1
|
Zong X, Liu P, Wang Z, Zhu H, Zhong C, Zhong P, Jiang H, Liu J, Ma Z, Liu X, Liu R, Ding Y. Structural insights into the binding of nanobodies to the Staphylococcal enterotoxin B. Int J Biol Macromol 2024; 276:133957. [PMID: 39029852 DOI: 10.1016/j.ijbiomac.2024.133957] [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: 04/16/2024] [Revised: 05/21/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Staphylococcal Enterotoxin Type B (SEB), produced by Staphylococcus aureus bacteria, is notorious for inducing severe food poisoning and toxic shock syndrome. While nanobody-based treatments hold promises for combating SEB-induced diseases, the lack of structural information between SEB and nanobodies has hindered the development of nanobody-based therapeutics. Here, we present crystal structures of SEB-Nb3, SEB-Nb6, SEB-Nb8, SEB-Nb11, and SEB-Nb20 at resolutions ranging from 1.59 Å to 2.33 Å. Crystallographic analysis revealed that Nb3, Nb8, Nb11, and Nb20 bind to SEB at the T-cell receptor (TCR) interface, while Nb6 binds at the major histocompatibility complex (MHC) interface, suggesting their potential to inhibit SEB function by disrupting interactions with TCR or MHC molecules. Molecular biological analyses confirmed the thermodynamic and kinetic parameters of Nb3, Nb5, Nb6, Nb8, Nb11, Nb15, Nb18, and Nb20 to SEB. The competitive inhibition was further confirmed by cell-based experiments demonstrating nanobody neutralization. These findings elucidate the structural basis for developing specific nanobodies to neutralize SEB threats, providing crucial insights into the underlying mechanisms and offering significant assistance for further optimization towards future therapeutic strategies.
Collapse
Affiliation(s)
- Xin Zong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Peng Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ziying Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Haoran Zhu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Chao Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Peiyu Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - He Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Jiayuan Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhiqiang Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xihuan Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Rui Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China; Quzhou Fudan Institute, Quzhou, Zhejiang 324002, China.
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China; Quzhou Fudan Institute, Quzhou, Zhejiang 324002, China.
| |
Collapse
|
2
|
Zeng J, Fang Y, Zhang Z, Lv Z, Wang X, Huang Q, Tian Z, Li J, Xu W, Zhu W, Yu J, Liu T, Qian Q. Antitumor activity of Z15-0-2, a bispecific nanobody targeting PD-1 and CTLA-4. Oncogene 2024; 43:2244-2252. [PMID: 38806619 PMCID: PMC11245388 DOI: 10.1038/s41388-024-03066-5] [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: 03/06/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
The combination of programmed cell death protein 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies has potential for enhancing clinical efficacy. We described the development and antitumor activity of Z15-0, a bispecific nanobody targeting both the PD-1 and CTLA-4 pathways simultaneously. We designed and optimized the mRNA sequence encoding Z15-0, referred to as Z15-0-2 and through a series of in vitro and in vivo experiments, we established that the optimized Z15-0-2 mRNA sequence significantly increased the expression of the bispecific nanobody. Administration of Z15-0-2 mRNA to tumor-bearing mice led to greater inhibition of tumor growth compared to controls. In aggregate, we introduced a novel bispecific nanobody and have re-engineered it to boost expression of mRNA, representing a new drug development paradigm.
Collapse
Affiliation(s)
- Jianyao Zeng
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Yuan Fang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zixuan Zhang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhenzhen Lv
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Xiaodie Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Qian Huang
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Zhidan Tian
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jiaguo Li
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Wenfeng Xu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Weimin Zhu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Jing Yu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China
| | - Tao Liu
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
| | - Qijun Qian
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Shanghai Cell Therapy Group Co., Ltd, Shanghai, 201805, China.
- Shanghai Mengchao Cancer Hospital, Shanghai University, Shanghai, 201805, China.
| |
Collapse
|
3
|
Ming K, Hu Y, Zhu M, Xing B, Mei M, Wei Z. Development of nanobodies against Staphylococcus enterotoxin B through yeast surface display. Int J Biol Macromol 2023; 253:126822. [PMID: 37703983 DOI: 10.1016/j.ijbiomac.2023.126822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023]
Abstract
Staphylococcus enterotoxin B (SEB) is one of the primary virulence factors of Staphylococcus aureus but there is still a lack of targeted drugs. SEB activates immune cells via interacting with MHC-II on antigen-presenting cells, leading to the production of large amounts of pro-inflammatory cytokines. Blocking the interaction between SEB and MHC-II can avert the overactivation of immune cells. Nanobodies are the smallest functional antibodies that can bind stably to antigens. In this study, an ideal approach to obtain specific nanobodies without immunizing camelids was introduced. We constructed a library containing up to 5 × 108 nanobodies, and then screened those targeting SEB by using yeast surface display (YSD) technique and fluorescence-activated cell sorting (FACS). A total of 8 nanobodies with divergent complementarity-determining regions (CDRs) sequences were identified and one candidate Nb8 with high affinity to SEB was isolated. In vitro study demonstrated that Nb8 significantly inhibited SEB-induced inflammatory response. Molecular docking simulation indicated that the unique CDR3 sequence contributed to the binding of Nb8 to the MHC-II binding domain of SEB and accordingly cut off the connection between SEB and MHC-II. Our efforts contributed to the development of specific nanobodies for eliminating the threats of SEB.
Collapse
Affiliation(s)
- Ke Ming
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China
| | - Yang Hu
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China
| | - Meijun Zhu
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China
| | - Banbin Xing
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China
| | - Meng Mei
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China
| | - Zigong Wei
- School of life sciences, Hubei University, Wuhan, Hubei, PR China; State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, PR China; National & Local Joint Engineering Research Center of High-throughput Drug Screening Technology, School of life sciences, Hubei University, Wuhan, Hubei, PR China.
| |
Collapse
|
4
|
Zhu Z, Hu Z, Li S, Fang R, Ono HK, Hu DL. Molecular Characteristics and Pathogenicity of Staphylococcus aureus Exotoxins. Int J Mol Sci 2023; 25:395. [PMID: 38203566 PMCID: PMC10778951 DOI: 10.3390/ijms25010395] [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: 11/24/2023] [Revised: 12/24/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Staphylococcus aureus stands as one of the most pervasive pathogens given its morbidity and mortality worldwide due to its roles as an infectious agent that causes a wide variety of diseases ranging from moderately severe skin infections to fatal pneumonia and sepsis. S. aureus produces a variety of exotoxins that serve as important virulence factors in S. aureus-related infectious diseases and food poisoning in both humans and animals. For example, staphylococcal enterotoxins (SEs) produced by S. aureus induce staphylococcal foodborne poisoning; toxic shock syndrome toxin-1 (TSST-1), as a typical superantigen, induces toxic shock syndrome; hemolysins induce cell damage in erythrocytes and leukocytes; and exfoliative toxin induces staphylococcal skin scalded syndrome. Recently, Panton-Valentine leucocidin, a cytotoxin produced by community-associated methicillin-resistant S. aureus (CA-MRSA), has been reported, and new types of SEs and staphylococcal enterotoxin-like toxins (SEls) were discovered and reported successively. This review addresses the progress of and novel insights into the molecular structure, biological activities, and pathogenicity of both the classic and the newly identified exotoxins produced by S. aureus.
Collapse
Affiliation(s)
- Zhihao Zhu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Zuo Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Shaowen Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Rendong Fang
- Joint International Research Laboratory of Animal Health and Animal Food Safety, College of Veterinary Medicine, Southwest University, Chongqing 400715, China;
| | - Hisaya K. Ono
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| | - Dong-Liang Hu
- Department of Zoonoses, Kitasato University School of Veterinary Medicine, Towada 034-8628, Japan; (Z.Z.); (Z.H.); (H.K.O.)
| |
Collapse
|
5
|
Kunkl M, Amormino C, Spallotta F, Caristi S, Fiorillo MT, Paiardini A, Kaempfer R, Tuosto L. Bivalent binding of staphylococcal superantigens to the TCR and CD28 triggers inflammatory signals independently of antigen presenting cells. Front Immunol 2023; 14:1170821. [PMID: 37207220 PMCID: PMC10189049 DOI: 10.3389/fimmu.2023.1170821] [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: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023] Open
Abstract
Staphylococcus aureus superantigens (SAgs) such as staphylococcal enterotoxin A (SEA) and B (SEB) are potent toxins stimulating T cells to produce high levels of inflammatory cytokines, thus causing toxic shock and sepsis. Here we used a recently released artificial intelligence-based algorithm to better elucidate the interaction between staphylococcal SAgs and their ligands on T cells, the TCR and CD28. The obtained computational models together with functional data show that SEB and SEA are able to bind to the TCR and CD28 stimulating T cells to activate inflammatory signals independently of MHC class II- and B7-expressing antigen presenting cells. These data reveal a novel mode of action of staphylococcal SAgs. By binding to the TCR and CD28 in a bivalent way, staphylococcal SAgs trigger both the early and late signalling events, which lead to massive inflammatory cytokine secretion.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Francesco Spallotta
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Silvana Caristi
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Rome, Italy
| | - Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Loretta Tuosto
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University, Rome, Italy
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
6
|
Boehm T, Morimoto R, Trancoso I, Aleksandrova N. Genetic conflicts and the origin of self/nonself-discrimination in the vertebrate immune system. Trends Immunol 2023; 44:372-383. [PMID: 36941153 DOI: 10.1016/j.it.2023.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/23/2023] [Accepted: 02/26/2023] [Indexed: 03/22/2023]
Abstract
Genetic conflicts shape the genomes of prokaryotic and eukaryotic organisms. Here, we argue that some of the key evolutionary novelties of adaptive immune systems of vertebrates are descendants of prokaryotic toxin-antitoxin (TA) systems. Cytidine deaminases and RAG recombinase have evolved from genotoxic enzymes to programmable editors of host genomes, supporting the astounding discriminatory capability of variable lymphocyte receptors of jawless vertebrates, as well as immunoglobulins and T cell receptors of jawed vertebrates. The evolutionarily recent lymphoid lineage is uniquely sensitive to mutations of the DNA maintenance methylase, which is an orphaned distant relative of prokaryotic restriction-modification systems. We discuss how the emergence of adaptive immunity gave rise to higher order genetic conflicts between genetic parasites and their vertebrate host.
Collapse
Affiliation(s)
- Thomas Boehm
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Ryo Morimoto
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Inês Trancoso
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | |
Collapse
|
7
|
Schlievert PM. Staphylococcal Enterotoxin B and C Mutants and Vaccine Toxoids. Microbiol Spectr 2023; 11:e0444622. [PMID: 36815779 PMCID: PMC10101070 DOI: 10.1128/spectrum.04446-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/26/2023] [Indexed: 02/24/2023] Open
Abstract
Three mutants individually of both staphylococcal enterotoxins B and C were prepared by site-specific mutagenesis of enterotoxin amino acids that contact host T lymphocyte immune cell receptor sites (N23A, Q210A, and N23A/Q210A); these amino acids are shared between the two enterotoxins, and mutations reduce the interaction with the variable part of the β-chain of the T lymphocyte receptor. The mutant proteins, as expressed in Staphylococcus aureus RN4220, lacked biological toxicity as measured by the loss of (i) stimulation of rabbit splenocyte proliferation, (ii) pyrogenicity, and (iii) the ability to enhance the lethality of endotoxin shock, compared to wild-type enterotoxins. In addition, the mutants were able to vaccinate rabbits against pyrogenicity, the enhancement of endotoxin shock, and lethality in a pneumonia model when animals were challenged with methicillin-resistant S. aureus. Three vaccine injections (one primary and two boosters) protected rabbits for at least 3.5 months postvaccination when challenged with wild-type enterotoxins (last time point tested). These mutant proteins have the potential to function as toxoid vaccines against these two causes of nonmenstrual toxic shock syndrome (TSS). IMPORTANCE Toxic shock syndrome toxin 1 (TSST-1) and staphylococcal enterotoxins B and C cause the majority of cases of staphylococcal toxic shock syndrome. Previously, vaccine toxoids of TSST-1 have been prepared. In this study, vaccine toxoids of enterotoxins B and C were prepared. The toxoids lost biological toxicity but were able to vaccinate rabbits against lethal TSS.
Collapse
Affiliation(s)
- Patrick M. Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
8
|
SARS-CoV-2 Spike Does Not Possess Intrinsic Superantigen-like Inflammatory Activity. Cells 2022; 11:cells11162526. [PMID: 36010602 PMCID: PMC9406418 DOI: 10.3390/cells11162526] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a rare hyperinflammatory disease occurring several weeks after SARS-CoV-2 infection. The clinical similarities between MIS-C and the toxic shock syndrome, together with the preferential expansion of T cells with a T-cell receptor variable β chain (TCRVβ) skewing, suggested a superantigen theory of MIS-C. For instance, recent in silico modelling evidenced the presence of a highly conserved motif within SARS-CoV-2 spike protein similar in structure to the superantigenic fragment of staphylococcal enterotoxin B (SEB). However, experimental data on the superantigenic activity of the SARS-CoV-2 spike have not yet been provided. Here, we assessed the superantigenic activity of the SARS-CoV-2 spike by analysing inflammatory cytokine production in both Jurkat cells and the peripheral blood CD4+ T cells stimulated with the SARS-CoV-2 spike or SEB as a control. We found that, unlike SEB, the SARS-CoV-2 spike does not exhibit an intrinsic superantigen-like activity.
Collapse
|
9
|
Bai G, Ge Y, Su Y, Chen S, Zeng X, Lu H, Ma B. Computational Construction of a Single-Chain Bi-Paratopic Antibody Allosterically Inhibiting TCR-Staphylococcal Enterotoxin B Binding. Front Immunol 2021; 12:732938. [PMID: 34887850 PMCID: PMC8649926 DOI: 10.3389/fimmu.2021.732938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 11/03/2021] [Indexed: 11/24/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) simultaneously crosslinks MHC class II antigen and TCR, promoting proliferation of T cells and releasing a large number of toxic cytokines. In this report, we computationally examined the possibility of using a single-chain biparatopic bispecific antibody to target SEB and prevent TCR binding. The design was inspired by the observation that mixing two anti-SEB antibodies 14G8 and 6D3 can block SEB-TCR activation, and we used 14G8-6D3-SEB tertiary crystal structure as a template. Twelve simulation systems were constructed to systematically examine the effects of the designed bispecific scFV MB102a, including isolated SEB, MB102a with different linkers, MB102a-SEB complex, MB102a-SEB-TCRβ complex, MB102a-SEB-TCR-MHC II complex, and MB102a-SEB-MHC II. Our all atom molecular dynamics simulations (total 18,900 ns) confirmed that the designed single-chain bispecific antibody may allosterically prevent SEB-TCRβ chain binding and inhibit SEB-TCR-MHC II formation. Subsequent analysis indicated that the binding of scFV to SEB correlates with SEB-TCR binding site motion and weakens SEB-TCR interactions.
Collapse
Affiliation(s)
- Ganggang Bai
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yanhong Ge
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhong Su
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Shuo Chen
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Xingcheng Zeng
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Huixia Lu
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Buyong Ma
- Engineering Research Center of Cell & Therapeutic Antibody (MOE), School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Molcell Biodesign, Inc., Frederick, MD, United States
| |
Collapse
|
10
|
Kunkl M, Amormino C, Caristi S, Tedeschi V, Fiorillo MT, Levy R, Popugailo A, Kaempfer R, Tuosto L. Binding of Staphylococcal Enterotoxin B (SEB) to B7 Receptors Triggers TCR- and CD28-Mediated Inflammatory Signals in the Absence of MHC Class II Molecules. Front Immunol 2021; 12:723689. [PMID: 34489975 PMCID: PMC8418141 DOI: 10.3389/fimmu.2021.723689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/02/2021] [Indexed: 01/19/2023] Open
Abstract
The inflammatory activity of staphylococcal enterotoxin B (SEB) relies on its capacity to trigger polyclonal T-cell activation by binding both T-cell receptor (TCR) and costimulatory receptor CD28 on T cells and MHC class II and B7 molecules on antigen presenting cells (APC). Previous studies highlighted that SEB may bind TCR and CD28 molecules independently of MHC class II, yet the relative contribution of these interactions to the pro-inflammatory function of SEB remained unclear. Here, we show that binding to MHC class II is dispensable for the inflammatory activity of SEB, whereas binding to TCR, CD28 and B7 molecules is pivotal, in both human primary T cells and Jurkat T cell lines. In particular, our finding is that binding of SEB to B7 molecules suffices to trigger both TCR- and CD28-mediated inflammatory signalling. We also provide evidence that, by strengthening the interaction between CD28 and B7, SEB favours the recruitment of the TCR into the immunological synapse, thus inducing lethal inflammatory signalling.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Silvana Caristi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Revital Levy
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Andrey Popugailo
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
11
|
Meilleur CE, Memarnejadian A, Shivji AN, Benoit JM, Tuffs SW, Mele TS, Singh B, Dikeakos JD, Topham DJ, Mu HH, Bennink JR, McCormick JK, Haeryfar SMM. Discordant rearrangement of primary and anamnestic CD8+ T cell responses to influenza A viral epitopes upon exposure to bacterial superantigens: Implications for prophylactic vaccination, heterosubtypic immunity and superinfections. PLoS Pathog 2020; 16:e1008393. [PMID: 32433711 PMCID: PMC7239382 DOI: 10.1371/journal.ppat.1008393] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Infection with (SAg)-producing bacteria may precede or follow infection with or vaccination against influenza A viruses (IAVs). However, how SAgs alter the breadth of IAV-specific CD8+ T cell (TCD8) responses is unknown. Moreover, whether recall responses mediating heterosubtypic immunity to IAVs are manipulated by SAgs remains unexplored. We employed wild-type (WT) and mutant bacterial SAgs, SAg-sufficient/deficient Staphylococcus aureus strains, and WT, mouse-adapted and reassortant IAV strains in multiple in vivo settings to address the above questions. Contrary to the popular view that SAgs delete or anergize T cells, systemic administration of staphylococcal enterotoxin B (SEB) or Mycoplasma arthritidis mitogen before intraperitoneal IAV immunization enlarged the clonal size of ‘select’ IAV-specific TCD8 and reshuffled the hierarchical pattern of primary TCD8 responses. This was mechanistically linked to the TCR Vβ makeup of the impacted clones rather than their immunodominance status. Importantly, SAg-expanded TCD8 retained their IFN-γ production and cognate cytolytic capacities. The enhancing effect of SEB on immunodominant TCD8 was also evident in primary responses to vaccination with heat-inactivated and live attenuated IAV strains administered intramuscularly and intranasally, respectively. Interestingly, in prime-boost immunization settings, the outcome of SEB administration depended strictly upon the time point at which this SAg was introduced. Accordingly, SEB injection before priming raised CD127highKLRG1low memory precursor frequencies and augmented the anamnestic responses of SEB-binding TCD8. By comparison, introducing SEB before boosting diminished recall responses to IAV-derived epitopes drastically and indiscriminately. This was accompanied by lower Ki67 and higher Fas, LAG-3 and PD-1 levels consistent with a pro-apoptotic and/or exhausted phenotype. Therefore, SAgs can have contrasting impacts on anti-IAV immunity depending on the naïve/memory status and the TCR composition of exposed TCD8. Finally, local administration of SEB or infection with SEB-producing S. aureus enhanced pulmonary TCD8 responses to IAV. Our findings have clear implications for superinfections and prophylactic vaccination. Exposure to bacterial superantigens (SAgs) is often a consequence of infection with common Gram-positive bacteria causing septic and toxic shock or food poisoning. How SAgs affect the magnitude, breadth and quality of infection/vaccine-elicited CD8+ T cell (TCD8) responses to respiratory viral pathogens, including influenza A viruses (IAVs), is far from clear. Also importantly, superinfections with IAVs and SAg-producing bacteria are serious clinical occurrences during seasonal and pandemic flu and require urgent attention. We demonstrate that two structurally distinct SAgs, including staphylococcal enterotoxin B (SEB), unexpectedly enhance primary TCD8 responses to ‘select’ IAV-derived epitopes depending on the TCR makeup of the responding clones. Intriguingly, the timing of exposure to SEB dictates the outcome of prime-boost immunization. Seeing a SAg before priming raises memory precursor frequencies and augments anamnestic TCD8 responses. Conversely, a SAg encounter before boosting renders TCD8 prone to death or exhaustion and impedes recall responses, thus likely compromising heterosubtypic immunity to IAVs. Finally, local exposure to SEB increases the pulmonary response of immunodominant IAV-specific TCD8. These findings shed new light on how bacterial infections and SAgs influence the effectiveness of anti-IAV TCD8 responses, and have, as such, wide-ranging implications for preventative vaccination and infection control.
Collapse
Affiliation(s)
- Courtney E. Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Arash Memarnejadian
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Adil N. Shivji
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jenna M. Benoit
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Stephen W. Tuffs
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Tina S. Mele
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Division of Critical Care Medicine, Department of Medicine, Western University, London, Ontario, Canada
| | - Bhagirath Singh
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - David J. Topham
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Hong-Hua Mu
- Division of Rheumatology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Jack R. Bennink
- Viral Immunology Section, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John K. McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Division of Clinical Immunology & Allergy, Department of Medicine, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
12
|
HIV gp120 Induces the Release of Proinflammatory, Angiogenic, and Lymphangiogenic Factors from Human Lung Mast Cells. Vaccines (Basel) 2020; 8:vaccines8020208. [PMID: 32375243 PMCID: PMC7349869 DOI: 10.3390/vaccines8020208] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Human lung mast cells (HLMCs) express the high-affinity receptor FcεRI for IgE and are involved in chronic pulmonary diseases occurring at high frequency among HIV-infected individuals. Immunoglobulin superantigens bind to the variable regions of either the heavy or light chain of immunoglobulins (Igs). Glycoprotein 120 (gp120) of HIV-1 is a typical immunoglobulin superantigen interacting with the heavy chain, variable 3 (VH3) region of human Igs. The present study investigated whether immunoglobulin superantigen gp120 caused the release of different classes of proinflammatory and immunoregulatory mediators from HLMCs. The results show that gp120 from different clades induced the rapid (30 min) release of preformed mediators (histamine and tryptase) from HLMCs. gp120 also caused the de novo synthesis of cysteinyl leukotriene C4 (LTC4) and prostaglandin D2 (PGD2) from HLMCs. Incubation (6 h) of HLMC with gp120 induced the release of angiogenic (VEGF-A) and lymphangiogenic (VEGF-C) factors from HLMCs. The activating property of gp120 was mediated through the interaction with IgE VH3+ bound to FcεRI. Our data indicate that HIV gp120 is a viral superantigen, which induces the release of different proinflammatory, angiogenic, and lymphangiogenic factors from HLMCs. These observations could contribute to understanding, at least in part, the pathophysiology of chronic pulmonary diseases in HIV-infected individuals.
Collapse
|
13
|
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
|
14
|
Varricchi G, Loffredo S, Borriello F, Pecoraro A, Rivellese F, Genovese A, Spadaro G, Marone G. Superantigenic Activation of Human Cardiac Mast Cells. Int J Mol Sci 2019; 20:ijms20081828. [PMID: 31013832 PMCID: PMC6514993 DOI: 10.3390/ijms20081828] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
B cell superantigens, also called immunoglobulin superantigens, bind to the variable regions of either the heavy or light chain of immunoglobulins mirroring the lymphocyte-activating properties of classical T cell superantigens. Protein A of Staphylococcus aureus, protein L of Peptostreptococcus magnus, and gp120 of HIV are typical immunoglobulin superantigens. Mast cells are immune cells expressing the high-affinity receptor for IgE (FcεRI) and are strategically located in the human heart, where they play a role in several cardiometabolic diseases. Here, we investigated whether immunoglobulin superantigens induced the activation of human heart mast cells (HHMCs). Protein A induced the de novo synthesis of cysteinyl leukotriene C4 (LTC4) from HHMCs through the interaction with IgE VH3+ bound to FcεRI. Protein L stimulated the production of prostaglandin D2 (PGD2) from HHMCs through the interaction with κ light chains of IgE. HIV glycoprotein gp120 induced the release of preformed (histamine) and de novo synthesized mediators, such as cysteinyl leukotriene C4 (LTC4), angiogenic (VEGF-A), and lymphangiogenic (VEGF-C) factors by interacting with the VH3 region of IgE. Collectively, our data indicate that bacterial and viral immunoglobulin superantigens can interact with different regions of IgE bound to FcεRI to induce the release of proinflammatory, angiogenic, and lymphangiogenic factors from human cardiac mast cells.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Francesco Borriello
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
- Division of Gastroenterology, Boston Children's Hospital and Harvard Medical School, Boston, 02115 MA, USA.
| | - Antonio Pecoraro
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
| | - Felice Rivellese
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, E1 4NS London, UK.
| | - Arturo Genovese
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80100 Naples, Italy.
- Center for Basic and Clinical Immunology Research (CISI), 80100 Naples, Italy.
- World Allergy Organization (WAO) Center of Excellence, 80100 Naples, Italy.
- Institute of Experimental Endocrinology and Oncology "Gaetano Salvatore", National Research Council (CNR), 80100 Naples, Italy.
| |
Collapse
|
15
|
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
|
16
|
Staphylococcal Superantigens: Pyrogenic Toxins Induce Toxic Shock. Toxins (Basel) 2019; 11:toxins11030178. [PMID: 30909619 PMCID: PMC6468478 DOI: 10.3390/toxins11030178] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 01/01/2023] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related superantigenic toxins produced by Staphylococcus aureus are potent activators of the immune system. These protein toxins bind to major histocompatibility complex (MHC) class II molecules and specific Vβ regions of T-cell receptors (TCRs), resulting in the activation of both monocytes/macrophages and T lymphocytes. The bridging of TCRs with MHC class II molecules by superantigens triggers an early “cytokine storm” and massive polyclonal T-cell proliferation. Proinflammatory cytokines, tumor necrosis factor α, interleukin 1 (IL-1), IL-2, interferon γ (IFNγ), and macrophage chemoattractant protein 1 elicit fever, inflammation, multiple organ injury, hypotension, and lethal shock. Upon MHC/TCR ligation, superantigens induce signaling pathways, including mitogen-activated protein kinase cascades and cytokine receptor signaling, which results in NFκB activation and the phosphoinositide 3-kinase/mammalian target of rapamycin pathways. In addition, gene profiling studies have revealed the essential roles of innate antimicrobial defense genes in the pathogenesis of SEB. The genes expressed in a murine model of SEB-induced shock include intracellular DNA/RNA sensors, apoptosis/DNA damage-related molecules, endoplasmic reticulum/mitochondrial stress responses, immunoproteasome components, and IFN-stimulated genes. This review focuses on the signaling pathways induced by superantigens that lead to the activation of inflammation and damage response genes. The induction of these damage response genes provides evidence that SEB induces danger signals in host cells, resulting in multiorgan injury and toxic shock. Therapeutics targeting both host inflammatory and cell death pathways can potentially mitigate the toxic effects of staphylococcal superantigens.
Collapse
|
17
|
Abstract
Menstrual toxic shock syndrome (TSS) is a serious infectious disease associated with vaginal colonization by Staphylococcus aureus producing the exotoxin TSS toxin 1 (TSST-1). We show that menstrual TSS occurs after TSST-1 interaction with an immune costimulatory molecule called CD40 on the surface of vaginal epithelial cells. Other related toxins, where the entire family is called the superantigen family, bind to CD40, but not with a high-enough apparent affinity to cause TSS; thus, TSST-1 is the only exotoxin superantigen associated. Once the epithelial cells become activated by TSST-1, they produce soluble molecules referred to as chemokines, which in turn facilitate TSST-1 activation of T lymphocytes and macrophages to cause the symptoms of TSS. Identification of small-molecule inhibitors of the interaction of TSST-1 with CD40 may be useful so that they may serve as additives to medical devices, such as tampons and menstrual cups, to reduce the incidence of menstrual TSS. Mucosal and skin tissues form barriers to infection by most bacterial pathogens. Staphylococcus aureus causes diseases across these barriers in part dependent on the proinflammatory properties of superantigens. We showed, through use of a CRISPR-Cas9 CD40 knockout, that the superantigens toxic shock syndrome toxin 1 (TSST-1) and staphylococcal enterotoxins (SEs) B and C stimulated chemokine production from human vaginal epithelial cells (HVECs) through human CD40. This response was enhanced by addition of antibodies against CD40 through an unknown mechanism. TSST-1 was better able to stimulate chemokine (IL-8 and MIP-3α) production by HVECs than SEB and SEC, suggesting this is the reason for TSST-1’s exclusive association with menstrual TSS. A mutant of TSST-1, K121A, caused TSS in a rabbit model when administered vaginally but not intravenously, emphasizing the importance of the local vaginal environment. Collectively, our data suggested that superantigens facilitate infections by disruption of mucosal barriers through their binding to CD40, with subsequent expression of chemokines. The chemokines facilitate TSS and possibly other epithelial conditions after attraction of the adaptive immune system to the local environment.
Collapse
|
18
|
Shannon BA, McCormick JK, Schlievert PM. Toxins and Superantigens of Group A Streptococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0054-2018. [PMID: 30737912 PMCID: PMC11590448 DOI: 10.1128/microbiolspec.gpp3-0054-2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Indexed: 02/07/2023] Open
Abstract
Streptococcus pyogenes (i.e., the group A Streptococcus) is a human-restricted and versatile bacterial pathogen that produces an impressive arsenal of both surface-expressed and secreted virulence factors. Although surface-expressed virulence factors are clearly vital for colonization, establishing infection, and the development of disease, the secreted virulence factors are likely the major mediators of tissue damage and toxicity seen during active infection. The collective exotoxin arsenal of S. pyogenes is rivaled by few bacterial pathogens and includes extracellular enzymes, membrane active proteins, and a variety of toxins that specifically target both the innate and adaptive arms of the immune system, including the superantigens; however, despite their role in S. pyogenes disease, each of these virulence factors has likely evolved with humans in the context of asymptomatic colonization and transmission. In this article, we focus on the biology of the true secreted exotoxins of the group A Streptococcus, as well as their roles in the pathogenesis of human disease.
Collapse
Affiliation(s)
- Blake A Shannon
- Department of Microbiology and Immunology, Western University and The Lawson Health Research Institute, London, Ontario, Canada N6A 4V2
| | - John K McCormick
- Department of Microbiology and Immunology, Western University and The Lawson Health Research Institute, London, Ontario, Canada N6A 4V2
| | - Patrick M Schlievert
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
19
|
Sadeghi J, Alizadeh N, Ahangar Oskouei M, Laghusi D, Savadi Oskouei D, Nikanfar M, Seyyed Mousavi MN. Frequency of superantigen encoding genes of Staphylococcus aureus isolates collected from multiple sclerosis (MS) patients and nasal carriers. Microb Pathog 2018; 127:316-319. [PMID: 30553909 DOI: 10.1016/j.micpath.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Bacterial superantigens are potent T cell activators that can have acute or chronic effects on the central nervous system. OBJECTIVES In this study, the role of enterotoxins, exfoliative toxins and toxic shock syndrome toxin of Staphylococcus aureus was investigated in MS patients and healthy nasal carriers. METHODS Three-hundred fifty nasal swabs were collected from healthy nasal carriers (n = 210) and MS (n = 140) patients. Staphylococcus aureus superantigens were detected by multiplex PCR. Antimicrobial susceptibility pattern was performed using disk diffusion method. RESULTS The highest rates of nasal colonization were seen in MS patients (46.42%). The rates of nasal colonization in the healthcare workers were 30.95%. The most commonly detected superantigens were SEA (31.5%), SEB (17.7%) and ETA (16.9%). The Staphylococcus aureus isolates had the highest levels of resistance against erythromycin (57.7%), clindamycin (55.4%) and co-trimoxazole (43.1%). All isolates were susceptible to vancomycin, linezolid, and mupirocin. CONCLUSION Our results revealed that the frequency of superantigen producing Staphylococcus aureus isolates is high in the MS patients. As well as these isolates are sensitive to mupirocin. Thus it is better to use of mupirocin for nasal decolonization of Staphylococcus aureus in the MS patients.
Collapse
Affiliation(s)
- Javid Sadeghi
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Naser Alizadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mahin Ahangar Oskouei
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Delara Laghusi
- Department of Social Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Daryush Savadi Oskouei
- Department of Neurology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Masoud Nikanfar
- Department of Neurology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mir Naser Seyyed Mousavi
- Department of Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
21
|
Jeong WH, Song DH, Hur GH, Jeong ST. Structure of the staphylococcal enterotoxin B vaccine candidate S19 showing eliminated superantigen activity. Acta Crystallogr F Struct Biol Commun 2017; 73:595-600. [PMID: 29095152 PMCID: PMC5683028 DOI: 10.1107/s2053230x17014844] [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: 07/26/2017] [Accepted: 10/13/2017] [Indexed: 11/29/2022] Open
Abstract
Four mutations (N23A, Y90A, R110A and F177A) were introduced into S19, a vaccine candidate for staphylococcal enterotoxin B (SEB), resulting in a lower binding affinity towards the T-cell receptor beta chain (TCB) and reducing its superantigen activity. The structure of S19 was solved and was superposed on the native or complex structure of SEB. In the superposition model, mutations that were introduced seemed to reduce the number of hydrogen bonds at the SEB-TCB interface. S19 also displayed an unexpected structural change around the flexible-loop region owing to the Y90A mutation. This local structural change provided evidence that the mutated form of S19 could have a lower affinity for major histocompatibility complex (MHC) class II than wild-type SEB.
Collapse
Affiliation(s)
- Woo Hyeon Jeong
- The 5th R&D Institute, Agency for Defense Development, Yuseong PO Box 35, Yuseong-gu, Daejeon 34188, Republic of Korea
| | - Dong Hyun Song
- The 5th R&D Institute, Agency for Defense Development, Yuseong PO Box 35, Yuseong-gu, Daejeon 34188, Republic of Korea
| | - Gyeung Haeng Hur
- The 5th R&D Institute, Agency for Defense Development, Yuseong PO Box 35, Yuseong-gu, Daejeon 34188, Republic of Korea
| | - Seong Tae Jeong
- The 5th R&D Institute, Agency for Defense Development, Yuseong PO Box 35, Yuseong-gu, Daejeon 34188, Republic of Korea
| |
Collapse
|
22
|
Nasopharyngeal infection by Streptococcus pyogenes requires superantigen-responsive Vβ-specific T cells. Proc Natl Acad Sci U S A 2017; 114:10226-10231. [PMID: 28794279 DOI: 10.1073/pnas.1700858114] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The globally prominent pathogen Streptococcus pyogenes secretes potent immunomodulatory proteins known as superantigens (SAgs), which engage lateral surfaces of major histocompatibility class II molecules and T-cell receptor (TCR) β-chain variable domains (Vβs). These interactions result in the activation of numerous Vβ-specific T cells, which is the defining activity of a SAg. Although streptococcal SAgs are known virulence factors in scarlet fever and toxic shock syndrome, mechanisms by how SAgs contribute to the life cycle of S. pyogenes remain poorly understood. Herein, we demonstrate that passive immunization against the Vβ8-targeting SAg streptococcal pyrogenic exotoxin A (SpeA), or active immunization with either wild-type or a nonfunctional SpeA mutant, protects mice from nasopharyngeal infection; however, only passive immunization, or vaccination with inactive SpeA, resulted in high-titer SpeA-specific antibodies in vivo. Mice vaccinated with wild-type SpeA rendered Vβ8+ T cells poorly responsive, which prevented infection. This phenotype was reproduced with staphylococcal enterotoxin B, a heterologous SAg that also targets Vβ8+ T cells, and rendered mice resistant to infection. Furthermore, antibody-mediated depletion of T cells prevented nasopharyngeal infection by S. pyogenes, but not by Streptococcus pneumoniae, a bacterium that does not produce SAgs. Remarkably, these observations suggest that S. pyogenes uses SAgs to manipulate Vβ-specific T cells to establish nasopharyngeal infection.
Collapse
|
23
|
Benkerroum N. Staphylococcal enterotoxins and enterotoxin-like toxins with special reference to dairy products: An overview. Crit Rev Food Sci Nutr 2017; 58:1943-1970. [DOI: 10.1080/10408398.2017.1289149] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Noreddine Benkerroum
- Department of Food Science and Agricultural Chemistry, Macdonald-Stewart Building, McGill University, Macdonald Campus, Sainte-Anne-de-Bellevue, Canada
| |
Collapse
|
24
|
A novel staphylococcal enterotoxin B subunit vaccine candidate elicits protective immune response in a mouse model. Toxicon 2017; 131:68-77. [PMID: 28359755 DOI: 10.1016/j.toxicon.2017.03.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 03/21/2017] [Accepted: 03/22/2017] [Indexed: 11/20/2022]
Abstract
Staphylococcal enterotoxin B (SEB), produced by the gram-positive bacterium Staphylococcus aureus, is responsible for food poisoning and toxic shock syndrome, and is considered a potential bioterrorism agent. Unfortunately, still now no approved vaccines are available against SEB. In this study, we constructed a series of nontoxic SEB mutants (mSEBs) and examined whether these mSEBs provide protective immunity against SEB challenge. These mSEB vaccine candidates did not demonstrate superantigen activity in mouse splenocyte cultures. Immunization with the vaccine candidates triggered the production of IgG-antibodies with neutralizing activity. In addition, increased production of IgG1 and IgG3 was observed after immunization, which signifies both Th1- and Th2-induced immune responses. Among the vaccine candidates tested, S9, a double mutant (N23A and Y90A) and S19, a quadruple mutant (N23A, Y90A, R110A, and F177A), demonstrated complete protection against a lethal SEB challenge. Altogether, our results strongly suggest that these mSEBs could be an effective recombinant SEB vaccine candidates for further/future preclinical and clinical studies.
Collapse
|
25
|
Rukkawattanakul T, Sookrung N, Seesuay W, Onlamoon N, Diraphat P, Chaicumpa W, Indrawattana N. Human scFvs That Counteract Bioactivities of Staphylococcus aureus TSST-1. Toxins (Basel) 2017; 9:toxins9020050. [PMID: 28218671 PMCID: PMC5331430 DOI: 10.3390/toxins9020050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 02/09/2017] [Indexed: 11/16/2022] Open
Abstract
Some Staphylococcus aureus isolates produced toxic shock syndrome toxin-1 (TSST-1) which is a pyrogenic toxin superantigen (PTSAg). The toxin activates a large fraction of peripheral blood T lymphocytes causing the cells to proliferate and release massive amounts of pro-inflammatory cytokines leading to a life-threatening multisystem disorder: toxic shock syndrome (TSS). PTSAg-mediated-T cell stimulation circumvents the conventional antigenic peptide presentation to T cell receptor (TCR) by the antigen-presenting cell (APC). Instead, intact PTSAg binds directly to MHC-II molecule outside peptide binding cleft and simultaneously cross-links TCR-Vβ region. Currently, there is neither specific TSS treatment nor drug that directly inactivates TSST-1. In this study, human single chain antibodies (HuscFvs) that bound to and neutralized bioactivities of the TSST-1 were generated using phage display technology. Three E. coli clones transfected with TSST-1-bound phages fished-out from the human scFv library using recombinant TSST-1 as bait expressed TSST-1-bound-HuscFvs that inhibited the TSST-1-mediated T cell activation and pro-inflammatory cytokine gene expressions and productions.Computerized simulation, verified by mutations of the residues of HuscFv complementarity determining regions (CDRs),predicted to involve in target binding indicated that the HuscFvs formed interface contact with the toxin residues important for immunopathogenesis. The HuscFvs have high potential for future therapeutic application.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/metabolism
- Antibodies, Neutralizing/pharmacology
- Bacterial Toxins/antagonists & inhibitors
- Bacterial Toxins/genetics
- Bacterial Toxins/immunology
- Bacterial Toxins/metabolism
- Cell Surface Display Techniques
- Cells, Cultured
- Cytokines/metabolism
- Enterotoxins/antagonists & inhibitors
- Enterotoxins/genetics
- Enterotoxins/immunology
- Enterotoxins/metabolism
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Histocompatibility Antigens Class II/metabolism
- Host-Pathogen Interactions
- Humans
- Inflammation Mediators/metabolism
- Lymphocyte Activation/drug effects
- Mutation
- Protein Binding
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Shock, Septic/immunology
- Shock, Septic/metabolism
- Shock, Septic/microbiology
- Shock, Septic/prevention & control
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/metabolism
- Single-Chain Antibodies/pharmacology
- Staphylococcal Infections/immunology
- Staphylococcal Infections/metabolism
- Staphylococcal Infections/microbiology
- Staphylococcal Infections/prevention & control
- Staphylococcus aureus/drug effects
- Staphylococcus aureus/genetics
- Staphylococcus aureus/immunology
- Staphylococcus aureus/metabolism
- Superantigens/genetics
- Superantigens/immunology
- Superantigens/metabolism
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/microbiology
Collapse
Affiliation(s)
- Thunchanok Rukkawattanakul
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Nitat Sookrung
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Watee Seesuay
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Nattawat Onlamoon
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Pornphan Diraphat
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand.
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
26
|
Two common structural motifs for TCR recognition by staphylococcal enterotoxins. Sci Rep 2016; 6:25796. [PMID: 27180909 PMCID: PMC4867771 DOI: 10.1038/srep25796] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/18/2016] [Indexed: 12/29/2022] Open
Abstract
Superantigens are toxins produced by Staphylococcus aureus, called staphylococcal enterotoxins (abbreviated SEA to SEU). They can cross-link the T cell receptor (TCR) and major histocompatibility complex class II, triggering a massive T cell activation and hence disease. Due to high stability and toxicity, superantigens are potential agents of bioterrorism. Hence, antagonists may not only be useful in the treatment of disease but also serve as countermeasures to biological warfare. Of particular interest are inhibitors against SEA and SEB. SEA is the main cause of food poisoning, while SEB is a common toxin manufactured as a biological weapon. Here, we present the crystal structures of SEA in complex with TCR and SEE in complex with the same TCR, complemented with computational alanine-scanning mutagenesis of SEA, SEB, SEC3, SEE, and SEH. We have identified two common areas that contribute to the general TCR binding for these superantigens. This paves the way for design of single antagonists directed towards multiple toxins.
Collapse
|
27
|
Rödström KEJ, Regenthal P, Lindkvist-Petersson K. Structure of Staphylococcal Enterotoxin E in Complex with TCR Defines the Role of TCR Loop Positioning in Superantigen Recognition. PLoS One 2015; 10:e0131988. [PMID: 26147596 PMCID: PMC4492778 DOI: 10.1371/journal.pone.0131988] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/09/2015] [Indexed: 11/18/2022] Open
Abstract
T cells are crucial players in cell-mediated immunity. The specificity of their receptor, the T cell receptor (TCR), is central for the immune system to distinguish foreign from host antigens. Superantigens are bacterial toxins capable of inducing a toxic immune response by cross-linking the TCR and the major histocompatibility complex (MHC) class II and circumventing the antigen specificity. Here, we present the structure of staphylococcal enterotoxin E (SEE) in complex with a human T cell receptor, as well as the unligated T cell receptor structure. There are clear structural changes in the TCR loops upon superantigen binding. In particular, the HV4 loop moves to circumvent steric clashes upon complex formation. In addition, a predicted ternary model of SEE in complex with both TCR and MHC class II displays intermolecular contacts between the TCR α-chain and the MHC, suggesting that the TCR α-chain is of importance for complex formation.
Collapse
Affiliation(s)
- Karin E. J. Rödström
- Department of Experimental Medical Science, Lund University, BMC C13, 22 184, Lund, Sweden
| | - Paulina Regenthal
- Department of Experimental Medical Science, Lund University, BMC C13, 22 184, Lund, Sweden
| | | |
Collapse
|
28
|
Li J, Yang J, Lu YW, Wu S, Wang MR, Zhu JM. Possible Role of Staphylococcal Enterotoxin B in the Pathogenesis of Autoimmune Diseases. Viral Immunol 2015; 28:354-9. [PMID: 26086678 DOI: 10.1089/vim.2015.0017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
As a member of superantigens (SAgs) produced by Staphylococcus aureus, staphylococcal enterotoxin B (SEB) is a exotoxin superantigen that can regulate the activity of immunomodulatory and pro-inflammatory cell types. In addition, SEB plays a critical role in the pathogenesis of autoimmune disorders either by initiating the autoimmune process or by inducing a relapse in an individual in clinical remission from an autoimmune disorder. SEB can directly activate T lymphocytes, leading to the release of cytokines, superoxides, or other mediators of inflammation either directly or indirectly, because of its unique ability to cross-link human major histocompatibility complex (MHC) class II and T cell receptors (TCR), forming a trimolecular complex. This review discusses the potential effects of SEB in the pathogenesis of autoimmune diseases such as multiple sclerosis, systemic lupus erythematosus, and rheumatoid arthritis, and explores some updated therapeutic medications to neutralize SEB.
Collapse
Affiliation(s)
- Jing Li
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Jie Yang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Yu-wei Lu
- 2 Department of Information, The Second Hospital of Anhui Medical University , Hefei, China
| | - Song Wu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ming-rui Wang
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| | - Ji-min Zhu
- 1 Department of Public Health and General Medicine, School of Integrated Traditional and Western Medicine, Anhui University of Chinese Medicine , Hefei, China
| |
Collapse
|
29
|
Dutta K, Varshney AK, Franklin MC, Goger M, Wang X, Fries BC. Mechanisms mediating enhanced neutralization efficacy of staphylococcal enterotoxin B by combinations of monoclonal antibodies. J Biol Chem 2015; 290:6715-30. [PMID: 25572397 DOI: 10.1074/jbc.m114.630715] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Staphylococcal enterotoxin B (SEB) is a superantigen that cross-links the major histocompatibility complex class II and specific V-β chains of the T-cell receptor, thus forming a ternary complex. Developing neutralizing mAb to disrupt the ternary complex and abrogate the resulting toxicity is a major therapeutic challenge because SEB is effective at very low concentrations. We show that combining two SEB-specific mAbs enhances their efficacy, even though one of the two mAbs by itself has no effect on neutralization. Crystallography was employed for fine-mapping conformational epitopes in binary and ternary complexes between SEB and Fab fragments. NMR spectroscopy was used to validate and identify subtle allosteric changes induced by mAbs binding to SEB. The mapping of epitopes established that a combination of different mAbs can enhance efficacy of mAb-mediated protection from SEB induced lethal shock by two different mechanisms: one mAb mixture promoted clearance of the toxin both in vitro and in vivo by FcR-mediated cross-linking and clearance, whereas the other mAb mixture induced subtle allosteric conformational changes in SEB that perturbed formation of the SEB·T-cell receptor·major histocompatibility complex class II trimer. Finally structural information accurately predicted mAb binding to other superantigens that share conformational epitopes with SEB. Fine mapping of conformational epitopes is a powerful tool to establish the mechanism and optimize the action of synergistic mAb combinations.
Collapse
Affiliation(s)
- Kaushik Dutta
- From the New York Structural Biology Center, New York, New York 10027,
| | - Avanish K Varshney
- the Department of Medicine and Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11790, and the Department of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | - Michael Goger
- From the New York Structural Biology Center, New York, New York 10027
| | - Xiaobo Wang
- the Department of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Bettina C Fries
- the Department of Medicine and Molecular Genetics and Microbiology, Stony Brook University, Stony Brook, New York 11790, and the Department of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
30
|
Kang H, Deng H, Shen M, He X, Xia Y, Li Y, Liang Z, Wang H, Huang J. Superantigenicity analysis of staphylococcal enterotoxins SElK and SElQ in a mouse model. RSC Adv 2015. [DOI: 10.1039/c4ra16649c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Staphylococcal enterotoxins (SEs) are superantigenic toxins secreted byStaphylococcus aureusthat is involved in causing food poisoning and human diseases.
Collapse
Affiliation(s)
- Hongzhi Kang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin
- China
| | - Hui Deng
- School of Life Sciences
- Tianjin University
- Tianjin
- China
| | - Menglu Shen
- School of Life Sciences
- Tianjin University
- Tianjin
- China
| | - Xianzhi He
- School of Life Sciences
- Tianjin University
- Tianjin
- China
| | - Yihe Xia
- School of Life Sciences
- Tianjin University
- Tianjin
- China
| | - Yi Li
- School of Life Sciences
- Tianjin University
- Tianjin
- China
| | - Zhixuan Liang
- Tianjin Center of Animal Disease Preventive and Control
- Tianjin
- China
| | - Hongjun Wang
- Tianjin Center of Animal Disease Preventive and Control
- Tianjin
- China
| | - Jinhai Huang
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin
- China
- School of Life Sciences
| |
Collapse
|
31
|
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
|
32
|
Benedik E, Skrt M, Podlipnik C, Ulrih NP. Binding of flavonoids to staphylococcal enterotoxin B. Food Chem Toxicol 2014; 74:1-8. [PMID: 25193263 DOI: 10.1016/j.fct.2014.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/29/2014] [Accepted: 08/21/2014] [Indexed: 10/24/2022]
Abstract
Staphylococcal enterotoxins are metabolic products of Staphylococcus aureus that are responsible for the second-most-commonly reported type of food poisoning. Polyphenols are known to interact with proteins to form complexes, the properties of which depend on the structures of both the polyphenols and the protein. In the present study, we investigated the binding of four flavonoid polyphenols to Staphylococcal enterotoxin B (SEB) at pH 7.5 and 25 °C: (-)-epigallocatechin-3-gallate (EGCG), (-)-epigallocatechin (EGC), kaempferol-3-glucoside (KAM-G) and kaempferol (KAM). Fluorescence emission spectrometry and molecular docking were applied to compare experimentally determined binding parameters with molecular modeling. EGCG showed an order of magnitude higher binding constant (1.4 × 10(5) M(-1)) than the other studied polyphenols. Our blind-docking results showed that EGCG and similar polyphenolic ligands is likely to bind to the channel at the surface of SEB that is responsible for the recognition of the T-cell beta chain fragment and influence the adhesion of SEB to T cells.
Collapse
Affiliation(s)
- Evgen Benedik
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Mihaela Skrt
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Crtomir Podlipnik
- Faculty of Chemistry and Chemical Technology, University of Ljubljana, Aškerčeva 5, 1000 Ljubljana, Slovenia
| | - Nataša Poklar Ulrih
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia; Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins (CipKeBiP), Jamova 39, Ljubljana, Slovenia.
| |
Collapse
|
33
|
Rödström KEJ, Elbing K, Lindkvist-Petersson K. Structure of the superantigen staphylococcal enterotoxin B in complex with TCR and peptide-MHC demonstrates absence of TCR-peptide contacts. THE JOURNAL OF IMMUNOLOGY 2014; 193:1998-2004. [PMID: 25015819 DOI: 10.4049/jimmunol.1401268] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Superantigens are immune-stimulatory toxins produced by Staphylococcus aureus, which are able to interact with host immune receptors to induce a massive release of cytokines, causing toxic shock syndrome and possibly death. In this article, we present the x-ray structure of staphylococcal enterotoxin B (SEB) in complex with its receptors, the TCR and MHC class II, forming a ternary complex. The structure, in combination with functional analyses, clearly shows how SEB adopts a wedge-like position when binding to the β-chain of TCR, allowing for an interaction between the α-chain of TCR and MHC. Furthermore, the binding mode also circumvents contact between TCR and the peptide presented by MHC, which enables SEB to initiate a peptide-independent activation of T cells.
Collapse
Affiliation(s)
- Karin E J Rödström
- Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Karin Elbing
- Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | | |
Collapse
|
34
|
Superantigens subvert the neutrophil response to promote abscess formation and enhance Staphylococcus aureus survival in vivo. Infect Immun 2014; 82:3588-98. [PMID: 24914221 DOI: 10.1128/iai.02110-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Staphylococcus aureus is a versatile bacterial pathogen that produces T cell-activating toxins known as superantigens (SAgs). Although excessive immune activation by SAgs can induce a dysregulated cytokine storm as a component of what is known as toxic shock syndrome (TSS), the contribution of SAgs to the staphylococcal infection process is not well defined. Here, we evaluated the role of the bacterial superantigen staphylococcal enterotoxin A (SEA) in a bacteremia model using humanized transgenic mice expressing SAg-responsive HLA-DR4 molecules. Infection with S. aureus Newman induced SEA-dependent Vβ skewing of T cells and enhanced bacterial survival in the liver compared with infection by sea knockout strain. SEA-induced gamma interferon, interleukin-12, and chemokine responses resulted in increased infiltration of CD11b(+) Ly6G(+) neutrophils into the liver, promoting the formation of abscesses that contained large numbers of viable staphylococci. Hepatic abscesses occurred significantly more frequently in S. aureus Newman-infected livers than in livers infected with the Newman sea knockout strain, promoting the survival of S. aureus in vivo. This represents a novel mechanism during infection whereby S. aureus utilizes SAgs to form a specialized niche and manipulate the immune system.
Collapse
|
35
|
Principato M, Qian BF. Staphylococcal enterotoxins in the etiopathogenesis of mucosal autoimmunity within the gastrointestinal tract. Toxins (Basel) 2014; 6:1471-89. [PMID: 24776983 PMCID: PMC4052247 DOI: 10.3390/toxins6051471] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/18/2014] [Accepted: 04/22/2014] [Indexed: 01/22/2023] Open
Abstract
The staphylococcal enterotoxins (SEs) are the products of Staphylococcus aureus and are recognized as the causative agents of classical food poisoning in humans following the consumption of contaminated food. While illness evoked by ingestion of the SE or its producer organism in tainted food are often self-limited, our current understanding regarding the evolution of S. aureus provokes the utmost concern. The organism and its associated toxins, has been implicated in a wide variety of disease states including infections of the skin, heart, sinuses, inflammatory gastrointestinal disease, toxic shock, and Sudden Infant Death Syndrome. The intricate relationship between the various subsets of immunocompetent T cells and accessory cells and the ingested material found within the gastrointestinal tract present daunting challenges to the maintenance of immunologic homeostasis. Dysregulation of the intricate balances within this environment has the potential for extreme consequences within the host, some of which are long-lived. The focus of this review is to evaluate the relevance of staphylococcal enterotoxin in the context of mucosal immunity, and the underlying mechanisms that contribute to the pathogenesis of gastrointestinal autoimmune disease.
Collapse
Affiliation(s)
- MaryAnn Principato
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| | - Bi-Feng Qian
- Commissioner's Fellowship Program, Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, 8301 Muirkirk Road, Laurel, MD 20708, USA.
| |
Collapse
|
36
|
Patterson KG, Dixon Pittaro JL, Bastedo PS, Hess DA, Haeryfar SMM, McCormick JK. Control of established colon cancer xenografts using a novel humanized single chain antibody-streptococcal superantigen fusion protein targeting the 5T4 oncofetal antigen. PLoS One 2014; 9:e95200. [PMID: 24736661 PMCID: PMC3988171 DOI: 10.1371/journal.pone.0095200] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 03/25/2014] [Indexed: 01/21/2023] Open
Abstract
Superantigens (SAgs) are microbial toxins that cross-link T cell receptors with major histocompatibility class II (MHC-II) molecules leading to the activation of large numbers of T cells. Herein, we describe the development and preclinical testing of a novel tumor-targeted SAg (TTS) therapeutic built using the streptococcal pyrogenic exotoxin C (SpeC) SAg and targeting cancer cells expressing the 5T4 tumor-associated antigen (TAA). To inhibit potentially harmful widespread immune cell activation, a SpeC mutation within the high-affinity MHC-II binding interface was generated (SpeCD203A) that demonstrated a pronounced reduction in mitogenic activity, yet this mutant could still induce immune cell-mediated cancer cell death in vitro. To target 5T4+ cancer cells, we engineered a humanized single chain variable fragment (scFv) antibody to recognize 5T4 (scFv5T4). Specific targeting of scFv5T4 was verified. SpeCD203A fused to scFv5T4 maintained the ability to activate and induce immune cell-mediated cytotoxicity of colorectal cancer cells. Using a xenograft model of established human colon cancer, we demonstrated that the SpeC-based TTS was able to control the growth and spread of large tumors in vivo. This required both TAA targeting by scFv5T4 and functional SAg activity. These studies lay the foundation for the development of streptococcal SAgs as ‘next-generation’ TTSs for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/genetics
- Antibodies, Monoclonal, Humanized/immunology
- Antigens, Neoplasm/immunology
- Cell Line, Tumor
- Cell Proliferation
- Cell Transformation, Neoplastic
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Humans
- Immunotherapy/methods
- Mice
- Models, Molecular
- Protein Conformation
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Single-Chain Antibodies/genetics
- Single-Chain Antibodies/immunology
- Streptococcus/immunology
- Superantigens/genetics
- Superantigens/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Kelcey G. Patterson
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | | | - Peter S. Bastedo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - David A. Hess
- Department of Physiology and Pharmacology, Western University, London Ontario, Canada
- Vascular Biology Research Group, Robarts Research Institute, London, Ontario, Canada
| | - S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| | - John K. McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
37
|
Fortin JS, Genève L, Gauthier C, Shoukry NH, Azar GA, Younes S, Yassine-Diab B, Sékaly RP, Fremont DH, Thibodeau J. MMTV superantigens coerce an unconventional topology between the TCR and MHC class II. THE JOURNAL OF IMMUNOLOGY 2014; 192:1896-906. [PMID: 24453254 DOI: 10.4049/jimmunol.1203130] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mouse mammary tumor virus superantigens (vSAGs) are notorious for defying structural characterization, and a consensus has yet to be reached regarding their ability to bridge the TCR to MHC class II (MHCII). In this study, we determined the topology of the T cell signaling complex by examining the respective relation of vSAG7 with the MHCII molecule, MHCII-associated peptide, and TCR. We used covalently linked peptide/MHCII complexes to demonstrate that vSAG presentation is tolerant to variation in the protruding side chains of the peptide, but can be sensitive to the nature of the protruding N-terminal extension. An original approach in which vSAG was covalently linked to either MHCII chain confirmed that vSAG binds outside the peptide binding groove. Also, whereas the C-terminal vSAG segment binds to the MHCII α-chain in a conformation-sensitive manner, the membrane-proximal N-terminal domain binds the β-chain. Because both moieties of the mature vSAG remain noncovalently associated after processing, our results suggest that vSAG crosslinks MHCII molecules. Comparing different T cell hybridomas, we identified key residues on the MHCII α-chain that are differentially recognized by the CDR3β when engaged by vSAG. Finally, we show that the highly conserved tyrosine residue found in the vSAg TGXY motif is required for T cell activation. Our results reveal a novel SAG/MHCII/TCR architecture in which vSAGs coerce a near-canonical docking between MHCII and TCR that allows eschewing of traditional CDR3 binding with the associated peptide in favor of MHCII α-chain binding. Our findings highlight the plasticity of the TCR CDRs.
Collapse
Affiliation(s)
- Jean-Simon Fortin
- Laboratoire d'Immunologie Moléculaire, Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec HC3 3J7, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Sharma P, Wang N, Kranz DM. Soluble T cell receptor Vβ domains engineered for high-affinity binding to staphylococcal or streptococcal superantigens. Toxins (Basel) 2014; 6:556-74. [PMID: 24476714 PMCID: PMC3942751 DOI: 10.3390/toxins6020556] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 11/29/2022] Open
Abstract
Staphylococcus aureus and group A Streptococcus secrete a collection of toxins called superantigens (SAgs), so-called because they stimulate a large fraction of an individual’s T cells. One consequence of this hyperactivity is massive cytokine release leading to severe tissue inflammation and, in some cases, systemic organ failure and death. The molecular basis of action involves the binding of the SAg to both a T cell receptor (TCR) on a T cell and a class II product of the major histocompatibility complex (MHC) on an antigen presenting cell. This cross-linking leads to aggregation of the TCR complex and signaling. A common feature of SAgs is that they bind with relatively low affinity to the variable region (V) of the beta chain of the TCR. Despite this low affinity binding, SAgs are very potent, as each T cell requires only a small fraction of their receptors to be bound in order to trigger cytokine release. To develop high-affinity agents that could neutralize the activity of SAgs, and facilitate the development of detection assays, soluble forms of the Vβ regions have been engineered to affinities that are up to 3 million-fold higher for the SAg. Over the past decade, six different Vβ regions against SAgs from S. aureus (SEA, SEB, SEC3, TSST-1) or S. pyogenes (SpeA and SpeC) have been engineered for high-affinity using yeast display and directed evolution. Here we review the engineering of these high-affinity Vβ proteins, structural features of the six different SAgs and the Vβ proteins, and the specific properties of the engineered Vβ regions that confer high-affinity and specificity for their SAg ligands.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA.
| | - Ningyan Wang
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA.
| | - David M Kranz
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA.
| |
Collapse
|
39
|
Abstract
SUMMARY This review begins with a discussion of the large family of Staphylococcus aureus and beta-hemolytic streptococcal pyrogenic toxin T lymphocyte superantigens from structural and immunobiological perspectives. With this as background, the review then discusses the major known and possible human disease associations with superantigens, including associations with toxic shock syndromes, atopic dermatitis, pneumonia, infective endocarditis, and autoimmune sequelae to streptococcal illnesses. Finally, the review addresses current and possible novel strategies to prevent superantigen production and passive and active immunization strategies.
Collapse
|
40
|
Mechanisms of staphylococcal enterotoxin-induced emesis. Eur J Pharmacol 2014; 722:95-107. [DOI: 10.1016/j.ejphar.2013.08.050] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 06/11/2013] [Accepted: 08/03/2013] [Indexed: 01/16/2023]
|
41
|
Spaulding AR, Salgado-Pabón W, Merriman JA, Stach CS, Ji Y, Gillman AN, Peterson ML, Schlievert PM. Vaccination against Staphylococcus aureus pneumonia. J Infect Dis 2013; 209:1955-62. [PMID: 24357631 DOI: 10.1093/infdis/jit823] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Staphylococcus aureus causes serious infections in both hospital and community settings. Attempts have been made to prevent human infection through vaccination against bacterial cell-surface antigens; thus far all have failed. Here we show that superantigens and cytolysins, when used in vaccine cocktails, provide protection from S. aureus USA100-USA400 intrapulmonary challenge. METHODS Rabbits were actively vaccinated (wild-type toxins or toxoids) or passively immunized (hyperimmune serum) against combinations of superantigens (toxic shock syndrome toxin 1, enterotoxins B and C, and enterotoxin-like X) and cytolysins (α-, β-, and γ-toxins) and challenged intrapulmonarily with multiple strains of S. aureus, both methicillin-sensitive and methicillin-resistant. RESULTS Active vaccination against a cocktail containing bacterial cell-surface antigens enhanced disease severity as tested by infective endocarditis. Active vaccination against secreted superantigens and cytolysins resulted in protection of 86 of 88 rabbits when challenged intrapulmonarily with 9 different S. aureus strains, compared to only 1 of 88 nonvaccinated animals. Passive immunization studies demonstrated that production of neutralizing antibodies was an important mechanism of protection. CONCLUSIONS The data suggest that vaccination against bacterial cell-surface antigens increases disease severity, but vaccination against secreted virulence factors provides protection against S. aureus. These results advance our understanding of S. aureus pathogenesis and have important implications in disease prevention.
Collapse
Affiliation(s)
- Adam R Spaulding
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City
| | - Wilmara Salgado-Pabón
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City
| | - Joseph A Merriman
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City
| | - Christopher S Stach
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City
| | - Yinduo Ji
- Department of Veterinary Biosciences, College of Veterinary Medicine
| | - Aaron N Gillman
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis
| | - Marnie L Peterson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis
| | - Patrick M Schlievert
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
42
|
Dembic Z. On recognizing 'shades-of-gray' (self-nonself discrimination) or 'colour' (Integrity model) by the immune system. Scand J Immunol 2013; 78:325-38. [PMID: 23819602 DOI: 10.1111/sji.12090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 06/27/2013] [Indexed: 01/31/2023]
Abstract
The aim is to discuss Cohn's T-cell receptor (TCR) Tritope model of recognition, propose a novel suggestion for prior-to-positive selection of thymocytes contributing to inherent major histocompatibility complex (MHC) reactivity of a T-cell repertoire and clarify the Integrity model about the function of the immune system. If we compare the perception of light with the recognition of nonself, we could imagine that the opacity might be a measure of docking interaction between specific receptors for antigen on T or B cells (TCR/peptide-MHC or BCR/antigen). From this viewpoint, the self-nonself discrimination (S-NS) metaphor would be perception of black (self) versus white (nonself). However, whereas detection of shades-of-gray suffices to describe S-NS discrimination principle, colour vision of the antigenic world portrays best the Integrity model. In concert with recognition of opacity, the Integrity model proposes detection of at least three colours (signals): red (harmful), blue (useful) and yellow (the rest, including homoeostatic ones). As a result, recognition of nonself is transferred into communication within self while deciding on type of the immune response. Hence, the S-NS discrimination model seems to be an oversimplification, because it fails to see colours and consequently lacks the need for suppressor/regulatory function. Similarly, the Danger model stops short of detecting being useful signals that confer immune asylum to helpful micro-organisms like commensals. I suggest that the immune system's repertoire for recognition, in general, has evolved by a novel drive called 'natural integrity' alongside natural selection, thus facilitating communication between cells of the immune system.
Collapse
Affiliation(s)
- Z Dembic
- Molecular Genetics Laboratory, Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
43
|
Sharma P, Postel S, Sundberg EJ, Kranz DM. Characterization of the Staphylococcal enterotoxin A: Vβ receptor interaction using human receptor fragments engineered for high affinity. Protein Eng Des Sel 2013; 26:781-9. [PMID: 24167300 DOI: 10.1093/protein/gzt054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Staphylococcal food poisoning is a gastrointestinal disorder caused by the consumption of food containing Staphylococcal enterotoxins. Staphylococcal enterotoxin A (SEA) is the most common enterotoxin recovered from food poisoning outbreaks in the USA. In addition to its enteric activity, SEA also acts as a potent superantigen through stimulation of T cells, although less is known about its interactions than the superantigens SEB, SEC and toxic shock syndrome toxin-1. To understand more about SEA:receptor interactions, and to develop toxin-detection systems for use in food testing, we engineered various SEA-binding receptor mutants. The extracellular domain of the receptor, a variable region of the beta chain (Vβ22) of the T-cell receptor, was engineered for stability as a soluble protein and for high affinity, using yeast-display technology. The highest affinity mutant was shown to bind SEA with a Kd value of 4 nM. This was a 25 000-fold improvement in affinity compared with the wild-type receptor, which bound to SEA with low affinity (Kd value of 100 µM), similar to other superantigen:Vβ interactions. The SEA:Vβ interface was centered around residues within the complementarity determining region 2 loop. The engineered receptor was specific for SEA, in that it did not bind to two other closely related enterotoxins SEE or SED, providing information on the SEA residues possibly involved in the interaction. The specificity and affinity of these high-affinity Vβ proteins also provide useful agents for the design of more sensitive and specific systems for SEA detection.
Collapse
Affiliation(s)
- P Sharma
- Department of Biochemistry, University of Illinois, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
44
|
Hedlund G, Eriksson H, Sundstedt A, Forsberg G, Jakobsen BK, Pumphrey N, Rödström K, Lindkvist-Petersson K, Björk P. The tumor targeted superantigen ABR-217620 selectively engages TRBV7-9 and exploits TCR-pMHC affinity mimicry in mediating T cell cytotoxicity. PLoS One 2013; 8:e79082. [PMID: 24194959 PMCID: PMC3806850 DOI: 10.1371/journal.pone.0079082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 09/26/2013] [Indexed: 01/20/2023] Open
Abstract
The T lymphocytes are the most important effector cells in immunotherapy of cancer. The conceptual objective for developing the tumor targeted superantigen (TTS) ABR-217620 (naptumomab estafenatox, 5T4Fab-SEA/E-120), now in phase 3 studies for advanced renal cell cancer, was to selectively coat tumor cells with cytotoxic T lymphocytes (CTL) target structures functionally similar to natural CTL pMHC target molecules. Here we present data showing that the molecular basis for the anti-tumor activity by ABR-217620 resides in the distinct interaction between the T cell receptor β variable (TRBV) 7-9 and the engineered superantigen (Sag) SEA/E-120 in the fusion protein bound to the 5T4 antigen on tumor cells. Multimeric but not monomeric ABR-217620 selectively stains TRBV7-9 expressing T lymphocytes from human peripheral blood similar to antigen specific staining of T cells with pMHC tetramers. SEA/E-120 selectively activates TRBV7-9 expressing T lymphocytes resulting in expansion of the subset. ABR-217620 selectively triggers TRBV7-9 expressing cytotoxic T lymphocytes to kill 5T4 positive tumor cells. Furthermore, ABR-217620 activates TRBV7-9 expressing T cell line cells in the presence of cell- and bead-bound 5T4 tumor antigen. Surface plasmon resonance analysis revealed that ABR-217620 binds to 5T4 with high affinity, to TRBV7-9 with low affinity and to MHC class II with very low affinity. The T lymphocyte engagement by ABR-217620 is constituted by displaying high affinity binding to the tumor cells (KD approximately 1 nM) and with the mimicry of natural productive immune TCR-pMHC contact using affinities of around 1 µM. This difference in kinetics between the two components of the ABR-217620 fusion protein will bias the binding towards the 5T4 target antigen, efficiently activating T-cells via SEA/E-120 only when presented by the tumor cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Karin Rödström
- Experimental Medical Science, Lund University, Lund, Sweden
| | | | | |
Collapse
|
45
|
Assessment of the functional regions of the superantigen staphylococcal enterotoxin B. Toxins (Basel) 2013; 5:1859-71. [PMID: 24152989 PMCID: PMC3813916 DOI: 10.3390/toxins5101859] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 11/17/2022] Open
Abstract
The functional activity of superantigens is based on capacity of these microbial proteins to bind to both the β-chain of the T cell receptor (TcR) and the major histocompatibility complex (MHC) class II dimer. We have previously shown that a subset of the bacterial superantigens also binds to a membrane protein, designated p85, which is expressed by renal epithelial cells. This binding activity is a property of SEB, SEC1, 2 and 3, but not SEA, SED, SEE or TSST. The crystal structure of the tri-molecular complex of the superantigen staphylococcal enterotoxin B (SEB) with both the TcR and class II has previously been reported. However, the relative contributions of regions of the superantigen to the overall functional activity of this superantigen remain undefined. In an effort to better define the molecular basis for the interaction of SEB with the TcR β-chain, we report studies here which show the comparative contributions of amino- and carboxy-terminal regions in the superantigen activity of SEB. Recombinant fusion proteins composed of bacterial maltose-binding protein linked to either full-length or truncated toxins in which the 81 N-terminal, or 19 or 34 C-terminal amino acids were deleted, were generated for these studies. This approach provides a determination of the relative strength of the functional activity of the various regions of the superantigen protein.
Collapse
|
46
|
Krakauer T. Update on staphylococcal superantigen-induced signaling pathways and therapeutic interventions. Toxins (Basel) 2013; 5:1629-54. [PMID: 24064719 PMCID: PMC3798877 DOI: 10.3390/toxins5091629] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/20/2022] Open
Abstract
Staphylococcal enterotoxin B (SEB) and related bacterial toxins cause diseases in humans and laboratory animals ranging from food poisoning, acute lung injury to toxic shock. These superantigens bind directly to the major histocompatibility complex class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in rapid hyper-activation of the host immune system. In addition to TCR and co-stimulatory signals, proinflammatory mediators activate signaling pathways culminating in cell-stress response, activation of NFκB and mammalian target of rapamycin (mTOR). This article presents a concise review of superantigen-activated signaling pathways and focuses on the therapeutic challenges against bacterial superantigens.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology, Integrated Toxicology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702 5011, USA.
| |
Collapse
|
47
|
Ganem MB, De Marzi MC, Fernández-Lynch MJ, Jancic C, Vermeulen M, Geffner J, Mariuzza RA, Fernández MM, Malchiodi EL. Uptake and intracellular trafficking of superantigens in dendritic cells. PLoS One 2013; 8:e66244. [PMID: 23799083 PMCID: PMC3682983 DOI: 10.1371/journal.pone.0066244] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 05/07/2013] [Indexed: 11/19/2022] Open
Abstract
Bacterial superantigens (SAgs) are exotoxins produced mainly by Staphylococcus aureus and Streptococcus pyogenes that can cause toxic shock syndrome (TSS). According to current paradigm, SAgs interact directly and simultaneously with T cell receptor (TCR) on the T cell and MHC class II (MHC-II) on the antigen-presenting cell (APC), thereby circumventing intracellular processing to trigger T cell activation. Dendritic cells (DCs) are professional APCs that coat nearly all body surfaces and are the most probable candidate to interact with SAgs. We demonstrate that SAgs are taken up by mouse DCs without triggering DC maturation. SAgs were found in intracellular acidic compartment of DCs as biologically active molecules. Moreover, SAgs co-localized with EEA1, RAB-7 and LAMP-2, at different times, and were then recycled to the cell membrane. DCs loaded with SAgs are capable of triggering in vitro lymphocyte proliferation and, injected into mice, stimulate T cells bearing the proper TCR in draining lymph nodes. Transportation and trafficking of SAgs in DCs might increase the local concentration of these exotoxins where they will produce the highest effect by promoting their encounter with both MHC-II and TCR in lymph nodes, and may explain how just a few SAg molecules can induce the severe pathology associated with TSS.
Collapse
Affiliation(s)
- María B. Ganem
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-UBA, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mauricio C. De Marzi
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-UBA, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Buenos Aires, Argentina
| | - María J. Fernández-Lynch
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-UBA, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Carolina Jancic
- Departamento de Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Mónica Vermeulen
- Departamento de Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Jorge Geffner
- Departamento de Inmunología, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Roy A. Mariuzza
- University of Maryland Institute for Bioscience and Biotechnology Research, W. M. Keck Laboratory for Structural Biology, Rockville, Maryland, United States of America
| | - Marisa M. Fernández
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-UBA, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Emilio L. Malchiodi
- Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), CONICET-UBA, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
48
|
Gu L, Yue J, Zheng Y, Zheng X, Wang J, Wang Y, Li J, Jiang Y, Jiang H. Evaluation of a recombinant double mutant of staphylococcal enterotoxin B (SEB-H32Q/K173E) with enhanced antitumor activity effects and decreased pyrexia. PLoS One 2013; 8:e55892. [PMID: 23405232 PMCID: PMC3566101 DOI: 10.1371/journal.pone.0055892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 01/07/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Immunotherapy has been used to improve patient immune function, inhibit tumor growth and has become a highly promising method of cancer treatment. Highly agglutinative staphylococcin (HAS), a mixture of Staphylococcus aureus culture filtrates, which include staphylococcal enterotoxin (SE) C as the active ingredient, has been used clinically as an immunomodifier in the treatment of a number of tumors for many years. However, the use of HAS has been associated with some unavoidable side-effects such as fever. Previous studies have shown that SEB stimulates a more potent activation of T lymphocytes than SEC3, and mutations of the histidine residues eliminated the toxicity of SEB. SE mutants with decreased side-effects and/or more potent antitumor activities are required. METHODOLOGY/PRINCIPAL FINDINGS We built a structural model of the MHC II-SEB-TCR complex and found that a mutation of SEB at Lys173 might decrease the repulsion force between the SEB-TCR, which would facilitate their interaction. From the above results, we designed SEB-H32Q/K173E (mSEB). Analysis of in vitro stimulation of the proliferation of human peripheral blood mononuclear cells (PBMCs), IFN-γ secretion and inhibition of the growth of various tumor cell lines demonstrated that mSEB exhibited higher antitumor activity compared with wild-type SEB (wtSEB). Notably, mSEB inhibited the growth of various tumors at an extremely low concentration with little cytotoxicity against normal cells. Three animal tumor models (C57BL/6 mouse, New Zealand rabbit and a humanized NOD/SCID mouse) were used to evaluate the in vivo immunotherapeutic effects. Compared with wtSEB, mSEB significantly enhanced antitumor effect in more than one animal model with reduced pyrexia toxicity and prolonged the survival of tumor-bearing mice. CONCLUSIONS/SIGNIFICANCE Our results suggest that SEB-H32Q/K173E retains superantigen (SAg) characteristics and enhances the host immune response to neoplastic diseases while reducing associated pyrogenic toxicity.
Collapse
MESH Headings
- Animals
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Enterotoxins/genetics
- Enterotoxins/immunology
- Enterotoxins/metabolism
- Female
- Fever/genetics
- Fever/immunology
- Fever/therapy
- Genes, MHC Class II/genetics
- Genes, MHC Class II/immunology
- Humans
- Immunotherapy
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, SCID
- Mutant Proteins/therapeutic use
- Mutation/genetics
- Rabbits
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Proteins/therapeutic use
- Staphylococcus aureus/metabolism
- Survival Rate
Collapse
Affiliation(s)
- Liwei Gu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Traditional Chinese Medicine Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Junjie Yue
- Beijing Institute of Biotechnology, Beijing, China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Xin Zheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Jun Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yanzi Wang
- Department of Pharmacy, Jiangsu Provincial Xuzhou Pharmaceutical Vocational College, Xuzhou, China
| | - Jianchun Li
- Department of Traditional Chinese Medicine Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
- * E-mail: (JCL); (YQJ); (HJ)
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- * E-mail: (JCL); (YQJ); (HJ)
| | - Hua Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- * E-mail: (JCL); (YQJ); (HJ)
| |
Collapse
|
49
|
Krakauer T. PI3K/Akt/mTOR, a pathway less recognized for staphylococcal superantigen-induced toxicity. Toxins (Basel) 2012; 4:1343-66. [PMID: 23202320 PMCID: PMC3509712 DOI: 10.3390/toxins4111343] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 11/12/2012] [Accepted: 11/13/2012] [Indexed: 12/27/2022] Open
Abstract
Immunostimulating staphylococcal enterotoxin B (SEB) and related superantigenic toxins cause diseases in humans and laboratory animals by activating cells of the immune system. These toxins bind directly to the major histocompatibility complex (MHC) class II molecules on antigen-presenting cells and specific Vβ regions of T-cell receptors (TCR), resulting in hyperactivation of both T lymphocytes and monocytes/macrophages. Activated host cells produce excessive amounts of proinflammatory cytokines and chemokines, especially tumor necrosis factor α, interleukin 1 (IL-1), IL-2, interferon γ (IFNγ), and macrophage chemoattractant protein 1 causing clinical symptoms of fever, hypotension, and shock. The well-explored signal transduction pathways for SEB-induced toxicity downstream from TCR/MHC ligation and interaction of cell surface co-stimulatory molecules include the mitogen-activated protein kinase cascades and cytokine receptor signaling, culminating in NFκB activation. Independently, IL-2, IFNγ, and chemokines from activated T cells signal via the phosphoinositide 3-kinase (PI3K), the serine/threonine kinases, Akt and mammalian target of rapamycin (mTOR) pathways. This article reviews the signaling molecules induced by superantigens in the activation of PI3K/Akt/mTOR pathways leading to staphylococcal superantigen-induced toxicity and updates potential therapeutics against superantigens.
Collapse
Affiliation(s)
- Teresa Krakauer
- Department of Immunology, Integrated Toxicology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
50
|
Kohler PL, Greenwood SD, Nookala S, Kotb M, Kranz DM, Schlievert PM. Staphylococcus aureus isolates encode variant staphylococcal enterotoxin B proteins that are diverse in superantigenicity and lethality. PLoS One 2012; 7:e41157. [PMID: 22815951 PMCID: PMC3397982 DOI: 10.1371/journal.pone.0041157] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 06/18/2012] [Indexed: 11/30/2022] Open
Abstract
Staphylococcus aureus produces superantigens (SAgs) that bind and cross-link T cells and APCs, leading to activation and proliferation of immune cells. SAgs bind to variable regions of the β-chains of T cell receptors (Vβ-TCRs), and each SAg binds a unique subset of Vβ-TCRs. This binding leads to massive cytokine production and can result in toxic shock syndrome (TSS). The most abundantly produced staphylococcal SAgs and the most common causes of staphylococcal TSS are TSS toxin-1 (TSST-1), and staphylococcal enterotoxins B and C (SEB and SEC, respectively). There are several characterized variants of humans SECs, designated SEC1-4, but only one variant of SEB has been described. Sequencing the seb genes from over 20 S. aureus isolates show there are at least five different alleles of seb, encoding forms of SEB with predicted amino acid substitutions outside of the predicted immune-cell binding regions of the SAgs. Examination of purified, variant SEBs indicates that these amino acid substitutions cause differences in proliferation of rabbit splenocytes in vitro. Additionally, the SEBs varied in lethality in a rabbit model of TSS. The SEBs were diverse in their abilities to cause proliferation of human peripheral blood mononuclear cells, and differed in their activation of subsets of T cells. A soluble, high-affinity Vβ-TCR, designed to neutralize the previously characterized variant of SEB (SEB1), was able to neutralize the variant SEBs, indicating that this high-affinity peptide may be useful in treating a variety of SEB-mediated illnesses.
Collapse
Affiliation(s)
- Petra L. Kohler
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Seth D. Greenwood
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Suba Nookala
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati Medical School, Cincinnati, Ohio, United States of America
| | - Malak Kotb
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati Medical School, Cincinnati, Ohio, United States of America
| | - David M. Kranz
- Department of Biochemistry, School of Molecular and Cellular Biology, University of Illinois, Urbana, Illinois, United States of America
| | - Patrick M. Schlievert
- Department of Microbiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|