1
|
Jarneborn A, Hu Z, Deshmukh M, Kopparapu PK, Jin T. Tofacitinib Treatment Suppresses CD4+ T-Cell Activation and Th1 Response, Contributing to Protection against Staphylococcal Toxic Shock. Int J Mol Sci 2024; 25:7456. [PMID: 39000566 PMCID: PMC11242597 DOI: 10.3390/ijms25137456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Staphylococcal toxic shock syndrome (STSS) is a rare, yet potentially fatal disease caused by Staphylococcus aureus (S. aureus) enterotoxins, known as superantigens, which trigger an intense immune response. Our previous study demonstrated the protective effect of tofacitinib against murine toxin-induced shock and a beneficial effect against S. aureus sepsis. In the current study, we examined the effects of tofacitinib on T-cell response in peripheral blood using a mouse model of enterotoxin-induced shock. Our data revealed that tofacitinib suppresses the activation of both CD4+ and CD8+ T cells in peripheral blood. Furthermore, both gene and protein levels of Th1 cytokines were downregulated by tofacitinib treatment in mice with enterotoxin-induced shock. Importantly, we demonstrated that CD4+ cells, but not CD8+ cells, are pathogenic in mice with enterotoxin-induced shock. In conclusion, our findings suggest that tofacitinib treatment suppresses CD4+ T-cell activation and Th1 response, thereby aiding in protection against staphylococcal toxic shock in mice. This insight may guide the future development of novel therapies for STSS.
Collapse
Affiliation(s)
- Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | - Zhicheng Hu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Meghshree Deshmukh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Pradeep Kumar Kopparapu
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 413 46 Gothenburg, Sweden; (A.J.); (Z.H.); (M.D.); (P.K.K.)
- Department of Rheumatology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| |
Collapse
|
2
|
Shi Y, Strasser A, Green DR, Latz E, Mantovani A, Melino G. Legacy of the discovery of the T-cell receptor: 40 years of shaping basic immunology and translational work to develop novel therapies. Cell Mol Immunol 2024; 21:790-797. [PMID: 38822079 PMCID: PMC11214623 DOI: 10.1038/s41423-024-01168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 06/02/2024] Open
Affiliation(s)
- Yufang Shi
- The Fourth Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, 215000, China.
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Bonn, 53127, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, 53175, Germany
| | | | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
3
|
Huang Y, Ji Y, Zheng M, Li X, Luan Y, Liu Y, Zhang X. Double-Enhanced Photothermal Lateral Flow Biosensor Based on Dual Gold Nanoparticle Conjugates. ACS Sens 2024; 9:2815-2825. [PMID: 38758028 DOI: 10.1021/acssensors.3c02329] [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] [Indexed: 05/18/2024]
Abstract
Bacterial toxins emerge as the primary triggers of foodborne illnesses, posing a significant threat to human health. To ensure food safety, it is imperative to implement point-of-care testing methods. Lateral flow biosensors (LFBs) based on gold nanoparticles (GNPs) have been commonly used for rapid detection, but their applicationis limited by low sensitivity. Based on the localized surface plasmon resonance and photothermal effect of dual gold nanoparticle conjugates (DGNPs), we developed a smartphone-integrated photothermal LFB (PLFB) with double-enhanced colorimetric and photothermal sensitivity. Through numerical simulations, we verified that DGNPs have significantly enhanced photothermal performance compared to single 15 nm GNPs (SGNPs), and applied DGNPs in PLFB for the detection of staphylococcus enterotoxin A (SEA). The colorimetric and photothermal limits of detection of DGNPs-based PLFB for SEA were 0.091 and 0.0038 ng mL-1, respectively. Compared with the colorimetric detection of the SGNPs-based LFB, the colorimetric detection sensitivity of the DGNPs-based PLFB was increased by 10.7 times, and the photothermal detection sensitivity was further improved by 255.3 times. Moreover, the PLFB exhibits robust reproducibility and exceptional specificity and is applicable for detecting SEA in milk samples. This smartphone-integrated PLFB based on DGNPs allows users to detect toxins simply, conveniently, and quickly and has huge application potential in the field of food safety.
Collapse
Affiliation(s)
- Yan Huang
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yongxiang Ji
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Mengxin Zheng
- SINOMACH Academy of Science and Technology Co., Ltd., Beijing 100083, China
| | - Xiujuan Li
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yi Luan
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Yang Liu
- Beijing Key Laboratory for Bioengineering and Sensing Technology, School of Chemistry and Biological Engineering, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xueji Zhang
- School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
4
|
Tuffs SW, Dufresne K, Rishi A, Walton NR, McCormick JK. Novel insights into the immune response to bacterial T cell superantigens. Nat Rev Immunol 2024; 24:417-434. [PMID: 38225276 DOI: 10.1038/s41577-023-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2023] [Indexed: 01/17/2024]
Abstract
Bacterial T cell superantigens (SAgs) are a family of microbial exotoxins that function to activate large numbers of T cells simultaneously. SAgs activate T cells by direct binding and crosslinking of the lateral regions of MHC class II molecules on antigen-presenting cells with T cell receptors (TCRs) on T cells; these interactions alter the normal TCR-peptide-MHC class II architecture to activate T cells in a manner that is independent of the antigen specificity of the TCR. SAgs have well-recognized, central roles in human diseases such as toxic shock syndrome and scarlet fever through their quantitative effects on the T cell response; in addition, numerous other consequences of SAg-driven T cell activation are now being recognized, including direct roles in the pathogenesis of endocarditis, bloodstream infections, skin disease and pharyngitis. In this Review, we summarize the expanding family of bacterial SAgs and how these toxins can engage highly diverse adaptive immune receptors. We highlight recent findings regarding how SAg-driven manipulation of the adaptive immune response may operate in multiple human diseases, as well as contributing to the biology and life cycle of SAg-producing bacterial pathogens.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada
| | - Karine Dufresne
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Aanchal Rishi
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Nicholas R Walton
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - John K McCormick
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
5
|
Liu CG, Li DY, Gao X, Ma T, Zhang K, Liu DY. Examining the causal relationship between circulating immune cells and the susceptibility to osteomyelitis: A Mendelian randomization study. Int Immunopharmacol 2024; 131:111815. [PMID: 38492335 DOI: 10.1016/j.intimp.2024.111815] [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: 01/14/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Osteomyelitis is considered as a deleterious inflammatory condition affecting the bone, primarily attributed to pathogenic infection. However, the underlying factors predisposing individuals to osteomyelitis remain incompletely elucidated. The immune system plays a multifaceted role in the progression of this condition, yet previous observational studies and randomized controlled trials investigating the association between circulating immune cell counts and osteomyelitis have been constrained. In order to address this knowledge gap, we conducted a Mendelian randomization (MR) analysis to evaluate the impact of diverse immune cell counts on the risk of developing osteomyelitis. METHODS In our study, we utilized single nucleotide polymorphisms (SNPs) that have been strongly linked to circulating immune cells or specific lymphocyte subtypes, as identified in large-scale genome-wide association studies (GWAS). These SNPs served as instrumental variables (IVs) for our MR analysis. We employed a more relaxed clumping threshold to conduct MR analysis on several related lymphocyte subtypes. To estimate causal effects, we utilized the Wald ratio, as well as the random-effects inverse variance weighted (IVW) and weighted median (WM) methods. To enhance the credibility of our results, we performed F-statistic calculations and a series of sensitivity analyses. RESULTS Our findings revealed a significant correlation between the absolute count of circulating lymphocytes and the risk of osteomyelitis [odds ratio(OR) 1.20;95 % confidence interval (CI), 1.08-1.32;P = 0.0005]. Furthermore, we identified a causal relationship between the absolute count of CD8+ T cells and susceptibility to osteomyelitis (OR 1.16; 95 % CI, 1.04-1.30; P = 0.0098). Importantly, these findings remained robust across a wide range of sensitivity analyses. CONCLUSION Through our MR analysis, we have provided evidence supporting a causal relationship between genetic predisposition to higher circulating immune cell counts and an increased risk of osteomyelitis. Specifically, our findings highlight the association between elevated CD8+ T cell counts and a heightened susceptibility to osteomyelitis. These results offer valuable insights for the future exploration of immunotherapy approaches in the management of osteomyelitis.
Collapse
Affiliation(s)
- Chun-Gui Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dong-Yang Li
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xi Gao
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Teng Ma
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kun Zhang
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - De-Yin Liu
- Severe & Poly-trauma Division, Orthopedic Trauma Department, Hong-Hui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
6
|
Romero LC, Silva LP, Teixeira NB, de Camargo KV, Del Masso Pereira MA, Corrente JE, Pereira VC, Ribeiro de Souza da Cunha MDL. Staphylococcus capitis Bloodstream Isolates: Investigation of Clonal Relationship, Resistance Profile, Virulence and Biofilm Formation. Antibiotics (Basel) 2024; 13:147. [PMID: 38391533 PMCID: PMC10885910 DOI: 10.3390/antibiotics13020147] [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: 12/15/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024] Open
Abstract
Staphylococcus capitis has been recognized as a relevant opportunistic pathogen, particularly its persistence in neonatal ICUs around the world. Therefore, the aim of this study was to describe the epidemiological profile of clinical isolates of S. capitis and to characterize the factors involved in the persistence and pathogenesis of these strains isolated from blood cultures collected in a hospital in the interior of the state of São Paulo, Brazil. A total of 141 S. capitis strains were submitted to detection of the mecA gene and SCCmec typing by multiplex PCR. Genes involved in biofilm production and genes encoding enterotoxins and hemolysins were detected by conventional PCR. Biofilm formation was evaluated by the polystyrene plate adherence test and phenotypic resistance was investigated by the disk diffusion method. Finally, pulsed-field gel electrophoresis (PFGE) was used to analyze the clonal relationship between isolates. The mecA gene was detected in 99 (70.2%) isolates, with this percentage reaching 100% in the neonatal ICU. SCCmec type III was the most prevalent type, detected in 31 (31.3%) isolates and co-occurrence of SCCmec was also observed. In vitro biofilm formation was detected in 46 (32.6%) isolates but was not correlated with the presence of the ica operon genes. Furthermore, biofilm production in ICU isolates was favored by hyperosmotic conditions, which are common in ICUs because of the frequent parenteral nutrition. Analysis of the clonal relationship between the isolates investigated in the present study confirms a homogeneous profile of S. capitis and the persistence of clones that are prevalent in the neonatal ICU and disseminated across the hospital. This study highlights the adaptation of isolates to specific hospital environments and their high clonality.
Collapse
Affiliation(s)
- Letícia Calixto Romero
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-691, Brazil
| | - Lucas Porangaba Silva
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-691, Brazil
| | - Nathalia Bibiana Teixeira
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-691, Brazil
| | - Karen Vilegas de Camargo
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-691, Brazil
| | | | - José Eduardo Corrente
- Department of Biostatistics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-900, Brazil
| | - Valéria Cataneli Pereira
- Microbiology Laboratory, Universidade do Oeste Paulista (UNOESTE), Presidente Prudente 18618-970, Brazil
| | | |
Collapse
|
7
|
Wang P, Fredj Z, Zhang H, Rong G, Bian S, Sawan M. Blocking Superantigen-Mediated Diseases: Challenges and Future Trends. J Immunol Res 2024; 2024:2313062. [PMID: 38268531 PMCID: PMC10807946 DOI: 10.1155/2024/2313062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/15/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024] Open
Abstract
Superantigens are virulence factors secreted by microorganisms that can cause various immune diseases, such as overactivating the immune system, resulting in cytokine storms, rheumatoid arthritis, and multiple sclerosis. Some studies have demonstrated that superantigens do not require intracellular processing and instated bind as intact proteins to the antigen-binding groove of major histocompatibility complex II on antigen-presenting cells, resulting in the activation of T cells with different T-cell receptor Vβ and subsequent overstimulation. To combat superantigen-mediated diseases, researchers have employed different approaches, such as antibodies and simulated peptides. However, due to the complex nature of superantigens, these approaches have not been entirely successful in achieving optimal therapeutic outcomes. CD28 interacts with members of the B7 molecule family to activate T cells. Its mimicking peptide has been suggested as a potential candidate to block superantigens, but it can lead to reduced T-cell activity while increasing the host's infection risk. Thus, this review focuses on the use of drug delivery methods to accurately target and block superantigens, while reducing the adverse effects associated with CD28 mimic peptides. We believe that this method has the potential to provide an effective and safe therapeutic strategy for superantigen-mediated diseases.
Collapse
Affiliation(s)
- Pengbo Wang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Zina Fredj
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Hongyong Zhang
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Guoguang Rong
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Sumin Bian
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| | - Mohamad Sawan
- CenBRAIN Neurotech, School of Engineering, Westlake University, Hangzhou 310030, China
| |
Collapse
|
8
|
Schoergenhofer C, Gelbenegger G, Hasanacevic D, Schöner L, Steiner MM, Firbas C, Buchtele N, Derhaschnig U, Tanzmann A, Model N, Larcher-Senn J, Drost M, Eibl MM, Roetzer A, Jilma B. A randomized, double-blind study on the safety and immunogenicity of rTSST-1 variant vaccine: phase 2 results. EClinicalMedicine 2024; 67:102404. [PMID: 38274114 PMCID: PMC10808908 DOI: 10.1016/j.eclinm.2023.102404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Background Toxic shock syndrome toxin-1 (TSST-1) is a superantigen produced by Staphylococcus aureus that causes the life-threatening toxic shock syndrome. The development of a safe and immunogenic vaccine against TSST-1 remains an unmet medical need. We investigated the safety, tolerability and immunogenicity of a recombinant TSST-1 variant vaccine (rTSST-1v) after 1-3 injections in healthy volunteers. Methods In this randomised, double-blind, adjuvant-controlled, parallel-group, phase 2 trial, healthy adults aged 18-64 were randomly allocated to undergo 1-3 injections of either 10 or 100 μg rTSST-1v or Al(OH)3. The primary endpoint was safety and tolerability of rTSST-1v in the intention-to-treat population. The per-protocol population was used for the immunogenicity analysis. The trial is registered with EudraCT#: 2015-003714-24; ClinicalTrials.gov#: NCT02814708. Findings Between April and November 2017,140 subjects were enrolled and 126 completed the trial. rTSST-1v showed a good safety and tolerability profile. A total of 855 systemic adverse events occurred, 280 of which were suspected related adverse events, without dose dependency. Two participants were discontinued early because of allergic reactions. Seroconversion occurred in >81% of subjects within 3 months of the first immunisation which was sustained until 18 months after the third immunisation in over 70% of subjects in the pooled low-dose group and in over 85% in the pooled high-dose group. Interpretation rTSST-1v in cumulative doses of up to 300 μg was safe, well-tolerated and highly immunogenic. Two immunisations with 100 μg rTSST-1v provided the most persistent immune response and may be evaluated in future trials. Funding Biomedizinische Forschung & Bio-Produkte AG funded this study.
Collapse
Affiliation(s)
| | - Georg Gelbenegger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Léa Schöner
- Biomedizinische Forschung & Bio-Produkte AG, Vienna, Austria
| | - Margarete M. Steiner
- Division of General Anesthesia and Intensive Care Medicine, Department of Anesthesia, Critical Care and Pain Medicine, Medical University of Vienna, Vienna, Austria
| | - Christa Firbas
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Nina Buchtele
- Division of Medicine I, Intensive Care Unit 13i2, Medical University of Vienna, Vienna, Austria
| | - Ulla Derhaschnig
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Andreas Tanzmann
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Nina Model
- Biomedizinische Forschung & Bio-Produkte AG, Vienna, Austria
| | | | - Manuel Drost
- Assign Data Management and Biostatistics GmbH, Innsbruck, Austria
| | - Martha M. Eibl
- Biomedizinische Forschung & Bio-Produkte AG, Vienna, Austria
| | - Andreas Roetzer
- Biomedizinische Forschung & Bio-Produkte AG, Vienna, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
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
|
10
|
Wan Y, Yang L, Li Q, Wang X, Zhou T, Chen D, Li L, Wang Y, Wang X. Stability and emetic activity of enterotoxin like X (SElX) with high carrier rate of food poisoning Staphylococcus aureus. Int J Food Microbiol 2023; 404:110352. [PMID: 37549593 DOI: 10.1016/j.ijfoodmicro.2023.110352] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/24/2023] [Accepted: 07/30/2023] [Indexed: 08/09/2023]
Abstract
In order to analyze and clarify the thermal stability of food poisoning Staphylococcus aureus (S. aureus) enterotoxin-like X (SElX) and the biological characteristics of digestive enzymes, and to evaluate the risk of S. aureus carrying selx gene in food poisoning, the selx gene carrying rates of 165 strains isolated from 95 food poisoning events from 2006 to 2019 were first statistically analyzed. Subsequently, the purified recombinant SElX protein was digested and heated, and the superantigen activity was verified with mouse spleen cells and peripheral blood mononuclear cells of kittens. At the same time, the emetic activity and toxicity of SElX were evaluated using the kitten vomiting animal model, mice toxin model and in vitro cell models. The results showed the selx gene carrying rate of 165 food poisoning S. aureus strains was 90.30 %. SElX had significant resistance to heat treatment and pepsin digestion (pH = 4.0 and pH = 4.5), and had good superantigen activity and emetic activity. However, there is no significant lethal effect on mice and no significant toxicity to cells. Importantly, we found that SElX had an inhibitory effect on acidic mucus of goblet cells in various segments of the small intestine. The present study investigated the stability of SElX, and confirmed the emetic activity of SElX by establishing a kitten vomiting model for the first time, suggesting that SElX is a high risk toxin of food poisoning, which will provide new ideas for the prevention and control of S. aureus food poisoning.
Collapse
Affiliation(s)
- Yangli Wan
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Liu Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Qianhong Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaowen Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ting Zhou
- College of Veterinary Medicine, Northwest A&F University, China
| | - Dishi Chen
- Sichuan Animal Disease Prevention and Control Center, Chengdu 610041, China
| | - Li Li
- Sichuan Animal Disease Prevention and Control Center, Chengdu 610041, China
| | - Yeru Wang
- Risk Assessment Division China National Center for Food Safety Risk Assessment, Chaoyang District, Beijing, China.
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
11
|
Turk S, Yanpar H, Baesmat AS, Canli SD, Cinar OE, Malkan UY, Turk C, Haznedaroglu IC, Ucar G. Enterotoxins A and B produced by Staphylococcus aureus increase cell proliferation, invasion and cytarabine resistance in acute myeloid leukemia cell lines. Heliyon 2023; 9:e19743. [PMID: 37810000 PMCID: PMC10559070 DOI: 10.1016/j.heliyon.2023.e19743] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023] Open
Abstract
As in the case of cancer, the risk of infection increases when the host's immune system is not working properly. It has been shown that toxins produced by the bacteria responsible for bacterial infections can alter the properties of cancer cells as well as their sensitivity to chemotherapy agents. Staphylococcus aureus (S. aureus) is one of the most prevalent pathogens in acute myeloid leukemia (AML) patients and it produces several virulence factors, including Staphylococcal enterotoxin A (SEA) and Staphylococcal enterotoxin B (SEB). Cytotoxicity, transwell migration, invasion assays, and various transcriptomic and gene set enrichment (GSE) analyses were used to determine how SEA and SEB alter cell proliferation, migration, invasion, and Cytarabine (Cyt) resistance in AML cell lines. The treatment of AML cell lines with SEA/SEB caused an increase in cell proliferation and Cyt resistance. Toxins enhanced the proclivity of cells to migrate and invade, with around 50% of cells in the presence of SEA and SEB. Transcriptomic and gene set enrichment analyses, and subsequent PCR validations showed dysregulation of immune related genes and genesets. Apparently, this allows AML cells to escape and survive the undesirable environment created by toxins, possibly via the ER stress signaling pathway. Therefore, SEA and SEB can significantly alter the characteristics of AML cancer cells and evaluation of alterations in responsible immune genes and pathways may be crucial for controlling the progression of cancer. In addition, our results suggest that there may be a strong interaction between the immune related pathways and the ER signaling pathway.
Collapse
Affiliation(s)
- Seyhan Turk
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Hatice Yanpar
- DS Nano and Biotechnology Product Tracing and Tracking Co., Ankara, Turkey
| | - Ayriana Safari Baesmat
- Department of Medical Microbiology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Secil Demirkol Canli
- Molecular Pathology Application and Research Center, Hacettepe University, Ankara, Turkey
- Tumor Pathology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Olgu Erkin Cinar
- Department of Hematology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Umit Yavuz Malkan
- Department of Hematology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Can Turk
- Department of Medical Microbiology, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | | | - Gulberk Ucar
- Department of Biochemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
12
|
Kuwatani M, Sakamoto N. Promising Highly Targeted Therapies for Cholangiocarcinoma: A Review and Future Perspectives. Cancers (Basel) 2023; 15:3686. [PMID: 37509347 PMCID: PMC10378186 DOI: 10.3390/cancers15143686] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
To overcome the poor prognosis of cholangiocarcinoma (CCA), highly targeted therapies, such as antibody-drug conjugates (ADCs), photodynamic therapy (PDT) with/without systemic chemotherapy, and experimental photoimmunotherapy (PIT), have been developed. Three preclinical trials have investigated the use of ADCs targeting specific antigens, namely HER2, MUC1, and glypican-1 (GPC1), for CCA. Trastuzumab emtansine demonstrated higher antiproliferative activity in CCA cells expressing higher levels of HER2. Similarly, "staphylococcal enterotoxin A-MUC1 antibody" and "anti-GPC1 antibody-monomethyl auristatin F" conjugates showed anticancer activity. PDT is effective in areas where appropriate photosensitizers and light coexist. Its mechanism involves photosensitizer excitation and subsequent reactive oxygen species production in cancer cells upon irradiation. Hematoporphyrin derivatives, temoporfin, phthalocyanine-4, talaporfin, and chlorine e6 derivatives have mainly been used clinically and preclinically in bile duct cancer. Currently, new forms of photosensitizers with nanotechnology and novel irradiation catheters are being developed. PIT is the most novel anti-cancer therapy developed in 2011 that selectively kills targeted cancer cells using a unique photosensitizer called "IR700" conjugated with an antibody specific for cancer cells. PIT is currently in the early stages of development for identifying appropriate CCA cell targets and irradiation devices. Future human and artificial intelligence collaboration has potential for overcoming challenges related to identifying universal CCA cell targets. This could pave the way for highly targeted therapies for CCA, such as ADC, PDT, and PIT.
Collapse
Affiliation(s)
- Masaki Kuwatani
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, North 14, West 5, Kita-ku, Sapporo 060-8648, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, North 14, West 5, Kita-ku, Sapporo 060-8648, Japan
| |
Collapse
|
13
|
Karbalaeimahdi M, Farajnia S, Bargahi N, Ghadiri-Moghaddam F, Rasouli Jazi HR, Bakhtiari N, Ghasemali S, Zarghami N. The Role of Interferons in Long Covid Infection. J Interferon Cytokine Res 2023; 43:65-76. [PMID: 36795973 DOI: 10.1089/jir.2022.0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Although the new generation of vaccines and anti-COVID-19 treatment regimens facilitated the management of acute COVID-19 infections, concerns about post-COVID-19 syndrome or Long Covid are rising. This issue can increase the incidence and morbidity of diseases such as diabetes, and cardiovascular, and lung infections, especially among patients suffering from neurodegenerative disease, cardiac arrhythmias, and ischemia. There are numerous risk factors that cause COVID-19 patients to experience post-COVID-19 syndrome. Three potential causes attributed to this disorder include immune dysregulation, viral persistence, and autoimmunity. Interferons (IFNs) are crucial in all aspects of post-COVID-19 syndrome etiology. In this review, we discuss the critical and double-edged role of IFNs in post-COVID-19 syndrome and how innovative biomedical approaches that target IFNs can reduce the occurrence of Long Covid infection.
Collapse
Affiliation(s)
- Mohammad Karbalaeimahdi
- Department of Medical Biotechnology, School of Advanced Medical Sciences, Tabriz, Iran.,Biotechnology Research Center, Tabriz, Iran
| | - Safar Farajnia
- Biotechnology Research Center, Tabriz, Iran.,Drug Applied Research Center, Tabriz, Iran
| | | | - Farzaneh Ghadiri-Moghaddam
- Drug Applied Research Center, Tabriz, Iran.,Department of Biology, Faculty of Science, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | | | | | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Noli Truant S, Redolfi DM, Sarratea MB, Malchiodi EL, Fernández MM. Superantigens, a Paradox of the Immune Response. Toxins (Basel) 2022; 14:toxins14110800. [PMID: 36422975 PMCID: PMC9692936 DOI: 10.3390/toxins14110800] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/19/2022] Open
Abstract
Staphylococcal enterotoxins are a wide family of bacterial exotoxins with the capacity to activate as much as 20% of the host T cells, which is why they were called superantigens. Superantigens (SAgs) can cause multiple diseases in humans and cattle, ranging from mild to life-threatening infections. Almost all S. aureus isolates encode at least one of these toxins, though there is no complete knowledge about how their production is triggered. One of the main problems with the available evidence for these toxins is that most studies have been conducted with a few superantigens; however, the resulting characteristics are attributed to the whole group. Although these toxins share homology and a two-domain structure organization, the similarity ratio varies from 20 to 89% among different SAgs, implying wide heterogeneity. Furthermore, every attempt to structurally classify these proteins has failed to answer differential biological functionalities. Taking these concerns into account, it might not be appropriate to extrapolate all the information that is currently available to every staphylococcal SAg. Here, we aimed to gather the available information about all staphylococcal SAgs, considering their functions and pathogenicity, their ability to interact with the immune system as well as their capacity to be used as immunotherapeutic agents, resembling the two faces of Dr. Jekyll and Mr. Hyde.
Collapse
|
15
|
Rani L, Singh J, Sharma A, Singh H, Verma I, Panda NK, Minz RW. Anti-staphylococcal responses and their relationship with HLA-DR-DQ polymorphism in granulomatosis with polyangiitis: a preliminary evidence of association with disease outcome. Clin Exp Med 2022:10.1007/s10238-022-00865-6. [PMID: 35881260 DOI: 10.1007/s10238-022-00865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/12/2022] [Indexed: 11/26/2022]
Abstract
Chronic nasal carriage of Staphylococcus aureus (S. aureus) is a risk factor for relapse of granulomatosis with polyangiitis (GPA), and genetic susceptibility to infections and autoimmune diseases is majorly affected by HLA genes. Previous studies have shown the association of HLA Class-II genes with GPA susceptibility. Here, we aim to assess immune responses of GPA patients against S. aureus antigens in relation to the HLA-DR-DQ genes polymorphism to determine the disease outcome. A total of 45 GPA patients and 128 healthy controls during 2010-2012 were included in this case-control study. HLA-DRB1/DQB1 allele typing was performed by polymerase chain reaction-sequence-specific primer (PCR-SSP) method. Immune responses against S. aureus antigens were investigated in 20 active vs. remitting GPA (after 6 months of cyclophosphamide and glucocorticoids) patients by Western blot. Statistical analysis was performed using χ2 test and Fisher's exact test. We observed a significant association of DRB1*08, DRB1*16 and DQB1*04 alleles with GPA susceptibility, whereas DRB1*15, DRB1*10 and DQB1*05 alleles were suggested as protective alleles. Among S. aureus antigens, active GPA patients' sera reacted more strongly with 34 and 24 kDa antigens of S. aureus than remitting and healthy control sera. Furthermore, we observed that the lack of DQB1*06 allele confers complete remission even in the presence of anti-S. aureus antibodies against 24 kDa protein. Our findings suggest that the presence of DQB1*06 allele and S. aureus infection may prolong active disease. Further, our study indicates the potential of using anti-staphylococcal medications for achieving remission in patients having HLA-DQB1*06 allele.
Collapse
Affiliation(s)
- Lekha Rani
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Jagdeep Singh
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Aman Sharma
- Department of Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Heera Singh
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Indu Verma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Naresh K Panda
- Department of Otolaryngology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ranjana W Minz
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
16
|
Zhu FC, Zeng H, Li JX, Wang B, Meng FY, Yang F, Gu J, Liang HY, Hu YM, Liu P, Peng LS, Hu XK, Zhuang Y, Fan M, Li HB, Tan ZM, Luo P, Zhang P, Chu K, Zhang JY, Zeng M, Zou QM. Evaluation of a recombinant five-antigen Staphylococcus aureus vaccine: The randomized, single-centre phase 1a/1b clinical trials. Vaccine 2022; 40:3216-3227. [DOI: 10.1016/j.vaccine.2022.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/10/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022]
|
17
|
Gan SKE, Derrick JP, Fraternali F. Editorial: Understanding and Engineering Antibody-Superantigen Interactions. Front Immunol 2022; 13:857339. [PMID: 35222446 PMCID: PMC8865624 DOI: 10.3389/fimmu.2022.857339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- Samuel Ken-En Gan
- Antibody & Product Development (APD) Lab, Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore.,APD SKEG Pte Ltd, Singapore, Singapore.,James Cook University, Singapore, Singapore
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Franca Fraternali
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom.,The Thomas Young Centre for Theory and Simulation of Materials, London, United Kingdom
| |
Collapse
|
18
|
Knopick P, Terman D, Riha N, Alvine T, Larson R, Badiou C, Lina G, Ballantyne J, Bradley D. Endogenous HLA-DQ8αβ programs superantigens (SEG/SEI) to silence toxicity and unleash a tumoricidal network with long-term melanoma survival. J Immunother Cancer 2021; 8:jitc-2020-001493. [PMID: 33109631 PMCID: PMC7592263 DOI: 10.1136/jitc-2020-001493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background As the most powerful T cell agonists known, superantigens (SAgs) have enormous potential for cancer immunotherapy. Their development has languished due to high incidence (60%–80%) of seroreactive neutralizing antibodies in humans and tumor necrosis factor-α (TNFα)-mediated cardiopulmonary toxicity. Such toxicity has narrowed their therapeutic index while neutralizing antibodies have nullified their therapeutic effects. Methods Female HLA-DQ8 (DQA*0301/DQB*0302) tg mice expressing the human major histocompatibility complex II (MHCII) HLA-DQ8 allele on a high proportion of PBL, spleen and lymph node cells were used. In the established tumor model, staphylococcal enterotoxin G and staphylococcal enterotoxin I (SEG/ SEI) (50 µg each) were injected on days 6 and 9 following tumor inoculation. Lymphoid, myeloid cells and tumor cell digests from tumor tissue were assayed using flow cytometry or quantitated using a cytometric bead array. Tumor density, necrotic and viable areas were quantitated using the ImageJ software. Results In a discovery-driven effort to address these problems we introduce a heretofore unrecognized binary complex comprizing SEG/SEI SAgs linked to the endogenous human MHCII HLA-DQ8 allele in humanized mice. By contrast to staphylococcal enterotoxin A (SEA) and staphylococcal enterotoxin B (SEB) deployed previously in clinical trials, SEG and SEI does not exhibit neutralizing antibodies in humans or TNFα-mediated toxicity in humanized HLA-DQ8 mice. In the latter model wherein SAg behavior is known to be ‘human-like’, SEG/SEI induced a powerful tumoricidal response and long-term survival against established melanoma in 82% of mice. Other SAgs deployed in the same model displayed toxic shock. Initially, HLA-DQ8 mediated melanoma antigen priming, after which SEG/SEI unleashed a broad CD4+ and CD8+ antitumor network marked by expansion of melanoma reactive T cells and interferon-γ (IFNy) in the tumor microenvironment (TME). SEG/SEI further initiated chemotactic recruitment of tumor reactive T cells to the TME converting the tumor from ‘cold’ to a ‘hot’. Long-term survivors displayed remarkable resistance to parental tumor rechallenge along with the appearance of tumor specific memory and tumor reactive T memory cells. Conclusions Collectively, these findings show for the first time that the SEG/SEI-(HLA-DQ8) empowers priming, expansion and recruitment of a population of tumor reactive T cells culminating in tumor specific memory and long-term survival devoid of toxicity. These properties distinguish SEG/SEI from other SAgs used previously in human tumor immunotherapy. Consolidation of these principles within the SEG/SEI-(HLA-DQ8) complex constitutes a conceptually new therapeutic weapon with compelling translational potential.
Collapse
Affiliation(s)
- Peter Knopick
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - David Terman
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Nathan Riha
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Travis Alvine
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Riley Larson
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| | - Cedric Badiou
- University of Lyon, Lyon, Auvergne-Rhône-Alpes, France
| | - Gerard Lina
- University of Lyon 1 University Institute of Tecnology Lyon 1, Villeurbanne, Auvergne-Rhône-Alpes, France
| | | | - David Bradley
- Biomedical Sciences, Universtiy of North Dakota School of Medicine, Grand Forks, North Dakota, USA
| |
Collapse
|
19
|
Hu DL, Li S, Fang R, Ono HK. Update on molecular diversity and multipathogenicity of staphylococcal superantigen toxins. ANIMAL DISEASES 2021. [DOI: 10.1186/s44149-021-00007-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AbstractStaphylococcal superantigen (SAg) toxins are the most notable virulence factors associated with Staphylococcus aureus, which is a pathogen associated with serious community and hospital acquired infections in humans and various diseases in animals. Recently, SAg toxins have become a superfamily with 29 types, including staphylococcal enterotoxins (SEs) with emetic activity, SE-like toxins (SEls) that do not induce emesis in primate models or have yet not been tested, and toxic shock syndrome toxin-1 (TSST-1). SEs and SEls can be subdivided into classical types (SEA to SEE) and novel types (SEG to SElY, SE01, SE02, SEl26 and SEl27). The genes of SAg toxins are located in diverse accessory genetic elements and share certain structural and biological properties. SAg toxins are heat-stable proteins that exhibit pyrogenicity, superantigenicity and capacity to induce lethal hypersensitivity to endotoxin in humans and animals. They have multiple pathogenicities that can interfere with normal immune function of host, increase the chances of survival and transmission of pathogenic bacteria in host, consequently contribute to the occurrence and development of various infections, persistent infections or food poisoning. This review focuses on the following aspects of SAg toxins: (1) superfamily members of classic and novelty discovered staphylococcal SAgs; (2) diversity of gene locations and molecular structural characteristics; (3) biological characteristics and activities; (4) multi-pathogenicity of SAgs in animal and human diseases, including bovine mastitis, swine sepsis, abscesses and skin edema in pig, arthritis and septicemia in poultry, and nosocomial infections and food-borne diseases in humans.
Collapse
|
20
|
Claeys E, Pauwels E, Humblet-Baron S, Provinciael B, Schols D, Waer M, Sprangers B, Vermeire K. Small Molecule Cyclotriazadisulfonamide Abrogates the Upregulation of the Human Receptors CD4 and 4-1BB and Suppresses In Vitro Activation and Proliferation of T Lymphocytes. Front Immunol 2021; 12:650731. [PMID: 33968048 PMCID: PMC8097030 DOI: 10.3389/fimmu.2021.650731] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/06/2021] [Indexed: 11/26/2022] Open
Abstract
The small molecule cyclotriazadisulfonamide (CADA) down-modulates the human CD4 receptor, an important factor in T cell activation. Here, we addressed the immunosuppressive potential of CADA using different activation models. CADA inhibited lymphocyte proliferation with low cellular toxicity in a mixed lymphocyte reaction, and when human PBMCs were stimulated with CD3/CD28 beads, phytohemagglutinin or anti-CD3 antibodies. The immunosuppressive effect of CADA involved both CD4+ and CD8+ T cells but was, surprisingly, most prominent in the CD8+ T cell subpopulation where it inhibited cell-mediated lympholysis. Immunosuppression by CADA was characterized by suppressed secretion of various cytokines, and reduced CD25, phosphoSTAT5 and CTPS-1 levels. We discovered a direct down-modulatory effect of CADA on 4-1BB (CD137) expression, a survival factor for activated CD8+ T cells. More specifically, CADA blocked 4‑1BB protein biosynthesis by inhibition of its co-translational translocation into the ER in a signal peptide-dependent way. Taken together, this study demonstrates that CADA, as potent down-modulator of human CD4 and 4‑1BB receptor, has promising immunomodulatory characteristics. This would open up new avenues toward chemotherapeutics that act as selective protein down-modulators to treat various human immunological disorders.
Collapse
Affiliation(s)
- Elisa Claeys
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Eva Pauwels
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Stephanie Humblet-Baron
- Department of Microbiology, Immunology and Transplantation, Laboratory of Adaptive Immunology, KU Leuven, Leuven, Belgium
| | - Becky Provinciael
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Dominique Schols
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Mark Waer
- Department of Microbiology, Immunology and Transplantation, Laboratory of Tracheal Transplantation, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, KU Leuven, Leuven, Belgium
| | - Kurt Vermeire
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| |
Collapse
|
21
|
Hashemzadeh MS, Tapeh BE, Mirhosseini SA. The Role of Bacterial Superantigens in the Immune Response: From Biology to Cancer Treatment. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666200812150402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Aims:
Encouraging results have been indicated preclinically and in patients using the
bacterial superantigen. This review article intends to summarize the role of the superantigens that
have been recently used in the treatment of cancer. In addition, the vector systems, including lentiviral
vectors, adeno-associated vector systems and retroviral vectors that are increasingly being
used in basic and applied research, were discussed. Most importantly, the new CRISPR technique
has also been discussed in this literature review.
Discussion:
More successful therapies can be achieved by manipulating bacterial vector systems
through incorporating genes related to the superantigens and cytokines. The products of SAg and
cytokine genes contribute to the strong stimulation of the immune system against tumor cells. They
bind to MHC II molecules as well as the V beta regions of TCR and lead to the production of IL2
and other cytokines, the activation of antigen-presenting cells and T lymphocytes. Additionally, superantigens
can be used to eradicate tumor cells. Better results in cancer treatment can be achieved
by transferring superantigen genes and subsequent strong immune stimulation along with other cancer
immunotherapy agents.
Conclusion:
Superantigens induce the proliferation of T lymphocytes and antigen-presenting cells
by binding to MHCII molecules and V beta regions in T cell receptors. Therefore, the presentation
of tumor cell antigens is increased. Additionally, the production of important cytokines by T cells
and APCs contributes to the stimulation of immune response against tumor cells. The manipulation
of bacterial vector systems through incorporating genesrelated to SAgs and other immune response
factors is a good strategy for the immune system stimulating and eradicating tumor cells along with
other immunotherapy agents.
Collapse
Affiliation(s)
- Mohammad S. Hashemzadeh
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behnam E.G. Tapeh
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed A. Mirhosseini
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Karauzum H, Venkatasubramaniam A, Adhikari RP, Kort T, Holtsberg FW, Mukherjee I, Mednikov M, Ortines R, Nguyen NTQ, Doan TMN, Diep BA, Lee JC, Aman MJ. IBT-V02: A Multicomponent Toxoid Vaccine Protects Against Primary and Secondary Skin Infections Caused by Staphylococcus aureus. Front Immunol 2021; 12:624310. [PMID: 33777005 PMCID: PMC7987673 DOI: 10.3389/fimmu.2021.624310] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus causes a wide range of diseases from skin infections to life threatening invasive diseases such as bacteremia, endocarditis, pneumonia, surgical site infections, and osteomyelitis. Skin infections such as furuncles, carbuncles, folliculitis, erysipelas, and cellulitis constitute a large majority of infections caused by S. aureus (SA). These infections cause significant morbidity, healthcare costs, and represent a breeding ground for antimicrobial resistance. Furthermore, skin infection with SA is a major risk factor for invasive disease. Here we describe the pre-clinical efficacy of a multicomponent toxoid vaccine (IBT-V02) for prevention of S. aureus acute skin infections and recurrence. IBT-V02 targets six SA toxins including the pore-forming toxins alpha hemolysin (Hla), Panton-Valentine leukocidin (PVL), leukocidin AB (LukAB), and the superantigens toxic shock syndrome toxin-1 and staphylococcal enterotoxins A and B. Immunization of mice and rabbits with IBT-V02 generated antibodies with strong neutralizing activity against toxins included in the vaccine, as well as cross-neutralizing activity against multiple related toxins, and protected against skin infections by several clinically relevant SA strains of USA100, USA300, and USA1000 clones. Efficacy of the vaccine was also shown in non-naïve mice pre-exposed to S. aureus. Furthermore, vaccination with IBT-V02 not only protected mice from a primary infection but also demonstrated lasting efficacy against a secondary infection, while prior challenge with the bacteria alone was unable to protect against recurrence. Serum transfer studies in a primary infection model showed that antibodies are primarily responsible for the protective response.
Collapse
Affiliation(s)
| | | | | | - Tom Kort
- Integrated BioTherapeutics, Rockville, MD, United States
| | | | | | - Mark Mednikov
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Roger Ortines
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Nhu T. Q. Nguyen
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Thien M. N. Doan
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Binh An Diep
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Jean C. Lee
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Rockville, MD, United States
| |
Collapse
|
23
|
Yan J, Yang R, Yu S, Zhao W. The application of the lytic domain of endolysin from Staphylococcus aureus bacteriophage in milk. J Dairy Sci 2020; 104:2641-2653. [PMID: 33358804 DOI: 10.3168/jds.2020-19456] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/09/2020] [Indexed: 11/19/2022]
Abstract
Staphylococcus aureus is a widespread foodborne pathogen that threatens human health. In particular, multidrug-resistant bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) are emerging problems in modern health care, food safety, and animal health, which require the development of new antimicrobials to replace overused conventional antibiotics. Dairy products can potentially act as vehicles for the transmission of S. aureus and other antibiotic-resistant strains from the farm into the general human population, and should be controlled during the production and storage process. Recently, bacteriophage endolysins, which degrade the cell wall that is indispensable for bacteria, have been deemed promising antimicrobial agents. In this study, one endolysin, LysGH15, demonstrated prominent antimicrobial efficacy against S. aureus, as did its catalytic domain, cysteine, histidine-dependent amidohydrolase/peptidases (CHAP)LysGH15 alone. The LysGH15 and CHAPLysGH15 exhibited different characteristics in one MRSA strain (MRSA 2701), reaching the highest activity under different conditions (35°C and pH 6.0 for LysGH15, 40°C and pH 9.0 for CHAPLysGH15). A difference in the sensitivity of LysGH15 and CHAPLysGH15 to NaCl concentration was found, where the lytic activity of LysGH15 depends strongly on its binding domain's binding capacity, which is positively correlated with the NaCl concentration, whereas the CHAPLysGH15 activity showed a negative correlation with the NaCl concentration. When the NaCl concentration was 450 mM, the lytic activity of LysGH15 reached its peak, whereas the lytic activity of CHAPLysGH15 was the highest in the absence of NaCl. The difference in NaCl sensitivity between LysGH15 and CHAPLysGH15 may be due to the sensitivity of the SH3b binding protein of LysGH15 to NaCl. The CHAPLysGH15 was tested as a biopreservative in whole and skim milk and exerted effective control against S. aureus (declined by approximately 2.5 log10 cfu/mL when incubated at 4°C for 8 h), which suggests promise for application in dairy products.
Collapse
Affiliation(s)
- Jiai Yan
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China
| | - Ruijin Yang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China
| | - Suhuai Yu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China.
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, P.R. China.
| |
Collapse
|
24
|
Bachert C, Marple B, Schlosser RJ, Hopkins C, Schleimer RP, Lambrecht BN, Bröker BM, Laidlaw T, Song WJ. Adult chronic rhinosinusitis. Nat Rev Dis Primers 2020; 6:86. [PMID: 33122665 DOI: 10.1038/s41572-020-00218-1] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Chronic rhinosinusitis (CRS) occurs in >10% of the adult population in Europe and the USA and can be differentiated into CRS without nasal polyps and CRS with nasal polyps (CRSwNP). Both phenotypes are characterized by a high disease burden and an overlapping spectrum of symptoms, with facial pain and loss of smell being the most differentiating. Great progress has been made in the understanding of CRS pathophysiology: from the epithelium and epithelial-mesenchymal transition to innate and adaptive immunity pathways and, finally, on the role of eosinophils and Staphylococcus aureus in the persistence of disease. Although clinical manifestations and diagnostic tools (including nasal endoscopy and imaging) have undergone major changes over the past few years, management (including pharmacotherapy, surgery and biologics) has experienced enormous progress based on the growing knowledge of key mediators in severe CRSwNP. The introduction of endotyping has led to a differentiation of 'tailored' surgical approaches, focusing on the mucosal concept in those with severe CRSwNP and on the identification of patients eligible for extended surgery and possibly biologics in the future.
Collapse
Affiliation(s)
- Claus Bachert
- Sun Yat-sen University, International Airway Research Center, First Affiliated Hospital, Guangzhou, China.
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium.
- Division of ENT diseases, CLINTEC, Karolinska Institute, University of Stockholm, Stockholm, Sweden.
| | - Bradley Marple
- University of Texas, Southwestern Medical Center, Department of Otolaryngology - Head and Neck Surgery, Dallas, TX, USA
| | - Rodney J Schlosser
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC, USA
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB-UGhent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, Netherlands
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Tanya Laidlaw
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Abstract
Staphylococcus aureus and Streptococcus pyogenes are common human pathogens, causing infections that include the skin. Both pathogens produce a family of secreted toxins called superantigens, which have been shown to be important in human diseases. The first cell types encountered by superantigens on skin are keratinocytes. Our studies demonstrated, that the human keratinocyte pathway, among other pathways, responds to superantigens with production of chemokines, setting off inflammation. This inflammatory response may be harmful, facilitating opening of the skin barrier. Staphylococcus aureus and Streptococcus pyogenes are significant human pathogens, causing infections at multiple body sites, including across the skin. Both are organisms that cause human diseases and secrete superantigens, including toxic shock syndrome toxin-1 (TSST-1), staphylococcal enterotoxins (SEs), and streptococcal pyrogenic exotoxins (SPEs). On the skin, human keratinocytes represent the first cell type to encounter these superantigens. We employed transcriptome sequencing (RNA-seq) to evaluate the human primary keratinocyte response to both TSST-1 and staphylococcal enterotoxin B (SEB) in triplicate analyses. Both superantigens caused large numbers of genes to be up- and downregulated. The genes that exhibited 2-fold differential gene expression compared to vehicle-treated cells, whether up- or downregulated, totaled 5,773 for TSST-1 and 4,320 for SEB. Of these, 4,482 were significantly upregulated by exposure of keratinocytes to TSST-1, whereas 1,291 were downregulated. For SEB, expression levels of 3,785 genes were upregulated, whereas those of 535 were downregulated. There was the expected high overlap in both upregulation (3,412 genes) and downregulation (400 genes). Significantly upregulated genes included those associated with chemokine production, with the possibility of stimulation of inflammation. We also tested an immortalized human keratinocyte line, from a different donor, for chemokine response to four superantigens. TSST-1 and SEB caused production of interleukin-8 (IL-8), MIP-3α, and IL-33. SPEA and SPEC were evaluated for stimulation of expression of IL-8 as a representative chemokine; both stimulated production of IL-8. IMPORTANCEStaphylococcus aureus and Streptococcus pyogenes are common human pathogens, causing infections that include the skin. Both pathogens produce a family of secreted toxins called superantigens, which have been shown to be important in human diseases. The first cell types encountered by superantigens on skin are keratinocytes. Our studies demonstrated, that the human keratinocyte pathway, among other pathways, responds to superantigens with production of chemokines, setting off inflammation. This inflammatory response may be harmful, facilitating opening of the skin barrier.
Collapse
|
26
|
Decolonization of Human Anterior Nares of Staphylococcus aureus with Use of a Glycerol Monolaurate Nonaqueous Gel. mSphere 2020; 5:5/4/e00552-20. [PMID: 32727862 PMCID: PMC7392545 DOI: 10.1128/msphere.00552-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
In this microflora study, we show that a 5% glycerol monolaurate nonaqueous gel is safe for use in the anterior nares. The gel was effective in reducing Staphylococcus aureus nasally, a highly significant hospital-associated pathogen. The gel may be a useful alternative or additive to mupirocin ointment for nasal use prior to surgery, noting that 80% of hospital-associated S. aureus infections are due to the same organism found in the nose. This gel also kills all enveloped viruses tested and should be considered for studies to reduce infection and transmission of coronaviruses and influenza viruses. Staphylococcus aureus is a highly significant infection problem in health care centers, particularly after surgery. It has been shown that nearly 80% of S. aureus infections following surgery are the same as those in the anterior nares of patients, suggesting that the anterior nares is the source of the infection strain. This has led to the use of mupirocin ointment being applied nasally to reduce infections; mupirocin resistance is being observed. This study was undertaken to determine whether gel composed of 5% glycerol monolaurate solubilized in a glycol-based, nonaqueous gel (5% GML gel) could be used as an alternative. In our study, 40 healthy human volunteers swabbed their anterior nares for 3 days with the 5% GML gel. Prior to swabbing and 8 to 12 h after swabbing, S. aureus and coagulase-negative staphylococcal CFU per milliliter were determined by plating the swabs on mannitol salt agar. Fourteen of the volunteers had S. aureus in their nares prior to 5% GML gel treatment, most persons with the organisms present in both nares; five had pure cultures of S. aureus. All participants without pure culture of S. aureus were cocolonized with S. aureus and coagulase-negative staphylococci. Five of the S. aureus strains produced the superantigens commonly associated with toxic shock syndrome, though none of the participants became ill. For both S. aureus and coagulase-negative staphylococci, the 5% GML gel treatment resulted in a 3-log-unit reduction in microorganisms. For S. aureus, the reduction persisted for 2 or 3 days. IMPORTANCE In this microflora study, we show that a 5% glycerol monolaurate nonaqueous gel is safe for use in the anterior nares. The gel was effective in reducing Staphylococcus aureus nasally, a highly significant hospital-associated pathogen. The gel may be a useful alternative or additive to mupirocin ointment for nasal use prior to surgery, noting that 80% of hospital-associated S. aureus infections are due to the same organism found in the nose. This gel also kills all enveloped viruses tested and should be considered for studies to reduce infection and transmission of coronaviruses and influenza viruses.
Collapse
|
27
|
Chung HY, Kim YT, Kwon JG, Im HH, Ko D, Lee JH, Choi SH. Molecular interaction between methicillin-resistant Staphylococcus aureus (MRSA) and chicken breast reveals enhancement of pathogenesis and toxicity for food-borne outbreak. Food Microbiol 2020; 93:103602. [PMID: 32912577 DOI: 10.1016/j.fm.2020.103602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 06/27/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022]
Abstract
To study pathogenesis and toxicity of Staphylococcus aureus in foods, FORC_062 was isolated from a human blood sample and complete genome sequence has a type II SCCmec gene cluster and a type II toxin-antitoxin system, indicating an MRSA strain. Its mobile gene elements has many pathogenic genes involved in host infection, biofilm formation, and various enterotoxin and hemolysin genes. Clinical MRSA is often found in animal foods and ingestion of MRSA-contaminated foods causes human infection. Therefore, it is very important to understand the role of contaminated foods. To elucidate the interaction between clinical MRSA FORC_062 and raw chicken breast, transcriptome analysis was conducted, showing that gene expressions of amino acid biosynthesis and metabolism were specifically down-regulated, suggesting that the strain may import and utilize amino acids from the chicken breast, but not able to synthesize them. However, toxin gene expressions were up-regulated, suggesting that human infection of S. aureus via contaminated food may be more fatal. In addition, the contaminated foods enhance multiple-antibiotic resistance activities and virulence factors in this clinical MRSA. Consequently, MRSA-contaminated food may play a role as a nutritional reservoir as well as in enhancing factor for pathogenesis and toxicity of clinical MRSA for severe food-borne outbreaks.
Collapse
Affiliation(s)
- Han Young Chung
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, And Center for Food Safety and Toxicology, Seoul National University, Seoul, 08826, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea
| | - You-Tae Kim
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 17104, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea
| | - Joon-Gi Kwon
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 17104, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea
| | - Han Hyeok Im
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, And Center for Food Safety and Toxicology, Seoul National University, Seoul, 08826, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea
| | - Duhyun Ko
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, And Center for Food Safety and Toxicology, Seoul National University, Seoul, 08826, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea
| | - Ju-Hoon Lee
- Department of Food Science and Biotechnology, Kyung Hee University, Yongin, 17104, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea.
| | - Sang Ho Choi
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology, And Center for Food Safety and Toxicology, Seoul National University, Seoul, 08826, South Korea; Food-borne Pathogen Omics Research Center (FORC), Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
28
|
Abstract
In the 1980s, menstrual toxic shock syndrome (mTSS) became a household topic, particularly among mothers and their daughters. The research performed at the time, and for the first time, exposed the American public as well as the biomedical community, in a major way, to understanding disease progression and investigation. Those studies led to the identification of the cause, Staphylococcus aureus and the pyrogenic toxin superantigen TSS toxin 1 (TSST-1), and many of the risk factors, for example, tampon use. Those studies in turn led to TSS warning labels on the outside and inside of tampon boxes and, as important, uniform standards worldwide of tampon absorbency labeling. This review addresses our understanding of the development and conclusions related to mTSS and risk factors. We leave the final message that even though mTSS is not commonly in the news today, cases continue to occur. Additionally, S. aureus strains cycle in human populations in roughly 10-year intervals, possibly dependent on immune status. TSST-1-producing S. aureus bacteria appear to be reemerging, suggesting that physician awareness of this emergence and mTSS history should be heightened.
Collapse
|
29
|
Interleukin-10, Interferon Gamma, Interleukin-2, and Soluble Interleukin-2 Receptor Alpha Detected during Peritonitis in the Dialysate and Serum of Patients on Continuous Ambulatory Peritoneal Dialysis. Perit Dial Int 2020. [DOI: 10.1177/089686089601600610] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective To analyze interleukin (IL)-10, interferon gamma (IFN-y), IL-2, and soluble IL-2 receptor alpha (sIL2Rα) in the dialysate and serum of patients on continuous ambulatory peritoneal dialysis (CAPD). Design and Patients Samples from dialysate bags were collected during the initial month of dialysis. During peritonitis, samples were collected from the first three bags on the day of admittance to the hospital and from the night bags on days 3 and 10. Serum samples were drawn on days 1 and 10. .Results: IL-10 was detected in all dialysate samples except one on the first day of infection, with a peak median level of 50 pg/mL and a slow decrease thereafter. In serum the median levels never exceeded detectable levels. Patients infected with Escherichia coli or Staphylococcus aureus had higher IL-10 levels in dialysate on day 3 as compared to the remaining patients (p < 0.05). If the catheter had to be drawn, because of persistent cloudy dialysate, the IL-10 levels remained elevated for a longer time (p<0.05). IFN-y and IL-2were detected only in the dialysate of patients infected with either S. aureusor S. epidermidis. Only one serum sample showed increased IFN-y. SIL-2Rα was found in all the serum and dialysis samples from the first day of infection. Contrary to the analyzed cytokines, the receptor showed several fold higher levels in serum as compared to the dialysate. During the infection the receptor levels in the dialysate increased, while they remained stationary in the serum, indicating a local production. Conclusion This is the first time IL-10 has been demonstrated in the dialysate during peritonitis in CAPD patients. In view of its role as a suppressor of the immune and inflammatory responses, it is a potentially important observation, which might have clinical implications in the future.
Collapse
|
30
|
Li Z, Li X, Jian M, Geleta GS, Wang Z. Two-Dimensional Layered Nanomaterial-Based Electrochemical Biosensors for Detecting Microbial Toxins. Toxins (Basel) 2019; 12:E20. [PMID: 31906152 PMCID: PMC7020412 DOI: 10.3390/toxins12010020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 01/04/2023] Open
Abstract
Toxin detection is an important issue in numerous fields, such as agriculture/food safety, environmental monitoring, and homeland security. During the past two decades, nanotechnology has been extensively used to develop various biosensors for achieving fast, sensitive, selective and on-site analysis of toxins. In particular, the two dimensional layered (2D) nanomaterials (such as graphene and transition metal dichalcogenides (TMDs)) and their nanocomposites have been employed as label and/or biosensing transducers to construct electrochemical biosensors for cost-effective detection of toxins with high sensitivity and specificity. This is because the 2D nanomaterials have good electrical conductivity and a large surface area with plenty of active groups for conjugating 2D nanomaterials with the antibodies and/or aptamers of the targeted toxins. Herein, we summarize recent developments in the application of 2D nanomaterial-based electrochemical biosensors for detecting toxins with a particular focus on microbial toxins including bacterial toxins, fungal toxins and algal toxins. The integration of 2D nanomaterials with some existing antibody/aptamer technologies into electrochemical biosensors has led to an unprecedented impact on improving the assaying performance of microbial toxins, and has shown great promise in public health and environmental protection.
Collapse
Affiliation(s)
- Zhuheng Li
- Jilin Provincial Institute of Education, Changchun 130022, China;
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China; (X.L.); (M.J.)
| | - Xiaotong Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China; (X.L.); (M.J.)
| | - Minghong Jian
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China; (X.L.); (M.J.)
| | - Girma Selale Geleta
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China; (X.L.); (M.J.)
- Department of Chemistry, College of Natural Sciences, Jimma University, Jimma 378, Ethiopia
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Science, Changchun 130022, China; (X.L.); (M.J.)
| |
Collapse
|
31
|
Czaja AJ. Examining pathogenic concepts of autoimmune hepatitis for cues to future investigations and interventions. World J Gastroenterol 2019; 25:6579-6606. [PMID: 31832000 PMCID: PMC6906207 DOI: 10.3748/wjg.v25.i45.6579] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/25/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Multiple pathogenic mechanisms have been implicated in autoimmune hepatitis, but they have not fully explained susceptibility, triggering events, and maintenance or escalation of the disease. Furthermore, they have not identified a critical defect that can be targeted. The goals of this review are to examine the diverse pathogenic mechanisms that have been considered in autoimmune hepatitis, indicate investigational opportunities to validate their contribution, and suggest interventions that might evolve to modify their impact. English abstracts were identified in PubMed by multiple search terms. Full length articles were selected for review, and secondary and tertiary bibliographies were developed. Genetic and epigenetic factors can affect susceptibility by influencing the expression of immune regulatory genes. Thymic dysfunction, possibly related to deficient production of programmed cell death protein-1, can allow autoreactive T cells to escape deletion, and alterations in the intestinal microbiome may help overcome immune tolerance and affect gender bias. Environmental factors may trigger the disease or induce epigenetic changes in gene function. Molecular mimicry, epitope spread, bystander activation, neo-antigen production, lymphocytic polyspecificity, and disturbances in immune inhibitory mechanisms may maintain or escalate the disease. Interventions that modify epigenetic effects on gene expression, alter intestinal dysbiosis, eliminate deleterious environmental factors, and target critical pathogenic mechanisms are therapeutic possibilities that might reduce risk, individualize management, and improve outcome. In conclusion, diverse pathogenic mechanisms have been implicated in autoimmune hepatitis, and they may identify a critical factor or sequence that can be validated and used to direct future management and preventive strategies.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, United States
| |
Collapse
|
32
|
The role of Staphylococcus aureus in atopic dermatitis: microbiological and immunological implications. Postepy Dermatol Alergol 2019; 36:485-491. [PMID: 31616226 PMCID: PMC6791154 DOI: 10.5114/ada.2018.77056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 06/07/2018] [Indexed: 12/13/2022] Open
Abstract
Introduction Atopic dermatitis (AD) is an inflammatory disease characterised by chronic and recurrent course. Its predominant symptom is skin pruritus. Therefore, many AD patients have recurrent skin infections and are susceptible to the colonisation of apparently healthy skin and nasal vestibule by Staphylococcus aureus (S. aureus). Some S. aureus strains are capable of producing exotoxins. Aim To assess the relation between the total IgE (tIgE) and asIgE targeted against SEA (SEA-sIgE) and SEB (SEB-sIgE), as indicators of the severity of the course of AD, and the presence of S. aureus on apparently healthy skin, in skin lesions and in the nasal vestibule. Material and methods The research was performed in a population of 134 AD patients (61 men and 73 women) aged 2–86 years. Three smears were collected for microbiological investigations: from the nasal vestibule, from the skin where lesions appeared at the moment of investigations and from the skin which was free from the eczema. On collection the material was cultured on solid and broth mediums. After incubation each medium was thoroughly analysed for the presence of S. aureus. Results There was a statistically significant correlation between healthy skin colonisation by S. aureus and increased SEA-sIgE. The same correlation was proved between healthy skin colonisation by S. aureus and increased SEB-sIgE. There was a statistically significant correlation between colonisation of the nasal vestibule by S. aureus and the SEA-sIgE and SEB-sIgE serum concentration. Conclusions It seems that the colonisation of the lesioned skin, healthy skin and the anterior nares by S. aureus is related with higher tIgE serum concentration, which translates to more severe course of the disease. Significantly increased SEA-IgE and SEB-IgE concentrations were observed in the patients whose tIgE serum concentration was statistically higher.
Collapse
|
33
|
The Role of Streptococcal and Staphylococcal Exotoxins and Proteases in Human Necrotizing Soft Tissue Infections. Toxins (Basel) 2019; 11:toxins11060332. [PMID: 31212697 PMCID: PMC6628391 DOI: 10.3390/toxins11060332] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 12/31/2022] Open
Abstract
Necrotizing soft tissue infections (NSTIs) are critical clinical conditions characterized by extensive necrosis of any layer of the soft tissue and systemic toxicity. Group A streptococci (GAS) and Staphylococcus aureus are two major pathogens associated with monomicrobial NSTIs. In the tissue environment, both Gram-positive bacteria secrete a variety of molecules, including pore-forming exotoxins, superantigens, and proteases with cytolytic and immunomodulatory functions. The present review summarizes the current knowledge about streptococcal and staphylococcal toxins in NSTIs with a special focus on their contribution to disease progression, tissue pathology, and immune evasion strategies.
Collapse
|
34
|
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: 32] [Impact Index Per Article: 6.4] [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
|
35
|
Staphylococcal Superantigens: Pyrogenic Toxins Induce Toxic Shock. Toxins (Basel) 2019; 11:toxins11030178. [PMID: 30909619 PMCID: PMC6468478 DOI: 10.3390/toxins11030178] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [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
|
36
|
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
|
37
|
Paik W, Alonzo F, Knight KL. Probiotic Exopolysaccharide Protects against Systemic Staphylococcus aureus Infection, Inducing Dual-Functioning Macrophages That Restrict Bacterial Growth and Limit Inflammation. Infect Immun 2019; 87:e00791-18. [PMID: 30396894 PMCID: PMC6300633 DOI: 10.1128/iai.00791-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus causes severe systemic infection with high mortality rates. We previously identified exopolysaccharide (EPS) from a probiotic, Bacillus subtilis, that induces anti-inflammatory macrophages with an M2 phenotype and protects mice from Citrobacter rodentium-induced colitis. We tested if EPS could protect from systemic infection induced by S. aureus and found that EPS-treated mice had enhanced survival as well as reduced weight loss, systemic inflammation, and bacterial burden. While macrophages from EPS-treated mice display an M2 phenotype, they also restrict growth of internalized S. aureus through reactive oxygen species (ROS), reminiscent of proinflammatory phagocytes. These EPS-induced macrophages also limit T cell activation by S. aureus superantigens, and EPS abrogates systemic induction of gamma interferon after infection. We conclude that B. subtilis EPS is an immunomodulatory agent that induces hybrid macrophages that bolster antibacterial immunity and simultaneously limit inflammation, reducing disease burden and promoting host survival.
Collapse
Affiliation(s)
- Wonbeom Paik
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Francis Alonzo
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois, USA
| |
Collapse
|
38
|
Rodrigues VCDC, de Oliveira IP, Bezerra RMN, Antunes AEC. Riscos microbiológicos de fórmulas para lactentes. BRAZILIAN JOURNAL OF FOOD TECHNOLOGY 2019. [DOI: 10.1590/1981-6723.05618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Resumo O objetivo desse trabalho foi discorrer sobre os principais perigos biológicos encontrados em fórmulas infantis a partir dos relatos da literatura, especialmente de registros de surtos. Este estudo é de cunho exploratório por meio de revisão bibliográfica, sendo utilizados como fontes de dados sites de busca científica. Dentre os principais micro-organismos causadores de doenças ligadas à ingestão de fórmulas infantis estão o Cronobacter sakazakii e a Salmonella enterica, porém outras bactérias, como Clostridium botulinum, Klebsiella pneumoniae, Staphylococcus aureus e Bacillus cereus, podem ser responsáveis por contaminações destas fórmulas. Visto que lactentes apresentam os sistemas imunológico e metabólico ainda em desenvolvimento, estes representam um público mais vulnerável a contaminantes, fazendo-se fundamental o oferecimento de alimentos seguros desde o processamento na indústria até a administração nas residências e unidades hospitalares.
Collapse
|
39
|
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: 1.0] [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
|
40
|
Wu X, Yang M, Fang X, Zhen S, Zhang J, Yang X, Qiao L, Yang Y, Zhang C. Expression and regulation of phenol-soluble modulins and enterotoxins in foodborne Staphylococcus aureus. AMB Express 2018; 8:187. [PMID: 30467730 PMCID: PMC6250609 DOI: 10.1186/s13568-018-0717-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/17/2018] [Indexed: 12/15/2022] Open
Abstract
Although high levels of staphylococcal phenol-soluble modulins (PSMs) in clinical methicillin-resistant Staphylococcus aureus (MRSA) has been shown to correlate with bacterial virulence, the PSMs expression in foodborne Staphylococcus aureus (S. aureus), as well as its association with staphylococcal food poisoning (SFP) was not yet clear. We collected a panel of 350 foodborne and 127 clinic-derived S. aureus strains and compared their PSMs expression. Overall, foodborne strains exhibited higher PSMs than clinical isolates, indicating a potential pathological significance of PSMs in staphylococcal food contamination. Furthermore, PSMs expression and staphylococcal enterotoxins (SEs) levels in relation to antibiotic sensitive and resistant strains were analysed. While the co-expression of PSMs and SEs was confirmed, one typical foodborne strain simultaneously yielding PSMs, SEB and SED was selected. By comparing this wildtype strain to a series of gene-deficient mutants, we concluded that PSMs and SEs expressions both relied on staphylococcal accessory regulator A initiation in the early stage of accessory gene regulator control, yet their succedent regulations differentiated to RNAIII-dependent and independent, respectively. These data provided preliminary insight into PSMs and SEs expression in foodborne S. aureus, and may guide the further studies on PSMs effects in SFP.
Collapse
|
41
|
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
42
|
Cavaillon JM. Historical links between toxinology and immunology. Pathog Dis 2018; 76:4923027. [PMID: 29718183 DOI: 10.1093/femspd/fty019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/01/2018] [Indexed: 01/28/2023] Open
Abstract
Research on bacterial toxins is closely linked to the birth of immunology. Our understanding of the interaction of bacterial protein toxins with immune cells has helped to decipher immunopathology, develop preventive and curative treatments for infections, and propose anti-cancer immunotherapies. The link started when Behring and Kitasato demonstrated that serotherapy was effective against 'the strangling angel', namely diphtheria, and its dreadful toxin discovered by Roux and Yersin. The antitoxin treatment helped to save thousands of children. Glenny demonstrated the efficacy of the secondary immune response compared to the primary one. Ramon described anatoxins that allowed the elaboration of effective vaccines and discovered the use of adjuvant to boost the antibody response. Similar approaches were later made for the tetanus toxin. Studying antitoxin antibodies Ehrlich demonstrated, for the first time, the transfer of immunity from mother to newborns. In 1989 Marrack and Kappler coined the concept of 'superantigens' to characterize protein toxins that induce T-lymphocyte proliferation, and cytokine release by both T-lymphocytes and antigen presenting cells. More recently, immunotoxins have been designed to kill cancer cells targeted by either specific antibodies or cytokines. Finally, the action of IgE antibodies against toxins may explain their persistence through evolution despite their side effect in allergy.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- Unit Cytokines and Inflammation, Institut Pasteur, 28 rue Dr. Roux, 75015 Paris, France
| |
Collapse
|
43
|
Kuschnaroff L, De Belder K, Vandeputte M, Waer M. Factors involved in peripheral T cell tolerance: the extent of clonal deletion or clonal anergy depends on the age of the tolerized lymphocytes. Transpl Int 2018. [DOI: 10.1111/tri.1992.5.s1.589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
44
|
Engebretsen K, Kezic S, Jakasa I, Hedengran A, Linneberg A, Skov L, Johansen J, Thyssen J. Effect of atopic skin stressors on natural moisturizing factors and cytokines in healthy adult epidermis. Br J Dermatol 2018; 179:679-688. [DOI: 10.1111/bjd.16487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2018] [Indexed: 12/29/2022]
Affiliation(s)
- K.A. Engebretsen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - S. Kezic
- Coronel Institute of Occupational Health; Academic Medical Center; Amsterdam Public Health Research Institute; University of Amsterdam; 1100 DE Amsterdam The Netherlands
| | - I. Jakasa
- Laboratory for Analytical Chemistry; Department of Chemistry and Biochemistry; Faculty of Food Technology and Biotechnology; University of Zagreb; Zagreb Croatia
| | - A. Hedengran
- Department of Clinical Biochemistry; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - A. Linneberg
- Research Centre for Prevention and Health; The Capital Region of Denmark; Copenhagen Denmark
- Department of Clinical Experimental Research; Rigshospitalet Glostrup Denmark
- Department of Clinical Medicine; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - L. Skov
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - J.D. Johansen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| | - J.P. Thyssen
- National Allergy Research Centre; Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
- Department of Dermatology and Allergy; Herlev and Gentofte Hospital; University of Copenhagen; Hellerup Denmark
| |
Collapse
|
45
|
Okano M, Takishita T, Yamamoto T, Hattori H, Yamashita Y, Nishioka S, Ogawa T, Nishizaki K. Presence and Characterization of Sensitization to Staphylococcal Enterotoxins in Patients with Allergic Rhinitis. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/194589240101500611] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Exotoxins derived from Staphylococcus aureus appear to be involved in the pathogenesis of allergic diseases, especially atopic dermatitis (AD). However, little is known about sensitization to enterotoxins in cases of respiratory allergies. Because the nasal cavity is a primary site of colonization by S. aureus, we sought to determine the prevalence and role of serum immunoglobulin E (IgE) antibodies against staphylococcal enterotoxin A (SEA) and SEB in patients with allergic rhinitis (AR). The presence of SEA- and SEB-specific IgE was determined in 40 patients with AR and 16 healthy control subjects. In the rhinitis group, nasal symptom score, total serum IgE, sensitization to other inhaled allergens and the presence of sinusitis were determined. Twenty-five percent of patients with AR were sensitized to SEA/SEB whereas 6.3% of controls were sensitized to the toxins. Sensitization to SEB was predominant relative to SEA. There were no significant differences in the severity of nasal symptoms and complications of other allergic diseases including AD and bronchial asthma, between patients with or without sensitization to SEA/SEB. However, patients sensitized to these toxins were likely to show increased total serum IgE and polyvalent sensitization, suggesting that exposure and subsequent sensitization to SEA/SEB may be involved in polyvalent sensitization.
Collapse
Affiliation(s)
- Mitsuhiro Okano
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Teruaki Takishita
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Takayoshi Yamamoto
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Hisashi Hattori
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Yasuhiko Yamashita
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Shinji Nishioka
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Teruhiro Ogawa
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| | - Kazunori Nishizaki
- Department of Otolaryngology–Head & Neck Surgery, Okayama University Medical School, Okayama, Japan
| |
Collapse
|
46
|
Fisher EL, Otto M, Cheung GYC. Basis of Virulence in Enterotoxin-Mediated Staphylococcal Food Poisoning. Front Microbiol 2018; 9:436. [PMID: 29662470 PMCID: PMC5890119 DOI: 10.3389/fmicb.2018.00436] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/26/2018] [Indexed: 12/17/2022] Open
Abstract
The Staphylococcus aureus enterotoxins are a superfamily of secreted virulence factors that share structural and functional similarities and possess potent superantigenic activity causing disruptions in adaptive immunity. The enterotoxins can be separated into two groups; the classical (SEA-SEE) and the newer (SEG-SElY and counting) enterotoxin groups. Many members from both these groups contribute to the pathogenesis of several serious human diseases, including toxic shock syndrome, pneumonia, and sepsis-related infections. Additionally, many members demonstrate emetic activity and are frequently responsible for food poisoning outbreaks. Due to their robust tolerance to denaturing, the enterotoxins retain activity in food contaminated previously with S. aureus. The genes encoding the enterotoxins are found mostly on a variety of different mobile genetic elements. Therefore, the presence of enterotoxins can vary widely among different S. aureus isolates. Additionally, the enterotoxins are regulated by multiple, and often overlapping, regulatory pathways, which are influenced by environmental factors. In this review, we also will focus on the newer enterotoxins (SEG-SElY), which matter for the role of S. aureus as an enteropathogen, and summarize our current knowledge on their prevalence in recent food poisoning outbreaks. Finally, we will review the current literature regarding the key elements that govern the complex regulation of enterotoxins, the molecular mechanisms underlying their enterotoxigenic, superantigenic, and immunomodulatory functions, and discuss how these activities may collectively contribute to the overall manifestation of staphylococcal food poisoning.
Collapse
Affiliation(s)
- Emilie L Fisher
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Gordon Y C Cheung
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
47
|
Activity of antimicrobial peptides and conventional antibiotics against superantigen positive Staphylococcus aureus isolated from patients with atopic dermatitis. Postepy Dermatol Alergol 2018; 35:74-82. [PMID: 29599675 PMCID: PMC5872235 DOI: 10.5114/ada.2018.62141] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 06/03/2016] [Indexed: 01/31/2023] Open
Abstract
Introduction Staphylococcus aureus causes a diverse array of diseases, ranging from relatively harmless localized skin infections to life-threatening systemic conditions. It secretes toxins directly associated with particular disease symptoms. Aim To determine the prevalence of methicillin-resistant S. aureus (MRSA) and methicillin-susceptible S. aureus (MSSA) colonization among patients with atopic dermatitis and to assess the antimicrobial susceptibility to conventional antibiotics and selected antimicrobial peptides among toxin-producing strains and nonproducing strains. Material and methods One hundred patients with atopic dermatitis and 50 healthy people were microbiologically assessed for the carriage of S. aureus. Antimicrobial susceptibility tests were performed using the broth microdilution method for conventional antibiotics and antimicrobial peptides (CAMEL, Citropin 1.1, LL-37, Temporin A). Detection of genes lukS/lukF-PV, tst, sea-sed, eta and etb by multiplex PCR was performed. Results Staphylococcus aureus strains were isolated from the majority of patients, from either the skin (75%) or the anterior nares (73%). Among the conventional antibiotics tested, the highest rates of resistance were observed for ampicillin, daptomycin, lincomycin and erythromycin. Antimicrobial peptides did not show significant diversity in activity. Among MSSA strains greater differentiation of secreted toxins was observed (sec, eta, pvl, tsst, etb, seb), while in the group of MRSA strains secretion of 3 toxins (pvl, eta, seb) was noted. Conclusions Antimicrobial resistance continues to evolve. It is important to monitor S. aureus infections. The profile of toxins produced by S. aureus strains is an important consideration in the selection of an antimicrobial agent to treat infections.
Collapse
|
48
|
Mittal P, Abblett R, Ryan JM, Hagymasi AT, Agyekum-Yamoah A, Svedova J, Reiner SL, St Rose MC, Hanley MP, Vella AT, Adler AJ. An Immunotherapeutic CD137 Agonist Releases Eomesodermin from ThPOK Repression in CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:1513-1526. [PMID: 29305435 DOI: 10.4049/jimmunol.1701039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Agonists to the TNF/TNFR costimulatory receptors CD134 (OX40) and CD137 (4-1BB) elicit antitumor immunity. Dual costimulation with anti-CD134 plus anti-CD137 is particularly potent because it programs cytotoxic potential in CD8+ and CD4+ T cells. Cytotoxicity in dual-costimulated CD4 T cells depends on the T-box transcription factor eomesodermin (Eomes), which we report is induced via a mechanism that does not rely on IL-2, in contrast to CD8+ CTL, but rather depends on the CD8 T cell lineage commitment transcription factor Runx3, which supports Eomes expression in mature CD8+ CTLs. Further, Eomes and Runx3 were indispensable for dual-costimulated CD4 T cells to mediate antitumor activity in an aggressive melanoma model. Runx3 is also known to be expressed in standard CD4 Th1 cells where it fosters IFN-γ expression; however, the CD4 T cell lineage commitment factor ThPOK represses transcription of Eomes and other CD8 lineage genes, such as Cd8a Hence, CD4 T cells can differentiate into Eomes+ cytotoxic CD4+CD8+ double-positive T cells by terminating ThPOK expression. In contrast, dual-costimulated CD4 T cells express Eomes, despite the continued expression of ThPOK and the absence of CD8α, indicating that Eomes is selectively released from ThPOK repression. Finally, although Eomes was induced by CD137 agonist, but not CD134 agonist, administered individually, CD137 agonist failed to induce CD134-/- CD4 T cells to express Eomes or Runx3, indicating that both costimulatory pathways are required for cytotoxic Th1 programming, even when only CD137 is intentionally engaged with a therapeutic agonist.
Collapse
Affiliation(s)
- Payal Mittal
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Rebecca Abblett
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Joseph M Ryan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam T Hagymasi
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Julia Svedova
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Steven L Reiner
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, New York, NY 10032; and.,Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Marie-Clare St Rose
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Matthew P Hanley
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam J Adler
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT 06030;
| |
Collapse
|
49
|
Nataf S. Autoimmunity as a Driving Force of Cognitive Evolution. Front Neurosci 2017; 11:582. [PMID: 29123465 PMCID: PMC5662758 DOI: 10.3389/fnins.2017.00582] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022] Open
Abstract
In the last decades, increasingly robust experimental approaches have formally demonstrated that autoimmunity is a physiological process involved in a large range of functions including cognition. On this basis, the recently enunciated “brain superautoantigens” theory proposes that autoimmunity has been a driving force of cognitive evolution. It is notably suggested that the immune and nervous systems have somehow co-evolved and exerted a mutual selection pressure benefiting to both systems. In this two-way process, the evolutionary-determined emergence of neurons expressing specific immunogenic antigens (brain superautoantigens) has exerted a selection pressure on immune genes shaping the T-cell repertoire. Such a selection pressure on immune genes has translated into the emergence of a finely tuned autoimmune T-cell repertoire that promotes cognition. In another hand, the evolutionary-determined emergence of brain-autoreactive T-cells has exerted a selection pressure on neural genes coding for brain superautoantigens. Such a selection pressure has translated into the emergence of a neural repertoire (defined here as the whole of neurons, synapses and non-neuronal cells involved in cognitive functions) expressing brain superautoantigens. Overall, the brain superautoantigens theory suggests that cognitive evolution might have been primarily driven by internal cues rather than external environmental conditions. Importantly, while providing a unique molecular connection between neural and T-cell repertoires under physiological conditions, brain superautoantigens may also constitute an Achilles heel responsible for the particular susceptibility of Homo sapiens to “neuroimmune co-pathologies” i.e., disorders affecting both neural and T-cell repertoires. These may notably include paraneoplastic syndromes, multiple sclerosis as well as autism, schizophrenia and neurodegenerative diseases. In the context of this theoretical frame, a specific emphasis is given here to the potential evolutionary role exerted by two families of genes, namely the MHC class II genes, involved in antigen presentation to T-cells, and the Foxp genes, which play crucial roles in language (Foxp2) and the regulation of autoimmunity (Foxp3).
Collapse
Affiliation(s)
- Serge Nataf
- CarMeN Laboratory, Bank of Tissues and Cells, Institut National de la Santé et de la Recherche Médicale 1060, INRA 1397, INSA Lyon, Lyon University Hospital (Hospices Civils de Lyon), Université Claude Bernard Lyon-1, Lyon, France
| |
Collapse
|
50
|
Cavaillon JM. Exotoxins and endotoxins: Inducers of inflammatory cytokines. Toxicon 2017; 149:45-53. [PMID: 29056305 DOI: 10.1016/j.toxicon.2017.10.016] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/26/2022]
Abstract
Endotoxins and exotoxins are among the most potent bacterial inducers of cytokines. During infectious processes, the production of inflammatory cytokines including tumor necrosis factor (TNF), interleukin-1β (IL-1β), gamma interferon (IFNγ) and chemokines orchestrates the anti-infectious innate immune response. However, an overzealous production, leading up to a cytokine storm, can be deleterious and contributes to mortality consecutive to sepsis or toxic shock syndrome. Endotoxins of Gram-negative bacteria (lipopolysaccharide, LPS) are particularly inflammatory because they generate auto-amplificatory loops after activation of monocytes/macrophages. LPS and numerous pore-forming exotoxins also activate the inflammasome, the molecular platform that allows the release of mature IL-1β and IL-18. Among exotoxins, some behave as superantigens, and as such activate the release of cytokines by T-lymphocytes. In most cases, pre-exposure to exotoxins enhances the cytokine production induced by LPS and its lethality, whereas pre-exposure to endotoxin usually results in tolerance. In this review we recall the various steps, which, from the very early discovery of pyrogenicity induced by bacterial products, ended to the discovery of the endogenous pyrogen. Furthermore, we compare the specific characteristics of endotoxins and exotoxins in their capacity to induce inflammatory cytokines.
Collapse
Affiliation(s)
- Jean-Marc Cavaillon
- Unit Cytokines & Inflammation, Institut Pasteur, 28 Rue Dr. Roux, 75015, Paris, France.
| |
Collapse
|