1
|
Shahraki PK, Kiani R, Siavash M, Bemani P. Design of a multi-epitope vaccine against Staphylococcus Aureus lukotoxin ED using in silico approaches. Sci Rep 2025; 15:14517. [PMID: 40280948 PMCID: PMC12032201 DOI: 10.1038/s41598-025-85147-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/01/2025] [Indexed: 04/29/2025] Open
Abstract
Development of a strategy to combat Staphylococcus aureus is a high priority for the World Health Organization. B cell and helper T lymphocyte (HTL) epitopes of leukotoxin ED (LukED) were predicted using computational tools. The predicted epitopes were screened for conservancy, allergenicity, toxicity, autoreactivity, and population coverage. The immunogenic regions of LukED were linked together and to Human β-defensin 3 (hBD3) as adjuvant with appropriate linkers. The predicted 3D structure of the vaccine validated by molecular dynamics (MD) simulations. Subsequently, the 3D structure was docked with the Toll-like receptor (TLR)1/2 to evaluate the binding capacity of the adjuvant. Finally, MD simulation was employed to characterizing the conformational dynamics and stability of this interaction. The predicted epitopes were found to be non-toxic and non-allergenic, with no homology to the human proteome. The vaccine demonstrated a population coverage of 65.15% globally. It was composed of the immunogenic regions of LukED. Molecular docking and MD simulation indicated a stable interaction between hBD3 in the vaccine and TLR1/2 during the simulation period. We have designed vaccine against S. aureus LukED that targets epitope-rich regions, which helps maintain a native-like conformation. This work lays the groundwork for further experimental studies to evaluate the vaccine's neutralizing effects.
Collapse
Affiliation(s)
- Parisa Kh Shahraki
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Razie Kiani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mansour Siavash
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Peyman Bemani
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Touati A, Ibrahim NA, Idres T. Disarming Staphylococcus aureus: Review of Strategies Combating This Resilient Pathogen by Targeting Its Virulence. Pathogens 2025; 14:386. [PMID: 40333163 PMCID: PMC12030135 DOI: 10.3390/pathogens14040386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Staphylococcus aureus is a formidable pathogen notorious for its antibiotic resistance and diverse virulence mechanisms, including toxin production, biofilm formation, and immune evasion. This article explores innovative anti-virulence strategies to disarm S. aureus by targeting critical virulence factors without exerting bactericidal pressure. Key approaches include inhibiting adhesion and biofilm formation, neutralizing toxins, disrupting quorum sensing (e.g., Agr system inhibitors), and blocking iron acquisition pathways. Additionally, interventions targeting two-component regulatory systems are highlighted. While promising, challenges such as strain variability, biofilm resilience, pharmacokinetic limitations, and resistance evolution underscore the need for combination therapies and advanced formulations. Integrating anti-virulence strategies with traditional antibiotics and host-directed therapies offers a sustainable solution to combat multidrug-resistant S. aureus, particularly methicillin-resistant strains (MRSA), and mitigate the global public health crisis.
Collapse
Affiliation(s)
- Abdelaziz Touati
- Laboratory of Microbial Ecology, FSNV, University of Bejaia, Bejaia 06000, Algeria
| | - Nasir Adam Ibrahim
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 13318, Saudi Arabia;
| | - Takfarinas Idres
- Laboratory for Livestock Animal Production and Health Research, Rabie Bouchama National Veterinary School of Algiers, Issad ABBAS Street, BP 161 Oued Smar, Algiers 16059, Algeria;
| |
Collapse
|
3
|
Wang WC, Lee CH, Wu CJ, Leu SJ, Kao PS, Tsai BY, Liu KJ, Chiang YW, Lo HJ, Mao YC, Yang YY. Phage Display Selected Chicken Antibodies Targeting Surface Alpha Enolase in Staphylococcus aureus. Biotechnol J 2025; 20:e70011. [PMID: 40165642 DOI: 10.1002/biot.70011] [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: 11/23/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Staphylococcus aureus, a prevalent gram-positive bacterium in human populations, poses a significant risk for causing serious opportunistic infections and increasing antibiotic resistance. Alpha-enolase in S. aureus plays important roles in extracellular matrix binding and biofilm formation. These functions enable S. aureus to invade host tissues and cause infections. The aim of this study was to develop specific alpha-enolase chicken antibodies through phage display technology targeting S. aureus surface proteins as a potential alternative to antibiotic therapy. A chicken was immunized with recombinant S. aureus alpha-enolase, leading to the construction of two phage display single-chain variable fragment libraries of 3.32 × 106 and 8.60×105 transformants with different linker lengths. After four rounds of biopanning, five single-chain variable fragment antibody clones, including three with high binding affinities (SaS1, SaS2, and SaL2), were selected. These clones exhibited distinct binding patterns in epitope mapping and cross-reaction assays, with SaS1 and SaS2 specifically recognizing S. aureus alpha-enolase and SaL2 cross-reacting with Streptococcus pneumoniae alpha-enolase. Furthermore, the specificity of these antibody clones toward clinical S. aureus strains, including methicillin-sensitive and methicillin-resistant strains, was validated through cell-based enzyme-linked immunosorbent assays (ELISA) and flow cytometry assays. The identification of SaS1, SaS2, and SaL2 underscores their diagnostic and therapeutic potential, offering promising alternatives to traditional antibiotic therapies.
Collapse
Affiliation(s)
- Wei-Chu Wang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chi-Hsin Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| | - Chao-Jung Wu
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Pei-Shih Kao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | | | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Yu-Wei Chiang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsiu-Jung Lo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Hsinchu, Miaoli County, Taiwan
| | - Yan-Chiao Mao
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taipei, Taichung, Taiwan
| | - Yi-Yuan Yang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
4
|
Al-Fetly DR, Rhyaf AG, Naji HA. Protective effects of Staphylococcal Enterotoxin B (SEB) toxoid on lung and liver tissue integrity in rats during systemic infection. IRANIAN JOURNAL OF MICROBIOLOGY 2025; 17:220-228. [PMID: 40337693 PMCID: PMC12053400 DOI: 10.18502/ijm.v17i2.18396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Background and Objectives Staphylococcal enterotoxin B (SEB), a potent superantigenic toxin produced by Staphylococcus aureus (S. aureus), plays a crucial role in S. aureus systemic infection. This investigation sought to determine whether immunising animals with SEB toxoid could protect against an experimental acute systemic infection caused by S. aureus. Materials and Methods This study involved three groups of animals: one group was administered with SEB toxoid, and the second group was administered with intramuscular injections of normal saline, after which both were subjected to systemic S. aureus infection. The third group served as the negative control. After two weeks, the outcomes of the experimental systemic infection demonstrated that SEB immunisation significantly shielded organs (lung and liver) from damage in comparison to the control group. Results Regarding the histopathological analysis of liver and lung tissues, the control group showed minimal alterations, indicating a normal tissue state. Infected individuals exhibited severe pathology, including inflammation, necrosis, and fibrosis. The immunised group displayed a mixed profile with elevated inflammation but lower necrosis and fibrosis. Immunisation mitigated pathological changes induced by infection, fostering a more controlled response. Conclusion SEB plays an important role in S. aureus pathogenesis and immunisation, and this toxoid might protect against fatal infections of S. aureus.
Collapse
Affiliation(s)
- Dhafer Rasheed Al-Fetly
- Department of Internal and Preventive Medicine, College of Veterinary Medicine, University of Al-Qadisiyah, Al-Diwaniyah, Iraq
| | - Atiaf Ghanim Rhyaf
- Department of Pathology, College of Veterinary Medicine, University of Al-Qadisiyah, Al-Diwaniyah, Iraq
| | - Hala Abbas Naji
- Department of Pathology, College of Veterinary Medicine, University of Al-Qadisiyah, Al-Diwaniyah, Iraq
| |
Collapse
|
5
|
Wang W, Li X, Ou Y, Zhou J, Gu Y, Liu B, Zheng Y, Wang Y, Zhang R, Zou Q, Zuo Q, Wang B. Potent human antibodies against SpA5 identified by high-throughput single-cell sequencing of phase I clinical volunteers' B cells. iScience 2025; 28:111627. [PMID: 39834865 PMCID: PMC11743104 DOI: 10.1016/j.isci.2024.111627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/09/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
The drug resistance problem of Staphylococcus aureus needs to be solved urgently. Here, we report the rapid identification of S. aureus human antibodies by high-throughput single-cell RNA and VDJ sequencing of memory B cells derived from 64 volunteers immunized with recombinant five-component S. aureus vaccine (clinical phase I). From 676 antigen-binding IgG1+ clonotypes, TOP10 sequences were selected for expression and characterization, with the most potent one, Abs-9, having nanomolar affinity for the pentameric form of the specific antigen S. aureus protein A. Abs-9 also demonstrated strong prophylactic efficacy in mice injected with lethal doses of a wide range of drug-resistant S. aureus strains. Additionally, the potential epitopes were predicted and validated based on Alphafold2 and molecular docking methods. In all, this study screened for a potent strain of antibody that prevents infection with antibiotic-resistant S. aureus, providing important data to guide the design of vaccines based on antibody architecture.
Collapse
Affiliation(s)
- Wenhao Wang
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xin Li
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing 400038, China
- Department of Cosmetology and Dermatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400021, China
| | - Yangxue Ou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, P.R. China
| | - Jinrui Zhou
- College of Medicine, Southwest Jiaotong University, Chengdu 610083, P.R. China
| | - Yaru Gu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400038, P.R. China
| | - Bixia Liu
- College of Medicine, Southwest Jiaotong University, Chengdu 610083, P.R. China
| | - Yan Zheng
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, P.R. China
| | - Ying Wang
- 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, 857000 Shigatse, China
| | - Rui Zhang
- Department of Clinical Laboratory, Chengdu Military General Hospital, Chengdu 610000, China
| | - Quanming Zou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, P.R. China
| | - Qianfei Zuo
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, P.R. China
| | - Bin Wang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| |
Collapse
|
6
|
Kerkman PF, de Vor L, van der Vaart TW, ten Doesschate T, Muts RM, Depelteau JS, Scheepmaker L, Ruyken M, de Haas CJ, Aerts PC, Marijnissen R, Schuurman J, Beurskens FJ, Gorlani A, Bardoel B, Rooijakkers SH. Single-cell Sequencing of Circulating Human Plasmablasts during Staphylococcus aureus Bacteremia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1644-1655. [PMID: 39451041 PMCID: PMC7616744 DOI: 10.4049/jimmunol.2300858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Staphylococcus aureus is the major cause of healthcare-associated infections, including life-threatening conditions as bacteremia, endocarditis, and implant-associated infections. Despite adequate antibiotic treatment, the mortality of S. aureus bacteremia remains high. This calls for different strategies to treat this infection. In past years, sequencing of Ab repertoires from individuals previously exposed to a pathogen emerged as a successful method to discover novel therapeutic monoclonal Abs and understand circulating B cell diversity during infection. In this paper, we collected peripheral blood from 17 S. aureus bacteremia patients to study circulating plasmablast responses. Using single-cell transcriptome gene expression combined with sequencing of variable heavy and light Ig genes, we retrieved sequences from >400 plasmablasts revealing a high diversity with >300 unique variable heavy and light sequences. More than 200 variable sequences were synthesized to produce recombinant IgGs that were analyzed for binding to S. aureus whole bacterial cells. This revealed four novel monoclonal Abs that could specifically bind to the surface of S. aureus in the absence of Ig-binding surface SpA. Interestingly, three of four mAbs showed cross-reactivity with Staphylococcus epidermidis. Target identification revealed that the S. aureus-specific mAb BC153 targets wall teichoic acid, whereas cross-reactive mAbs BC019, BC020, and BC021 target lipoteichoic acid. All mAbs could induce Fc-dependent phagocytosis of staphylococci by human neutrophils. Altogether, we characterize the active B cell responses to S. aureus in infected patients and identify four functional mAbs against the S. aureus surface, of which three cross-react with S. epidermidis.
Collapse
Affiliation(s)
- Priscilla F. Kerkman
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisanne de Vor
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Thomas W. van der Vaart
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Amsterdam Institute for Infection and Immunity, Infectious Diseases, Amsterdam, The Netherlands
| | - Thijs ten Doesschate
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Department of Internal Medicine, Jeroen Bosch Hospital, Den Bosch, The Netherlands
| | - Remy M. Muts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jamie S. Depelteau
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lisette Scheepmaker
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Maartje Ruyken
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carla J.C. de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Piet C. Aerts
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | | | | | - Bart Bardoel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Suzan H.M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
7
|
Zhang L, Zheng B, Lu J, Wu H, Wu H, Zhang Q, Jiao L, Pan H, Zhou J. Evaluation of human antibodies from vaccinated volunteers for protection against Yersinia pestis infection. Microbiol Spectr 2024; 12:e0105424. [PMID: 39189763 PMCID: PMC11448073 DOI: 10.1128/spectrum.01054-24] [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: 04/26/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Yersinia pestis has a broad host range and has caused lethal bubonic and pneumonic plague in humans. With the emergence of multiple resistant strains and the potential for biothreat use, there is an urgent need for new therapeutic strategies that can protect populations from natural or deliberate infection. Targeting F1 has been proven to be the main strategy for developing vaccines and therapeutic antibodies, but data on anti-F1 antibodies, especially in humans, are scarce. To date, three human anti-F1 monoclonal antibodies (m252, αF1Ig2, and αF1Ig8) from naive populations have been reported. Here, we constructed an antibody library from vaccinees immunized with the plague subunit vaccine IIa by phage display. The genetic basis, epitopes, and biological functions of the obtained mAbs were assessed and evaluated in plague-challenged mice. Three human mAbs, namely, F3, F19, and F23, were identified. Their biolayer responses were 0.4, 0.6, and 0.6 nm, respectively. The dissociation constants (KD) of the F1 antigen were 1 pM, 0.165 nM, and 1 pM, respectively. Although derived from distinct Ab lineages, that is, VH3-30-D3-10-JH4 (F3&F23) and VH3-43-D6-19-JH4 (F19), these mAbs share similar binding sites in F1 with some overlap with αF1Ig8 but are distinct from αF1Ig2. Each of them provided a significant protective effect for Balb/c mice against a 100 median lethal dose (MLD) challenge of a virulent Y. pestis strain when administered at a dose of 100 µg. No synergistic or antagonistic effects were observed among them. These mAbs are novel and excellent candidates for further drug development and use in clinical practice.IMPORTANCEIn this study, we identified three human monoclonal antibodies with a high affinity to F1 protein of Yersinia pestis. We discovered that they have relatively lower somatic hypermutations compared with antibodies, m252, αF1Ig2, and αF1Ig8, derived from the naive library reported previously. We also observed that these mAbs share similar binding sites in F1 with some overlapping with αF1Ig8 but distinct from that of αF1Ig2. Furthermore, each of them could provide complete protection for mice against a lethal dose of Yersinia pestis challenge. Our data provided new insights into the anti-F1 Ab repertories and their associated epitopes during vaccination in humans. The findings support the additional novel protective human anti-F1Abs for potential therapeutics against plaque.
Collapse
Affiliation(s)
- Li Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Binyang Zheng
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jing Lu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Haisheng Wu
- Qinghai Institute for Endemic Disease Control and Prevention, Xining, China
| | - Hailian Wu
- Qinghai Institute for Endemic Disease Control and Prevention, Xining, China
| | - Qi Zhang
- Qinghai Institute for Endemic Disease Control and Prevention, Xining, China
| | - Lei Jiao
- Lanzhou Institute of Biological Products Co., Ltd., State Key Laboratory of Novel Vaccines for Emerging Infectious Diseases, Lanzhou, China
| | - Hongxing Pan
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jianfang Zhou
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
8
|
Karlsson A, Alarcón LA, Piñeiro-Iglesias B, Jacobsson G, Skovbjerg S, Moore ERB, Kopparapu PK, Jin T, Karlsson R. Surface-Shaving of Staphylococcus aureus Strains and Quantitative Proteomic Analysis Reveal Differences in Protein Abundance of the Surfaceome. Microorganisms 2024; 12:1725. [PMID: 39203567 PMCID: PMC11357550 DOI: 10.3390/microorganisms12081725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Staphylococcus aureus is a pathogen known to cause a wide range of infections. To find new targets for identification and to understand host-pathogen interactions, many studies have focused on surface proteins. We performed bacterial-cell surface-shaving, followed by tandem mass tag for quantitative mass spectrometry proteomics, to examine the surfaceome of S. aureus. Two steps were performed, the first step including surface protein-deficient mutants of S. aureus Newman strain lacking important virulence genes (clfA and spa, important for adhesion and immune evasion and srtAsrtB, linking surface-associated virulence factors to the surface) and the second step including isolates of different clinical origin. All strains were compared to the Newman strain. In Step 1, altogether, 7880 peptides were identified, corresponding to 1290 proteins. In Step 2, 4949 peptides were identified, corresponding to 919 proteins and for each strain, approximately 20 proteins showed differential expression compared to the Newman strain. The identified surface proteins were related to host-cell-adherence and immune-system-evasion, biofilm formation, and survival under harsh conditions. The results indicate that surface-shaving of intact S. aureus bacterial strains in combination with quantitative proteomics is a useful tool to distinguish differences in protein abundance of the surfaceome, including the expression of virulence factors.
Collapse
Affiliation(s)
| | - Leonarda Achá Alarcón
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (L.A.A.); (B.P.-I.); (S.S.); (E.R.B.M.)
| | - Beatriz Piñeiro-Iglesias
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (L.A.A.); (B.P.-I.); (S.S.); (E.R.B.M.)
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
| | - Gunnar Jacobsson
- Department of Infectious Diseases, Skaraborg Hospital, 54185 Skövde, Sweden;
- Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, 40530 Gothenburg, Sweden
| | - Susann Skovbjerg
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (L.A.A.); (B.P.-I.); (S.S.); (E.R.B.M.)
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, 40530 Gothenburg, Sweden
| | - Edward R. B. Moore
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (L.A.A.); (B.P.-I.); (S.S.); (E.R.B.M.)
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, 40530 Gothenburg, Sweden
- Culture Collection of the University of Gothenburg (CCUG), Sahlgrenska Academy, 41390 Gothenburg, Sweden
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden; (P.K.K.); (T.J.)
- Department of Rheumatology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Roger Karlsson
- Nanoxis Consulting AB, 40016 Gothenburg, Sweden;
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 40530 Gothenburg, Sweden; (L.A.A.); (B.P.-I.); (S.S.); (E.R.B.M.)
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Region Västra Götaland, 41345 Gothenburg, Sweden
- Centre for Antibiotic Resistance Research (CARe), University of Gothenburg, 40530 Gothenburg, Sweden
| |
Collapse
|
9
|
Hou WT, Shen CR, Peng J, Jiang LW, Guo SY, Qiu XR, Zhang Y, Shen H, Jiang YY, An MM. Mechanism of Action for an All-in-One Monoclonal Antibody Against Staphylococcus aureus Infection. J Infect Dis 2023; 228:1789-1799. [PMID: 37335928 DOI: 10.1093/infdis/jiad215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/22/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023] Open
Abstract
Staphylococcus aureus is a major human pathogen associated with high mortality rates. The extensive use of antibiotics is associated with the rise of drug resistance, and exotoxins are not targeted by antibiotics. Therefore, monoclonal antibody (mAb) therapy has emerged as a promising solution to solve the clinical problems caused by refractory S aureus. Recent research suggests that the synergistic effects of several cytotoxins, including bicomponent toxins, are critical to the pathogenesis of S aureus. By comparing the amino acid sequences, researchers found that α-toxin and bicomponent toxins have high homology. Therefore, we aimed to screen an antibody, designated an all-in-one mAb, that could neutralize α-toxin and bicomponent toxins through hybridoma fusion. We found that this mAb has a significant pharmacodynamic effect within in vivo mouse models and in vitro experiments.
Collapse
Affiliation(s)
- Wei-Tong Hou
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen-Rui Shen
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ji Peng
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li-Wen Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shi-Yu Guo
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi-Ran Qiu
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Zhang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Shen
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuan-Ying Jiang
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mao-Mao An
- Department of Pharmacology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Wang W, Gu Y, Ou Y, Zhou J, Liu B, Zuo H, Du Y, Wang Y, Tang T, Zou Q, Zuo Q. Human monoclonal antibodies against Staphylococcus aureus A protein identified by high-throughput single-cell sequencing of phase I clinical volunteers' B cells. Clin Immunol 2023; 257:109843. [PMID: 37981106 DOI: 10.1016/j.clim.2023.109843] [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: 09/19/2023] [Revised: 11/05/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023]
Abstract
Methicillin-resistant Staphylococcus aureus, poses a significant threat through infections in both community and hospital settings. To address this challenge, we conducted a phase I clinical trial study involving a recombinant Staphylococcus aureus vaccine. Utilizing peripheral blood lymphocytes from 64 subjects, we isolated antigen-specific memory B cells for subsequent single-cell sequencing. Among the 676 identified antigen-binding IgG1+ clones, we selected the top 10 antibody strains for construction within expression vectors. Successful expression and purification of these monoclonal antibodies led to the discovery of a highly expressed human antibody, designated as IgG-6. This antibody specifically targets the pentameric form of the Staphylococcus aureus protein A (SpA5). In vivo assessments revealed that IgG-6 provided prophylactic protection against MRSA252 infection. This study underscores the potential of human antibodies as an innovative strategy against Staphylococcus aureus infections, offering a promising avenue for further research and clinical development.
Collapse
Affiliation(s)
- WenHao Wang
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - YaRu Gu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400038, PR China
| | - YangXue Ou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - JinRui Zhou
- College of Medicine, Southwest Jiaotong University, Chengdu 610083, PR China
| | - BiXia Liu
- College of Medicine, Southwest Jiaotong University, Chengdu 610083, PR China
| | - HouYi Zuo
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - YeXiang Du
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China
| | - Ying Wang
- 953th Hospital, Shigatse Branch, Xinqiao Hospital, Army Medical University, 857000 Shigatse, China
| | - TengQian Tang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing 400038, PR China.
| | - QuanMing Zou
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| | - QianFei Zuo
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing 400038, PR China.
| |
Collapse
|
11
|
He J, Yang X, Yang K, Xu H, Chen C, Wang J, Zeng J. TPST2-mediated receptor tyrosine sulfation enhances leukocidin cytotoxicity and S. aureus infection. Front Immunol 2023; 14:1242330. [PMID: 37671153 PMCID: PMC10476081 DOI: 10.3389/fimmu.2023.1242330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 07/31/2023] [Indexed: 09/07/2023] Open
Abstract
Background An essential fact underlying the severity of Staphylococcus aureus (S. aureus) infection is the bicomponent leukocidins released by the pathogen to target and lyse host phagocytes through specific binding cell membrane receptors. However, little is known about the impact of post-transcriptional modification of receptors on the leukocidin binding. Method In this study, we used small interfering RNA library (Horizon/Dharmacon) to screen potential genes that affect leukocidin binding on receptors. The cell permeability was investigated through flow cytometry measuring the internalization of 4',6-diamidino-2-phenylindole. Expression of C5a anaphylatoxin chemotactic receptor 1 (C5aR1), sulfated C5aR1 in, and binding of 6x-His-tagged Hemolysin C (HlgC) and Panton-Valentine leukocidin (PVL) slow-component to THP-1 cell lines was detected and analyzed via flow cytometry. Bacterial burden and Survival analysis experiment was conducted in WT and myeloid TPST-cko C57BL/6N mice. Results After short hairpin RNA (shRNA) knockdown of TPST2 gene in THP-1, HL-60, and RAW264.7, the cytotoxicity of HlgAB, HlgCB, and Panton-Valentine leukocidin on THP-1 or HL-60 cells was decreased significantly, and the cytotoxicity of HlgAB on RAW264.7 cells was also decreased significantly. Knockdown of TPST2 did not affect the C5aR1 expression but downregulated cell surface C5aR1 tyrosine sulfation on THP-1. In addition, we found that the binding of HlgC and LukS-PV on cell surface receptor C5aR1 was impaired in C5aR1+TPST2- and C5aR1-TPST2- cells. Phagocyte knockout of TPST2 protects mice from S. aureus infection and improves the survival of mice infected with S. aureus. Conclusion These results indicate that phagocyte TPST2 mediates the bicomponent leukocidin cytotoxicity by promoting cell membrane receptor sulfation modification that facilitates its binding to leukocidin S component.
Collapse
Affiliation(s)
- Jie He
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xianggui Yang
- Department of Laboratory Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Kai Yang
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | | | | | | | - Jun Zeng
- Division of Pulmonary and Critical Care Medicine, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| |
Collapse
|
12
|
Bernardino PN, Bhattacharya M, Chen X, Jenkins J, Missiakas D, Thammavongsa V. A humanized monoclonal antibody targeting protein a promotes opsonophagocytosis of Staphylococcus aureus in human umbilical cord blood. Vaccine 2023; 41:5079-5084. [PMID: 37455161 PMCID: PMC10412981 DOI: 10.1016/j.vaccine.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/03/2023] [Accepted: 07/09/2023] [Indexed: 07/18/2023]
Abstract
Low and very-low-birth-weight (V/LBW) neonates are highly susceptible to bacterial sepsis and meningitis. Bacterial infections caused by Staphylococcus aureus can be particularly dangerous for neonates and can result in high mortality and long-term disabilities.Antibody-based strategies have been attempted to protect V/LBW neonates against staphylococcal disease. However, these efforts have so far been unsuccessful. Failures were attributed to the immaturity of the neonatal immune system but did not account for the anti-opsonic activity of Staphylococcal protein A (SpA). Here we show that monoclonal antibody 3F6, which blocks SpA activity, promotes complement-dependent cell-mediated phagocytosis of S. aureus in human umbilical cord blood. A substitution in the crystallizable fragment (Fc) region of 3F6 that enhances recruitment of complement component C1q further increases the phagocytic activity of cord blood. Our data demonstrate that the neonatal immune system possesses bactericidal activity that can be harnessed by antibodies that circumvent a key innate immune strategy of S. aureus.
Collapse
Affiliation(s)
- Paola Nol Bernardino
- The University of Chicago, Department of Microbiology, Howard Taylor Ricketts Laboratory, Lemont, IL 60439, USA
| | - Mohini Bhattacharya
- The University of Chicago, Department of Microbiology, Howard Taylor Ricketts Laboratory, Lemont, IL 60439, USA
| | - Xinhai Chen
- The University of Chicago, Department of Microbiology, Howard Taylor Ricketts Laboratory, Lemont, IL 60439, USA
| | - Julia Jenkins
- The University of Chicago, Department of Microbiology, Howard Taylor Ricketts Laboratory, Lemont, IL 60439, USA
| | - Dominique Missiakas
- The University of Chicago, Department of Microbiology, Howard Taylor Ricketts Laboratory, Lemont, IL 60439, USA
| | | |
Collapse
|
13
|
Kumar G, Kiran Tudu A. Tackling multidrug-resistant Staphylococcus aureus by natural products and their analogues acting as NorA efflux pump inhibitors. Bioorg Med Chem 2023; 80:117187. [PMID: 36731248 DOI: 10.1016/j.bmc.2023.117187] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/29/2023]
Abstract
Staphylococcus aureus (S. aureus) is a pathogen responsible for various community and hospital-acquired infections with life-threatening complications like bacteraemia, endocarditis, meningitis, liver abscess, and spinal cord epidural abscess. Antibiotics have been used to treat microbial infections since the introduction of penicillin in 1940. In recent decades, the abuse and misuse of antibiotics in humans, animals, plants, and fungi, including the treatment of non-microbial diseases, have led to the rapid emergence of multidrug-resistant pathogens with increased virulence. Bacteria have developed several complementary mechanisms to avoid the effects of antibiotics. These mechanisms include chemical transformations and enzymatic inactivation of antibiotics, modification of antibiotics' target site, and reduction of intracellular antibiotics concentration by changes in membrane permeability or by the overexpression of efflux pumps (EPs). The strategy to check antibiotic resistance includes synthesis of the antibiotic analogues, or antibiotics are given in combination with the adjuvant. The inhibitors of multidrug EPs are considered promising alternative therapeutic options with the potential to revive the effects of antibiotics and reduce bacterial virulence. Natural products played a vital role in drug discovery and significantly contributed to the area of infectious diseases. Also, natural products provide lead compounds that sometimes need modification based on structural and biological properties to meet the drug criteria. This review discusses natural products and their derived compounds as NorA efflux pump inhibitors (EPIs).
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India.
| | - Asha Kiran Tudu
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Balanagar, Telangana 500037, India
| |
Collapse
|
14
|
Liu X, Wang Y, Zou J, Wang H, Li X, Huang Y, Hu E, Guan Z, Quan D, Liu J, Zhang W. Quasi-opsonin conjugated lipase-sensitive micelles activate macrophages against facultative intracellular bacterial infection. J Mater Chem B 2023; 11:865-878. [PMID: 36594907 DOI: 10.1039/d2tb01802k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance caused by facultative intracellular bacteria such as Salmonella typhimurium (S. typhimurium) is still a tough challenge. Bacteria phagocytosed by macrophages have evolved a variety of mechanisms to defend against host attack, and the poor entry of antibiotics into infected macrophages is conducive to the survival of intracellular bacteria. In this report, we prepared a quasi-opsonized chloramphenicol (Chl)-loaded micellar system (B-mLBP-M/Chl) assembled by a bacterial lipase-sensitive polymer with a conjugate of lipopolysaccharide-binding protein (LBP) analog and biotin (B) as a ligand, which could eliminate drug-resistant S. typhimurium with quasi-opsonization via 3 steps: (i) target and release antibiotics on bacteria lipase, (ii) opsonize S. typhimurium to be digested by the macrophage, and (iii) activate the macrophage for fighting. The B-mLBP-M/Chl could target bacterial LPS through mLBP by simulating the N-terminal sequence of native LBP, exhibiting a high ability to target the localized infection site in mice. It could also activate the phagocytosis of macrophages via coupled biotin, cooperating with antibiotics and effectively improving the survival of mice with little pathological damage to tissues. Moreover, compared with native opsonin, B-mLBP does not cause an excessive inflammatory response and could recover homeostasis after exerting the quasi-opsonization by regulating the levels of pro-inflammatory cytokines and anti-inflammatory cytokines. With a universal target site for Gram-negative bacteria and macrophage activation, this B-mLBP-M/Chl could be applied to other bacterial infections in the future. In particular, this analog may also serve as a useful template to design safe artificial opsonin, which could be a ligand for drug delivery systems or prodrugs.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Yajie Wang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Jiahui Zou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Hui Wang
- Department of Microbiology, China Pharmaceutical University, Jiangsu 210009, P. R. China
| | - Xuechun Li
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Ying Huang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Enshi Hu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Zhiyu Guan
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, P. R. China
| | - Danyi Quan
- Institute of Advanced Drug Delivery Technology, Jiangsu 210032, P. R. China.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu 210009, P. R. China.
| |
Collapse
|
15
|
Grace PS, Gunn BM, Lu LL. Engineering the supernatural: monoclonal antibodies for challenging infectious diseases. Curr Opin Biotechnol 2022; 78:102818. [PMID: 36242952 PMCID: PMC9612313 DOI: 10.1016/j.copbio.2022.102818] [Citation(s) in RCA: 6] [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: 07/22/2022] [Revised: 08/31/2022] [Accepted: 09/04/2022] [Indexed: 12/14/2022]
Abstract
The COVID-19 pandemic demonstrated that monoclonal antibodies can be deployed faster than antimicrobials and vaccines. However, the majority of mAbs treat cancer and autoimmune diseases, whereas a minority treat infection. This is in part because targeting a single antigen by the antibody Fab domain is insufficient to stop the dynamic microbial life cycle. Thus, finding the 'right' antigens remains the focus of intense investigations. Equally important is the antibody-Fc domain that has the capacity to induce immune responses that enhance neutralization, and limit pathology and transmission. While Fc-effector functions have been less deeply studied, conceptual and technical advances reveal previously underappreciated antibody potential to combat diseases from microbes difficult to address with current diagnostics, therapeutics, and vaccines, including S. aureus, P. aeruginosa, P. falciparum, and M. tuberculosis. What is learned about engineering antibodies for these challenging organisms will enhance our approach to new and emerging infectious diseases.
Collapse
Affiliation(s)
- Patricia S Grace
- Harvard T.H. Chan School of Public Health, Boston, MA, United States; Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States
| | - Bronwyn M Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, WA, United States
| | - Lenette L Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, United States; Department of Immunology, UT Southwestern Medical Center, Dallas, TX, United States; Parkland Health & Hospital System, United States.
| |
Collapse
|
16
|
Piri-Gavgani S, Ghanei M, Fateh A, Siadat SD, Nematollahi L, Rahimi-Jamnani F. Identification of two neutralizing human single-chain variable fragment antibodies targeting Staphylococcus aureus alpha-hemolysin. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1207-1214. [PMID: 36311199 PMCID: PMC9588317 DOI: 10.22038/ijbms.2022.64103.14253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/06/2022] [Indexed: 01/24/2023]
Abstract
OBJECTIVES The inability of the host immune system to defeat Staphylococcus aureus is due to various secreted virulent factors such as leukocidins, superantigens, and hemolysins, which interrupt the function of immune components. Alpha-hemolysin is one of the most studied cytolysins due to its pronounced effect on developing staphylococcal infections. Alpha-hemolysin-neutralizing antibodies are among the best candidates for blocking the toxin activity and preventing S. aureus pathogenesis. MATERIALS AND METHODS A human single-chain variable fragment (scFv) phage display library was biopanned against alpha-hemolysin. The selected phage clones were assessed based on their binding ability to alpha-hemolysin. The binding specificity and affinity of two scFvs (designated SP192 and SP220) to alpha-hemolysin were determined by enzyme-linked immunosorbent assay. Furthermore, the neutralizing activity of SP192 and SP220 was examined by concurrent incubation of rabbit red blood cells (RBCs) with alpha-hemolysin and scFvs. RESULTS SP192 and SP220 showed significant binding to alpha-hemolysin compared with the control proteins, including bovine serum albumin, human adiponectin, and toxic shock syndrome toxin-1. Besides, both scFvs showed high-affinity binding to alpha-hemolysin in the nanomolar range (Kaff: 0.9 and 0.7 nM-1, respectively), leading to marked inhibition of alpha-hemolysin-mediated lysis of rabbit RBCs (73% and 84% inhibition; respectively). CONCLUSION SP192 and SP220 scFvs can potentially be used as alpha-hemolysin-neutralizing agents in conjunction with conventional antibiotics to combat S. aureus infections.
Collapse
Affiliation(s)
- Somayeh Piri-Gavgani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Nematollahi
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rahimi-Jamnani
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
Visperas A, Santana D, Klika AK, Higuera‐Rueda CA, Piuzzi NS. Current treatments for biofilm-associated periprosthetic joint infection and new potential strategies. J Orthop Res 2022; 40:1477-1491. [PMID: 35437846 PMCID: PMC9322555 DOI: 10.1002/jor.25345] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 04/11/2022] [Accepted: 04/16/2022] [Indexed: 02/04/2023]
Abstract
Periprosthetic joint infection (PJI) remains a devastating complication after total joint arthroplasty. Bacteria involved in these infections are notorious for adhering to foreign implanted surfaces and generating a biofilm matrix. These biofilms protect the bacteria from antibiotic treatment and the immune system making eradication difficult. Current treatment strategies including debridement, antibiotics, and implant retention, and one- and two-stage revisions still present a relatively high overall failure rate. One of the main shortcomings that has been associated with this high failure rate is the lack of a robust approach to treating bacterial biofilm. Therefore, in this review, we will highlight new strategies that have the potential to combat PJI by targeting biofilm integrity, therefore giving antibiotics and the immune system access to the internal network of the biofilm structure. This combination antibiofilm/antibiotic therapy may be a new strategy for PJI treatment while promoting implant retention.
Collapse
Affiliation(s)
- Anabelle Visperas
- Department of Orthopaedic SurgeryCleveland Clinic FoundationClevelandOhioUSA
| | - Daniel Santana
- Department of Orthopaedic SurgeryCleveland Clinic FoundationClevelandOhioUSA
- Cleveland Clinic Lerner College of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Alison K. Klika
- Department of Orthopaedic SurgeryCleveland Clinic FoundationClevelandOhioUSA
| | | | - Nicolas S. Piuzzi
- Department of Orthopaedic SurgeryCleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
18
|
Kember M, Grandy S, Raudonis R, Cheng Z. Non-Canonical Host Intracellular Niche Links to New Antimicrobial Resistance Mechanism. Pathogens 2022; 11:pathogens11020220. [PMID: 35215166 PMCID: PMC8876822 DOI: 10.3390/pathogens11020220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/04/2022] Open
Abstract
Globally, infectious diseases are one of the leading causes of death among people of all ages. The development of antimicrobials to treat infectious diseases has been one of the most significant advances in medical history. Alarmingly, antimicrobial resistance is a widespread phenomenon that will, without intervention, make currently treatable infections once again deadly. In an era of widespread antimicrobial resistance, there is a constant and pressing need to develop new antibacterial drugs. Unraveling the underlying resistance mechanisms is critical to fight this crisis. In this review, we summarize some emerging evidence of the non-canonical intracellular life cycle of two priority antimicrobial-resistant bacterial pathogens: Pseudomonas aeruginosa and Staphylococcus aureus. The bacterial factors that modulate this unique intracellular niche and its implications in contributing to resistance are discussed. We then briefly discuss some recent research that focused on the promises of boosting host immunity as a combination therapy with antimicrobials to eradicate these two particular pathogens. Finally, we summarize the importance of various strategies, including surveillance and vaccines, in mitigating the impacts of antimicrobial resistance in general.
Collapse
|
19
|
Liu X, Wu Y, Mao C, Shen J, Zhu K. Host-acting antibacterial compounds combat cytosolic bacteria. Trends Microbiol 2022; 30:761-777. [DOI: 10.1016/j.tim.2022.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/22/2021] [Accepted: 01/12/2022] [Indexed: 01/25/2023]
|
20
|
Iskandar K, Murugaiyan J, Hammoudi Halat D, Hage SE, Chibabhai V, Adukkadukkam S, Roques C, Molinier L, Salameh P, Van Dongen M. Antibiotic Discovery and Resistance: The Chase and the Race. Antibiotics (Basel) 2022; 11:182. [PMID: 35203785 PMCID: PMC8868473 DOI: 10.3390/antibiotics11020182] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/23/2022] [Accepted: 01/26/2022] [Indexed: 12/14/2022] Open
Abstract
The history of antimicrobial resistance (AMR) evolution and the diversity of the environmental resistome indicate that AMR is an ancient natural phenomenon. Acquired resistance is a public health concern influenced by the anthropogenic use of antibiotics, leading to the selection of resistant genes. Data show that AMR is spreading globally at different rates, outpacing all efforts to mitigate this crisis. The search for new antibiotic classes is one of the key strategies in the fight against AMR. Since the 1980s, newly marketed antibiotics were either modifications or improvements of known molecules. The World Health Organization (WHO) describes the current pipeline as bleak, and warns about the scarcity of new leads. A quantitative and qualitative analysis of the pre-clinical and clinical pipeline indicates that few antibiotics may reach the market in a few years, predominantly not those that fit the innovative requirements to tackle the challenging spread of AMR. Diversity and innovation are the mainstays to cope with the rapid evolution of AMR. The discovery and development of antibiotics must address resistance to old and novel antibiotics. Here, we review the history and challenges of antibiotics discovery and describe different innovative new leads mechanisms expected to replenish the pipeline, while maintaining a promising possibility to shift the chase and the race between the spread of AMR, preserving antibiotic effectiveness, and meeting innovative leads requirements.
Collapse
Affiliation(s)
- Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon;
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | - Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University–AP, Amaravati 522502, India; (J.M.); (S.A.)
| | - Dalal Hammoudi Halat
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese International University, Bekaa Campus, Beirut 1103, Lebanon
| | - Said El Hage
- Faculty of Medicine, Lebanese University, Beirut 6573, Lebanon;
| | - Vindana Chibabhai
- Division of Clinical Microbiology and Infectious Diseases, School of Pathology, University of the Witwatersrand, Johannesburg 2193, South Africa;
- Microbiology Laboratory, National Health Laboratory Service, Charlotte Maxeke Johannesburg Academic Hospital, Johannesburg 2193, South Africa
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University–AP, Amaravati 522502, India; (J.M.); (S.A.)
| | - Christine Roques
- Laboratoire de Génie Chimique, Department of Bioprocédés et Systèmes Microbiens, Université Paul Sabtier, Toulouse III, UMR 5503, 31330 Toulouse, France;
| | - Laurent Molinier
- Department of Medical Information, Centre Hospitalier Universitaire, INSERM, UMR 1295, Université Paul Sabatier Toulouse III, 31000 Toulouse, France;
| | - Pascale Salameh
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon;
- Faculty of Medicine, Lebanese University, Beirut 6573, Lebanon;
- Department of Primary Care and Population Health, University of Nicosia Medical School, Nicosia 2408, Cyprus
| | | |
Collapse
|
21
|
Engineered human antibodies for the opsonization and killing of Staphylococcus aureus. Proc Natl Acad Sci U S A 2022; 119:2114478119. [PMID: 35058363 PMCID: PMC8795526 DOI: 10.1073/pnas.2114478119] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 01/13/2023] Open
Abstract
Staphylococcus aureus invariably acquires resistance mechanisms against new antibiotics. The persistent colonization with S. aureus is the key risk factor for invasive disease and a driver for the evolution of antibiotic resistant isolates. Anti-S. aureus antibodies that could promote decolonization, prevent infection, or treat disease would alleviate the selection for drug resistance. The successful development of such antibodies is complicated by Staphylococcal protein A (SpA) in the envelope of S. aureus. SpA captures immunoglobulins via their constant region, preventing antibodies from initiating anti-staphylococcal activities. Here, we demonstrate that therapeutic anti-S. aureus antibodies can be engineered to avoid sequestration by SpA. Such antibodies display extended half-lives and improve bacterial uptake and killing by immune cells. Gram-positive organisms with their thick envelope cannot be lysed by complement alone. Nonetheless, antibody-binding on the surface can recruit complement and mark these invaders for uptake and killing by phagocytes, a process known as opsonophagocytosis. The crystallizable fragment of immunoglobulins (Fcγ) is key for complement recruitment. The cell surface of S. aureus is coated with Staphylococcal protein A (SpA). SpA captures the Fcγ domain of IgG and interferes with opsonization by anti-S. aureus antibodies. In principle, the Fcγ domain of therapeutic antibodies could be engineered to avoid the inhibitory activity of SpA. However, the SpA-binding site on Fcγ overlaps with that of the neonatal Fc receptor (FcRn), an interaction that is critical for prolonging the half-life of serum IgG. This evolutionary adaptation poses a challenge for the exploration of Fcγ mutants that can both weaken SpA–IgG interactions and retain stability. Here, we use both wild-type and transgenic human FcRn mice to identify antibodies with enhanced half-life and increased opsonophagocytic killing in models of S. aureus infection and demonstrate that antibody-based immunotherapy can be improved by modifying Fcγ. Our experiments also show that by competing for FcRn-binding, staphylococci effectively reduce the half-life of antibodies during infection. These observations may have profound impact in treating cancer, autoimmune, and asthma patients colonized or infected with S. aureus and undergoing monoclonal antibody treatment.
Collapse
|
22
|
de Vor L, van Dijk B, van Kessel K, Kavanaugh JS, de Haas C, Aerts PC, Viveen MC, Boel EC, Fluit AC, Kwiecinski JM, Krijger GC, Ramakers RM, Beekman FJ, Dadachova E, Lam MGEH, Vogely HC, van der Wal BCH, van Strijp JAG, Horswill AR, Weinans H, Rooijakkers SHM. Human monoclonal antibodies against Staphylococcus aureus surface antigens recognize in vitro and in vivo biofilm. eLife 2022; 11:e67301. [PMID: 34989676 PMCID: PMC8751199 DOI: 10.7554/elife.67301] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 12/06/2021] [Indexed: 12/22/2022] Open
Abstract
Implant-associated Staphylococcus aureus infections are difficult to treat because of biofilm formation. Bacteria in a biofilm are often insensitive to antibiotics and host immunity. Monoclonal antibodies (mAbs) could provide an alternative approach to improve the diagnosis and potential treatment of biofilm-related infections. Here, we show that mAbs targeting common surface components of S. aureus can recognize clinically relevant biofilm types. The mAbs were also shown to bind a collection of clinical isolates derived from different biofilm-associated infections (endocarditis, prosthetic joint, catheter). We identify two groups of antibodies: one group that uniquely binds S. aureus in biofilm state and one that recognizes S. aureus in both biofilm and planktonic state. Furthermore, we show that a mAb recognizing wall teichoic acid (clone 4497) specifically localizes to a subcutaneously implanted pre-colonized catheter in mice. In conclusion, we demonstrate the capacity of several human mAbs to detect S. aureus biofilms in vitro and in vivo.
Collapse
Affiliation(s)
- Lisanne de Vor
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Bruce van Dijk
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Kok van Kessel
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Carla de Haas
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Piet C Aerts
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Marco C Viveen
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Edwin C Boel
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Ad C Fluit
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Jakub M Kwiecinski
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
| | - Gerard C Krijger
- Department of Radiology and Nuclear Medicine, University Medical Centre UtrechtUtrechtNetherlands
| | - Ruud M Ramakers
- MILabs B.VUtrechtNetherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical CenterUtrechtNetherlands
- Department of Radiation Science and Technology, Delft University of TechnologyDelftNetherlands
| | - Freek J Beekman
- MILabs B.VUtrechtNetherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical CenterUtrechtNetherlands
- Department of Radiation Science and Technology, Delft University of TechnologyDelftNetherlands
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of SaskatchewanSaskatoonCanada
| | - Marnix GEH Lam
- Department of Radiology and Nuclear Medicine, University Medical Centre UtrechtUtrechtNetherlands
| | - H Charles Vogely
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Bart CH van der Wal
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
| | - Jos AG van Strijp
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of MedicineAuroraUnited States
- Department of Veterans Affairs, Eastern Colorado Health Care SystemDenverUnited States
| | - Harrie Weinans
- Department of Orthopedics, University Medical Centre UtrechtUtrechtNetherlands
- Department of Biomechanical engineering, TU DelftDelftNetherlands
| | - Suzan HM Rooijakkers
- Department of Medical Microbiology, University Medical Centre UtrechtUtrechtNetherlands
| |
Collapse
|
23
|
Soleimanpour Moghadam N, Azadmehr A, Hezarkhani A. Improving the 6-Aminopenicillanic acid release process using vermiculite-alginate biocomposite bead on drug delivery system. Drug Dev Ind Pharm 2021; 47:1489-1501. [PMID: 34806923 DOI: 10.1080/03639045.2021.2001492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The present study deals with developing vermiculite (VMT)-alginate (Alg) composites with different cross-linker concentrations (CaCl2) to deliver the controlled 6-aminopenicillin acid (6-APA). The Characterization of synthesized composites was conducted by Fourier transform infrared spectroscopy (FTIR) and X-ray diffraction (XRD) analyses. Optimization attempts were explored via the response surface method (RSM) to best predict the actual amount of compound. The adsorption capacity of 6-APA onto this adsorbent was found to be 208.33 mg/g, which was higher than that for other clays. The equilibrium and Kinetic studies (chemical reaction and diffusion-based models) indicated that drug absorption on VMT-Alg is homogeneous with chemical interaction. An increase in cross-linker (CaCl2) concentration leads to improvement in the drug encapsulation efficiency while having no significant effect on loading efficiency. The in-vitro release of the pure drug shows a rapid burst release followed by 100% cumulative release within 6 h. Whereas, the synthesized drug with Alg substantially showed less release of 43% after 8 h. Release experiments revealed that the presence of the CaCl2 delayed the release of the 6-APA less than 35% after 12 h. The kinetic release of 6-APA is followed by the Korsmeyer-Peppas model based on Fick's law mechanism due to the kinetic exponent (n < 0.5). All studied composites antibacterial activity after 24 h exposure against E. Coli and S. aureus. The antibacterial activities of composites were evaluated by the halo of no growth. The results showed that the VMT-Alg-6APA composite had strong activity against Gram-positive and Gram-negative bacteria.
Collapse
Affiliation(s)
| | - Amirreza Azadmehr
- Department of Mining & Metallurgical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Ardeshir Hezarkhani
- Department of Mining & Metallurgical Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
24
|
Soltanmohammadi B, Piri‐Gavgani S, Basardeh E, Ghanei M, Azizi M, Khaksar Z, Sharifzadeh Z, Badmasti F, Soezi M, Fateh A, Azimi P, Siadat SD, Shooraj F, Bouzari S, Omrani MD, Rahimi‐Jamnani F. Bactericidal fully human single-chain fragment variable antibodies protect mice against methicillin-resistant Staphylococcus aureus bacteraemia. Clin Transl Immunology 2021; 10:e1302. [PMID: 34221401 PMCID: PMC8240403 DOI: 10.1002/cti2.1302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/01/2021] [Accepted: 05/30/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES The increasing prevalence of antibiotic-resistant Staphylococcus aureus, besides the inadequate numbers of effective antibiotics, emphasises the need to find new therapeutic agents against this lethal pathogen. METHODS In this study, to obtain antibody fragments against S. aureus, a human single-chain fragment variable (scFv) library was enriched against living methicillin-resistant S. aureus (MRSA) cells, grown in three different conditions, that is human peripheral blood mononuclear cells with plasma, whole blood and biofilm. The antibacterial activity of scFvs was evaluated by the growth inhibition assay in vitro. Furthermore, the therapeutic efficacy of anti-S. aureus scFvs was appraised in a mouse model of bacteraemia. RESULTS Three scFv antibodies, that is MEH63, MEH158 and MEH183, with unique sequences, were found, which exhibited significant binding to S. aureus and reduced the viability of S. aureus in in vitro inhibition assays. Based on the results, MEH63, MEH158 and MEH183, in addition to their combination, could prolong the survival rate, reduce the bacterial burden in the blood and prevent inflammation and tissue destruction in the kidneys and spleen of mice with MRSA bacteraemia compared with the vehicle group (treated with normal saline). CONCLUSION The combination therapy with anti-S. aureus scFvs and conventional antibiotics might shed light on the treatment of patients with S. aureus infections.
Collapse
Affiliation(s)
- Behnoush Soltanmohammadi
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Somayeh Piri‐Gavgani
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Eilnaz Basardeh
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Mostafa Ghanei
- Chemical Injuries Research CenterSystems Biology and Poisoning InstituteBaqiyatallah University of Medical SciencesTehranIran
| | - Masoumeh Azizi
- Molecular Medicine Department, Biotechnology Research CenterPasteur Institute of IranTehranIran
| | - Zabihollah Khaksar
- Department of Basic SciencesSchool of Veterinary MedicineShiraz UniversityShirazIran
| | | | - Farzad Badmasti
- Department of BacteriologyPasteur Institute of IranTehranIran
| | - Mahdieh Soezi
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Parisa Azimi
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Fahimeh Shooraj
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| | - Saeid Bouzari
- Molecular Biology DepartmentPasteur Institute of IranTehranIran
| | - Mir Davood Omrani
- Department of Medical GeneticsSchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fatemeh Rahimi‐Jamnani
- Department of Mycobacteriology and Pulmonary ResearchPasteur Institute of IranTehranIran
- Microbiology Research CenterPasteur Institute of IranTehranIran
| |
Collapse
|
25
|
Zwarthoff SA, Widmer K, Kuipers A, Strasser J, Ruyken M, Aerts PC, de Haas CJC, Ugurlar D, den Boer MA, Vidarsson G, van Strijp JAG, Gros P, Parren PWHI, van Kessel KPM, Preiner J, Beurskens FJ, Schuurman J, Ricklin D, Rooijakkers SHM. C1q binding to surface-bound IgG is stabilized by C1r 2s 2 proteases. Proc Natl Acad Sci U S A 2021; 118:e2102787118. [PMID: 34155115 PMCID: PMC8256010 DOI: 10.1073/pnas.2102787118] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Complement is an important effector mechanism for antibody-mediated clearance of infections and tumor cells. Upon binding to target cells, the antibody's constant (Fc) domain recruits complement component C1 to initiate a proteolytic cascade that generates lytic pores and stimulates phagocytosis. The C1 complex (C1qr2s2) consists of the large recognition protein C1q and a heterotetramer of proteases C1r and C1s (C1r2s2). While interactions between C1 and IgG-Fc are believed to be mediated by the globular heads of C1q, we here find that C1r2s2 proteases affect the capacity of C1q to form an avid complex with surface-bound IgG molecules (on various 2,4-dinitrophenol [DNP]-coated surfaces and pathogenic Staphylococcus aureus). The extent to which C1r2s2 contributes to C1q-IgG stability strongly differs between human IgG subclasses. Using antibody engineering of monoclonal IgG, we reveal that hexamer-enhancing mutations improve C1q-IgG stability, both in the absence and presence of C1r2s2 In addition, hexamer-enhanced IgGs targeting S. aureus mediate improved complement-dependent phagocytosis by human neutrophils. Altogether, these molecular insights into complement binding to surface-bound IgGs could be important for optimal design of antibody therapies.
Collapse
Affiliation(s)
- Seline A Zwarthoff
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Kevin Widmer
- Pharmaceutical Sciences, University of Basel, 4001 Basel, Switzerland
| | - Annemarie Kuipers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Jürgen Strasser
- Nano Structuring and Bio-Analytics Group, TIMed Center, of Applied Sciences Upper Austria, 4020 Linz, Austria
| | - Maartje Ruyken
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Piet C Aerts
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Carla J C de Haas
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Deniz Ugurlar
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Maurits A den Boer
- Biomolecular Mass Spectrometry & Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Center, 3584 CH, Utrecht, The Netherlands
| | - Gestur Vidarsson
- Experimental Immunohematology, Sanquin Research, 1066 CX Amsterdam, The Netherlands
| | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Piet Gros
- Structural Biochemistry, Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Paul W H I Parren
- Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Lava Therapeutics, 3584 CM Utrecht, The Netherlands
| | - Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Johannes Preiner
- Nano Structuring and Bio-Analytics Group, TIMed Center, of Applied Sciences Upper Austria, 4020 Linz, Austria
| | | | | | - Daniel Ricklin
- Pharmaceutical Sciences, University of Basel, 4001 Basel, Switzerland
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| |
Collapse
|
26
|
Cruz AR, Boer MAD, Strasser J, Zwarthoff SA, Beurskens FJ, de Haas CJC, Aerts PC, Wang G, de Jong RN, Bagnoli F, van Strijp JAG, van Kessel KPM, Schuurman J, Preiner J, Heck AJR, Rooijakkers SHM. Staphylococcal protein A inhibits complement activation by interfering with IgG hexamer formation. Proc Natl Acad Sci U S A 2021; 118:e2016772118. [PMID: 33563762 PMCID: PMC7896290 DOI: 10.1073/pnas.2016772118] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Immunoglobulin (Ig) G molecules are essential players in the human immune response against bacterial infections. An important effector of IgG-dependent immunity is the induction of complement activation, a reaction that triggers a variety of responses that help kill bacteria. Antibody-dependent complement activation is promoted by the organization of target-bound IgGs into hexamers that are held together via noncovalent Fc-Fc interactions. Here we show that staphylococcal protein A (SpA), an important virulence factor and vaccine candidate of Staphylococcus aureus, effectively blocks IgG hexamerization and subsequent complement activation. Using native mass spectrometry and high-speed atomic force microscopy, we demonstrate that SpA blocks IgG hexamerization through competitive binding to the Fc-Fc interaction interface on IgG monomers. In concordance, we show that SpA interferes with the formation of (IgG)6:C1q complexes and prevents downstream complement activation on the surface of S. aureus. Finally, we demonstrate that IgG3 antibodies against S. aureus can potently induce complement activation and opsonophagocytic killing even in the presence of SpA. Together, our findings identify SpA as an immune evasion protein that specifically blocks IgG hexamerization.
Collapse
Affiliation(s)
- Ana Rita Cruz
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Maurits A den Boer
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Jürgen Strasser
- Nano Structuring and Bio-Analytics Group, TIMed Center, University of Applied Sciences Upper Austria, 4020 Linz, Austria
| | - Seline A Zwarthoff
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | - Carla J C de Haas
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Piet C Aerts
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Guanbo Wang
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
- School of Chemistry and Materials Science, Nanjing Normal University, 210023 Nanjing, China
| | | | | | - Jos A G van Strijp
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Kok P M van Kessel
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | | | - Johannes Preiner
- Nano Structuring and Bio-Analytics Group, TIMed Center, University of Applied Sciences Upper Austria, 4020 Linz, Austria
| | - Albert J R Heck
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Suzan H M Rooijakkers
- Medical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| |
Collapse
|
27
|
Genotypic and Phenotypic Characterization of Staphylococcus aureus Isolates from the Respiratory Tract in Mechanically-Ventilated Patients. Toxins (Basel) 2021; 13:toxins13020122. [PMID: 33562023 PMCID: PMC7915691 DOI: 10.3390/toxins13020122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is a commensal and frequent colonizer of the upper respiratory tract. When mechanical ventilation disrupts natural defenses, S. aureus is frequently isolated from the lower airways, but distinguishing between colonization and infection is difficult. The objectives of this study were (1) to investigate the bacterial genome sequence in consecutive isolates in order to identify changes related to the pathological adaptation to the lower respiratory tract and (2) to explore the relationship between specific phenotypic and genotypic features with the patient’s study group, persistence of the clinical isolate and clinical outcome. A set of 94 clinical isolates were selected and corresponded to 34 patients that were classified as having pneumonia (10), tracheobronchitis (11) and bronchial colonization (13). Clinical strains were phenotypically characterized by conventional identification and susceptibility testing methods. Isolates underwent whole genome sequencing using Illumina HiSeq4000. Genotypic characterization was performed with an in-house pipeline (BacterialTyper). Genomic variation arising within-host was determined by comparing mapped sequences and de novo assemblies. Virulence factors important in staphylococcal colonization and infection were characterized using previously established functional assays. (1) Toxin production was assessed using a THP-1 cytotoxicity assay, which reports on the gross cytotoxicity of individual isolates. In addition, we investigated the expression of the major virulence factor, alpha-toxin (Hla) by Western blot. (2) Adhesion to the important extracellular matrix molecule, fibronectin, was determined using a standardized microtitre plate assay. Finally, invasion experiments using THP-1 and A539 cell lines and selected clinical strains were also performed. Repeated isolation of S. aureus from endotracheal aspirate usually reflects persistence of the same strain. Within-host variation is detectable in this setting, but it shows no evidence of pathological adaptation related to virulence, resistance or niche adaptations. Cytotoxicity was variable among isolates with 14 strains showing no cytotoxicity, with these latter presenting an unaltered Fn binding capacity. No changes on cytotoxicity were reported when comparing study groups. Fn binding capacity was reported for almost all strains, with the exception of two strains that presented the lowest values. Strains isolated from patients with pneumonia presented a lower capacity of adhesion in comparison to those isolated during tracheobronchitis (p = 0.002). Hla was detected in 71 strains (75.5%), with most of the producer strains in pneumonia and bronchial colonization group (p = 0.06). In our cohort, Hla expression (presence or absence) in sequential isolates was usually preserved (70%) although in seven cases the expression varied over time. No relationship was found between low cytotoxicity and intracellular persistence in invasion experiments. In our study population, persistent S. aureus isolation from airways in ventilated patients does not reflect pathological adaptation. There is an important diversity of sequence types. Cytotoxicity is variable among strains, but no association with study groups was found, whereas isolates from patients with pneumonia had lower adhesion capability. Favorable clinical outcome correlated with increased bacterial adhesion in vitro. Most of the strains isolated from the lower airways were Hla producers and no correlation with an adverse outcome was reported. The identification of microbial factors that contribute to virulence is relevant to optimize patient management during lower respiratory tract infections.
Collapse
|
28
|
Hou X, Zhang W, Meng Y, Zhou W, Peng G, Lei J, Cong S, Song M, Li G, Li X. A New Anti‐Immune Evasion Strategy against Methicillin‐Resistant
Staphylococcus Aureus
(MRSA) Infections: Simulating Complement Immunotherapy Based on Complement‐Mimic Antibiotic Delivery System. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Xucheng Hou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
- Institute of Medicinal Biotechnology Chinese Academy of Medical Science and Peking Union Medical College Beijing 100050 China
| | - Wenxi Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Yansha Meng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Wenkai Zhou
- Institute of Medicinal Biotechnology Chinese Academy of Medical Science and Peking Union Medical College Beijing 100050 China
| | - Guanghua Peng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Jiongxi Lei
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Shuangchen Cong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Maoyuan Song
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| | - Guiling Li
- Institute of Medicinal Biotechnology Chinese Academy of Medical Science and Peking Union Medical College Beijing 100050 China
| | - Xinru Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug System, School of Pharmaceutical Sciences Peking University Health Science Center Beijing 100191 China
| |
Collapse
|
29
|
Zeng H, Zhang J, Song X, Zeng J, Yuan Y, Chen Z, Xu L, Gou Q, Yang F, Zeng N, Zhang Y, Peng L, Zhao L, Zhu J, Liu Y, Luo P, Zou Q, Zhao Z. An Immunodominant Epitope-Specific Monoclonal Antibody Cocktail Improves Survival in a Mouse Model of Staphylococcus aureus Bacteremia. J Infect Dis 2020; 223:1743-1752. [PMID: 32959055 DOI: 10.1093/infdis/jiaa602] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/18/2020] [Indexed: 01/22/2023] Open
Abstract
To date, no vaccine or monoclonal antibody (mAb) against Staphylococcus aureus has been approved for use in humans. Our laboratory has developed a 5-antigen S. aureus vaccine (rFSAV), which is now under efficacy evaluation in a phase 2 clinical trial. In the current study, using overlapping peptides and antiserum from rFSAV-immunized volunteers, we identified 7 B-cell immunodominant epitopes on 4 antigens in rFSAV, including 5 novel epitopes (Hla48-65, IsdB402-419, IsdB432-449, SEB78-95, and MntC7-24). Ten immunodominant epitope mAbs were generated against these epitopes, and all of them exhibited partial protection in a mouse sepsis model. Four robust mAbs were used together as an mAb cocktail to prevent methicillin-resistant S. aureus strain 252 infection. The results showed that the mAb cocktail was efficient in combating S. aureus infection and that its protective efficacy correlated with a reduced bacterial burden and decreased infection pathology, which demonstrates that the mAb cocktail is a promising S. aureus vaccine candidate.
Collapse
Affiliation(s)
- Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Xu Song
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Jiangmin Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Yue Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Zhifu Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Limin Xu
- Chengdu Olymvax Biotechnology Co, Ltd, Chengdu, Sichuan, People's Republic of China
| | - Qiang Gou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Feng Yang
- Chengdu Olymvax Biotechnology Co, Ltd, Chengdu, Sichuan, People's Republic of China
| | - Ni Zeng
- Chengdu Olymvax Biotechnology Co, Ltd, Chengdu, Sichuan, People's Republic of China
| | - Yi Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Liusheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Liqun Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Jiang Zhu
- Department of Pathology, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yuanyuan Liu
- Medical Corps Department, Unit 69016, Chinese People's Liberation Army, Xinjiang, People's Republic of China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, People's Republic of China
| |
Collapse
|
30
|
Pecetta S, Finco O, Seubert A. Quantum leap of monoclonal antibody (mAb) discovery and development in the COVID-19 era. Semin Immunol 2020; 50:101427. [PMID: 33277154 PMCID: PMC7670927 DOI: 10.1016/j.smim.2020.101427] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/16/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
In recent years the global market for monoclonal antibodies (mAbs) became a multi-billion-dollar business. This success is mainly driven by treatments in the oncology and autoimmune space. Instead, development of effective mAbs against infectious diseases has been lagging behind. For years the high production cost and limited efficacy have blocked broader application of mAbs in the infectious disease space, which instead has been dominated for almost a century by effective and cheap antibiotics and vaccines. Only very few mAbs against RSV, anthrax, Clostridium difficile or rabies have reached the market. This is about to change. The development of urgently needed and highly effective mAbs as preventive and therapeutic treatments against a variety of pathogens is gaining traction. Vast advances in mAb isolation, engineering and production have entirely shifted the cost-efficacy balance. MAbs against devastating diseases like Ebola, HIV and other complex pathogens are now within reach. This trend is further accelerated by ongoing or imminent health crises like COVID-19 and antimicrobial resistance (AMR), where antibodies could be the last resort. In this review we will retrace the history of antibodies from the times of serum therapy to modern mAbs and lay out how the current run for effective treatments against COVID-19 will lead to a quantum leap in scientific, technological and health care system innovation around mAb treatments for infectious diseases.
Collapse
|
31
|
Dreisbach A, Wang M, van der Kooi-Pol MM, Reilman E, Koedijk DGAM, Mars RAT, Duipmans J, Jonkman M, Benschop JJ, Bonarius HPJ, Groen H, Hecker M, Otto A, Bäsell K, Bernhardt J, Back JW, Becher D, Buist G, van Dijl JM. Tryptic Shaving of Staphylococcus aureus Unveils Immunodominant Epitopes on the Bacterial Cell Surface. J Proteome Res 2020; 19:2997-3010. [DOI: 10.1021/acs.jproteome.0c00043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Annette Dreisbach
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Magdalena M. van der Kooi-Pol
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Ewoud Reilman
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Dennis G. A. M. Koedijk
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Ruben A. T. Mars
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - José Duipmans
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Marcel Jonkman
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700 RB Groningen, the Netherlands
| | - Joris J. Benschop
- Pepscan Therapeutics BV, P. O. Box 2098, 8203 AB Lelystad, the Netherlands
| | | | - Herman Groen
- IQ Therapeutics, Rozenburglaan 13a, 9727 DL Groningen, the Netherlands
| | - Michael Hecker
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Andreas Otto
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Katrin Bäsell
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Jörg Bernhardt
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Jaap Willem Back
- Pepscan Therapeutics BV, P. O. Box 2098, 8203 AB Lelystad, the Netherlands
| | - Dörte Becher
- Institut für Mikrobiologie, Ernst-Moritz-Arndt Universität Greifswald, Friedrich-Ludwig-Jahn-Str. 15, D-17489 Greifswald, Germany
| | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P. O. Box 30001, 9700 RB Groningen, the Netherlands
| |
Collapse
|
32
|
Polyketides from marine-derived Aspergillus welwitschiae inhibit Staphylococcus aureus virulence factors and potentiate vancomycin antibacterial activity in vivo. Microb Pathog 2020; 143:104066. [DOI: 10.1016/j.micpath.2020.104066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 12/11/2022]
|
33
|
Ohsawa H, Baba T, Enami J, Hiramatsu K. Protective activity of anti-lipoteichoic acid monoclonal antibody in single or combination therapies in methicillin-resistant Staphylococcus aureus-induced murine sepsis models. J Infect Chemother 2020; 26:520-522. [DOI: 10.1016/j.jiac.2019.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/20/2019] [Accepted: 12/26/2019] [Indexed: 10/25/2022]
|
34
|
Liu J, Kozhaya L, Torres VJ, Unutmaz D, Lu M. Structure-based discovery of a small-molecule inhibitor of methicillin-resistant Staphylococcus aureus virulence. J Biol Chem 2020; 295:5944-5959. [PMID: 32179646 PMCID: PMC7196633 DOI: 10.1074/jbc.ra120.012697] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Indexed: 01/07/2023] Open
Abstract
The rapid emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains poses a major threat to public health. MRSA possesses an arsenal of secreted host-damaging virulence factors that mediate pathogenicity and blunt immune defenses. Panton-Valentine leukocidin (PVL) and α-toxin are exotoxins that create lytic pores in the host cell membrane. They are recognized as being important for the development of invasive MRSA infections and are thus potential targets for antivirulence therapies. Here, we report the high-resolution X-ray crystal structures of both PVL and α-toxin in their soluble, monomeric, and oligomeric membrane-inserted pore states in complex with n-tetradecylphosphocholine (C14PC). The structures revealed two evolutionarily conserved phosphatidylcholine-binding mechanisms and their roles in modulating host cell attachment, oligomer assembly, and membrane perforation. Moreover, we demonstrate that the soluble C14PC compound protects primary human immune cells in vitro against cytolysis by PVL and α-toxin and hence may serve as the basis for the development of an antivirulence agent for managing MRSA infections.
Collapse
Affiliation(s)
- Jie Liu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Lina Kozhaya
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Min Lu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, To whom correspondence should be addressed:
Public Health Research Institute, Dept. of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Newark, NJ 07103. Tel.:
973-854-3260; E-mail:
| |
Collapse
|
35
|
Zurawski DV, McLendon MK. Monoclonal Antibodies as an Antibacterial Approach Against Bacterial Pathogens. Antibiotics (Basel) 2020; 9:antibiotics9040155. [PMID: 32244733 PMCID: PMC7235762 DOI: 10.3390/antibiotics9040155] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
In the beginning of the 21st century, the frequency of antimicrobial resistance (AMR) has reached an apex, where even 4th and 5th generation antibiotics are becoming useless in clinical settings. In turn, patients are suffering from once-curable infections, with increases in morbidity and mortality. The root cause of many of these infections are the ESKAPEE pathogens (Enterococcus species, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species, and Escherichia coli), which thrive in the nosocomial environment and are the bacterial species that have seen the largest rise in the acquisition of antibiotic resistance genes. While traditional small-molecule development still dominates the antibacterial landscape for solutions to AMR, some researchers are now turning to biological approaches as potential game changers. Monoclonal antibodies (mAbs)—more specifically, human monoclonal antibodies (Hu-mAbs)—have been highly pursued in the anti-cancer, autoimmune, and antiviral fields with many success stories, but antibody development for bacterial infection is still just scratching the surface. The untapped potential for Hu-mAbs to be used as a prophylactic or therapeutic treatment for bacterial infection is exciting, as these biologics do not have the same toxicity hurdles of small molecules, could have less resistance as they often target virulence proteins rather than proteins required for survival, and are narrow spectrum (targeting just one pathogenic species), therefore avoiding the disruption of the microbiome. This mini-review will highlight the current antibacterial mAbs approved for patient use, the success stories for mAb development, and new Hu-mAb products in the antibacterial pipeline.
Collapse
|
36
|
Unbiased Identification of Immunogenic Staphylococcus aureus Leukotoxin B-Cell Epitopes. Infect Immun 2020; 88:IAI.00785-19. [PMID: 32014894 DOI: 10.1128/iai.00785-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/26/2020] [Indexed: 12/26/2022] Open
Abstract
Unbiased identification of individual immunogenic B-cell epitopes in major antigens of a pathogen remains a technology challenge for vaccine discovery. We therefore developed a platform for rapid phage display screening of deep recombinant libraries consisting of as few as one major pathogen antigen. Using the bicomponent pore-forming leukocidin (Luk) exotoxins of the major pathogen Staphylococcus aureus as a prototype, we randomly fragmented and separately ligated the hemolysin gamma A (HlgA) and LukS genes into a custom-built phage display system, termed pComb-Opti8. Deep sequence analysis of barcoded amplimers of the HlgA and LukS gene fragment libraries demonstrated that biopannng against a cross-reactive anti-Luk monoclonal antibody (MAb) recovered convergent molecular clones with short overlapping homologous sequences. We thereby identified an 11-amino-acid sequence that is highly conserved in four Luk toxin subunits and is ubiquitous in representation within S. aureus clinical isolates. The isolated 11-amino-acid peptide probe was predicted to retain the native three-dimensional (3D) conformation seen within the Luk holotoxin. Indeed, this peptide was recognized by the selecting anti-Luk MAb, and, using mutated peptides, we showed that a particular amino acid side chain was essential for these interactions. Furthermore, murine immunization with this peptide elicited IgG responses that were highly reactive with both the autologous synthetic peptide and the full-length Luk toxin homologues. Thus, using a gene fragment- and phage display-based pipeline, we have identified and validated immunogenic B-cell epitopes that are cross-reactive between members of the pore-forming leukocidin family. This approach could be harnessed to identify novel epitopes for a much-needed S. aureus-protective subunit vaccine.
Collapse
|
37
|
Chan R, Buckley PT, O'Malley A, Sause WE, Alonzo F, Lubkin A, Boguslawski KM, Payne A, Fernandez J, Strohl WR, Whitaker B, Lynch AS, Torres VJ. Identification of biologic agents to neutralize the bicomponent leukocidins of Staphylococcus aureus. Sci Transl Med 2020; 11:11/475/eaat0882. [PMID: 30651319 DOI: 10.1126/scitranslmed.aat0882] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 01/22/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022]
Abstract
A key aspect underlying the severity of infections caused by Staphylococcus aureus is the abundance of virulence factors that the pathogen uses to thwart critical components of the human immune response. One such mechanism involves the destruction of host immune cells by cytolytic toxins secreted by S. aureus, including five bicomponent leukocidins: PVL, HlgAB, HlgCB, LukED, and LukAB. Purified leukocidins can lyse immune cells ex vivo, and systemic injections of purified LukED or HlgAB can acutely kill mice. Here, we describe the generation and characterization of centyrins that bind S. aureus leukocidins with high affinity and protect primary human immune cells from toxin-mediated cytolysis. Centyrins are small protein scaffolds derived from the fibronectin type III-binding domain of the human protein tenascin-C. Although centyrins are potent in tissue culture assays, their short serum half-lives limit their efficacies in vivo. By extending the serum half-lives of centyrins through their fusion to an albumin-binding consensus domain, we demonstrate the in vivo efficacy of these biologics in a murine intoxication model and in models of both prophylactic and therapeutic treatment of live S. aureus systemic infections. These biologics that target S. aureus virulence factors have potential for treating and preventing serious staphylococcal infections.
Collapse
Affiliation(s)
- Rita Chan
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Peter T Buckley
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Aidan O'Malley
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - William E Sause
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Francis Alonzo
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Kristina M Boguslawski
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA
| | - Angela Payne
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Jeffrey Fernandez
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - William R Strohl
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Brian Whitaker
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA
| | - Anthony Simon Lynch
- Janssen Research & Development, 1400 McKean Road, Spring House, PA, 19477, USA.
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, Alexandria Center for Life Science, 430 East 29th Street, New York, NY 10016, USA.
| |
Collapse
|
38
|
Wang Z, Wen Z, Liu L, Zhu X, Shen B, Yan X, Duan Y, Huang Y. Yangpumicins F and G, Enediyne Congeners from Micromonospora yangpuensis DSM 45577. JOURNAL OF NATURAL PRODUCTS 2019; 82:2483-2488. [PMID: 31490685 PMCID: PMC7170010 DOI: 10.1021/acs.jnatprod.9b00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Enediyne natural products are among the most cytotoxic small molecules and thus excellent payload candidates for the development of antibody-drug conjugates (ADCs). Here we report the isolation and structural elucidation of two new 10-membered anthraquinone-fused enediynes, yangpumicins (YPM) F (6) and G (7), together with five known congeners, YPM A-E (1-5), from Micromonospora yangpuensis DSM 45577. YPM F (6) and G (7) showed strong cytotoxicity against the tested human cancer cell lines, as well as activity against several Gram-positive and Gram-negative pathogens. The 1,2-diols in 6 and 7 promise to enable new linker chemistry for the development of YPM-based ADCs.
Collapse
Affiliation(s)
- Zilong Wang
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Zhongqing Wen
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Ling Liu
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
| | - Xiangcheng Zhu
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, Hunan 410011, China
| | - Ben Shen
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Natural Products Library Initiative at The Scripps Research Institute, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Xiaohui Yan
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| | - Yanwen Duan
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- Hunan Engineering Research Center of Combinatorial Biosynthesis and Natural Product Drug Discovery, Changsha, Hunan 410011, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| | - Yong Huang
- Xiangya International Academy of Translational Medicine at Central South University, Changsha, Hunan 410013, China
- National Engineering Research Center of Combinatorial Biosynthesis for Drug Discovery, Changsha, Hunan 410011, China
| |
Collapse
|
39
|
S. aureus Evades Macrophage Killing through NLRP3-Dependent Effects on Mitochondrial Trafficking. Cell Rep 2019; 22:2431-2441. [PMID: 29490278 PMCID: PMC7160668 DOI: 10.1016/j.celrep.2018.02.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 01/05/2018] [Accepted: 02/06/2018] [Indexed: 12/28/2022] Open
Abstract
Clinical severity of Staphylococcus aureus respiratory infection correlates with alpha toxin (AT) expression. AT activates the NLRP3 inflammasome; deletion of Nlrp3, or AT neutralization, protects mice from lethal S. aureus pneumonia. We tested the hypothesis that this protection is not due to a reduction in inflammasome-dependent cytokines (IL-1β/IL-18) but increased bactericidal function of macrophages. In vivo, neutralization of AT or NLRP3 improved bacterial clearance and survival, while blocking IL-1β/IL-18 did not. Primary human monocytes were used in vitro to determine the mechanism through which NLRP3 alters bacterial killing. In cells treated with small interfering RNA (siRNA) targeting NLRP3 or infected with AT-null S. aureus, mitochondria co-localize with bacterial-containing phagosomes. Mitochondrial engagement activates caspase-1, a process dependent on complex II of the electron transport chain, near the phagosome, promoting its acidification. These data demonstrate a mechanism utilized by S. aureus to sequester itself from antimicrobial processes within the cell.
Collapse
|
40
|
Freitas MAA, Pereira AHC, Pinto JG, Casas A, Ferreira-Strixino J. Bacterial viability after antimicrobial photodynamic therapy with curcumin on multiresistant Staphylococcus aureus. Future Microbiol 2019; 14:739-748. [DOI: 10.2217/fmb-2019-0042] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Staphylococcus aureus are multiresistant pathogens that causes superficial and systemic infections. Antimicrobial photodynamic therapy (APDT) is an alternative in the treatment of diseases caused by these bacteria. Aim: In this study the APDT response on growth, viability, formation of reactive oxygen species and adhesion of methicillin-sensitive strains of Staphylococcus aureus, strains of methicillin-resistant S. aureus and American-type culture collection (ATCC) of S. aureus were evaluated in vitro, after incubation with curcumin for 20 min, and irradiated with LED. Materials & methods: Bacterial growth was assessed by the number of colony-forming units, viability and adhesion were evaluated by confocal microscopy and ROS quantification was performed by fluorimetry. Results: Was observed increase in the production of ROS in APDT groups, besides a decrease in the 4 log growth and loss of the bacterial adhesion. Conclusion: APDT with Curcumin may be an interesting therapeutic alternative, due to its in vitro response, in the control multiresistant clinical S. aureus strains.
Collapse
Affiliation(s)
- Mirian AA Freitas
- Photodynamic Therapy Laboratory - Research & Development Institute – R&DI, University of Vale do Paraíba, Univap. Shishima Hifumi Avenue, 2911, 12244–000, São José dos Campos, São Paulo, Brazil
| | - André HC Pereira
- Photodynamic Therapy Laboratory - Research & Development Institute – R&DI, University of Vale do Paraíba, Univap. Shishima Hifumi Avenue, 2911, 12244–000, São José dos Campos, São Paulo, Brazil
| | - Juliana G Pinto
- Photodynamic Therapy Laboratory - Research & Development Institute – R&DI, University of Vale do Paraíba, Univap. Shishima Hifumi Avenue, 2911, 12244–000, São José dos Campos, São Paulo, Brazil
| | - Adriana Casas
- Centro de Investigaciones sobre Porfirinas y Porfirias (CIPYP), University of Buenos Aires- CONICET-Htal de Clínicas Gral. José de San Martín, Ciudad de Buenos Aires, Argentina
| | - Juliana Ferreira-Strixino
- Photodynamic Therapy Laboratory - Research & Development Institute – R&DI, University of Vale do Paraíba, Univap. Shishima Hifumi Avenue, 2911, 12244–000, São José dos Campos, São Paulo, Brazil
| |
Collapse
|
41
|
Abstract
Community-acquired pneumonia (CAP) is a leading cause of morbidity and mortality worldwide. Despite broad literature including basic and translational scientific studies, many gaps in our understanding of host-pathogen interactions remain. In this review, pathogen virulence factors that drive lung infection and injury are discussed in relation to their associated host immune pathways. CAP epidemiology is considered, with a focus on Staphylococcus aureus and Streptococcus pneumoniae as primary pathogens. Bacterial factors involved in nasal colonization and subsequent virulence are illuminated. A particular emphasis is placed on bacterial pore-forming toxins, host cell death, and inflammasome activation. Identified host-pathogen interactions are then examined by linking pathogen factors to aberrant host response pathways in the context of acute lung injury in both primary and secondary infection. While much is known regarding bacterial virulence and host immune responses, CAP management is still limited to mostly supportive care. It is likely that improvements in therapy will be derived from combinatorial targeting of both pathogen virulence factors and host immunomodulation.
Collapse
|
42
|
Krüger W, Vielreicher S, Kapitan M, Jacobsen ID, Niemiec MJ. Fungal-Bacterial Interactions in Health and Disease. Pathogens 2019; 8:E70. [PMID: 31117285 PMCID: PMC6630686 DOI: 10.3390/pathogens8020070] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/02/2019] [Accepted: 05/16/2019] [Indexed: 12/28/2022] Open
Abstract
Fungi and bacteria encounter each other in various niches of the human body. There, they interact directly with one another or indirectly via the host response. In both cases, interactions can affect host health and disease. In the present review, we summarized current knowledge on fungal-bacterial interactions during their commensal and pathogenic lifestyle. We focus on distinct mucosal niches: the oral cavity, lung, gut, and vagina. In addition, we describe interactions during bloodstream and wound infections and the possible consequences for the human host.
Collapse
Affiliation(s)
- Wibke Krüger
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Sarah Vielreicher
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| | - Ilse D Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
- Institute of Microbiology, Friedrich Schiller University, Jena 07743, Germany.
| | - Maria Joanna Niemiec
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| |
Collapse
|
43
|
de Jong NWM, van Kessel KPM, van Strijp JAG. Immune Evasion by Staphylococcus aureus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0061-2019. [PMID: 30927347 PMCID: PMC11590434 DOI: 10.1128/microbiolspec.gpp3-0061-2019] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus has become a serious threat to human health. In addition to having increased antibiotic resistance, the bacterium is a master at adapting to its host by evading almost every facet of the immune system, the so-called immune evasion proteins. Many of these immune evasion proteins target neutrophils, the most important immune cells in clearing S. aureus infections. The neutrophil attacks pathogens via a plethora of strategies. Therefore, it is no surprise that S. aureus has evolved numerous immune evasion strategies at almost every level imaginable. In this review we discuss step by step the aspects of neutrophil-mediated killing of S. aureus, such as neutrophil activation, migration to the site of infection, bacterial opsonization, phagocytosis, and subsequent neutrophil-mediated killing. After each section we discuss how S. aureus evasion molecules are able to resist the neutrophil attack of these different steps. To date, around 40 immune evasion molecules of S. aureus are known, but its repertoire is still expanding due to the discovery of new evasion proteins and the addition of new functions to already identified evasion proteins. Interestingly, because the different parts of neutrophil attack are redundant, the evasion molecules display redundant functions as well. Knowing how and with which proteins S. aureus is evading the immune system is important in understanding the pathophysiology of this pathogen. This knowledge is crucial for the development of therapeutic approaches that aim to clear staphylococcal infections.
Collapse
Affiliation(s)
- Nienke W M de Jong
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Kok P M van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
44
|
Belete TM. Novel targets to develop new antibacterial agents and novel alternatives to antibacterial agents. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.humic.2019.01.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
45
|
Chen Y, Zhang Y, Chen M, Zhuang J, Fang RH, Gao W, Zhang L. Biomimetic Nanosponges Suppress In Vivo Lethality Induced by the Whole Secreted Proteins of Pathogenic Bacteria. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1804994. [PMID: 30637970 DOI: 10.1002/smll.201804994] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/13/2018] [Indexed: 05/18/2023]
Abstract
Polymeric nanoparticles coated with membrane of intact red blood cells have emerged as biomimetic toxin nanosponges (RBC-NS) that absorb and neutralize bacterial virulence factors associated with numerous bacterial infections. Despite its promise, a clear correlation between in vitro neutralization of complex bacterial toxins and in vivo therapeutic efficacy remains elusive. In this study, the whole secreted proteins (wSP) of methicillin-resistant Staphylococcus aureus (MRSA) are collected to induce lethality in mice. The wSP preserve the complexity of bacterial virulence profile while avoiding the intricacy and dynamics of infections by live bacteria. RBC-NS are first quantified for their neutralization capacity against the hemolytic activity of MRSA wSP in vitro. Using a mouse model, in vivo studies further demonstrate that, by neutralizing the hemolytic activity, RBC-NS confer significant survival benefits against wSP-induced lethality. Furthermore, when mice are challenged with a sublethal dosage of MRSA supernatant, RBC-NS reduce lung damages and inhibit the activation of nuclear factor kappa B in the spleen. These results provide a systematic evaluation of RBC-NS toward the treatment of severe MRSA infections such as MRSA bacteremia and MRSA-induced sepsis.
Collapse
Affiliation(s)
- Yijie Chen
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yue Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mengchun Chen
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jia Zhuang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
46
|
Naran K, Nundalall T, Chetty S, Barth S. Principles of Immunotherapy: Implications for Treatment Strategies in Cancer and Infectious Diseases. Front Microbiol 2018; 9:3158. [PMID: 30622524 PMCID: PMC6308495 DOI: 10.3389/fmicb.2018.03158] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/05/2018] [Indexed: 12/13/2022] Open
Abstract
The advances in cancer biology and pathogenesis during the past two decades, have resulted in immunotherapeutic strategies that have revolutionized the treatment of malignancies, from relatively non-selective toxic agents to specific, mechanism-based therapies. Despite extensive global efforts, infectious diseases remain a leading cause of morbidity and mortality worldwide, necessitating novel, innovative therapeutics that address the current challenges of increasing antimicrobial resistance. Similar to cancer pathogenesis, infectious pathogens successfully fashion a hospitable environment within the host and modulate host metabolic functions to support their nutritional requirements, while suppressing host defenses by altering regulatory mechanisms. These parallels, and the advances made in targeted therapy in cancer, may inform the rational development of therapeutic interventions for infectious diseases. Although "immunotherapy" is habitually associated with the treatment of cancer, this review accentuates the evolving role of key targeted immune interventions that are approved, as well as those in development, for various cancers and infectious diseases. The general features of adoptive therapies, those that enhance T cell effector function, and ligand-based therapies, that neutralize or eliminate diseased cells, are discussed in the context of specific diseases that, to date, lack appropriate remedial treatment; cancer, HIV, TB, and drug-resistant bacterial and fungal infections. The remarkable diversity and versatility that distinguishes immunotherapy is emphasized, consequently establishing this approach within the armory of curative therapeutics, applicable across the disease spectrum.
Collapse
Affiliation(s)
- Krupa Naran
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Trishana Nundalall
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Shivan Chetty
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Stefan Barth
- Medical Biotechnology and Immunotherapy Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
47
|
Alanine Scanning Mutagenesis of the MEDI4893 (Suvratoxumab) Epitope Reduces Alpha Toxin Lytic Activity In Vitro and Staphylococcus aureus Fitness in Infection Models. Antimicrob Agents Chemother 2018; 62:AAC.01033-18. [PMID: 30150481 PMCID: PMC6201083 DOI: 10.1128/aac.01033-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/18/2018] [Indexed: 02/06/2023] Open
Abstract
Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. Alpha toxin (AT) is a cytolytic pore-forming toxin that plays a key role in Staphylococcus aureus pathogenesis; consequently, extensive research was undertaken to understand the AT mechanism of action and its utility as a target for novel prophylaxis and treatment strategies against S. aureus infections. MEDI4893 (suvratoxumab) is a human anti-AT IgG1 monoclonal antibody (MAb) that targets AT and is currently in phase 2 clinical development. As shown previously, the MEDI4893-binding epitope on AT is comprised of the highly conserved amino acid regions 177 to 200 and 261 to 271, suggesting these amino acids are important for AT function. To test this hypothesis and gain insight into the effect of mutations in the epitope on AT neutralization by MEDI4893, nine MEDI4893 contact residues in AT were individually mutated to alanine. Consistent with our hypothesis, 8 out of 9 mutants exhibited >2-fold loss in lytic activity resulting from a defect in cell binding and pore formation. MEDI4893 binding affinity was reduced >2-fold (2- to 27-fold) for 7 out of 9 mutants, and no binding was detected for the W187A mutant. MEDI4893 effectively neutralized all of the lytic mutants in vitro and in vivo. When the defective mutants were introduced into an S. aureus clinical isolate, the mutant-expressing strains exhibited less severe disease in mouse models and were effectively neutralized by MEDI4893. These results indicate the MEDI4893 epitope is highly conserved due in part to its role in AT pore formation and bacterial fitness, thereby decreasing the likelihood for the emergence of MAb-resistant variants.
Collapse
|
48
|
Lysostaphin Lysibody Leads to Effective Opsonization and Killing of Methicillin-Resistant Staphylococcus aureus in a Murine Model. Antimicrob Agents Chemother 2018; 62:AAC.01056-18. [PMID: 30038041 DOI: 10.1128/aac.01056-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022] Open
Abstract
The cell wall of Gram-positive bacteria contains abundant surface-exposed carbohydrate structures that are highly conserved. While these properties make surface carbohydrates ideal targets for immunotherapy, carbohydrates elicit a poor immune response that results primarily in low-affinity IgM antibodies. In a previous publication, we introduced the lysibody approach to address this shortcoming. Lysibodies are engineered molecules that combine a high-affinity carbohydrate-binding domain of bacterial or bacteriophage origin and an Fc effector portion of a human IgG antibody, thus directing effective immunity to conserved bacterial surface carbohydrates. Here, we describe the first example of a lysibody containing the binding domain from a bacteriocin, lysostaphin. We also describe the creation of five lysibodies with binding domains derived from phage lysins, directed against Staphylococcus aureus The lysostaphin and LysK lysibodies showed the most promise and were further characterized. Both lysibodies bound a range of clinically important staphylococcal strains, fixed complement on the staphylococcal surface, and induced phagocytosis of S. aureus by macrophages and human neutrophils. The lysostaphin lysibody had superior in vitro activity compared to that of the LysK lysibody, as well as that of the previously characterized ClyS lysibody, and it effectively protected mice in a kidney abscess/bacteremia model. These results further demonstrate that the lysibody approach is a reproducible means of creating antibacterial antibodies that cannot be produced by conventional means. Lysibodies therefore are a promising solution for opsonic antibodies that may be used passively to both treat and prevent infection by drug-resistant pathogens.
Collapse
|
49
|
Schüller SS, Kramer BW, Villamor E, Spittler A, Berger A, Levy O. Immunomodulation to Prevent or Treat Neonatal Sepsis: Past, Present, and Future. Front Pediatr 2018; 6:199. [PMID: 30073156 PMCID: PMC6060673 DOI: 10.3389/fped.2018.00199] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Despite continued advances in neonatal medicine, sepsis remains a leading cause of death worldwide in neonatal intensive care units. The clinical presentation of sepsis in neonates varies markedly from that in older children and adults, and distinct acute inflammatory responses results in age-specific inflammatory and protective immune response to infection. This review first provides an overview of the neonatal immune system, then covers current mainstream, and experimental preventive and adjuvant therapies in neonatal sepsis. We also discuss how the distinct physiology of the perinatal period shapes early life immune responses and review strategies to reduce neonatal sepsis-related morbidity and mortality. A summary of studies that characterize immune ontogeny and neonatal sepsis is presented, followed by discussion of clinical trials assessing interventions such as breast milk, lactoferrin, probiotics, and pentoxifylline. Finally, we critically appraise future treatment options such as stem cell therapy, other antimicrobial protein and peptides, and targeting of pattern recognition receptors in an effort to prevent and/or treat sepsis in this highly vulnerable neonatal population.
Collapse
Affiliation(s)
- Simone S. Schüller
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Boris W. Kramer
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Centre (MUMC+), Maastricht, Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, Netherlands
| | - Andreas Spittler
- Department of Surgery, Research Labs & Core Facility Flow Cytometry, Medical University of Vienna, Vienna, Austria
| | - Angelika Berger
- Division of Neonatology, Pediatric Intensive Care & Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Department of Medicine, Boston Children's Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Boston, MA, United States
| |
Collapse
|
50
|
Lacoma A, Gomes-Fernandes M, Mesalles E, Arméstar F, Prat C. Persistent Isolation of Staphylococcus aureus in Mechanically-ventilated Patients: Impact of Host-Pathogen Factors on Outcome. Arch Bronconeumol 2018; 55:158-160. [PMID: 29980308 DOI: 10.1016/j.arbres.2018.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Alicia Lacoma
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Spain
| | - Meissiner Gomes-Fernandes
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Spain; CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Eduard Mesalles
- Intensive Care Unit, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Fernando Arméstar
- Intensive Care Unit, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Cristina Prat
- Microbiology Department, Hospital Universitari Germans Trias i Pujol, Institut d'Investigació Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain; CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Spain.
| |
Collapse
|