1
|
Weiss S, Holtfreter S, Meyer TC, Schmiedeke F, Cammann C, Dörr M, Felix SB, Grabe HJ, Homuth G, Kohler C, Mahncke C, Michalik S, Nauck M, Friedrich N, Samietz S, Völzke H, Völker U, Bröker BM. Toxin exposure and HLA alleles determine serum antibody binding to toxic shock syndrome toxin 1 (TSST-1) of Staphylococcus aureus. Front Immunol 2023; 14:1229562. [PMID: 37731490 PMCID: PMC10507260 DOI: 10.3389/fimmu.2023.1229562] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
Life-threatening toxic shock syndrome is often caused by the superantigen toxic shock syndrome toxin-1 (TSST-1) produced by Staphylococcus aureus. A well-known risk factor is the lack of neutralizing antibodies. To identify determinants of the anti-TSST-1 antibody response, we examined 976 participants of the German population-based epidemiological Study of Health in Pomerania (SHIP-TREND-0). We measured anti-TSST-1 antibody levels, analyzed the colonization with TSST-1-encoding S. aureus strains, and performed a genome-wide association analysis of genetic risk factors. TSST-1-specific serum IgG levels varied over a range of 4.2 logs and were elevated by a factor of 12.3 upon nasal colonization with TSST-1-encoding S. aureus. Moreover, the anti-TSST-1 antibody levels were strongly associated with HLA class II gene loci. HLA-DRB1*03:01 and HLA-DQB1*02:01 were positively, and HLA-DRB1*01:01 as well as HLA-DQB1*05:01 negatively associated with the anti-TSST-1 antibody levels. Thus, both toxin exposure and HLA alleles affect the human antibody response to TSST-1.
Collapse
Affiliation(s)
- Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Tanja C. Meyer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Frieder Schmiedeke
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Clemens Cammann
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Stephan B. Felix
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Cedric Mahncke
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nele Friedrich
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Stefanie Samietz
- Department of Prosthetic Dentistry, Gerodontology and Biomaterials, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
2
|
Soezi M, Piri-Gavgani S, Ghanei M, Omrani MD, Soltanmohammadi B, Bagheri KP, Cohan RA, Vaziri F, Siadat SD, Fateh A, Khatami S, Azizi M, Rahimi-Jamnani F. Identification of a novel fully human anti-toxic shock syndrome toxin (TSST)-1 single-chain variable fragment antibody averting TSST-1-induced mitogenesis and cytokine secretion. BMC Biotechnol 2022; 22:31. [PMID: 36307814 PMCID: PMC9617332 DOI: 10.1186/s12896-022-00760-8] [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: 02/09/2022] [Revised: 08/18/2022] [Accepted: 10/07/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Staphylococcal superantigens are virulence factors that help the pathogen escape the immune system and develop an infection. Toxic shock syndrome toxin (TSST)-1 is one of the most studied superantigens whose role in toxic shock syndrome and some particular disorders have been demonstrated. Inhibiting TSST-1 production with antibiotics and targeting TSST-1 with monoclonal antibodies might be one of the best strategies to prevent TSST-1-induced cytokines storm followed by lethality. RESULTS A novel single-chain variable fragment (scFv), MS473, against TSST-1 was identified by selecting an scFv phage library on the TSST-1 protein. The MS473 scFv showed high affinity and specificity for TSST-1. Moreover, MS473 could significantly prevent TSST-1-induced mitogenicity (the IC50 value: 1.5 µM) and cytokine production. CONCLUSION Using traditional antibiotics with an anti-TSST-1 scFv as a safe and effective agent leads to deleting the infection source and preventing the detrimental effects of the toxin disseminated into the whole body.
Collapse
Affiliation(s)
- Mahdieh Soezi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - 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, Systems Biology and Poisoning Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnoush Soltanmohammadi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Farzam Vaziri
- 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
| | - Abolfazl Fateh
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
- Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shohreh Khatami
- Department of Biochemistry, Pasteur Institute of Iran, Tehran, Iran
| | - Masoumeh Azizi
- Molecular Medicine Department, 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
|
3
|
Liu Y, Song Z, Ge S, Zhang J, Xu L, Yang F, Lu D, Luo P, Gu J, Zou Q, Zeng H. Determining the immunological characteristics of a novel human monoclonal antibody developed against staphylococcal enterotoxin B. Hum Vaccin Immunother 2020; 16:1708-1718. [PMID: 32275466 DOI: 10.1080/21645515.2020.1744362] [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] [Indexed: 10/24/2022] Open
Abstract
Staphylococci are the main cause of nosocomial infections globally. The exotoxin staphylococcal enterotoxin B (SEB) produced by methicillin-resistant Staphylococcus aureus is a major cause of pathology after a staphylococcal infection. We previously isolated an anti-SEB human monoclonal antibody designated as M0313. Here we further characterize this antibody in vitro and in vivo. Immunoblotting analysis and ELISA results indicated that M0313 accurately recognized and bound to SEB. Its binding affinity to native SEB was measured at the low nM level. M0313 effectively inhibited SEB from inducing mouse splenic lymphocyte and human peripheral blood mononuclear cell proliferation and cytokine release in cell culture. M0313 also neutralized SEB toxicity in BALB/c female mice. Most importantly, M0313 promoted the survival of mice treated with SEB-expressing bacteria. In-vivo imaging revealed that M0313 treatment significantly reduced the replication of SEB-expressing bacteria in mice. The neutralization capacity of M0313 correlated with its ability to block SEB from binding to major histocompatibility complex II and T-cell receptor by binding to the SEB residues 85-102 and 90-92. Thus, the monoclonal antibody M0313 may be developed into a therapeutic agent.
Collapse
Affiliation(s)
- Yuanyuan Liu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Zhen Song
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China.,Clinical Laboratory Department, Army 954th Hospital, General Hospital of Tibet Military Region , Tibet, PR China
| | - Shuang Ge
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Limin Xu
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Feng Yang
- Research and Development Department, Chengdu Olymvax Biotechnology Co., Ltd ., Chengdu, Sichuan, PR China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University , Chongqing, PR China
| |
Collapse
|
4
|
Chen G, Karauzum H, Long H, Carranza D, Holtsberg FW, Howell KA, Abaandou L, Zhang B, Jarvik N, Ye W, Liao GC, Gross ML, Leung DW, Amarasinghe GK, Aman MJ, Sidhu SS. Potent Neutralization of Staphylococcal Enterotoxin B In Vivo by Antibodies that Block Binding to the T-Cell Receptor. J Mol Biol 2019; 431:4354-4367. [PMID: 30928493 DOI: 10.1016/j.jmb.2019.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/15/2022]
Abstract
To develop an antibody (Ab) therapeutic against staphylococcal enterotoxin B (SEB), a potential incapacitating bioterrorism agent and a major cause of food poisoning, we developed a "class T" anti-SEB neutralizing Ab (GC132) targeting an epitope on SEB distinct from that of previously developed "class M" Abs. A systematic engineering approach was applied to affinity-mature Ab GC132 to yield an optimized therapeutic candidate (GC132a) with sub-nanomolar binding affinity. Mapping of the binding interface by hydrogen-deuterium exchange coupled to mass spectrometry revealed that the class T epitope on SEB overlapped with the T-cell receptor binding site, whereas other evidence suggested that the class M epitope overlapped with the binding site for the major histocompatibility complex. In the IgG format, GC132a showed ∼50-fold more potent toxin-neutralizing efficacy than the best class M Ab in vitro, and fully protected mice from lethal challenge in a toxic shock post-exposure model. We also engineered bispecific Abs (bsAbs) that bound tetravalently by utilizing two class M binding sites and two class T binding sites. The bsAbs displayed enhanced toxin neutralization efficacy compared with the respective monospecific Ab subunits as well as a mixture of the two, indicating that enhanced efficacy was due to heterotypic tetravalent binding to two non-overlapping epitopes on SEB. Together, these results suggest that class T anti-SEB Ab GC132a is an excellent candidate for clinical development and for bsAb engineering.
Collapse
Affiliation(s)
- Gang Chen
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | | | - Hua Long
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Danielle Carranza
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | | | - Katie A Howell
- Integrated Biotherapeutics, Inc., Rockville, MD 20850, USA
| | - Laura Abaandou
- Integrated Biotherapeutics, Inc., Rockville, MD 20850, USA
| | - Bojie Zhang
- Department of Chemistry, Washington University in St. Louis, St Louis, MO 63130, USA
| | - Nick Jarvik
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Wei Ye
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Grant C Liao
- Integrated Biotherapeutics, Inc., Rockville, MD 20850, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, St Louis, MO 63130, USA; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - M Javad Aman
- Integrated Biotherapeutics, Inc., Rockville, MD 20850, USA.
| | - Sachdev S Sidhu
- Banting and Best Department of Medical Research, Department of Molecular Genetics, and the Terrence Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada.
| |
Collapse
|
5
|
Minasyan H. Sepsis: mechanisms of bacterial injury to the patient. Scand J Trauma Resusc Emerg Med 2019; 27:19. [PMID: 30764843 PMCID: PMC6376788 DOI: 10.1186/s13049-019-0596-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 12/17/2022] Open
Abstract
In bacteremia the majority of bacterial species are killed by oxidation on the surface of erythrocytes and digested by local phagocytes in the liver and the spleen. Sepsis-causing bacteria overcome this mechanism of human innate immunity by versatile respiration, production of antioxidant enzymes, hemolysins, exo- and endotoxins, exopolymers and other factors that suppress host defense and provide bacterial survival. Entering the bloodstream in different forms (planktonic, encapsulated, L-form, biofilm fragments), they cause different types of sepsis (fulminant, acute, subacute, chronic, etc.). Sepsis treatment includes antibacterial therapy, support of host vital functions and restore of homeostasis. A bacterium killing is only one of numerous aspects of antibacterial therapy. The latter should inhibit the production of bacterial antioxidant enzymes and hemolysins, neutralize bacterial toxins, modulate bacterial respiration, increase host tolerance to bacterial products, facilitate host bactericidal mechanism and disperse bacterial capsule and biofilm.
Collapse
|
6
|
Vitamin D inhibits the Staphylococcal enterotoxin B-induced expression of tumor necrosis factor in microglial cells. Immunol Res 2017; 65:913-919. [DOI: 10.1007/s12026-017-8930-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
7
|
Abstract
Staphylococcal enterotoxin B is one of the most potent bacterial superantigens that exerts profound toxic effects upon the immune system, leading to stimulation of cytokine release and inflammation. It is associated with food poisoning, nonmenstrual toxic shock, atopic dermatitis, asthma, and nasal polyps in humans. Currently, there is no treatment or vaccine available. Passive immunotherapy using monoclonal antibodies made in several different species has shown significant inhibition in in vitro studies and reduction in staphylococcal enterotoxin B-induced lethal shock in in vivo studies. This should encourage future endeavors to develop these antibodies as therapeutic reagents.
Collapse
|
8
|
Xia T, Liang S, Wang H, Hu S, Sun Y, Yu X, Han J, Li J, Guo S, Dai J, Lou Z, Guo Y. Structural basis for the neutralization and specificity of Staphylococcal enterotoxin B against its MHC Class II binding site. MAbs 2014; 6:119-29. [PMID: 24423621 DOI: 10.4161/mabs.27106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Staphylococcal enterotoxin (SE) B is among the potent toxins produced by Staphylococcus aureus that cause toxic shock syndrome (TSS), which can result in multi-organ failure and death. Currently, neutralizing antibodies have been shown to be effective immunotherapeutic agents against this toxin, but the structural basis of the neutralizing mechanism is still unknown. In this study, we generated a neutralizing monoclonal antibody, 3E2, against SEB, and analyzed the crystal structure of the SEB-3E2 Fab complex. Crystallographic analysis suggested that the neutralizing epitope overlapped with the MHC II molecule binding site on SEB, and thus 3E2 could inhibit SEB function by preventing interaction with the MHC II molecule. Mutagenesis studies were done on SEB, as well as the related Staphylococcus aureus toxins SEA and SEC. These studies revealed that tyrosine (Y)46 and lysine (K)71 residues of SEB are essential to specific antibody-antigen recognition and neutralization. Substitution of Y at SEA glutamine (Q)49, which corresponds to SEB Y46, increased both 3E2's binding to SEA in vitro and the neutralization of SEA in vivo. These results suggested that SEB Y46 is responsible for distinguishing SEB from SEA. These findings may be helpful for the development of antibody-based therapy for SEB-induced TSS.
Collapse
Affiliation(s)
- Tian Xia
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| | - Shuaiyi Liang
- Laboratory of Structural Biology and MOE Laboratory of Protein Science; School of Medicine and Life Science; Tsinghua University; Beijing, P.R. China
| | - Huajing Wang
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering and Antibody; Shanghai, P.R. China
| | - Shi Hu
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering and Antibody; Shanghai, P.R. China
| | - Yuna Sun
- National Laboratory of Macromolecules; Institute of Biophysics; Chinese Academy of Science; Beijing, P.R. China
| | - Xiaojie Yu
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering and Antibody; Shanghai, P.R. China
| | - Jun Han
- College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| | - Jun Li
- College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| | - Shangjing Guo
- College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| | - Jianxin Dai
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering and Antibody; Shanghai, P.R. China; College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| | - Zhiyong Lou
- Laboratory of Structural Biology and MOE Laboratory of Protein Science; School of Medicine and Life Science; Tsinghua University; Beijing, P.R. China
| | - Yajun Guo
- International Joint Cancer Institute; Second Military Medical University; Shanghai, P.R. China; State Key Laboratory of Antibody Medicine and Targeting Therapy and Shanghai Key Laboratory of Cell Engineering and Antibody; Shanghai, P.R. China; College of Pharmacy; Liaocheng University; Liaocheng, P.R. China
| |
Collapse
|
9
|
Lindsay CD, Griffiths GD. Addressing bioterrorism concerns: options for investigating the mechanism of action of Staphylococcus aureus enterotoxin B. Hum Exp Toxicol 2013; 32:606-19. [PMID: 23023027 DOI: 10.1177/0960327112458941] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Staphylococcal enterotoxin B (SEB) is of concern to military and civilian populations as a bioterrorism threat agent. It is a highly potent toxin produced by Staphylococcus aureus and is stable in storage and under aerosolisation; it is able to produce prolonged highly incapacitating illness at very low-inhaled doses and death at elevated doses. Concerns regarding SEB are compounded by the lack of effective medical countermeasures for mass treatment of affected populations. This article considers the mechanism of action of SEB, the availability of appropriate experimental models for evaluating the efficacy of candidate medical countermeasures with particular reference to the need to realistically model SEB responses in man and the availability of candidate countermeasures (with an emphasis on commercial off-the-shelf options). The proposed in vitro approaches would be in keeping with Dstl’s commitment to reduction, refinement and replacement of animal models in biomedical research, particularly in relation to identifying valid alternatives to the use of nonhuman primates in experimental studies.
Collapse
Affiliation(s)
- C D Lindsay
- Biomedical Sciences Department, Dstl Porton Down, Salisbury, Wiltshire, UK.
| | | |
Collapse
|
10
|
Chow SK, Casadevall A. Monoclonal antibodies and toxins--a perspective on function and isotype. Toxins (Basel) 2012; 4:430-54. [PMID: 22822456 PMCID: PMC3398419 DOI: 10.3390/toxins4060430] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 06/06/2012] [Accepted: 06/07/2012] [Indexed: 11/16/2022] Open
Abstract
Antibody therapy remains the only effective treatment for toxin-mediated diseases. The development of hybridoma technology has allowed the isolation of monoclonal antibodies (mAbs) with high specificity and defined properties, and numerous mAbs have been purified and characterized for their protective efficacy against different toxins. This review summarizes the mAb studies for 6 toxins—Shiga toxin, pertussis toxin, anthrax toxin, ricin toxin, botulinum toxin, and Staphylococcal enterotoxin B (SEB)—and analyzes the prevalence of mAb functions and their isotypes. Here we show that most toxin-binding mAbs resulted from immunization are non-protective and that mAbs with potential therapeutic use are preferably characterized. Various common practices and caveats of protection studies are discussed, with the goal of providing insights for the design of future research on antibody-toxin interactions.
Collapse
Affiliation(s)
- Siu-Kei Chow
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA;
| | - Arturo Casadevall
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA;
- Division of Infectious Diseases of the Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-718-430-2811; Fax: +1-718-430-8711
| |
Collapse
|
11
|
Drozdowski B, Zhou Y, Kline B, Spidel J, Chan YY, Albone E, Turchin H, Chao Q, Henry M, Balogach J, Routhier E, Bavari S, Nicolaides NC, Sass PM, Grasso L. Generation and characterization of high affinity human monoclonal antibodies that neutralize staphylococcal enterotoxin B. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2010; 8:9. [PMID: 21176153 PMCID: PMC3022601 DOI: 10.1186/1476-8518-8-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 12/21/2010] [Indexed: 11/10/2022]
Abstract
Background Staphylococcal enterotoxins are considered potential biowarfare agents that can be spread through ingestion or inhalation. Staphylococcal enterotoxin B (SEB) is a widely studied superantigen that can directly stimulate T-cells to release a massive amount of proinflammatory cytokines by bridging the MHC II molecules on an antigen presenting cell (APC) and the Vβ chains of the T-cell receptor (TCR). This potentially can lead to toxic, debilitating and lethal effects. Currently, there are no preventative measures for SEB exposure, only supportive therapies. Methods To develop a potential therapeutic candidate to combat SEB exposure, we have generated three human B-cell hybridomas that produce human monoclonal antibodies (HuMAbs) to SEB. These HuMAbs were screened for specificity, affinity and the ability to block SEB activity in vitro as well as its lethal effect in vivo. Results The high-affinity HuMAbs, as determined by BiaCore analysis, were specific to SEB with minimal crossreactivity to related toxins by ELISA. In an immunoblotting experiment, our HuMAbs bound SEB mixed in a cell lysate and did not bind any of the lysate proteins. In an in vitro cell-based assay, these HuMAbs could inhibit SEB-induced secretion of the proinflammatory cytokines (INF-γ and TNF-α) by primary human lymphocytes with high potency. In an in vivo LPS-potentiated mouse model, our lead antibody, HuMAb-154, was capable of neutralizing up to 100 μg of SEB challenge equivalent to 500 times over the reported LD50 (0.2 μg) , protecting mice from death. Extended survival was also observed when HuMAb-154 was administered after SEB challenge. Conclusion We have generated high-affinity SEB-specific antibodies capable of neutralizing SEB in vitro as well as in vivo in a mouse model. Taken together, these results suggest that our antibodies hold the potential as passive immunotherapies for both prophylactic and therapeutic countermeasures of SEB exposure.
Collapse
|
12
|
Potent neutralization of staphylococcal enterotoxin B by synergistic action of chimeric antibodies. Infect Immun 2010; 78:2801-11. [PMID: 20308304 DOI: 10.1128/iai.01121-09] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Staphylococcal enterotoxin B (SEB), a shock-inducing exotoxin synthesized by Staphylococcus aureus, is an important cause of food poisoning and is a class B bioterrorism agent. SEB mediates antigen-independent activation of a major subset of the T-cell population by cross-linking T-cell receptors (TCRs) with class II major histocompatibility complex (MHC-II) molecules of antigen-presenting cells, resulting in the induction of antigen independent proliferation and cytokine secretion by a significant fraction of the T-cell population. Neutralizing antibodies inhibit SEB-mediated T-cell activation by blocking the toxin's interaction with the TCR or MHC-II and provide protection against the debilitating effects of this superantigen. We derived and searched a set of monoclonal mouse anti-SEB antibodies to identify neutralizing anti-SEB antibodies that bind to different sites on the toxin. A pair of non-cross-reactive, neutralizing anti-SEB monoclonal antibodies (MAbs) was found, and a combination of these antibodies inhibited SEB-induced T-cell proliferation in a synergistic rather than merely additive manner. In order to engineer antibodies more suitable than mouse MAbs for use in humans, the genes encoding the VL and VH gene segments of a synergistically acting pair of mouse MAbs were grafted, respectively, onto genes encoding the constant regions of human Igkappa and human IgG1, transfected into mammalian cells, and used to generate chimeric versions of these antibodies that had affinity and neutralization profiles essentially identical to their mouse counterparts. When tested in cultures of human peripheral blood mononuclear cells or splenocytes derived from HLA-DR3 transgenic mice, the chimeric human-mouse antibodies synergistically neutralized SEB-induced T-cell activation and cytokine production.
Collapse
|
13
|
Verkaik NJ, Boelens HA, de Vogel CP, Tavakol M, Bode LGM, Verbrugh HA, van Belkum A, van Wamel WJB. Heterogeneity of the humoral immune response following Staphylococcus aureus bacteremia. Eur J Clin Microbiol Infect Dis 2010; 29:509-18. [PMID: 20186449 PMCID: PMC2854366 DOI: 10.1007/s10096-010-0888-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 02/01/2010] [Indexed: 12/18/2022]
Abstract
Expanding knowledge on the humoral immune response in Staphylococcus aureus-infected patients is a mandatory step in the development of vaccines and immunotherapies. Here, we present novel insights into the antibody responses following S. aureus bacteremia. Fifteen bacteremic patients were followed extensively from diagnosis onwards (median 29 days, range 9–74). S. aureus strains (median 3, range 1–6) and serial serum samples (median 16, range 6–27) were collected. Strains were genotyped by pulsed-field gel electrophoresis (PFGE) and genes encoding 19 staphylococcal proteins were detected by polymerase chain reaction (PCR). The levels of IgG, IgA, and IgM directed to these proteins were determined using bead-based flow cytometry. All strains isolated from individual patients were PFGE-identical. The genes encoding clumping factor (Clf) A, ClfB, and iron-responsive surface-determinant (Isd) A were detected in all isolates. Antigen-specific IgG levels increased more frequently than IgA or IgM levels. In individual patients, different proteins induced an immune response and the dynamics clearly differed. Anti-ClfB, anti-IsdH, and anti-fibronectin-binding protein A IgG levels increased in 7 of 13 adult patients (p < 0.05). The anti-IsdA IgG level increased in 12 patients (initial to peak level: 1.13–10.72 fold; p < 0.01). Peak level was reached 7–37 days after diagnosis. In a bacteremic 5-day-old newborn, antistaphylococcal IgG levels declined from diagnosis onwards. In conclusion, each bacteremic patient develops a unique immune response directed to different staphylococcal proteins. Therefore, vaccines should be based on multiple components. IsdA is immunogenic and, therefore, produced in nearly all bacteremic patients. This suggests that IsdA might be a useful component of a multivalent staphylococcal vaccine.
Collapse
Affiliation(s)
- N J Verkaik
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, 's Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Park JS, Kim JS, Yi J, Kim EC. [Production and characterization of anti-staphylococcal toxic shock syndrome toxin-1 monoclonal antibody]. Korean J Lab Med 2009; 28:449-56. [PMID: 19127110 DOI: 10.3343/kjlm.2008.28.6.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Recently the association between the virulence factors of Staphylococcus aureus and the outcome of the patients infected with the organism appears to be the subject of active investigation. Toxic shock syndrome toxin-1 (TSST-1) is thought to be a clinically more significant virulence factor than other staphylococcal toxins. We attempted to produce and characterize monoclonal antibodies to staphylococcal TSST-1. METHODS An important epitope of TSST-1, amino acids 1-15 region, was synthesized into a peptide antigen, and Balb/c mice were immunized by intraperitoneal injection of the synthetic antigen. Hybridomas were produced by fusing immunized murine splenocytes with immortal myeloma cells. Hybridomas were cloned through a limiting dilution method. Stable cultured hybridoma was injected into the peritoneal cavity of Balb/c mice, and peritoneal fluid containing the monoclonal antibody was produced. RESULTS One IgG(2b) type monoclonal antibody and two IgM type monoclonal antibodies were obtained. The IgG(2b) type monoclonal antibody was able to detect 5 microg of TSST-1 with Western blot analysis and showed a strong reactivity to TSST-1 with ELISA. CONCLUSIONS Highly immunoreactive anti-TSST-1 monoclonal antibody was produced by the use of synthesized peptide antigen. Diagnostic and protective capacity of this monoclonal antibody should be evaluated in the future.
Collapse
Affiliation(s)
- Jeong-Su Park
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
15
|
Immunogenicity in peptide-immunotherapy: from self/nonself to similar/dissimilar sequences. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:198-207. [PMID: 19065793 DOI: 10.1007/978-0-387-09789-3_15] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The nature of the relationship between an antigenic amino acid sequence and its capability to evoke an immune response is still an unsolved problem. Although experiments indicate that specific (dis)continuous amino acid sequences may determine specific immune responses, how immunogenic properties and recognition informations are mapped onto a non-linear sequence is not understood. Immunology has invoked the concept of self/nonself discrimination in order to explain the capability of the organism to selectively immunoreact. However, no clear, logical and rational pathway has emerged to relate a structure and its immuno-nonreactivity. It cannot yet be dismissed what Koshland wrote in 1990: "Of all the mysteries of modern science, the mechanism of self versus nonself recognition in the immune system ranks at or near the top". This chapter reviews the concept of self/nonself discrimination in the immune system starting from the historical perspective and the conceptual framework that underlie immune reaction pattern. It also introduces future research directions based on a proteomic dissection of the immune unit, qualitatively defined as a low-similarity sequence and quantitatively delimitated by the minimum amino acid requisite able to evoke an immune response, independently ofany, microbial or viral, "foreignness".
Collapse
|
16
|
Kanduc D. Epitopic peptides with low similarity to the host proteome: towards biological therapies without side effects. Expert Opin Biol Ther 2008; 9:45-53. [DOI: 10.1517/14712590802614041] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
17
|
Yang X, Buonpane RA, Moza B, Rahman AKMNU, Wang N, Schlievert PM, McCormick JK, Sundberg EJ, Kranz DM. Neutralization of multiple staphylococcal superantigens by a single-chain protein consisting of affinity-matured, variable domain repeats. J Infect Dis 2008; 198:344-8. [PMID: 18522504 PMCID: PMC2649774 DOI: 10.1086/589776] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Staphylococcus aureus secretes various toxins that act as superantigens by stimulating a large fraction of the host's T cells. Toxin binding to variable domains of T cell receptor beta chains (Vbeta) leads to massive release of inflammatory molecules and potentially to toxic shock syndrome (TSS). Previously, we generated soluble forms of different Vbeta domains with a high affinity for binding superantigens. However, a broader spectrum antagonist is required for the neutralization of multiple toxins. In the present study, we expressed Vbeta domains in tandem as a single-chain protein and neutralized the clinically important superantigens staphylococcal enterotoxin B and TSS toxin-1 with a single agent.
Collapse
Affiliation(s)
- Xi Yang
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | | | - Beenu Moza
- Boston Biomedical Research Institute, Watertown, Massachusetts
| | - A. K. M. Nur-ur Rahman
- Department of Microbiology and Immunology, University of Western Ontario, and Lawson Health Research Institute, London, Ontario, Canada
| | - Ningyan Wang
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| | - Patrick M. Schlievert
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - John K. McCormick
- Department of Microbiology and Immunology, University of Western Ontario, and Lawson Health Research Institute, London, Ontario, Canada
| | | | - David M. Kranz
- Department of Biochemistry, University of Illinois, Urbana, Illinois
| |
Collapse
|
18
|
Sundberg EJ, Deng L, Mariuzza RA. TCR recognition of peptide/MHC class II complexes and superantigens. Semin Immunol 2007; 19:262-71. [PMID: 17560120 PMCID: PMC2949352 DOI: 10.1016/j.smim.2007.04.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 04/23/2007] [Accepted: 04/23/2007] [Indexed: 11/21/2022]
Abstract
Major histocompatibility complex (MHC) class II molecules display peptides to the T cell receptor (TCR). The ability of the TCR to discriminate foreign from self-peptides presented by MHC molecules is a requirement of an effective adaptive immune response. Dysregulation of this molecular recognition event often leads to a disease state. Recently, a number of structural studies have provided significant insight into several such dysregulated interactions between peptide/MHC complexes and TCR molecules. These include TCR recognition of self-peptides, which results in autoimmune reactions, and of mutant self-peptides, common in the immunosurveillance of tumors, as well as the engagement of TCRs by superantigens, a family of bacterial toxins responsible for toxic shock syndrome.
Collapse
Affiliation(s)
- Eric J Sundberg
- Boston Biomedical Research Institute, Watertown, MA 02472, USA.
| | | | | |
Collapse
|
19
|
Moza B, Varma AK, Buonpane RA, Zhu P, Herfst CA, Nicholson MJ, Wilbuer AK, Seth NP, Wucherpfennig KW, McCormick JK, Kranz DM, Sundberg EJ. Structural basis of T-cell specificity and activation by the bacterial superantigen TSST-1. EMBO J 2007; 26:1187-97. [PMID: 17268555 PMCID: PMC1852840 DOI: 10.1038/sj.emboj.7601531] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2006] [Accepted: 12/07/2006] [Indexed: 01/12/2023] Open
Abstract
Superantigens (SAGs) bind simultaneously to major histocompatibility complex (MHC) and T-cell receptor (TCR) molecules, resulting in the massive release of inflammatory cytokines that can lead to toxic shock syndrome (TSS) and death. A major causative agent of TSS is toxic shock syndrome toxin-1 (TSST-1), which is unique relative to other bacterial SAGs owing to its structural divergence and its stringent TCR specificity. Here, we report the crystal structure of TSST-1 in complex with an affinity-matured variant of its wild-type TCR ligand, human T-cell receptor beta chain variable domain 2.1. From this structure and a model of the wild-type complex, we show that TSST-1 engages TCR ligands in a markedly different way than do other SAGs. We provide a structural basis for the high TCR specificity of TSST-1 and present a model of the TSST-1-dependent MHC-SAG-TCR T-cell signaling complex that is structurally and energetically unique relative to those formed by other SAGs. Our data also suggest that protein plasticity plays an exceptionally significant role in this affinity maturation process that results in more than a 3000-fold increase in affinity.
Collapse
Affiliation(s)
- Beenu Moza
- Boston Biomedical Research Institute, Watertown, MA, USA
| | - Ashok K Varma
- Boston Biomedical Research Institute, Watertown, MA, USA
| | | | - Penny Zhu
- Boston Biomedical Research Institute, Watertown, MA, USA
| | - Christine A Herfst
- Department of Microbiology and Immunology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Melissa J Nicholson
- Department of Cancer Immunology and AIDS, Dana Farber Cancer Research Institute, Harvard Medical School, Boston, MA, USA
| | - Anne-Kathrin Wilbuer
- Department of Cancer Immunology and AIDS, Dana Farber Cancer Research Institute, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Nilufer P Seth
- Department of Cancer Immunology and AIDS, Dana Farber Cancer Research Institute, Harvard Medical School, Boston, MA, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and AIDS, Dana Farber Cancer Research Institute, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - John K McCormick
- Department of Microbiology and Immunology, Lawson Health Research Institute, University of Western Ontario, London, Ontario, Canada
| | - David M Kranz
- Department of Biochemistry, University of Illinois, Urbana, IL, USA
| | - Eric J Sundberg
- Boston Biomedical Research Institute, Watertown, MA, USA
- Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02472, USA. Tel.: +1 617 658 7882; Fax: +1 617 972 1761; E-mail:
| |
Collapse
|
20
|
Bavoso A, Ostuni A, De Vendel J, Pollaro F, Armentano F, Knight T, Makker S, Tramontano A. Aldehyde modification of peptide immunogen enhances protein-reactive antibody response to toxic shock syndrome toxin-1. J Pept Sci 2007; 12:843-9. [PMID: 17131300 DOI: 10.1002/psc.821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Introduction of aldehyde groups into protein conjugates enhanced the immune response to a coupled peptide without the use of strong adjuvants. Synthetic peptides representing the N-terminal (residues 1-16) and internal (residues 53-65) epitopes of toxic shock syndrome toxin-1 (TSST-1) were coupled to carrier protein, and carbonyl tags were introduced by Amadori reaction with glycolaldehyde. Modified and unmodified antigens in alum were used to immunize rabbits and the reactivities of antisera were compared. Aldehyde modification augmented the response detected by ELISA, which included enhanced binding to peptides and to native TSST-1. In western blot, TSST-1 was detected by antiserum elicited to the N-terminal peptide, but not that generated to the peptide representing the internal sequence. The same antiserum also neutralized TSST-1 activity in a lymphocyte proliferation assay. The circular dichroism spectrum of the N-terminal peptide indicated a propensity for helical conformation, similar to the structure at the corresponding sequence of the native protein. These data suggest that aldehyde modification can boost immunogenicity of peptide-based vaccines, generating epitope-specific immune responses against the cognate protein antigens without using potent adjuvants.
Collapse
Affiliation(s)
- Alfonso Bavoso
- Universita' degli Studi della Basilicata, Department of Chemistry, Potenza, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
&NA;. Early identification and treatment fundamental for good outcome in children with toxic shock syndrome. DRUGS & THERAPY PERSPECTIVES 2005. [DOI: 10.2165/00042310-200521120-00004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
22
|
Casadevall A, Pirofski LA. The potential of antibody-mediated immunity in the defence against biological weapons. Expert Opin Biol Ther 2005; 5:1359-72. [PMID: 16197341 DOI: 10.1517/14712598.5.10.1359] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibody-mediated immunity (AMI) has been used for the treatment and prevention of infectious diseases for > 100 years, and has a remarkable record of safety, efficacy and versatility. AMI can be used for defence against a wide variety of biological weapons, and passive antibody (Ab) therapy has the potential to provide immediate immunity to susceptible individuals. Recent advances in the Ab field make it possible to generate Abs with enhanced antimicrobial functions. There are significant gaps in our understanding of Ab function, such that the development of Ab-based strategies remains a largely empirical exercise. Nevertheless, the advantages inherent in the therapeutic and prophylactic use of AMI provide a powerful rationale for continued development that will undoubtedly yield many new vaccines and therapeutic Abs.
Collapse
Affiliation(s)
- Arturo Casadevall
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
23
|
Abstract
Toxic shock syndrome (TSS) is an acute, toxin-mediated illness, like endotoxic shock, and is characterized by fever, rash, hypotension, multiorgan involvement, and desquamation. TSS reflects the most severe form of the disease caused by Staphylococcus aureus and Streptococcus pyogenes. A case definition for staphylococcal TSS was well established in the early 1980s and helped in defining the epidemiology. Since the late 1980s, a resurgence of highly invasive streptococcal infections, including a toxic shock-like syndrome, was noted worldwide and a consensus case definition for streptococcal TSS was subsequently proposed in 1993. Both TSS and the toxic shock-like syndrome occur at a lower incidence in children than in adults. Changes in the manufacturing and use of tampons led to a decline in staphylococcal TSS over the past decade, while the incidence of nonmenstrual staphylococcal TSS increased. Nonmenstrual TSS and menstrual TSS are now reported with almost equal frequency. The incidence of streptococcal TSS remains constant after its resurgence, but varies with geographic location. Streptococcal TSS occurs most commonly following varicella or during the use of NSAIDs. Sites of infection in streptococcal TSS are much deeper than in staphylococcal TSS, such as infection caused by blunt trauma, and necrotizing fasciitis. Bacteremia is more common in streptococcal TSS than in staphylococcal TSS. Mortality associated with streptococcal TSS is 5-10% in children, much lower than in adults (30-80%), and is 3-5% for staphylococcal TSS in children.TSS is thought to be a superantigen-mediated disease. Toxins produced by staphylococci and streptococci act as superantigens that can activate the immune system by bypassing the usual antigen-mediated immune-response sequence. The host-pathogen interaction, virulence factors, and the absence or presence of host immunity determines the epidemiology, clinical syndrome, and outcome. Early recognition of this disease is important, because the clinical course is fulminant and the outcome depends on the prompt institution of therapy. Management of a child with TSS includes hemodynamic stabilization and appropriate antimicrobial therapy to eradicate the bacteria. Supportive therapy, aggressive fluid resuscitation, and vasopressors remain the main elements. An adjuvant therapeutic strategy may include agents that can block superantigens, such as intravenous immunoglobulin that contains superantigen neutralizing antibodies.
Collapse
Affiliation(s)
- Yu-Yu Chuang
- Department of Pediatrics, St. Mary's Hospital, LoTung, Taiwan.
| | | | | |
Collapse
|
24
|
de Carvalho Uhl MV, Bottecchia RJ, Azevedo-Silva J, Antonio DL, Vieira-da-Motta O, Mittmann J, Ribeiro PD, de Souza Campos Fernandes RC, Távora N, Medina-Acosta E. Suitability of a recombinant Staphylococcus aureus enterotoxin C bovine variant for immunodiagnostics and therapeutic vaccine development. Vaccine 2004; 22:4191-202. [PMID: 15474709 DOI: 10.1016/j.vaccine.2004.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2003] [Accepted: 05/04/2004] [Indexed: 11/22/2022]
Abstract
Recombinant bovine variant of staphylococcal enterotoxin C (SECbovine), produced as a NH2-terminal histidine hexamer fusion protein (His6-tagged SECbovine), expressed at high levels (25%) in Escherichia coli and affinity purified to homogeneity (99.9%), was tested for its diagnostic and therapeutic potentials. His6-tagged SECbovine is antigenically authentic to native SECbovine across host species, as confirmed by antibody-based capture detection assays using human, mouse, rabbit and chicken hyperimmune sera. His6-tagged SECbovine showed significant T-cell stimulation activity in vitro. His6-tagged SECbovine was immunogenic for IgG in mice (intragastric and intravenous routes) and rabbits (intramuscular and subcutaneous routes), dispensing immunoadjuvant coadministration. The formation of neutralizing antibodies reduced the severity of intoxication symptoms in immunized rabbits. Purified anti-recombinant SECbovine rabbit polyclonal IgG neutralized the pyrexic and diarrhoeagenic effects of native SEC/SED and recombinant SEC, tested by the kitten and rabbit bioassays, respectively.
Collapse
Affiliation(s)
- Marcelle Vianna de Carvalho Uhl
- Laboratório de Biotecnologia, Universidade Estadual do Norte Fluminense Darcy Ribeiro, Avenida Alberto Lamego 2000, Parque Califórnia, CEP 28013-602 Campos dos Goytacazes, RJ, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Binder P, Attre O, Boutin JP, Cavallo JD, Debord T, Jouan A, Vidal D. Medical management of biological warfare and bioterrorism: place of the immunoprevention and the immunotherapy. Comp Immunol Microbiol Infect Dis 2003; 26:401-21. [PMID: 12818625 DOI: 10.1016/s0147-9571(03)00023-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biological weapons are considered as mass destruction and terror weapons. Terrorism including bioterrorism is the major threat in the future conflicts for our nations. The aim of bioterrorism is more related to the potential disorganisation of the society than to the lethal effects of the agents used. The dramatic consequences cannot be discarded, especially if contagious agents such viral are used. The preparation of specific defence measures is a major challenge for our countries. The knowledge acquired from the struggle against natural infectious diseases and recent events are essential to improve behaviours to face the biological weapon threats. The defence attitude is based on the anticipation of the threat, the management of the victims, and the restoration of the operational capabilities. This global defence attitude implies six important functions: (i) alert, (ii) detection and diagnosis, (iii) availability of pharmaceutical countermeasures such as vaccine, sera and anti-infectious medicine and products, (iv) medical management of victims, (v) training and information, (vi) research and development. Passive and active immunoprevention and immuntherapy belong to the approaches discussed in the context of bioterrorism countermeasures. Further researches might be focused on these topics.
Collapse
Affiliation(s)
- Patrice Binder
- Direction centrale du service de santé des Armées, BP 125, 00459, Armees, France.
| | | | | | | | | | | | | |
Collapse
|
26
|
Hong-Geller E, Gupta G. Therapeutic approaches to superantigen-based diseases: a review. J Mol Recognit 2003; 16:91-101. [PMID: 12720278 DOI: 10.1002/jmr.612] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Superantigens secreted by the bacterial pathogen Staphyloccocus aureus are extremely potent toxins that overstimulate the host immune system by binding to the MHC class II and T cell receptors and activating a large population of T cells. Superantigen infection has been shown to be the causative agents in acute diseases, food poisoning and toxic shock syndrome, and in more chronic conditions such as inflammatory skin diseases. In addition to the toll on public health, S. aureus superantigens also represent a potential biothreat to our national security. To address these risks, a number of different therapeutic strategies have been developed that target different aspects of the pathogenic mechanism of S. aureus and superantigen infection. These therapies, which encompass strategies as diverse as production of neutralizing antibodies, inhibitory peptide/receptor design and blockage of superantigen gene transcription, are being tested for treatment of established S. aureus infections in pre- and post-exposure scenarios. In this review, we will describe these different strategies and their efficacies in inhibition of superantigen-induced effects in the host, and present the future outlook for successfully producing therapies for superantigen-based disease.
Collapse
Affiliation(s)
- Elizabeth Hong-Geller
- Los Alamos National Laboratory, Biosciences Division, HRL-1, MS-M888, Los Alamos, NM 87545, USA
| | | |
Collapse
|
27
|
Cho K, Adamson LK, Park J, Greenhalgh DG. Burn injury-mediated alterations in cell cycle progression in lymphoid organs of mice. Shock 2003; 19:138-43. [PMID: 12578122 DOI: 10.1097/00024382-200302000-00009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A key event in the cellular and molecular pathogenesis of multiple organ failure (MOF) after burn injury may be the change in profiles of the cell cycle progression in affected organs. We investigated the effects of burn injury on cell cycle progression in immune organs. Cell cycle analysis in the lymphoid tissues of mice after 18% burn injury revealed that S phase entry was temporarily arrested in the thymus 1 day after injury, whereas the spleen had substantially increased S phase entry at day 8. This mode of cell cycle regulation was reproduced in different age groups and strains of mice. Furthermore, the reactivity to the Ki-67 antibody (indicative of proliferation) was markedly reduced in the thymic cortex at day 1. There was a distinct pattern of hematopoietic foci formation and increased reactivities to the Ki-67 antibody in myelogenous cells in the red pulp of spleen at day 7, consistent with the elevated S phase entry. These data suggest that differential regulation of cell cycle progression may play a crucial role in the phenotypic changes in immune organs after burn injury.
Collapse
Affiliation(s)
- Kiho Cho
- Bum Surgery, Shriners Hospitals for Children Northern California, Sacramento, California 95817, USA
| | | | | | | |
Collapse
|
28
|
Gampfer JM, Samstag A, Waclavicek M, Wolf HM, Eibl MM, Gulle H. Epitope mapping of neutralizing TSST-1 specific antibodies induced by immunization with toxin or toxoids. Vaccine 2002; 20:3675-84. [PMID: 12399195 DOI: 10.1016/s0264-410x(02)00400-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Toxic shock syndrome toxin-1 (TSST-1), a superantigen produced by Staphylococcus aureus, is a potent stimulator of the immune system. T-cells are activated by crosslinking of MHC class II molecules on antigen presenting cells with T-cell receptors (TCR). TSST-1 is associated with the majority of the cases of menstrual staphylococcal toxic shock, a severe and life-threatening multisystem disorder. Even though antibody mediated protection has been studied, information on antibody specificity directed to individual antigenic determinants of the protein is incomplete. To obtain immunogens with low toxicity, we generated a double-site mutant (dmTSST-1), modified at solvent-exposed residues predicted to be important for both MHC class II and TCR binding, and detoxified recombinantly expressed TSST-1 (rTSST-1) as well as native TSST-1 (nTSST-1) isolated from Staphylococcus aureus by treatment with formaldehyde. Rabbits were immunized with rTSST-1, nTSST-1, dmTSST-1, and formaldehyde inactivated toxoids. The sera obtained were used to map the antigen-reactive regions of the molecule and to identify specificities of antibodies induced by immunization with the different antigens. To detect linear antigenic epitopes of TSST-1 the reactivity of the sera with 11-meric peptides having an overhang of four residues, covering the entire molecule of TSST-1, have been studied. We found that sera of TSST-1 immunized rabbits predominantly reacted with N-terminal residues 1-15, while sera generated with formaldehyde inactivated toxoid recognized a total of 7 regions located at the N- and C-terminus and internal sites of TSST-1. Despite different specificities all sera were able to inhibit TSST-1 induced proliferation of human mononuclear cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/biosynthesis
- Antibodies, Bacterial/blood
- Antibodies, Bacterial/chemistry
- Antibody Specificity
- Antigens, Bacterial/blood
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Toxins
- Enterotoxins/chemistry
- Enterotoxins/genetics
- Enterotoxins/immunology
- Enzyme-Linked Immunosorbent Assay
- Epitope Mapping/methods
- Female
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed/genetics
- Mutagenesis, Site-Directed/immunology
- Polyethylene/immunology
- Polyethylene/metabolism
- Protein Structure, Quaternary
- Protein Structure, Tertiary
- Rabbits
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Staphylococcal Vaccines/administration & dosage
- Staphylococcal Vaccines/immunology
- Superantigens
- Toxoids/immunology
- Vaccines, Synthetic/genetics
Collapse
Affiliation(s)
- Jörg M Gampfer
- Biomedizinische Forschungsgesellschaft mbH, Schwarzspanierstrasse 15/1/19, A-1090, Vienna, Austria.
| | | | | | | | | | | |
Collapse
|