glycoconjugate vaccines against microbial infections.
Collapse
Affiliation(s)
- Dongwei Li
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Jianjun Wang
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Xianyang Wang
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Zhi Qiao
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Lingjun Wang
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Peng Wang
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Ni Song
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Ming Li
- Molecular Synthesis Center, Key Laboratory of Marine Medicine, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, National Laboratory for Marine Science and Technology, Qingdao 266237, China
- Shandong Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
4
|
Balducci E, Papi F, Capialbi DE, Del Bino L. Polysaccharides' Structures and Functions in Biofilm Architecture of Antimicrobial-Resistant (AMR) Pathogens. Int J Mol Sci 2023; 24:ijms24044030. [PMID: 36835442 PMCID: PMC9965654 DOI: 10.3390/ijms24044030] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023] Open
Abstract
Bacteria and fungi have developed resistance to the existing therapies such as antibiotics and antifungal drugs, and multiple mechanisms are mediating this resistance. Among these, the formation of an extracellular matrix embedding different bacterial cells, called biofilm, is an effective strategy through which bacterial and fungal cells are establishing a relationship in a unique environment. The biofilm provides them the possibility to transfer genes conferring resistance, to prevent them from desiccation and to impede the penetration of antibiotics or antifungal drugs. Biofilms are formed of several constituents including extracellular DNA, proteins and polysaccharides. Depending on the bacteria, different polysaccharides form the biofilm matrix in different microorganisms, some of them involved in the first stage of cells' attachment to surfaces and to each other, and some responsible for giving the biofilm structure resistance and stability. In this review, we describe the structure and the role of different polysaccharides in bacterial and fungal biofilms, we revise the analytical methods to characterize them quantitatively and qualitatively and finally we provide an overview of potential new antimicrobial therapies able to inhibit biofilm formation by targeting exopolysaccharides.
Collapse
Affiliation(s)
| | | | - Daniela Eloisa Capialbi
- GSK, 53100 Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | | |
Collapse
|
5
|
Sorieul C, Dolce M, Romano MR, Codée J, Adamo R. Glycoconjugate vaccines against antimicrobial resistant pathogens. Expert Rev Vaccines 2023; 22:1055-1078. [PMID: 37902243 DOI: 10.1080/14760584.2023.2274955] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/20/2023] [Indexed: 10/31/2023]
Abstract
INTRODUCTION Antimicrobial resistance (AMR) is responsible for the death of millions worldwide and stands as a major threat to our healthcare systems, which are heavily reliant on antibiotics to fight bacterial infections. The development of vaccines against the main pathogens involved is urgently required as prevention remains essential against the rise of AMR. AREAS COVERED A systematic research review was conducted on MEDLINE database focusing on the six AMR pathogens defined as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli), which are considered critical or high priority pathogens by the World Health Organization (WHO) and the Centers for Disease Control and Prevention (CDC). The analysis was intersecated with the terms carbohydrate, glycoconjugate, bioconjugate, glyconanoparticle, and multiple presenting antigen system vaccines. EXPERT OPINION Glycoconjugate vaccines have been successful in preventing meningitis and pneumoniae, and there are high expectations that they will play a key role in fighting AMR. We herein discuss the recent technological, preclinical, and clinical advances, as well as the challenges associated with the development of carbohydrate-based vaccines against leading AMR bacteria, with focus on the ESKAPE pathogens. The need of innovative clinical and regulatory approaches to tackle these targets is also highlighted.
Collapse
Affiliation(s)
- Charlotte Sorieul
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Marta Dolce
- GSK, Via Fiorentina 1, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Jeroen Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | | |
Collapse
|
6
|
Han J, Poma A. Molecular Targets for Antibody-Based Anti-Biofilm Therapy in Infective Endocarditis. Polymers (Basel) 2022; 14:3198. [PMID: 35956712 PMCID: PMC9370930 DOI: 10.3390/polym14153198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Infective endocarditis (IE) is a heart disease caused by the infection of heart valves, majorly caused by Staphilococcus aureus. IE is initiated by bacteria entering the blood circulation in favouring conditions (e.g., during invasive procedures). So far, the conventional antimicrobial strategies based on the usage of antibiotics remain the major intervention for treating IE. Nevertheless, the therapeutic efficacy of antibiotics in IE is limited not only by the bacterial drug resistance, but also by the formation of biofilms, which resist the penetration of antibiotics into bacterial cells. To overcome these drawbacks, the development of anti-biofilm treatments that can expose bacteria and make them more susceptible to the action of antibiotics, therefore resulting in reduced antimicrobial resistance, is urgently required. A series of anti-biofilm strategies have been developed, and this review will focus in particular on the development of anti-biofilm antibodies. Based on the results previously reported in the literature, several potential anti-biofilm targets are discussed, such as bacterial adhesins, biofilm matrix and bacterial toxins, covering their antigenic properties (with the identification of potential promising epitopes), functional mechanisms, as well as the antibodies already developed against these targets and, where feasible, their clinical translation.
Collapse
Affiliation(s)
- Jiahe Han
- UCL Institute of Cardiovascular Science, The Rayne Building, 5 University Street, London WC1E 6JF, UK
| | - Alessandro Poma
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Hospital, UCL Medical School, Rowland Hill Street, London NW3 2PF, UK
| |
Collapse
|
7
|
An Overview of Biofilm Formation-Combating Strategies and Mechanisms of Action of Antibiofilm Agents. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081110. [PMID: 35892912 PMCID: PMC9394423 DOI: 10.3390/life12081110] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/19/2022]
Abstract
Biofilm formation on surfaces via microbial colonization causes infections and has become a major health issue globally. The biofilm lifestyle provides resistance to environmental stresses and antimicrobial therapies. Biofilms can cause several chronic conditions, and effective treatment has become a challenge due to increased antimicrobial resistance. Antibiotics available for treating biofilm-associated infections are generally not very effective and require high doses that may cause toxicity in the host. Therefore, it is essential to study and develop efficient anti-biofilm strategies that can significantly reduce the rate of biofilm-associated healthcare problems. In this context, some effective combating strategies with potential anti-biofilm agents, including plant extracts, peptides, enzymes, lantibiotics, chelating agents, biosurfactants, polysaccharides, organic, inorganic, and metal nanoparticles, etc., have been reviewed to overcome biofilm-associated healthcare problems. From their extensive literature survey, it can be concluded that these molecules with considerable structural alterations might be applied to the treatment of biofilm-associated infections, by evaluating their significant delivery to the target site of the host. To design effective anti-biofilm molecules, it must be assured that the minimum inhibitory concentrations of these anti-biofilm compounds can eradicate biofilm-associated infections without causing toxic effects at a significant rate.
Collapse
|
8
|
Colonization and Infection of Indwelling Medical Devices by Staphylococcus aureus with an Emphasis on Orthopedic Implants. Int J Mol Sci 2022; 23:ijms23115958. [PMID: 35682632 PMCID: PMC9180976 DOI: 10.3390/ijms23115958] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/08/2023] Open
Abstract
The use of indwelling medical devices has constantly increased in recent years and has revolutionized the quality of life of patients affected by different diseases. However, despite the improvement of hygiene conditions in hospitals, implant-associated infections remain a common and serious complication in prosthetic surgery, mainly in the orthopedic field, where infection often leads to implant failure. Staphylococcus aureus is the most common cause of biomaterial-centered infection. Upon binding to the medical devices, these bacteria proliferate and develop dense communities encased in a protective matrix called biofilm. Biofilm formation has been proposed as occurring in several stages-(1) attachment; (2) proliferation; (3) dispersal-and involves a variety of host and staphylococcal proteinaceous and non-proteinaceous factors. Moreover, biofilm formation is strictly regulated by several control systems. Biofilms enable staphylococci to avoid antimicrobial activity and host immune response and are a source of persistent bacteremia as well as of localized tissue destruction. While considerable information is available on staphylococcal biofilm formation on medical implants and important results have been achieved on the treatment of biofilms, preclinical and clinical applications need to be further investigated. Thus, the purpose of this review is to gather current studies about the mechanism of infection of indwelling medical devices by S. aureus with a special focus on the biochemical factors involved in biofilm formation and regulation. We also provide a summary of the current therapeutic strategies to combat biomaterial-associated infections and highlight the need to further explore biofilm physiology and conduct research for innovative anti-biofilm approaches.
Collapse
|
9
|
Dollery SJ, Harro JM, Wiggins TJ, Wille BP, Kim PC, Tobin JK, Bushnell RV, Tasker NJPER, MacLeod DA, Tobin GJ. Select Whole-Cell Biofilm-Based Immunogens Protect against a Virulent Staphylococcus Isolate in a Stringent Implant Model of Infection. Vaccines (Basel) 2022; 10:vaccines10060833. [PMID: 35746441 PMCID: PMC9231243 DOI: 10.3390/vaccines10060833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Many microbes of concern to human health remain without vaccines. We have developed a whole-microbe inactivation technology that enables us to rapidly inactivate large quantities of a pathogen while retaining epitopes that were destroyed by previous inactivation methods. The method that we call UVC-MDP inactivation can be used to make whole-cell vaccines with increased potency. We and others are exploring the possibility of using improved irradiation-inactivation technologies to develop whole-cell vaccines for numerous antibiotic-resistant microbes. Here, we apply UVC-MDP to produce candidate MRSA vaccines which we test in a stringent tibia implant model of infection challenged with a virulent MSRA strain. We report high levels of clearance in the model and observe a pattern of protection that correlates with the immunogen protein profile used for vaccination.
Collapse
Affiliation(s)
- Stephen J. Dollery
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
- Correspondence:
| | - Janette M. Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Taralyn J. Wiggins
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Brendan P. Wille
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - Peter C. Kim
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; (J.M.H.); (B.P.W.); (P.C.K.)
| | - John K. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Ruth V. Bushnell
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Naomi J. P. E. R. Tasker
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - David A. MacLeod
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| | - Gregory J. Tobin
- Biological Mimetics, Inc., Frederick, MD 21702, USA; (T.J.W.); (J.K.T.); (R.V.B.); (N.J.P.E.R.T.); (D.A.M.); (G.J.T.)
| |
Collapse
|
10
|
Liu S, Wu X, Chandra S, Lyon C, Ning B, jiang L, Fan J, Hu TY. Extracellular vesicles: Emerging tools as therapeutic agent carriers. Acta Pharm Sin B 2022; 12:3822-3842. [PMID: 36213541 PMCID: PMC9532556 DOI: 10.1016/j.apsb.2022.05.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are secreted by both eukaryotes and prokaryotes, and are present in all biological fluids of vertebrates, where they transfer DNA, RNA, proteins, lipids, and metabolites from donor to recipient cells in cell-to-cell communication. Some EV components can also indicate the type and biological status of their parent cells and serve as diagnostic targets for liquid biopsy. EVs can also natively carry or be modified to contain therapeutic agents (e.g., nucleic acids, proteins, polysaccharides, and small molecules) by physical, chemical, or bioengineering strategies. Due to their excellent biocompatibility and stability, EVs are ideal nanocarriers for bioactive ingredients to induce signal transduction, immunoregulation, or other therapeutic effects, which can be targeted to specific cell types. Herein, we review EV classification, intercellular communication, isolation, and characterization strategies as they apply to EV therapeutics. This review focuses on recent advances in EV applications as therapeutic carriers from in vitro research towards in vivo animal models and early clinical applications, using representative examples in the fields of cancer chemotherapeutic drug, cancer vaccine, infectious disease vaccines, regenerative medicine and gene therapy. Finally, we discuss current challenges for EV therapeutics and their future development.
Collapse
|
11
|
Caldara M, Belgiovine C, Secchi E, Rusconi R. Environmental, Microbiological, and Immunological Features of Bacterial Biofilms Associated with Implanted Medical Devices. Clin Microbiol Rev 2022; 35:e0022120. [PMID: 35044203 PMCID: PMC8768833 DOI: 10.1128/cmr.00221-20] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The spread of biofilms on medical implants represents one of the principal triggers of persistent and chronic infections in clinical settings, and it has been the subject of many studies in the past few years, with most of them focused on prosthetic joint infections. We review here recent works on biofilm formation and microbial colonization on a large variety of indwelling devices, ranging from heart valves and pacemakers to urological and breast implants and from biliary stents and endoscopic tubes to contact lenses and neurosurgical implants. We focus on bacterial abundance and distribution across different devices and body sites and on the role of environmental features, such as the presence of fluid flow and properties of the implant surface, as well as on the interplay between bacterial colonization and the response of the human immune system.
Collapse
Affiliation(s)
- Marina Caldara
- Interdepartmental Center on Safety, Technologies, and Agri-food Innovation (SITEIA.PARMA), University of Parma, Parma, Italy
| | - Cristina Belgiovine
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Scuola di Specializzazione in Microbiologia e Virologia, Università degli Studi di Pavia, Pavia, Italy
| | - Eleonora Secchi
- Institute of Environmental Engineering, ETH Zürich, Zürich, Switzerland
| | - Roberto Rusconi
- IRCCS Humanitas Research Hospital, Rozzano–Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele–Milan, Italy
| |
Collapse
|
12
|
Synthetic carbohydrate-based cell wall components from Staphylococcus aureus. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 38:35-43. [PMID: 34895639 DOI: 10.1016/j.ddtec.2021.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/17/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022]
Abstract
Glycopolymers are found surrounding the outer layer of many bacterial species. The first uses as immunogenic component in vaccines are reported since the beginning of the XX century, but it is only in the last decades that glycoconjugate based vaccines have been effectively applied for controlling and preventing several infectious diseases, such as H. influenzae type b (Hib), N. meningitidis, S. pneumoniae or group B Streptococcus. Methicillin resistant S. aureus (MRSA) strains has been appointed by the WHO as one of those pathogens, for which new treatments are urgently needed. Herein we present an overview of the carbohydrate-based cell wall polymers associated with different S. aureus strains and the related affords to deliver well-defined fragments through synthetic chemistry.
Collapse
|
13
|
Taus NS, Cywes-Bentley C, Johnson WC, Pier GB, Fry LM, Mousel MR, Ueti MW. Immunization against a Conserved Surface Polysaccharide Stimulates Bovine Antibodies with Opsonic Killing Activity but Does Not Protect against Babesia bovis Challenge. Pathogens 2021; 10:pathogens10121598. [PMID: 34959553 PMCID: PMC8709247 DOI: 10.3390/pathogens10121598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
Arthropod-borne apicomplexan pathogens remain a great concern and challenge for disease control in animals and humans. In order to prevent Babesia infection, the discovery of antigens that elicit protective immunity is essential to establish approaches to stop disease dissemination. In this study, we determined that poly-N-acetylglucosamine (PNAG) is conserved among tick-borne pathogens including B. bovis, B. bigemina, B. divergens, B. microti, and Babesia WA1. Calves immunized with synthetic ß-(1→6)-linked glucosamine oligosaccharides conjugated to tetanus toxoid (5GlcNH2-TT) developed antibodies with in vitro opsonophagocytic activity against Staphylococcus aureus. Sera from immunized calves reacted to B. bovis. These results suggest strong immune responses against PNAG. However, 5GlcNH2-TT-immunized bovines challenged with B. bovis developed acute babesiosis with the cytoadhesion of infected erythrocytes to brain capillary vessels. While this antigen elicited antibodies that did not prevent disease, we are continuing to explore other antigens that may mitigate these vector-borne diseases for the cattle industry.
Collapse
Affiliation(s)
- Naomi S. Taus
- Animal Disease Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Pullman, WA 99164, USA; (W.C.J.); (L.M.F.); (M.R.M.); (M.W.U.)
- Correspondence: ; Tel.: +(509)-335-6318; Fax: +(509)-335-8328
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.C.-B.); (G.B.P.)
| | - Wendell C. Johnson
- Animal Disease Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Pullman, WA 99164, USA; (W.C.J.); (L.M.F.); (M.R.M.); (M.W.U.)
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (C.C.-B.); (G.B.P.)
| | - Lindsay M. Fry
- Animal Disease Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Pullman, WA 99164, USA; (W.C.J.); (L.M.F.); (M.R.M.); (M.W.U.)
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, Pullman, WA 99164, USA
| | - Michelle R. Mousel
- Animal Disease Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Pullman, WA 99164, USA; (W.C.J.); (L.M.F.); (M.R.M.); (M.W.U.)
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| | - Massaro W. Ueti
- Animal Disease Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Pullman, WA 99164, USA; (W.C.J.); (L.M.F.); (M.R.M.); (M.W.U.)
- Program in Vector-Borne Diseases, Department of Veterinary Microbiology and Pathology, Pullman, WA 99164, USA
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
14
|
Mirzaei B, Babaei R, Zeighami H, Dadar M, Soltani A. Staphylococcus aureus Putative Vaccines Based on the Virulence Factors: A Mini-Review. Front Microbiol 2021; 12:704247. [PMID: 34539603 PMCID: PMC8447878 DOI: 10.3389/fmicb.2021.704247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022] Open
Abstract
Since the 1960s, the frequency of methicillin-resistant Staphylococcus aureus as a recurrent cause of nosocomial infections has increased. Since multidrug-resistant Staphylococcus has overcome antimicrobial treatment, the development of putative vaccines based on virulence factors could be a great help in controlling the infections caused by bacteria and are actively being pursued in healthcare settings. This mini-review provides an overview of the recent progress in vaccine development, immunogenicity, and therapeutic features of some S. aureus macromolecules as putative vaccine candidates and their implications against human S. aureus-related infections. Based on the reviewed experiments, multivalent vaccines could prevent the promotion of the diseases caused by this bacterium and enhance the prevention chance of S. aureus infections.
Collapse
Affiliation(s)
- Bahman Mirzaei
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ryhaneh Babaei
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Habib Zeighami
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Dadar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | - Ali Soltani
- Department of English Language, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
15
|
Suresh MK, Vasudevan AK, Biswas L, Biswas R. Protective efficacy of Alum adjuvanted Amidase protein vaccine against Staphylococcus aureus infection in multiple mouse models. J Appl Microbiol 2021; 132:1422-1434. [PMID: 34487603 DOI: 10.1111/jam.15291] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/05/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
AIMS Staphylococcus aureus is an opportunistic pathogen of humans. No commercial vaccine is available to combat S. aureus infections. In this study, we have investigated the protective immune response generated by S. aureus non-covalently associated cell wall surface protein N-acetylmuramoyl-L-alanine amidase (AM) in combination with Alum (Al) and heat-killed S. aureus (hkSA) using murine models. METHODS AND RESULTS BALB/c mice were immunized with increasing concentrations of AM antigen or hkSA to determine their optimum concentration for vaccination. Fifty micrograms of AM and hkSA each were found to generate maximum anti-AM IgG antibody production. BALB/c mice were immunized next with 50 µg of AM, 50 µg of hKSA and 1 mg Al vaccine formulation. Vaccine efficacy was validated by challenging immunized BALB/c mice with S. aureus Newman and three clinical methicillin-resistant S. aureus strains. AM-hkSA-Al-immunized mice generated high anti-AM IgG antibody response with IgG1 and IgG2b as the predominant immunoglobulin subtypes. Increased survival (60%-90%) with decreased clinical disease symptoms was observed in the vaccinated BALB/c mice group. A significantly lower bacterial load and decreased kidney abscess formation was observed following the challenge with S. aureus in the vaccinated BALB/c mice group. Furthermore, the efficacy of AM-hkSA-Al vaccine was also validated using C57 BL/6 and Swiss albino mice. CONCLUSIONS Using murine infection models, we have demonstrated that AM-hkSA-Al vaccine would be effective in preventing S. aureus infections. SIGNIFICANCE AND IMPACT OF STUDY AM-hkSA-Al vaccine elicited strong immune response and may be considered for future vaccine design against S. aureus infections.
Collapse
Affiliation(s)
- Maneesha K Suresh
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Anil Kumar Vasudevan
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, AIMS - Ponekkara, Cochin, India
| | - Lalitha Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | - Raja Biswas
- Center for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
16
|
Basu N, Ghosh R. Recent chemical syntheses of bacteria related oligosaccharides using modern expeditious approaches. Carbohydr Res 2021; 507:108295. [PMID: 34271477 DOI: 10.1016/j.carres.2021.108295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/15/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022]
Abstract
Apart from some essential and crucial roles in life processes carbohydrates also are involved in a few detrimental courses of action related to human health, like infections by pathogenic microbes, cancer metastasis, transplanted tissue rejection, etc. Regarding management of pathogenesis by microbes, keeping in mind of multi drug-resistant bacteria and epidemic or endemic incidents, preventive measure by vaccination is the best pathway as also recommended by the WHO; by vaccination, eradication of bacterial diseases is also possible. Although some valid vaccines based on attenuated bacterial cells or isolated pure polysaccharide-antigens or the corresponding conjugates thereof are available in the market for prevention of several bacterial diseases, but these are not devoid of some disadvantages also. In order to develop improved conjugate T-cell dependent vaccines oligosaccharides related to bacterial antigens are synthesized and converted to the corresponding carrier protein conjugates. Marketed Cuban Quimi-Hib is such a vaccine being used since 2004 to resist Haemophilus influenza b infections. During nearly the past two decades research is going on worldwide for improved synthesis of bacteria related oligosaccharides or polysaccharides towards development of such semisynthetic or synthetic glycoconjugate vaccines. The present dissertation is an endeavour to encompass the recent syntheses of several pathogenic bacterial oligosaccharides or polysaccharides, made during the past ten-eleven years with special reference to modern expeditious syntheses.
Collapse
Affiliation(s)
- Nabamita Basu
- Department of Chemistry, Nabagram Hiralal Paul College, Konnagar, Hoogly, West Bengal, 712246, India
| | - Rina Ghosh
- Department of Chemistry, Jadavpur University, Kolkata, 700 032, India.
| |
Collapse
|
17
|
Rainard P, Gilbert FB, Germon P, Foucras G. Invited review: A critical appraisal of mastitis vaccines for dairy cows. J Dairy Sci 2021; 104:10427-10448. [PMID: 34218921 DOI: 10.3168/jds.2021-20434] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/23/2021] [Indexed: 11/19/2022]
Abstract
Infections of the mammary gland remain a frequent disease of dairy ruminants that negatively affect animal welfare, milk quality, farmer serenity, and farming profitability and cause an increase in use of antimicrobials. There is a need for efficacious vaccines to alleviate the burden of mastitis in dairy farming, but this need has not been satisfactorily fulfilled despite decades of research. A careful appraisal of past and current research on mastitis vaccines reveals the peculiarities but also the commonalities among mammary gland infections associated with the major mastitis pathogens Escherichia coli, Staphylococcus aureus, Streptococcus uberis, Streptococcus agalactiae, or Streptococcus dysgalactiae. A major pitfall is that the immune mechanisms of effective protection have not been fully identified. Until now, vaccine development has been directed toward the generation of antibodies. In this review, we drew up an inventory of the main approaches used to design vaccines that aim at the major pathogens for the mammary gland, and we critically appraised the current and tentative vaccines. In particular, we sought to relate efficacy to vaccine-induced defense mechanisms to shed light on some possible reasons for current vaccine shortcomings. Based on the lessons learned from past attempts and the recent results of current research, the design of effective vaccines may take a new turn in the years to come.
Collapse
Affiliation(s)
- Pascal Rainard
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France.
| | - Florence B Gilbert
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France
| | - Pierre Germon
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Tours, Infectiologie et Santé Publique, 37380 Nouzilly, France
| | - Gilles Foucras
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Université de Toulouse, École Nationale Vétérinaire de Toulouse, Interactions Hôtes-Agents Pathogènes, 31076 Toulouse, France
| |
Collapse
|
18
|
Dorado‐Morales P, Martínez I, Rivero‐Buceta V, Díaz E, Bähre H, Lasa I, Solano C. Elevated c-di-GMP levels promote biofilm formation and biodesulfurization capacity of Rhodococcus erythropolis. Microb Biotechnol 2021; 14:923-937. [PMID: 33128507 PMCID: PMC8085952 DOI: 10.1111/1751-7915.13689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/29/2022] Open
Abstract
Bacterial biofilms provide high cell density and a superior adaptation and protection from stress conditions compared to planktonic cultures, making them a very promising approach for bioremediation. Several Rhodococcus strains can desulfurize dibenzothiophene (DBT), a major sulphur pollutant in fuels, reducing air pollution from fuel combustion. Despite multiple efforts to increase Rhodococcus biodesulfurization activity, there is still an urgent need to develop better biocatalysts. Here, we implemented a new approach that consisted in promoting Rhodococcus erythropolis biofilm formation through the heterologous expression of a diguanylate cyclase that led to the synthesis of the biofilm trigger molecule cyclic di-GMP (c-di-GMP). R. erythropolis biofilm cells displayed a significantly increased DBT desulfurization activity when compared to their planktonic counterparts. The improved biocatalyst formed a biofilm both under batch and continuous flow conditions which turns it into a promising candidate for the development of an efficient bioreactor for the removal of sulphur heterocycles present in fossil fuels.
Collapse
Affiliation(s)
- Pedro Dorado‐Morales
- Laboratory of Microbial PathogenesisNavarrabiomed‐Universidad Pública de Navarra (UPNA)‐Complejo Hospitalario de Navarra (CHN)IdiSNAIrunlarrea 3PamplonaNavarra31008Spain
| | - Igor Martínez
- Department of Systems BiologyCentro Nacional de BiotecnologíaAgencia Estatal Consejo Superior de Investigaciones CientíficasDarwin 3Madrid28049Spain
| | - Virginia Rivero‐Buceta
- Department of Microbial and Plant BiotechnologyCentro de Investigaciones Biológicas Margarita SalasAgencia Estatal Consejo Superior de Investigaciones CientíficasRamiro de Maeztu 9Madrid28040Spain
| | - Eduardo Díaz
- Department of Microbial and Plant BiotechnologyCentro de Investigaciones Biológicas Margarita SalasAgencia Estatal Consejo Superior de Investigaciones CientíficasRamiro de Maeztu 9Madrid28040Spain
| | - Heike Bähre
- Research Core Unit MetabolomicsHannover Medical SchoolCarl‐Neuberg‐Straße 1Hannover30625Germany
| | - Iñigo Lasa
- Laboratory of Microbial PathogenesisNavarrabiomed‐Universidad Pública de Navarra (UPNA)‐Complejo Hospitalario de Navarra (CHN)IdiSNAIrunlarrea 3PamplonaNavarra31008Spain
| | - Cristina Solano
- Laboratory of Microbial PathogenesisNavarrabiomed‐Universidad Pública de Navarra (UPNA)‐Complejo Hospitalario de Navarra (CHN)IdiSNAIrunlarrea 3PamplonaNavarra31008Spain
| |
Collapse
|
19
|
Abstract
Introduction: As a result of progress in medical care, a huge number of medical devices are used in the treatment of human diseases. In turn, biofilm-related infection has become a growing threat due to the tolerance of biofilms to antimicrobials, a problem magnified by the development of antimicrobial resistance worldwide. As a result, successful treatment of biofilm-disease using only antimicrobials is problematic.Areas covered: We summarize some alternative approaches to classic antimicrobials for the treatment of biofilm disease. This review is not intended to be exhaustive but to give a clinical picture of alternatives to antimicrobial agents to manage biofilm disease. We highlight those strategies that may be closer to application in clinical practice.Expert opinion: There are a number of outstanding challenges in the development of novel antibiofilm therapies. Screening for effective antibiofilm compounds requires models relevant to all clinical scenarios. Although in vitro research of anti-biofilm strategies has progressed significantly over the past decade, there is a lack of in vivo research. In addition, the complexity of biofilm biology makes it difficult to develop a compound that is likely to provide the single 'magic bullet'. The multifaceted nature of biofilms imposes the need for multi-targeted or combinatorial therapies.
Collapse
Affiliation(s)
- Jose L Del Pozo
- Infectious Diseases Division, Clínica Universidad De Navarra, Pamplona, Spain.,Department of Microbiology, Clínica Universidad De Navarra, Pamplona, Spain.,Laboratory of Microbial Biofilms, Clínica Universidad De Navarra, Pamplona, Spain
| |
Collapse
|
20
|
Pirolli NH, Bentley WE, Jay SM. Bacterial Extracellular Vesicles and the Gut-Microbiota Brain Axis: Emerging Roles in Communication and Potential as Therapeutics. Adv Biol (Weinh) 2021; 5:e2000540. [PMID: 33857347 DOI: 10.1002/adbi.202000540] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 03/24/2021] [Indexed: 12/20/2022]
Abstract
Bacterial extracellular vesicles (BEVs) have emerged as candidate signaling vectors for long-distance interkingdom communication within the gut-microbiota brain axis. Most bacteria release these nanosized vesicles, capable of signaling to the brain via their abundant protein and small RNA cargo, possibly directly via crossing the blood-brain barrier. BEVs have been shown to regulate brain gene expression and induce pathology at most stages of neuroinflammation and neurodegeneration, and thus they may play a causal role in diseases such as Alzheimer's, Parkinson's, and depression/anxiety. On the other hand, BEVs have intrinsic therapeutic properties that may be relevant to probiotic therapy and can also be engineered to function as drug delivery vehicles and vaccines. Thus, BEVs may be both a cause of and solution to neuropathological conditions. In this review, current knowledge of the physiological roles of BEVs as well as state of the art pertaining to the development of therapeutic BEVs in the context of the microbiome-gut-brain axis are summarized.
Collapse
Affiliation(s)
- Nicholas H Pirolli
- Fischell Department of Bioengineering, University of Maryland, 3102 A. James Clark Hall, College Park, MD, 20742, USA
| | - William E Bentley
- Fischell Department of Bioengineering, Robert E. Fischell Institute, and Institute for Bioscience and Biotechnology Research, University of Maryland, 5120A A. James Clark Hall, College Park, MD, 20742, USA
| | - Steven M Jay
- Fischell Department of Bioengineering and Program in Molecular and Cell Biology, University of Maryland, 3116 A. James Clark Hall, College Park, MD, 20742, USA
| |
Collapse
|
21
|
Mirzaei B, Babaei R, Haghshenas MR, Mohammadi F, Homayoni P, Shafaei E. PIA and rSesC Mixture Arisen Antibodies Could Inhibit the Biofilm-Formation in Staphylococcus aureus. Rep Biochem Mol Biol 2021; 10:1-12. [PMID: 34277863 PMCID: PMC8279720 DOI: 10.52547/rbmb.10.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/11/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Staphylococcus aureus as a causative agent of hospital-acquired infections has been considered as the primary concern in biomaterial-related infections (BAIs). METHODS Following the purification of polysaccharide intercellular adhesion (PIA) as an efficient macromolecule in biofilm formation in the native condition, recombinant S. epidermidis surface-exposed rSesC protein, with the most homology to clumping factor A (ClfA) in S. aureus was cloned and expressed in a prokaryotic host as well. Fourier transform infrared spectrometry (FTIR) and Western blotting procedure analyzed purified PIA and protein, respectively. Then, the immune response was evaluated by measuring total IgG titers. Moreover, the capacity of Anti-biofilm forming activity of arisen antibodies to a biofilm-forming S. aureus strains was assessed by the semi-quantitative micro-plate procedure. RESULTS Data showed that the total IgGs were boosted in mice immunized sera. By performing an inhibition assay, the biofilm inhibitory effect of secreted antibodies to test strain was observed. Arisen antibodies against the mixture significantly were more potent than PIA and rSesC, when comparing individual antigens in a biofilm inhibition assay. CONCLUSION immunization of mice with mentioned antigens especially a mixture of them, could eliminate the biofilm formation process in S. aureus. Hopefully, this study corresponds to the suggestion that the immunization of mice with PIA and rSesC candidate vaccines could protect against S. aureus infection.
Collapse
Affiliation(s)
- Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences.
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science.
| | - Ryhane Babaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences.
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science.
| | - Mohammad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences.
| | - Fatemeh Mohammadi
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science.
| | - Pegah Homayoni
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science.
| | - Ebrahim Shafaei
- Infectious diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| |
Collapse
|
22
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
23
|
Mirzaei B, Babaei R, Valinejad S. Staphylococcal Vaccine Antigens related to biofilm formation. Hum Vaccin Immunother 2021; 17:293-303. [PMID: 32498595 PMCID: PMC7872035 DOI: 10.1080/21645515.2020.1767449] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/05/2020] [Indexed: 10/24/2022] Open
Abstract
The number and frequency of multidrug-resistant (MDR) strains as a frequent cause of nosocomial infections have increased, especially for Methicillin-resistant Staphylococcus aureus and Staphylococcus epidermidis, in part due to device-related infections. The transition to antibiotic-resistance in related bacterial genes and the capability for immune escape have increased the sustainability of biofilms produced by these bacteria. The formation and changes in biofilms have been suggested as a target to prevent or treat staphylococcal infections. Thus, this study reviews the development of candidate staphylococcal vaccines by database searching, and evaluates the immunogenicity and efficacy profiles of bacterial components involved in biofilms. The literature suggests that using common staphylococcal vaccine antigens and multivalent vaccines should further enhance vaccine efficacy.
Collapse
Affiliation(s)
- Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Ryhaneh Babaei
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Sina Valinejad
- Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| |
Collapse
|
24
|
The staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020. [PMID: 33240473 DOI: 10.1016/jcsbj202010027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Exopolysaccharide is a key part of the extracellular matrix that contributes to important mechanisms of bacterial pathogenicity, most notably biofilm formation and immune evasion. In the human pathogens Staphylococcus aureus and S. epidermidis, as well as in many other staphylococcal species, the only exopolysaccharide is polysaccharide intercellular adhesin (PIA), a cationic, partially deacetylated homopolymer of N-acetylglucosamine, whose biosynthetic machinery is encoded in the ica locus. PIA production is strongly dependent on environmental conditions and controlled by many regulatory systems. PIA contributes significantly to staphylococcal biofilm formation and immune evasion mechanisms, such as resistance to antimicrobial peptides and ingestion and killing by phagocytes, and presence of the ica genes is associated with infectivity. Due to its role in pathogenesis, PIA has raised considerable interest as a potential vaccine component or target.
Collapse
|
25
|
Nguyen HTT, Nguyen TH, Otto M. The staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020; 18:3324-3334. [PMID: 33240473 PMCID: PMC7674160 DOI: 10.1016/j.csbj.2020.10.027] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/20/2022] Open
Abstract
PIA is a key extracellular matrix component in staphylococci and other bacteria. PIA is a cationic, partially deacetylated N-acetylglucosamine polymer. PIA has a major role in bacterial biofilms and biofilm-associated infection.
Exopolysaccharide is a key part of the extracellular matrix that contributes to important mechanisms of bacterial pathogenicity, most notably biofilm formation and immune evasion. In the human pathogens Staphylococcus aureus and S. epidermidis, as well as in many other staphylococcal species, the only exopolysaccharide is polysaccharide intercellular adhesin (PIA), a cationic, partially deacetylated homopolymer of N-acetylglucosamine, whose biosynthetic machinery is encoded in the ica locus. PIA production is strongly dependent on environmental conditions and controlled by many regulatory systems. PIA contributes significantly to staphylococcal biofilm formation and immune evasion mechanisms, such as resistance to antimicrobial peptides and ingestion and killing by phagocytes, and presence of the ica genes is associated with infectivity. Due to its role in pathogenesis, PIA has raised considerable interest as a potential vaccine component or target.
Collapse
Affiliation(s)
- Hoai T T Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA.,School of Biotechnology, International University, Vietnam National University of Ho Chi Minh City, Khu Pho 6, Thu Duc, Ho Chi Minh City, Viet Nam
| | - Thuan H Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| |
Collapse
|
26
|
Gening ML, Pier GB, Nifantiev NE. Broadly protective semi-synthetic glycoconjugate vaccine against pathogens capable of producing poly-β-(1→6)-N-acetyl-d-glucosamine exopolysaccharide. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 35-36:13-21. [PMID: 33388124 DOI: 10.1016/j.ddtec.2020.09.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 11/15/2022]
Abstract
Poly-β-(1→6)-N-acetylglucosamine (PNAG) was first discovered as a major component of biofilms formed by Staphylococcus aureus and some other staphylococci but later this exopolysaccharide was also found to be produced by pathogens of various nature. This common antigen is considered as a promising target for construction of a broadly protective vaccine. Extensive studies of PNAG, its de-N-acetylated derivative (dPNAG, containing around 15% of residual N-acetates) and their conjugates with Tetanus Toxoid (TT) revealed the crucial role of de-N-acetylated glucosamine units for the induction of protective immunity. Conjugates of synthetic penta- (5GlcNH2) and nona-β-(1→6)-d-glucosamines (9GlcNH2) were tested in vitro and in different animal models and proved to be effective in passive and active protection against different microbial pathogens. Presently conjugate 5GlcNH2-TT is being produced under GMP conditions and undergoes safety and effectiveness evaluation in humans and economically important animals. Current review summarizes all stages of this long-termed study.
Collapse
Affiliation(s)
- Marina L Gening
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, Boston, MA 02115, USA.
| | - Nikolay E Nifantiev
- Laboratory of Glycoconjugate Chemistry, N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia.
| |
Collapse
|
27
|
Cohen ND, Cywes-Bentley C, Kahn SM, Bordin AI, Bray JM, Wehmeyer SG, Pier GB. Vaccination of yearling horses against poly-N-acetyl glucosamine fails to protect against infection with Streptococcus equi subspecies equi. PLoS One 2020; 15:e0240479. [PMID: 33057397 PMCID: PMC7561144 DOI: 10.1371/journal.pone.0240479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/27/2020] [Indexed: 12/04/2022] Open
Abstract
Strangles is a common disease of horses with worldwide distribution caused by the bacterium Streptococcus equi subspecies equi (SEE). Although vaccines against strangles are available commercially, these products have limitations in safety and efficacy. The microbial surface antigen β 1→6 poly-N-acetylglucosamine (PNAG) is expressed by SEE. Here we show that intramuscular (IM) injection alone or a combination of IM plus intranasal (IN) immunization generated antibodies to PNAG that functioned to deposit complement and mediate opsonophagocytic killing of SEE ex vivo. However, immunization strategies targeting PNAG either by either IM only injection or a combination of IM and IN immunizations failed to protect yearling horses against infection following contact with infected horses in an experimental setting. We speculate that a protective vaccine against strangles will require additional components, such as those targeting SEE enzymes that degrade or inactivate equine IgG.
Collapse
Affiliation(s)
- Noah D. Cohen
- Equine Infectious Disease Laboratory, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
- * E-mail: (NDC); (GBP)
| | - Colette Cywes-Bentley
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
| | - Susanne M. Kahn
- Equine Infectious Disease Laboratory, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Angela I. Bordin
- Equine Infectious Disease Laboratory, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Jocelyne M. Bray
- Equine Infectious Disease Laboratory, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - S. Garrett Wehmeyer
- Equine Infectious Disease Laboratory, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Gerald B. Pier
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
- * E-mail: (NDC); (GBP)
| |
Collapse
|
28
|
Laverde D, Romero-Saavedra F, Argunov DA, Enotarpi J, Krylov VB, Kalfopoulou E, Martini C, Torelli R, van der Marel GA, Sanguinetti M, Codée JDC, Nifantiev NE, Huebner J. Synthetic Oligomers Mimicking Capsular Polysaccharide Diheteroglycan are Potential Vaccine Candidates against Encapsulated Enterococcal Infections. ACS Infect Dis 2020; 6:1816-1826. [PMID: 32364376 DOI: 10.1021/acsinfecdis.0c00063] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Infections caused by Enterococcus spp. are a major concern in the clinical setting. In Enterococcus faecalis, the capsular polysaccharide diheteroglycan (DHG), composed of ß-d-galactofuranose-(1 → 3)-ß-d-glucopyranose repeats, has been described as an important virulence factor and as a potential vaccine candidate against encapsulated strains. Synthetic structures emulating immunogenic polysaccharides present many advantages over native polysaccharides for vaccine development. In this work, we described the synthesis of a library of DHG oligomers, differing in length and order of the monosaccharide constituents. Using suitably protected thioglycoside building blocks, oligosaccharides up to 8-mer in length built up from either Galf-Glcp or Glcp-Galf dimers were generated, and we evaluated their immunoreactivity with antibodies raised against DHG. After the screening, we selected two octasaccharides, having either a galactofuranose or glucopyranose terminus, which were conjugated to a carrier protein for the production of polyclonal antibodies. The resulting antibodies were specific toward the synthetic structures and mediated in vitro opsonophagocytic killing of different encapsulated E. feacalis strains. The evaluated oligosaccharides are the first synthetic structures described to elicit antibodies that target encapsulated E. faecalis strains and are, therefore, promising candidates for the development of a well-defined enterococcal glycoconjugate vaccine.
Collapse
Affiliation(s)
- D. Laverde
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich 80337, Germany
| | - F. Romero-Saavedra
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich 80337, Germany
| | - D. A. Argunov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119334, Russia
| | - J. Enotarpi
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, Netherlands
| | - V. B. Krylov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119334, Russia
| | - E. Kalfopoulou
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich 80337, Germany
| | - C. Martini
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - R. Torelli
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome 00168, Italy
| | - G. A. van der Marel
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, Netherlands
| | - M. Sanguinetti
- Istituto di Microbiologia, Università Cattolica del Sacro Cuore, Rome 00168, Italy
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome 00168, Italy
| | - J. D. C. Codée
- Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, Netherlands
| | - N. E. Nifantiev
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow 119334, Russia
| | - J. Huebner
- Division of Paediatric Infectious Diseases, Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich 80337, Germany
| |
Collapse
|
29
|
Menendez-Gil P, Caballero CJ, Catalan-Moreno A, Irurzun N, Barrio-Hernandez I, Caldelari I, Toledo-Arana A. Differential evolution in 3'UTRs leads to specific gene expression in Staphylococcus. Nucleic Acids Res 2020; 48:2544-2563. [PMID: 32016395 PMCID: PMC7049690 DOI: 10.1093/nar/gkaa047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 12/05/2019] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
The evolution of gene expression regulation has contributed to species differentiation. The 3' untranslated regions (3'UTRs) of mRNAs include regulatory elements that modulate gene expression; however, our knowledge of their implications in the divergence of bacterial species is currently limited. In this study, we performed genome-wide comparative analyses of mRNAs encoding orthologous proteins from the genus Staphylococcus and found that mRNA conservation was lost mostly downstream of the coding sequence (CDS), indicating the presence of high sequence diversity in the 3'UTRs of orthologous genes. Transcriptomic mapping of different staphylococcal species confirmed that 3'UTRs were also variable in length. We constructed chimeric mRNAs carrying the 3'UTR of orthologous genes and demonstrated that 3'UTR sequence variations affect protein production. This suggested that species-specific functional 3'UTRs might be specifically selected during evolution. 3'UTR variations may occur through different processes, including gene rearrangements, local nucleotide changes, and the transposition of insertion sequences. By extending the conservation analyses to specific 3'UTRs, as well as the entire set of Escherichia coli and Bacillus subtilis mRNAs, we showed that 3'UTR variability is widespread in bacteria. In summary, our work unveils an evolutionary bias within 3'UTRs that results in species-specific non-coding sequences that may contribute to bacterial diversity.
Collapse
Affiliation(s)
- Pilar Menendez-Gil
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-Gobierno de Navarra, 31192-Mutilva, Navarra, Spain
| | - Carlos J Caballero
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-Gobierno de Navarra, 31192-Mutilva, Navarra, Spain
| | - Arancha Catalan-Moreno
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-Gobierno de Navarra, 31192-Mutilva, Navarra, Spain
| | - Naiara Irurzun
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-Gobierno de Navarra, 31192-Mutilva, Navarra, Spain
| | - Inigo Barrio-Hernandez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Isabelle Caldelari
- Université de Strasbourg, CNRS, Architecture et Réactivité de l’ARN, UPR9002, F-67000-Strasbourg, France
| | - Alejandro Toledo-Arana
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-Gobierno de Navarra, 31192-Mutilva, Navarra, Spain
| |
Collapse
|
30
|
Gholami SA, Goli HR, Haghshenas MR, Mirzaei B. Evaluation of polysaccharide intercellular adhesion (PIA) and glycerol teichoic acid (Gly-TA) arisen antibodies to prevention of biofilm formation in Staphylococcus aureus and Staphylococcus epidermidis strains. BMC Res Notes 2019; 12:691. [PMID: 31653277 PMCID: PMC6815028 DOI: 10.1186/s13104-019-4736-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/15/2019] [Indexed: 02/21/2023] Open
Abstract
Objective Staphylococcus aureus and S. epidermidis as opportunistic pathogens, notable for their frequency and severity of infections are recognized as the most usual reasons for medical device-associated infections that strike hospitalized patients and also immunocompromised individuals. In this study, the polysaccharide intercellular adhesion (PIA) and Glycerol teichoic acid) Gly-TA) as two major macromolecules in the biofilm formation process were purified under the native condition and their structure was analyzed by using colorimetric assays and Fourier Transform Infrared spectroscopy (FTIR). Afterward, the immune response of macromolecules and the mixture of them were assessed by measuring total IgG titers. Subsequently, biofilm inhibitory effects of raising antibodies to biofilm former S. aureus and S. epidermidis were evaluated. Results Obtained data were shown a significant rise in levels of antibodies in immunized mice with mentioned antibodies in comparison with the control group. According to the obtained findings, mentioned antibodies could eliminate S. aureus and S. epidermidis biofilm formation in vitro assays. This survey confirms the proposal that immunization of mice with a mixture of Gly-TA and PIA vaccine could be secure and protected against S. epidermidis and S. aureus infection.
Collapse
Affiliation(s)
- Sanaz Amir Gholami
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Mohammad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science, Sari, Iran. .,Department of Medical Microbiology and Virology, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran.
| |
Collapse
|
31
|
Caballero CJ, Menendez-Gil P, Catalan-Moreno A, Vergara-Irigaray M, García B, Segura V, Irurzun N, Villanueva M, Ruiz de Los Mozos I, Solano C, Lasa I, Toledo-Arana A. The regulon of the RNA chaperone CspA and its auto-regulation in Staphylococcus aureus. Nucleic Acids Res 2019; 46:1345-1361. [PMID: 29309682 PMCID: PMC5815144 DOI: 10.1093/nar/gkx1284] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022] Open
Abstract
RNA-binding proteins (RBPs) are essential to fine-tune gene expression. RBPs containing the cold-shock domain are RNA chaperones that have been extensively studied. However, the RNA targets and specific functions for many of them remain elusive. Here, combining comparative proteomics and RBP-immunoprecipitation-microarray profiling, we have determined the regulon of the RNA chaperone CspA of Staphylococcus aureus. Functional analysis revealed that proteins involved in carbohydrate and ribonucleotide metabolism, stress response and virulence gene expression were affected by cspA deletion. Stress-associated phenotypes such as increased bacterial aggregation and diminished resistance to oxidative-stress stood out. Integration of the proteome and targetome showed that CspA post-transcriptionally modulates both positively and negatively the expression of its targets, denoting additional functions to the previously proposed translation enhancement. One of these repressed targets was its own mRNA, indicating the presence of a negative post-transcriptional feedback loop. CspA bound the 5′UTR of its own mRNA disrupting a hairpin, which was previously described as an RNase III target. Thus, deletion of the cspA 5′UTR abrogated mRNA processing and auto-regulation. We propose that CspA interacts through a U-rich motif, which is located at the RNase III cleavage site, portraying CspA as a putative RNase III-antagonist.
Collapse
Affiliation(s)
- Carlos J Caballero
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Pilar Menendez-Gil
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Arancha Catalan-Moreno
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Marta Vergara-Irigaray
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain.,Navarrabiomed-Universidad Pública de Navarra (UPNA)-Complejo Hospitalario de Navarra (CHN), IDISNA. 31008 Pamplona, Navarra, Spain
| | - Begoña García
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain.,Navarrabiomed-Universidad Pública de Navarra (UPNA)-Complejo Hospitalario de Navarra (CHN), IDISNA. 31008 Pamplona, Navarra, Spain
| | - Víctor Segura
- Genomics, Proteomics and Bioinformatics Unit. Center for Applied Medical Research. University of Navarra. 31008 Pamplona, Spain
| | - Naiara Irurzun
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Maite Villanueva
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Igor Ruiz de Los Mozos
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| | - Cristina Solano
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain.,Navarrabiomed-Universidad Pública de Navarra (UPNA)-Complejo Hospitalario de Navarra (CHN), IDISNA. 31008 Pamplona, Navarra, Spain
| | - Iñigo Lasa
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain.,Navarrabiomed-Universidad Pública de Navarra (UPNA)-Complejo Hospitalario de Navarra (CHN), IDISNA. 31008 Pamplona, Navarra, Spain
| | - Alejandro Toledo-Arana
- Instituto de Agrobiotecnología. IDAB, CSIC-UPNA-Gobierno de Navarra. 31192-Mutilva, Navarra, Spain
| |
Collapse
|
32
|
Bahonar S, Ghazvinian M, Haghshenas MR, Goli HR, Mirzaei B. Purification of PIA and rSesC as Putative Vaccine Candidates Against Staphylococcus aureus. Rep Biochem Mol Biol 2019; 8:161-167. [PMID: 31832440 PMCID: PMC6844615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/10/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Staphylococcus aureus is predominant at sites of biomaterial-associated infection (BAI) and frequently infects hospitalized individuals. METHODS The polysaccharide intercellular adhesin (PIA) and S. epidermidis rSesC protein, major macromolecules in biofilm formation, were purified under native conditions and cloned and expressed in a prokaryotic host. RESULTS LPurification of the macromolecules was confirmed by FTIR and Western blotting. CONCLUSION The S. epidermidis SesC protein and PIA were uccessfully purified. Both are considered as vaccine candidates.
Collapse
Affiliation(s)
- Sara Bahonar
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
- Molecular and cell biology Research center, Faculty of Medicine, Mazandaran University of Medical sciences, Sari, Iran.
| | - Maryam Ghazvinian
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Mohamad Reza Haghshenas
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Hamid Reza Goli
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
| | - Bahman Mirzaei
- Department of Medical Microbiology and Virology, Faculty of Medicine, Mazandaran University of Medical Science.
- Molecular and cell biology Research center, Faculty of Medicine, Mazandaran University of Medical sciences, Sari, Iran.
| |
Collapse
|
33
|
σ B Inhibits Poly- N-Acetylglucosamine Exopolysaccharide Synthesis and Biofilm Formation in Staphylococcus aureus. J Bacteriol 2019; 201:JB.00098-19. [PMID: 30858304 DOI: 10.1128/jb.00098-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/07/2019] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus clinical strains are able to produce at least two distinct types of biofilm matrixes: biofilm matrixes made of the polysaccharide intercellular adhesin (PIA) or poly-N-acetylglucosamine (PNAG), whose synthesis is mediated by the icaADBC locus, and biofilm matrixes built of proteins (polysaccharide independent). σB is a conserved alternative sigma factor that regulates the expression of more than 100 genes in response to changes in environmental conditions. While numerous studies agree that σB is required for polysaccharide-independent biofilms, controversy persists over the role of σB in the regulation of PIA/PNAG-dependent biofilm development. Here, we show that genetically unrelated S. aureus σB-deficient strains produced stronger biofilms under both static and flow conditions and accumulated higher levels of PIA/PNAG exopolysaccharide than their corresponding wild-type strains. The increased accumulation of PIA/PNAG in the σB mutants correlated with a greater accumulation of the IcaC protein showed that it was not due to adjustments in icaADBC operon transcription and/or icaADBC mRNA stability. Overall, our results reveal that in the presence of active σB, the turnover of Ica proteins is accelerated, reducing the synthesis of PIA/PNAG exopolysaccharide and consequently the PIA/PNAG-dependent biofilm formation capacity.IMPORTANCE Due to its multifaceted lifestyle, Staphylococcus aureus needs a complex regulatory network to connect environmental signals with cellular physiology. One particular transcription factor, named σB (SigB), is involved in the general stress response and the expression of virulence factors. For many years, great confusion has existed about the role of σB in the regulation of the biofilm lifestyle in S. aureus Our study demonstrated that σB is not necessary for exopolysaccharide-dependent biofilms and, even more, that S. aureus produces stronger biofilms in the absence of σB The increased accumulation of exopolysaccharide correlates with higher stability of the proteins responsible for its synthesis. The present findings reveal an additional regulatory layer to control biofilm exopolysaccharide synthesis under stress conditions.
Collapse
|
34
|
Folmar CN, Cywes-Bentley C, Bordin AI, Rocha JN, Bray JM, Kahn SK, Schuckert AE, Pier GB, Cohen ND. In vitro evaluation of complement deposition and opsonophagocytic killing of Rhodococcus equi mediated by poly-N-acetyl glucosamine hyperimmune plasma compared to commercial plasma products. J Vet Intern Med 2019; 33:1493-1499. [PMID: 31034109 PMCID: PMC6524092 DOI: 10.1111/jvim.15511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/17/2019] [Indexed: 11/29/2022] Open
Abstract
Background The bacterium Rhodococcus equi can cause severe pneumonia in foals. The absence of a licensed vaccine and limited effectiveness of commercial R. equi hyperimmune plasma (RE‐HIP) create a great need for improved prevention of this disease. Hypothesis Plasma hyperimmune to the capsular polysaccharide poly‐N‐acetyl glucosamine (PNAG) would be significantly more effective than RE‐HIP at mediating complement deposition and opsonophagocytic killing (OPK) of R. equi. Animals Venipuncture was performed on 9 Quarter Horses. Methods The ability of the following plasma sources to mediate complement component 1 (C1) deposition onto either PNAG or R. equi was determined by ELISA: (1) PNAG hyperimmune plasma (PNAG‐HIP), (2) RE‐HIP, and (3) standard non‐hyperimmune commercial plasma (SP). For OPK, each plasma type was combined with R. equi, equine complement, and neutrophils isolated from horses (n = 9); after 4 hours, the number of R. equi in each well was determined by quantitative culture. Data were analyzed using linear mixed‐effects regression with significance set at P < .05. Results The PNAG‐HIP and RE‐HIP were able to deposit significantly (P < .05) more complement onto their respective targets than the other plasmas. The mean proportional survival of R. equi opsonized with PNAG‐HIP was significantly (P < .05) less (14.7%) than that for SP (51.1%) or RE‐HIP (42.2%). Conclusions and Clinical Importance Plasma hyperimmune to PNAG is superior to RE‐HIP for opsonizing and killing R. equi in vitro. Comparison of these 2 plasmas in field trials is warranted because of the reported incomplete effectiveness of RE‐HIP.
Collapse
Affiliation(s)
- Chelsea N Folmar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Colette Cywes-Bentley
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Angela I Bordin
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Joana N Rocha
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Jocelyne M Bray
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Susanne K Kahn
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Amanda E Schuckert
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Gerald B Pier
- Department of Medicine, Harvard Medical School, Harvard University, Boston, Massachusetts
| | - Noah D Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas
| |
Collapse
|
35
|
Bronesky D, Desgranges E, Corvaglia A, François P, Caballero CJ, Prado L, Toledo-Arana A, Lasa I, Moreau K, Vandenesch F, Marzi S, Romby P, Caldelari I. A multifaceted small RNA modulates gene expression upon glucose limitation in Staphylococcus aureus. EMBO J 2019; 38:e99363. [PMID: 30760492 PMCID: PMC6418428 DOI: 10.15252/embj.201899363] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 12/17/2018] [Accepted: 01/21/2019] [Indexed: 01/10/2023] Open
Abstract
Pathogenic bacteria must rapidly adapt to ever-changing environmental signals resulting in metabolism remodeling. The carbon catabolite repression, mediated by the catabolite control protein A (CcpA), is used to express genes involved in utilization and metabolism of the preferred carbon source. Here, we have identified RsaI as a CcpA-repressed small non-coding RNA that is inhibited by high glucose concentrations. When glucose is consumed, RsaI represses translation initiation of mRNAs encoding a permease of glucose uptake and the FN3K enzyme that protects proteins against damage caused by high glucose concentrations. RsaI also binds to the 3' untranslated region of icaR mRNA encoding the transcriptional repressor of exopolysaccharide production and to sRNAs induced by the uptake of glucose-6 phosphate or nitric oxide. Furthermore, RsaI expression is accompanied by a decreased transcription of genes involved in carbon catabolism pathway and an activation of genes involved in energy production, fermentation, and nitric oxide detoxification. This multifaceted RNA can be considered as a metabolic signature when glucose becomes scarce and growth is arrested.
Collapse
Affiliation(s)
- Delphine Bronesky
- Architecture et Réactivité de l'ARN, CNRS, Université de Strasbourg, Strasbourg, France
| | - Emma Desgranges
- Architecture et Réactivité de l'ARN, CNRS, Université de Strasbourg, Strasbourg, France
| | - Anna Corvaglia
- Genomic Research Laboratory, Department of Medical Specialties, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Patrice François
- Genomic Research Laboratory, Department of Medical Specialties, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | | | - Laura Prado
- Instituto de Agrobiotecnología (IdAB), CSIC-UPNA-GN, Navarra, Spain
| | | | - Inigo Lasa
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
| | - Karen Moreau
- CIRI, Centre international de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Univ Lyon, Lyon, France
| | - François Vandenesch
- CIRI, Centre international de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Hospices Civils de Lyon, Univ Lyon, Lyon, France
| | - Stefano Marzi
- Architecture et Réactivité de l'ARN, CNRS, Université de Strasbourg, Strasbourg, France
| | - Pascale Romby
- Architecture et Réactivité de l'ARN, CNRS, Université de Strasbourg, Strasbourg, France
| | - Isabelle Caldelari
- Architecture et Réactivité de l'ARN, CNRS, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
36
|
Raafat D, Otto M, Reppschläger K, Iqbal J, Holtfreter S. Fighting Staphylococcus aureus Biofilms with Monoclonal Antibodies. Trends Microbiol 2019; 27:303-322. [PMID: 30665698 DOI: 10.1016/j.tim.2018.12.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/10/2018] [Accepted: 12/18/2018] [Indexed: 02/07/2023]
Abstract
Staphylococcus aureus (S. aureus) is a notorious pathogen and one of the most frequent causes of biofilm-related infections. The treatment of S. aureus biofilms is hampered by the ability of the biofilm structure to shield bacteria from antibiotics as well as the host's immune system. Therefore, new preventive and/or therapeutic interventions, including the use of antibody-based approaches, are urgently required. In this review, we describe the mechanisms by which anti-S. aureus antibodies can help in combating biofilms, including an up-to-date overview of monoclonal antibodies currently in clinical trials. Moreover, we highlight ongoing efforts in passive vaccination against S. aureus biofilm infections, with special emphasis on promising targets, and finally indicate the direction into which future research could be heading.
Collapse
Affiliation(s)
- Dina Raafat
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Egypt; Current affiliation: Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, US National Institutes of Health, Bethesda, MD, USA
| | - Kevin Reppschläger
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jawad Iqbal
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Department of Immunology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
37
|
Micoli F, Costantino P, Adamo R. Potential targets for next generation antimicrobial glycoconjugate vaccines. FEMS Microbiol Rev 2018; 42:388-423. [PMID: 29547971 PMCID: PMC5995208 DOI: 10.1093/femsre/fuy011] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Cell surface carbohydrates have been proven optimal targets for vaccine development. Conjugation of polysaccharides to a carrier protein triggers a T-cell-dependent immune response to the glycan moiety. Licensed glycoconjugate vaccines are produced by chemical conjugation of capsular polysaccharides to prevent meningitis caused by meningococcus, pneumococcus and Haemophilus influenzae type b. However, other classes of carbohydrates (O-antigens, exopolysaccharides, wall/teichoic acids) represent attractive targets for developing vaccines. Recent analysis from WHO/CHO underpins alarming concern toward antibiotic-resistant bacteria, such as the so called ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.) and additional pathogens such as Clostridium difficile and Group A Streptococcus. Fungal infections are also becoming increasingly invasive for immunocompromised patients or hospitalized individuals. Other emergencies could derive from bacteria which spread during environmental calamities (Vibrio cholerae) or with potential as bioterrorism weapons (Burkholderia pseudomallei and mallei, Francisella tularensis). Vaccination could aid reducing the use of broad-spectrum antibiotics and provide protection by herd immunity also to individuals who are not vaccinated. This review analyzes structural and functional differences of the polysaccharides exposed on the surface of emerging pathogenic bacteria, combined with medical need and technological feasibility of corresponding glycoconjugate vaccines.
Collapse
Affiliation(s)
- Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), Via Fiorentina 1, 53100 Siena
| | | | | |
Collapse
|
38
|
Nagy E, Nagy G, Power CA, Badarau A, Szijártó V. Anti-bacterial Monoclonal Antibodies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1053:119-153. [PMID: 29549638 DOI: 10.1007/978-3-319-72077-7_7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The failing efficacy of antibiotics and the high mortality rate among high-risk patients calls for new treatment modalities for bacterial infections. Due to the vastly divergent pathogenesis of human pathogens, each microbe requires a tailored approach. The main modes of action of anti-bacterial antibodies are virulence factor neutralization, complement-mediated bacterial lysis and enhancement of opsonophagocytic uptake and killing (OPK). Gram-positive bacteria cannot be lysed by complement and their pathogenesis often involves secreted toxins, therefore typically toxin-neutralization and OPK activity are required to prevent and ameliorate disease. In fact, the success stories in terms of approved products, in the anti-bacterial mAb field are based on toxin neutralization (Bacillus anthracis, Clostridium difficile). In contrast, Gram-negative bacteria are vulnerable to antibody-dependent complement-mediated lysis, while their pathogenesis rarely relies on secreted exotoxins, and involves the pro-inflammatory endotoxin (lipopolysaccharide). Given the complexity of bacterial pathogenesis, antibody therapeutics are expected to be most efficient upon targeting more than one virulence factor and/or combining different modes of action. The improved understanding of bacterial pathogenesis combined with the versatility and maturity of antibody discovery technologies available today are pivotal for the design of novel anti-bacterial therapeutics. The intensified research generating promising proof-of-concept data, and the increasing number of clinical programs with anti-bacterial mAbs, indicate that the field is ready to fulfill its promise in the coming years.
Collapse
Affiliation(s)
- Eszter Nagy
- Arsanis Biosciences GmbH/Arsanis, Inc, Vienna, Austria.
| | - Gábor Nagy
- Arsanis Biosciences GmbH/Arsanis, Inc, Vienna, Austria
| | | | | | | |
Collapse
|
39
|
Cywes-Bentley C, Rocha JN, Bordin AI, Vinacur M, Rehman S, Zaidi TS, Meyer M, Anthony S, Lambert M, Vlock DR, Giguère S, Cohen ND, Pier GB. Antibody to Poly-N-acetyl glucosamine provides protection against intracellular pathogens: Mechanism of action and validation in horse foals challenged with Rhodococcus equi. PLoS Pathog 2018; 14:e1007160. [PMID: 30024986 PMCID: PMC6053243 DOI: 10.1371/journal.ppat.1007160] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/15/2018] [Indexed: 12/16/2022] Open
Abstract
Immune correlates of protection against intracellular bacterial pathogens are largely thought to be cell-mediated, although a reasonable amount of data supports a role for antibody-mediated protection. To define a role for antibody-mediated immunity against an intracellular pathogen, Rhodococcus equi, that causes granulomatous pneumonia in horse foals, we devised and tested an experimental system relying solely on antibody-mediated protection against this host-specific etiologic agent. Immunity was induced by vaccinating pregnant mares 6 and 3 weeks prior to predicted parturition with a conjugate vaccine targeting the highly conserved microbial surface polysaccharide, poly-N-acetyl glucosamine (PNAG). We ascertained antibody was transferred to foals via colostrum, the only means for foals to acquire maternal antibody. Horses lack transplacental antibody transfer. Next, a randomized, controlled, blinded challenge was conducted by inoculating at ~4 weeks of age ~106 cfu of R. equi via intrabronchial challenge. Eleven of 12 (91%) foals born to immune mares did not develop clinical R. equi pneumonia, whereas 6 of 7 (86%) foals born to unvaccinated controls developed pneumonia (P = 0.0017). In a confirmatory passive immunization study, infusion of PNAG-hyperimmune plasma protected 100% of 5 foals against R. equi pneumonia whereas all 4 recipients of normal horse plasma developed clinical disease (P = 0.0079). Antibodies to PNAG mediated killing of extracellular and intracellular R. equi and other intracellular pathogens. Killing of intracellular organisms depended on antibody recognition of surface expression of PNAG on infected cells, along with complement deposition and PMN-assisted lysis of infected macrophages. Peripheral blood mononuclear cells from immune and protected foals released higher levels of interferon-γ in response to PNAG compared to controls, indicating vaccination also induced an antibody-dependent cellular release of this critical immune cytokine. Overall, antibody-mediated opsonic killing and interferon-γ release in response to PNAG may protect against diseases caused by intracellular bacterial pathogens. Development of effective vaccines for diseases such as tuberculosis, brucellosis and others caused by intracellular pathogens has proved challenging, as data exist supporting both antibody and cellular immune effectors as mediators of protection. To address this problem against an important, and representative, equine intracellular pathogen, we chose to test a vaccine candidate for the ability to protect horse foals challenged at 4 weeks of age with Rhodococcus equi. To make these foals immune, their pregnant mares were immunized with a vaccine targeting the conserved surface antigen found on many microbes, termed PNAG. Antibody in the pregnant mares was transferred to their foals and, after the foals were challenged, 91% of those born to vaccinated mares were protected against R. equi pneumonia. Meanwhile, 86% of the non-vaccinated controls developed pneumonia. We also showed antibody to PNAG could kill various bacteria that produce this antigen when residing inside of human macrophage cells, a new mechanism for antibody-mediated immunity to intracellular bacteria. These results support the development of PNAG as a vaccine for intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Colette Cywes-Bentley
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
| | - Joana N. Rocha
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Angela I. Bordin
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Mariana Vinacur
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
| | - Safia Rehman
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
| | - Tanweer S. Zaidi
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
| | - Mark Meyer
- Mg Biologics, Ames, IA, United States of America
| | | | | | | | - Steeve Giguère
- College of Veterinary Medicine, University of Georgia, Athens, GA, United States of America
| | - Noah D. Cohen
- College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
- * E-mail: (NDC); (GBP)
| | - Gerald B. Pier
- Harvard Medical School, Brigham & Women’s Hospital, Boston, MA, United States of America
- * E-mail: (NDC); (GBP)
| |
Collapse
|
40
|
Zheng Y, He L, Asiamah TK, Otto M. Colonization of medical devices by staphylococci. Environ Microbiol 2018; 20:3141-3153. [PMID: 29633455 DOI: 10.1111/1462-2920.14129] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/16/2018] [Accepted: 04/03/2018] [Indexed: 12/19/2022]
Abstract
The use of medical devices in modern medicine is constantly increasing. Despite the multiple precautionary strategies that are being employed in hospitals, which include increased hygiene and sterilization measures, bacterial infections on these devices still happen frequently. Staphylococci are among the major causes of medical device infection. This is mostly due to the strong capacity of those bacteria to form device-associated biofilms, which provide resistance to chemical and physical treatments as well as attacks by the host's immune system. Biofilm development is a multistep process with specific factors participating in each step. It is tightly regulated to provide a balance between biofilm expansion and detachment. Detachment from a biofilm on a medical device can lead to severe systemic infection, such as bacteremia and sepsis. While our understanding of staphylococcal biofilm formation has increased significantly and staphylococcal biofilm formation on medical devices is among the best understood biofilm-associated infections, the extensive effort put in preclinical studies with the goal to find novel therapies against staphylococcal device-associated infections has not yet resulted in efficient, applicable therapeutic options for that difficult-to-treat type of disease.
Collapse
Affiliation(s)
- Yue Zheng
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Lei He
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Titus K Asiamah
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
41
|
Immunization with outer membrane vesicles displaying conserved surface polysaccharide antigen elicits broadly antimicrobial antibodies. Proc Natl Acad Sci U S A 2018; 115:E3106-E3115. [PMID: 29555731 DOI: 10.1073/pnas.1718341115] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Many microbial pathogens produce a β-(1→6)-linked poly-N-acetyl-d-glucosamine (PNAG) surface capsule, including bacterial, fungal, and protozoan cells. Broadly protective immune responses to this single conserved polysaccharide antigen in animals are possible but only when a deacetylated poly-N-acetyl-d-glucosamine (dPNAG; <30% acetate) glycoform is administered as a conjugate to a carrier protein. Unfortunately, conventional methods for natural extraction or chemical synthesis of dPNAG and its subsequent conjugation to protein carriers can be technically demanding and expensive. Here, we describe an alternative strategy for creating broadly protective vaccine candidates that involved coordinating recombinant poly-N-acetyl-d-glucosamine (rPNAG) biosynthesis with outer membrane vesicle (OMV) formation in laboratory strains of Escherichia coli The glycosylated outer membrane vesicles (glycOMVs) released by these engineered bacteria were decorated with the PNAG glycopolymer and induced high titers of PNAG-specific IgG antibodies after immunization in mice. When a Staphylococcus aureus enzyme responsible for PNAG deacetylation was additionally expressed in these cells, glycOMVs were generated that elicited antibodies to both highly acetylated PNAG (∼95-100% acetate) and a chemically deacetylated dPNAG derivative (∼15% acetate). These antibodies mediated efficient in vitro killing of two distinct PNAG-positive bacterial species, namely S. aureus and Francisella tularensis subsp. holarctica, and mice immunized with PNAG-containing glycOMVs developed protective immunity against these unrelated pathogens. Collectively, our results reveal the potential of glycOMVs for targeting this conserved polysaccharide antigen and engendering protective immunity against the broad range of pathogens that produce surface PNAG.
Collapse
|
42
|
Human Immunoglobulin G Cannot Inhibit Fibrinogen Binding by the Genetically Diverse A Domain of Staphylococcus aureus Fibronectin-Binding Protein A. mSphere 2018; 3:mSphere00590-17. [PMID: 29564394 PMCID: PMC5853482 DOI: 10.1128/msphere.00590-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/14/2018] [Indexed: 11/20/2022] Open
Abstract
The fibronectin-binding protein A (FnBPA) is a cell surface-associated protein of Staphylococcus aureus which mediates adherence to the host extracellular matrix and is important for bacterial virulence. Previously, substantial sequence diversity was found among strains in the fibrinogen-binding A domain of this protein, and 7 different isotypes were described. The effect of this sequence diversity on the human antibody response, in terms of both antibody production and antibody function, remains unclear. In this study, we identify five different FnBPA A domain isotypes based on the sequence results of 22 clinical S. aureus isolates, obtained from the same number of patients suffering from bacteremia. Using a bead-based Luminex technique, we measure the patients' total immunoglobulin G (IgG) against the 7 FnBPA isotypes at the onset and during the time course of bacteremia (median of 10 serum samples per patient over a median of 35 days). A significant increase in IgG against the FnBPA A domain, including the isotype carried by the infecting strain, is observed in only three out of 22 patients (14%) after the onset of bacteremia. Using a Luminex-based FnBPA-fibrinogen-binding assay, we find that preincubation of recombinant FnBPA isotypes with IgG from diverse patients does not interfere with binding to fibrinogen. This observation is confirmed using an alternative Luminex-based assay and enzyme-linked immunosorbent assay (ELISA). IMPORTANCE Despite the many in vitro and murine in vivo studies involving FnBPA, the actual presence of this virulence factor during human infection is less well established. Furthermore, it is currently unknown to what extent sequence variation in such a virulence factor affects the human antibody response and the ability of antibodies to interfere with FnBPA function. This study sheds new light on these issues. First, the uniform presence of a patient's IgG against FnBPA indicates the presence and importance of this virulence factor during S. aureus pathogenesis. Second, the absence of an increase in antibody production in most patients following bacteremia indicates the complexity of S. aureus-host interactions, possibly involving immune evasion or lack of expression of FnBPA during invasive infection. Finally, we provide new insights into the inability of human antibodies to interfere with FnBPA-fibrinogen binding. These observations should be taken into account during the development of novel vaccination approaches.
Collapse
|
43
|
Sanchez-Larrayoz AF, Elhosseiny NM, Chevrette MG, Fu Y, Giunta P, Spallanzani RG, Ravi K, Pier GB, Lory S, Maira-Litrán T. Complexity of Complement Resistance Factors Expressed by Acinetobacter baumannii Needed for Survival in Human Serum. THE JOURNAL OF IMMUNOLOGY 2017; 199:2803-2814. [PMID: 28855313 DOI: 10.4049/jimmunol.1700877] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/07/2017] [Indexed: 11/19/2022]
Abstract
Acinetobacter baumannii is a bacterial pathogen with increasing impact in healthcare settings, due in part to this organism's resistance to many antimicrobial agents, with pneumonia and bacteremia as the most common manifestations of disease. A significant proportion of clinically relevant A. baumannii strains are resistant to killing by normal human serum (NHS), an observation supported in this study by showing that 12 out of 15 genetically diverse strains of A. baumannii are resistant to NHS killing. To expand our understanding of the genetic basis of A. baumannii serum resistance, a transposon (Tn) sequencing (Tn-seq) approach was used to identify genes contributing to this trait. An ordered Tn library in strain AB5075 with insertions in every nonessential gene was subjected to selection in NHS. We identified 50 genes essential for the survival of A. baumannii in NHS, including already known serum resistance factors, and many novel genes not previously associated with serum resistance. This latter group included the maintenance of lipid asymmetry genetic pathway as a key determinant in protecting A. baumannii from the bactericidal activity of NHS via the alternative complement pathway. Follow-up studies validated the role of eight additional genes identified by Tn-seq in A. baumannii resistance to killing by NHS but not by normal mouse serum, highlighting the human species specificity of A. baumannii serum resistance. The identification of a large number of genes essential for serum resistance in A. baumannii indicates the degree of complexity needed for this phenotype, which might reflect a general pattern that pathogens rely on to cause serious infections.
Collapse
Affiliation(s)
- Amaro F Sanchez-Larrayoz
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Noha M Elhosseiny
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Marc G Chevrette
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Yang Fu
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Peter Giunta
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Raúl G Spallanzani
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Keerthikka Ravi
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| | - Stephen Lory
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Tomás Maira-Litrán
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115; and
| |
Collapse
|
44
|
The Bordetella Bps Polysaccharide Is Required for Biofilm Formation and Enhances Survival in the Lower Respiratory Tract of Swine. Infect Immun 2017; 85:IAI.00261-17. [PMID: 28559403 DOI: 10.1128/iai.00261-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/18/2017] [Indexed: 12/21/2022] Open
Abstract
Bordetella bronchiseptica is pervasive in swine populations and plays multiple roles in respiratory disease. Additionally, B. bronchiseptica is capable of establishing long-term or chronic infections in swine. Bacterial biofilms are increasingly recognized as important contributors to chronic bacterial infections. Recently the polysaccharide locus bpsABCD has been demonstrated to serve a critical role in the development of mature biofilms formed by the sequenced laboratory strain of B. bronchiseptica We hypothesized that swine isolates would also have the ability to form mature biofilms and the bpsABCD locus would serve a key role in this process. A mutant containing an in-frame deletion of the bpsABCD structural genes was constructed in a wild-type swine isolate and found to be negative for poly-N-acetylglucosamine (PNAG)-like material by immunoblot assay. Further, the bpsABCD locus was found to be required for the development and maintenance of the three-dimensional structures under continuous-flow conditions. To investigate the contribution of the bpsABCD locus to the pathogenesis of B. bronchiseptica in swine, the KM22Δbps mutant was compared to the wild-type swine isolate for the ability to colonize and cause disease in pigs. The bpsABCD locus was found to not be required for persistence in the upper respiratory tract of swine. Additionally, the bpsABCD locus did not affect the development of anti-Bordetella humoral immunity, did not contribute to disease severity, and did not mediate protection from complement-mediated killing. However, the bpsABCD locus was found to enhance survival in the lower respiratory tract of swine.
Collapse
|
45
|
Zhao G, Zaidi TS, Bozkurt-Guzel C, Zaidi TH, Lederer JA, Priebe GP, Pier GB. Efficacy of Antibody to PNAG Against Keratitis Caused by Fungal Pathogens. Invest Ophthalmol Vis Sci 2017; 57:6797-6804. [PMID: 28002842 PMCID: PMC5215555 DOI: 10.1167/iovs.16-20358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose Developing immunotherapies for fungal eye infections is a high priority. We analyzed fungal pathogens for expression of the surface polysaccharide, poly-N-acetyl glucosamine (PNAG), and used a mouse model of ocular keratitis caused by Aspergillus flavus, A. fumigatus, or Fusarium solani to determine if PNAG was an immunotherapy target and requirements for ancillary cellular and molecular immune effectors. Methods Enzyme-linked immunosorbent assay (ELISA) or immunofluorescence was used to detect PNAG on fungal cells. Keratitis was induced by scratching corneas of C57BL/6, IL-17R KO, RAG-1 KO, or IL-22 KO mice followed by inoculation with fungal pathogens. Goat antibodies to PNAG, a PNAG-specific human IgG1 monoclonal antibody, or control antibodies were injected either prophylactically plus therapeutically or therapeutically only, and corneal pathology and fungal levels determined in infected eyes at 24 or 48 hours after infection. Results All tested fungal species produced PNAG. Prophylactic or therapeutic treatment by intraperitoneal (IP) injection of antibody to PNAG combined with post-infection topical application of antibody, the latter also used for A. fumigatus, led to reduced fungal levels, corneal pathology, and cytokine expression. Topical administration only of the PNAG monoclonal antibodies (MAb) reduced fungal loads and corneal pathology. There was no antibody protection in IL-17R KO, RAG-1 KO, or IL-22 KO mice. Conclusions Poly-N-acetyl glucosamine is produced by clinically important fungal ocular pathogens. Antibody to PNAG demonstrated protection against Aspergillus and Fusarium keratitis, requiring T cells producing IL-17 and IL-22. These findings indicate the potential to prevent or treat fungal infections by vaccines and immunotherapeutics to PNAG.
Collapse
Affiliation(s)
- Ge Zhao
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Tanweer S Zaidi
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Cagla Bozkurt-Guzel
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Tauqeer H Zaidi
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - James A Lederer
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Gregory P Priebe
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States 3Division of Critical Care Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston, Massachusetts, United States
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
46
|
Echeverz M, García B, Sabalza A, Valle J, Gabaldón T, Solano C, Lasa I. Lack of the PGA exopolysaccharide in Salmonella as an adaptive trait for survival in the host. PLoS Genet 2017; 13:e1006816. [PMID: 28542593 PMCID: PMC5464674 DOI: 10.1371/journal.pgen.1006816] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 06/08/2017] [Accepted: 05/12/2017] [Indexed: 12/22/2022] Open
Abstract
Many bacteria build biofilm matrices using a conserved exopolysaccharide named PGA or PNAG (poly-β-1,6-N-acetyl-D-glucosamine). Interestingly, while E. coli and other members of the family Enterobacteriaceae encode the pgaABCD operon responsible for PGA synthesis, Salmonella lacks it. The evolutionary force driving this difference remains to be determined. Here, we report that Salmonella lost the pgaABCD operon after the divergence of Salmonella and Citrobacter clades, and previous to the diversification of the currently sequenced Salmonella strains. Reconstitution of the PGA machinery endows Salmonella with the capacity to produce PGA in a cyclic dimeric GMP (c-di-GMP) dependent manner. Outside the host, the PGA polysaccharide does not seem to provide any significant benefit to Salmonella: resistance against chlorine treatment, ultraviolet light irradiation, heavy metal stress and phage infection remained the same as in a strain producing cellulose, the main biofilm exopolysaccharide naturally produced by Salmonella. In contrast, PGA production proved to be deleterious to Salmonella survival inside the host, since it increased susceptibility to bile salts and oxidative stress, and hindered the capacity of S. Enteritidis to survive inside macrophages and to colonize extraintestinal organs, including the gallbladder. Altogether, our observations indicate that PGA is an antivirulence factor whose loss may have been a necessary event during Salmonella speciation to permit survival inside the host. During bacterial evolution, specific traits that optimize the organism’s fitness are selected. The production of exopolysaccharides is widespread among bacteria in which they play a protective shielding role as main constituents of biofilms. In contrast to closely related siblings, Salmonella has lost the capacity to produce the exopolysaccharide PGA. Our study reveals that Salmonella lost pga genes, and that the driving force for such a loss may have been the detrimental impact that PGA has during Salmonella invasion of internal organs where it augments the susceptibility to bile salts and oxygen radicals, reducing bacterial survival inside macrophages and rendering Salmonella avirulent. These results suggest that gene-loss has played an important role during Salmonella evolution.
Collapse
Affiliation(s)
- Maite Echeverz
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
| | - Begoña García
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
| | - Amaia Sabalza
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
| | - Jaione Valle
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
| | - Toni Gabaldón
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Cristina Solano
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
- * E-mail: (CS); (IL)
| | - Iñigo Lasa
- Navarrabiomed-Universidad Pública de Navarra-Departamento de Salud, IDISNA, Pamplona, Spain
- * E-mail: (CS); (IL)
| |
Collapse
|
47
|
Skurnik D, Cywes-Bentley C, Pier GB. The exceptionally broad-based potential of active and passive vaccination targeting the conserved microbial surface polysaccharide PNAG. Expert Rev Vaccines 2016; 15:1041-53. [PMID: 26918288 PMCID: PMC4985264 DOI: 10.1586/14760584.2016.1159135] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/24/2016] [Indexed: 11/08/2022]
Abstract
A challenging component of vaccine development is the large serologic diversity of protective antigens. Remarkably, there is a conserved surface/capsular polysaccharide, one of the most effective vaccine targets, expressed by a large number of bacterial, fungal and eukaryotic pathogens: poly-N-acetyl glucosamine (PNAG). Natural antibodies to PNAG are poorly effective at mediating in vitro microbial killing or in vivo protection. Removing most of the acetate substituents to produce a deacetylated glycoform, or using synthetic oligosaccharides of poly-β-1-6-linked glucosamine conjugated to carrier proteins, results in vaccines that elicit high levels of broad-based immunity. A fully human monoclonal antibody is highly active in laboratory and preclinical studies and has been successfully tested in a phase-I setting. Both the synthetic oligosaccharide conjugate vaccine and MAb will be further tested in humans starting in 2016; but, even if effective against only a fraction of the PNAG-producing pathogens, a major advance in vaccine-preventable diseases will occur.
Collapse
Affiliation(s)
- David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 181 Longwood Ave., Boston, MA 02115, Phone: 617-525-2269; FAX: 617-525-2510
| |
Collapse
|
48
|
Guillard T, Pons S, Roux D, Pier GB, Skurnik D. Antibiotic resistance and virulence: Understanding the link and its consequences for prophylaxis and therapy. Bioessays 2016; 38:682-93. [PMID: 27248008 DOI: 10.1002/bies.201500180] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
"Antibiotic resistance is usually associated with a fitness cost" is frequently accepted as common knowledge in the field of infectious diseases. However, with the advances in high-throughput DNA sequencing that allows for a comprehensive analysis of bacterial pathogenesis at the genome scale, including antibiotic resistance genes, it appears that this paradigm might not be as solid as previously thought. Recent studies indicate that antibiotic resistance is able to enhance bacterial fitness in vivo with a concomitant increase in virulence during infections. As a consequence, strategies to minimize antibiotic resistance turn out to be not as simple as initially believed. Indeed, decreased antibiotic use may not be sufficient to let susceptible strains outcompete the resistant ones. Here, we put in perspective these findings and review alternative approaches, such as preventive and therapeutic anti-bacterial immunotherapies that have the potential to by-pass the classic antibiotics.
Collapse
Affiliation(s)
- Thomas Guillard
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Laboratoire de Bactériologie-Virologie-Hygiène hospitalière, Hôpital Robert Debré - CHU de Reims, UFR de Médecine, Université de Reims Champagne-Ardenne, Reims, France
| | - Stéphanie Pons
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Damien Roux
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,INSERM, IAME, UMR 1137, Paris, France.,Univ Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Louis Mourier, Service de Réanimation Médico-Chirurgicale, Colombes, France
| | - Gerald B Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David Skurnik
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
49
|
Diversity of Virulence Factors Associated with West Australian Methicillin-Sensitive Staphylococcus aureus Isolates of Human Origin. BIOMED RESEARCH INTERNATIONAL 2016; 2016:8651918. [PMID: 27247944 PMCID: PMC4876210 DOI: 10.1155/2016/8651918] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/01/2015] [Accepted: 07/12/2015] [Indexed: 11/23/2022]
Abstract
An extensive array of virulence factors associated with S. aureus has contributed significantly to its success as a major nosocomial pathogen in hospitals and community causing variety of infections in affected patients. Virulence factors include immune evading capsular polysaccharides, poly-N-acetyl glucosamine, and teichoic acid in addition to damaging toxins including hemolytic toxins, enterotoxins, cytotoxins, exfoliative toxin, and microbial surface components recognizing adhesive matrix molecules (MSCRAMM). In this investigation, 31 West Australian S. aureus isolates of human origin and 6 controls were analyzed for relative distribution of virulence-associated genes using PCR and/or an immunoassay kit and MSCRAMM by PCR-based typing. Genes encoding MSCRAMM, namely, Spa, ClfA, ClfB, SdrE, SdrD, IsdA, and IsdB, were detected in >90% of isolates. Gene encoding α-toxin was detected in >90% of isolates whereas genes encoding β-toxin and SEG were detectable in 50–60% of isolates. Genes encoding toxin proteins, namely, SEA, SEB, SEC, SED, SEE, SEH, SEI, SEJ, TSST, PVL, ETA, and ETB, were detectable in >50% of isolates. Use of RAPD-PCR for determining the virulence factor-based genetic relatedness among the isolates revealed five cluster groups confirming genetic diversity among the MSSA isolates, with the greatest majority of the clinical S. aureus (84%) isolates clustering in group IIIa.
Collapse
|
50
|
Selle M, Hertlein T, Oesterreich B, Klemm T, Kloppot P, Müller E, Ehricht R, Stentzel S, Bröker BM, Engelmann S, Ohlsen K. Global antibody response to Staphylococcus aureus live-cell vaccination. Sci Rep 2016; 6:24754. [PMID: 27103319 PMCID: PMC4840433 DOI: 10.1038/srep24754] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/05/2016] [Indexed: 02/06/2023] Open
Abstract
The pathogen Staphylococcus aureus causes a broad range of severe diseases and is feared for its ability to rapidly develop resistance to antibiotic substances. The increasing number of highly resistant S. aureus infections has accelerated the search for alternative treatment options to close the widening gap in anti-S. aureus therapy. This study analyses the humoral immune response to vaccination of Balb/c mice with sublethal doses of live S. aureus. The elicited antibody pattern in the sera of intravenously and intramuscularly vaccinated mice was determined using of a recently developed protein array. We observed a specific antibody response against a broad set of S. aureus antigens which was stronger following i.v. than i.m. vaccination. Intravenous but not intramuscular vaccination protected mice against an intramuscular challenge infection with a high bacterial dose. Vaccine protection was correlated with the strength of the anti-S. aureus antibody response. This study identified novel vaccine candidates by using protein microarrays as an effective tool and showed that successful vaccination against S. aureus relies on the optimal route of administration.
Collapse
Affiliation(s)
- Martina Selle
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Tobias Hertlein
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Babett Oesterreich
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Theresa Klemm
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| | - Peggy Kloppot
- University Greifswald, Institute for Microbiology, Greifswald, Germany
| | - Elke Müller
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Ralf Ehricht
- Alere Technologies GmbH, Jena, Germany.,InfectoGnostics Research Campus Jena, Germany
| | - Sebastian Stentzel
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Barbara M Bröker
- University Medicine Greifswald, Department of Immunology, Greifswald, Germany
| | - Susanne Engelmann
- Technical University Braunschweig, Institute for Microbiology, Braunschweig, Germany.,Helmholtz-Zentrum für Infektionsforschung, Mikrobielle Proteomik, Braunschweig, Germany
| | - Knut Ohlsen
- University Würzburg, Institute for Molecular Infection Biology, Würzburg, Germany
| |
Collapse
|