1
|
Bergersen KV, Zheng Y, Rossetti M, Ruffin F, Pickering H, Parmar R, Sunga G, Chan LC, Gjertson D, Fowler VG, Yeaman MR, Reed EF. Early cytokine signatures and clinical phenotypes discriminate persistent from resolving MRSA bacteremia. BMC Infect Dis 2025; 25:231. [PMID: 39966757 PMCID: PMC11834594 DOI: 10.1186/s12879-025-10620-3] [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: 11/13/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Staphylococcus aureus bacteremia (SAB) is a prevalent life-threatening infection often caused by methicillin-resistant S. aureus (MRSA). Up to 30% of SAB patients fail to clear infection even with gold-standard anti-MRSA antibiotics. This phenomenon is termed antibiotic-persistent MRSA bacteremia (APMB). The mechanisms driving APMB are complex and involve host phenotypes significantly impacting the immune response. Thus, defining early immune signatures and clinical phenotypes that differentiate APMB from antibiotic resolving (AR)MB could aid therapeutic success. METHODS We assessed 38 circulating cytokines and chemokines using affinity proteomics in 74 matched pairs of vancomycin-treated SAB cases identified as ARMB or APMB after 5 days of blood culture. RESULTS Unsupervised hierarchical clustering segregated APMB from ARMB based on differential levels of IL-10, IL-12p40, IL-13, CCL4, and TGFα. Additionally, CXCL1, CCL22 and IL-17A significantly differed between APMB and ARMB when correlated with diabetes, dialysis, metastatic infection, or cardiac vegetation. Combining immune signatures with these relevant clinical phenotypes sharply increased accuracy of discriminating APMB outcome to 79.1% via logistic regression modeling. Finally, classification-regression tree analysis revealed explicit analyte thresholds associated with APMB outcome at presentation especially in patients with metastatic infection. CONCLUSIONS Collectively, this study identifies previously unrecognized cytokine and chemokine signatures that distinguish APMB and ARMB at presentation and in the context of host clinical characteristics associated with increased disease severity. Validation of a biomarker signature that accurately predicts outcomes could guide early therapeutic strategies and interventions to reduce risks of persistent SAB that are associated with worsened morbidity and mortality.
Collapse
Affiliation(s)
- Kristina V Bergersen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Ying Zheng
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Maura Rossetti
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Felicia Ruffin
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gemalene Sunga
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA
| | - Liana C Chan
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David Gjertson
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University School of Medicine, 2301 Erwin Road, Durham, NC, 27710, USA.
- Duke Clinical Research Institute, Duke University, Durham, NC, USA.
| | - Michael R Yeaman
- Institute for Infection and Immunity, Lundquist Institute at Harbor UCLA Medical Center, Torrance, CA, USA.
- Division of Molecular Medicine, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Division of Infectious Diseases, Los Angeles County Harbor-UCLA Medical Center, Torrance, CA, USA.
- Divisions of Molecular Medicine and Infectious Diseases, David Geffen School of Medicine and Harbor-UCLA Medical Center, 1124 West Carson Street, Building MRL / 250, Torrance, CA, 90502, USA.
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 1000 Veteran Ave, Los Angeles, CA, 90095, USA.
- UCLA Immunogenetics Center, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
3
|
Fu S, Li J, You J, Liu S, Dong Q, Fu Y, Luo R, Sun Y, Tian X, Liu W, Zhang J, Ding Y, Zhang Y, Wang W, Guo L, Qiu Y. Baicalin attenuates PD-1/PD-L1 axis-induced immunosuppression in piglets challenged with Glaesserella parasuis by inhibiting the PI3K/Akt/mTOR and RAS/MEK/ERK signalling pathways. Vet Res 2024; 55:95. [PMID: 39075562 PMCID: PMC11285455 DOI: 10.1186/s13567-024-01355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Infection of piglets with Glaesserella parasuis (G. parasuis) induces host immunosuppression. However, the mechanism underlying the immunosuppression of piglets remains unclear. Activation of the PD-1/PD-L1 axis has been shown to trigger host immunosuppression. Baicalin possesses anti-inflammatory and immunomodulatory functions. However, whether baicalin inhibits PD-1/PD-L1 activation and thus alleviates host immunosuppression has not been investigated. In this study, the effect of baicalin on the attenuation of piglet immunosuppression induced by G. parasuis was evaluated. Seventy piglets were randomly divided into the control group, infection group, levamisole group, BMS-1 group, 25 mg/kg baicalin group, 50 mg/kg baicalin group and 100 mg/kg baicalin group. Following pretreatment with levamisole, BMS-1 or baicalin, the piglets were challenged with 1 × 108 CFU of G. parasuis. Our results showed that baicalin, levamisole and BMS-1 modified routine blood indicators and biochemical parameters; downregulated IL-1β, IL-10, IL-18, TNF-α and IFN-γ mRNA expression; and upregulated IL-2 and IL-8 mRNA expression in blood. Baicalin, levamisole and BMS-1 increased the proportions of CD3+ T cells, CD3+CD4+ T cells, CD3+CD8+ T cells and CD3-CD21+ B cells in the splenocyte population, increased the proportions of CD3+ T cells, CD3+CD4+ T cells and CD3+CD8+ T cells in the blood, and inhibited PD-1/PD-L1 and TIM-3 activation. Baicalin, levamisole and BMS-1 reduced p-PI3K, p-Akt, and p-mTOR expression, the p-MEK1/2/MEK1/2 and p-ERK1/2/ERK1/2 ratios and increased RAS expression. Baicalin, levamisole and BMS-1 provided substantial protection against G. parasuis challenge and relieved tissue histopathological damage. Our findings might provide new strategies for controlling G. parasuis infection and other immunosuppressive diseases.
Collapse
Affiliation(s)
- Shulin Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Jingyang Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Jiarui You
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Siyu Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Qiaoli Dong
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Yunjian Fu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Ronghui Luo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Yamin Sun
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Xinyue Tian
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Wei Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China
| | - Jingyi Zhang
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yu Ding
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yitian Zhang
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Wutao Wang
- School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Ling Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, China.
| | - Yinsheng Qiu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, 430023, China.
- Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety, Wuhan, 430023, 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
|
Chen Y, Liu Z, Lin Z, Lu M, Fu Y, Liu G, Yu B. The effect of Staphylococcus aureus on innate and adaptive immunity and potential immunotherapy for S. aureus-induced osteomyelitis. Front Immunol 2023; 14:1219895. [PMID: 37744377 PMCID: PMC10517662 DOI: 10.3389/fimmu.2023.1219895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Osteomyelitis is a chronic inflammatory bone disease caused by infection of open fractures or post-operative implants. Particularly in patients with open fractures, the risk of osteomyelitis is greatly increased as the soft tissue damage and bacterial infection are often more severe. Staphylococcus aureus, one of the most common pathogens of osteomyelitis, disrupts the immune response through multiple mechanisms, such as biofilm formation, virulence factor secretion, and metabolic pattern alteration, which attenuates the effectiveness of antibiotics and surgical debridement toward osteomyelitis. In osteomyelitis, immune cells such as neutrophils, macrophages and T cells are activated in response to pathogenic bacteria invasion with excessive inflammatory factor secretion, immune checkpoint overexpression, and downregulation of immune pathway transcription factors, which enhances osteoclastogenesis and results in bone destruction. Therefore, the study of the mechanisms of abnormal immunity will be a new breakthrough in the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Yingqi Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zixian Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Zexin Lin
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Mincheng Lu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Yong Fu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
- Trauma Center, Department of Orthopaedic Trauma, The Second Affiliated Hospital of Hengyang Medical College, South China University, Hengyang, China
| | - Guanqiao Liu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Southern Medical University Nanfang Hospital, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Cartilage Regenerative Medicine, Southern Medical University Nanfang Hospital, Guangzhou, China
| |
Collapse
|
6
|
Mata Forsberg M, Arasa C, van Zwol W, Uzunçayir S, Schönbichler A, Regenthal P, Schelin J, Lindkvist-Petersson K, Björkander S, Sverremark-Ekström E. Activation of human γδ T cells and NK cells by Staphylococcal enterotoxins requires both monocytes and conventional T cells. J Leukoc Biol 2021; 111:597-609. [PMID: 34114693 DOI: 10.1002/jlb.3a1020-630rr] [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] [Indexed: 12/20/2022] Open
Abstract
Staphylococcal enterotoxins (SE) pose a great threat to human health due to their ability to bypass antigen presentation and activate large amounts of conventional T cells resulting in a cytokine storm potentially leading to toxic shock syndrome. Unconventional T- and NK cells are also activated by SE but the mechanisms remain poorly understood. In this study, the authors aimed to explore the underlying mechanism behind SE-mediated activation of MAIT-, γδ T-, and NK cells in vitro. CBMC or PBMC were stimulated with the toxins SEA, SEH, and TSST-1, and cytokine and cytotoxic responses were analyzed with ELISA and flow cytometry. All toxins induced a broad range of cytokines, perforin and granzyme B, although SEH was not as potent as SEA and TSST-1. SE-induced IFN-γ expression in MAIT-, γδ T-, and NK cells was clearly reduced by neutralization of IL-12, while cytotoxic compounds were not affected at all. Kinetic assays showed that unconventional T cell and NK cell-responses are secondary to the response in conventional T cells. Furthermore, co-cultures of isolated cell populations revealed that the ability of SEA to activate γδ T- and NK cells was fully dependent on the presence of both monocytes and αβ T cells. Lastly, it was found that SE provoked a reduced and delayed cytokine response in infants, particularly within the unconventional T and NK cell populations. This study provides novel insights regarding the activation of unconventional T- and NK cells by SE, which contribute to understanding the vulnerability of young children towards Staphylococcus aureus infections.
Collapse
Affiliation(s)
- Manuel Mata Forsberg
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Claudia Arasa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Willemien van Zwol
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sibel Uzunçayir
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anna Schönbichler
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Paulina Regenthal
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jenny Schelin
- Division of Applied Microbiology, Department of Chemistry, Lund University, Lund, Sweden
| | | | - Sophia Björkander
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
7
|
Guan Z, Liu Y, Liu C, Wang H, Feng J, Yang G. Staphylococcus aureus β-Hemolysin Up-Regulates the Expression of IFN-γ by Human CD56 bright NK Cells. Front Cell Infect Microbiol 2021; 11:658141. [PMID: 33854984 PMCID: PMC8039520 DOI: 10.3389/fcimb.2021.658141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/11/2021] [Indexed: 01/12/2023] Open
Abstract
IFN-γ is produced upon stimulation with S. aureus and may play a detrimental role during infection. However, whether hemolysins play a role in the mechanism of IFN-γ production has not been fully characterized. In this study, we demonstrated that Hlb, one of the major hemolysins of S. aureus, upregulated IFN-γ production by CD56bright NK cells from human peripheral blood mononuclear cells (PBMCs). Further investigation showed that Hlb increased calcium influx and induced phosphorylation of ERK1/2. Either blocking calcium or specifically inhibiting phosphorylation of ERK1/2 decreased the production of IFN-γ induced by Hlb. Moreover, we found that this process was dependent on the sphingomyelinase activity of Hlb. Our findings revealed a novel mechanism of IFN-γ production in NK cells induced by Hlb, which may be involved in the pathogenesis of S. aureus.
Collapse
Affiliation(s)
- Zhangchun Guan
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Yu Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Chenghua Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Huiting Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Jiannan Feng
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| | - Guang Yang
- Beijing Institute of Pharmacology and Toxicology, Beijing, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China
| |
Collapse
|
8
|
Uebele J, Habenicht K, Ticha O, Bekeredjian-Ding I. Staphylococcus aureus Protein A Induces Human Regulatory T Cells Through Interaction With Antigen-Presenting Cells. Front Immunol 2020; 11:581713. [PMID: 33117390 PMCID: PMC7560526 DOI: 10.3389/fimmu.2020.581713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Despite continuous exposure and development of specific immunity, Staphylococcus aureus (Sa) remains one of the leading causes of severe infections worldwide. Although innate immune defense mechanisms are well understood, the role of the T cell response has not been fully elucidated. Here, we demonstrate that Sa and one of its major virulence factors protein A (SpA) induce human regulatory T cells (Tregs), key players in immune tolerance. In human PBMC and MoDC/T cell cocultures CD4+CD25+CD127dim Tregs were induced upon stimulation with Sa and to a lower extent with SpA alone. Treg induction was strongly, but not exclusively, dependent on SpA, and independent of antigen presentation or T cell epitope recognition. Lastly, soluble factors in the supernatant of SpA-stimulated MoDC were sufficient to trigger Treg formation, while supernatants of MoDC/T cell cocultures containing Sa-triggered Tregs displayed T cell suppressive activity. In summary, our findings identify a new immunosuppressory function of SpA, which leads to release of soluble, Treg-inducing factors and might be relevant to establish colonization.
Collapse
Affiliation(s)
- Julia Uebele
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Olga Ticha
- Division of Microbiology, Paul-Ehrlich-Institut, Langen, Germany
| | | |
Collapse
|
9
|
Han P, Yosinski S, Kobos ZA, Chaudhury R, Lee JS, Fahmy TM, Reed MA. Continuous Label-Free Electronic Discrimination of T Cells by Activation State. ACS NANO 2020; 14:8646-8657. [PMID: 32530598 DOI: 10.1021/acsnano.0c03018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The sensitivity and speed with which the immune system reacts to host disruption is unrivaled by any detection method for pathogenic biomarkers or infectious signatures. Engagement of cellular immunity in response to infections or cancer is contingent upon activation and subsequent cytotoxic activity by T cells. Thus, monitoring T cell activation can reliably serve as a metric for disease diagnosis as well as therapeutic prognosis. Rapid and direct quantification of T cell activation states, however, has been hindered by challenges associated with antigen target identification, labeling requirements, and assay duration. Here we present an electronic, label-free method for simultaneous separation and evaluation of T cell activation states. Our device utilizes a microfluidic design integrated with nanolayered electrode structures for dielectrophoresis (DEP)-driven discrimination of activated vs naïve T cells at single-cell resolution and demonstrates rapid (<2 min) separation of T cells at high single-pass efficiency as quantified by an on-chip Coulter counter module. Our device represents a microfluidic tool for electronic assessment of immune activation states and, hence, a portable diagnostic for quantitative evaluation of immunity and disease state. Further, its ability to achieve label-free enrichment of activated immune cells promises clinical utility in cell-based immunotherapies.
Collapse
Affiliation(s)
- Patrick Han
- Department of Chemical & Environmental Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Shari Yosinski
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Zachary A Kobos
- Department of Electrical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Rabib Chaudhury
- Department of Chemical & Environmental Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Jung Seok Lee
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Tarek M Fahmy
- Department of Chemical & Environmental Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| | - Mark A Reed
- Department of Electrical Engineering, School of Engineering and Applied Sciences, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
10
|
Nowicka D, Grywalska E. Staphylococcus aureus and Host Immunity in Recurrent Furunculosis. Dermatology 2019; 235:295-305. [DOI: 10.1159/000499184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus is one of the severest and most persistent bacterial pathogens. The most frequent S. aureus infections include impetigo, folliculitis, furuncles, furunculosis, abscesses, hidradenitis suppurativa, and mastitis. S. aureus produces a great variety of cellular and extracellular factors responsible for its invasiveness and ability to cause pathological lesions. Their expression depends on the growth phase, environmental factors, and location of the infection. Susceptibility to staphylococcal infections is rooted in multiple mechanisms of host immune responses and reactions to bacterial colonization. Immunological and inflammatory processes of chronic furunculosis are based on the pathogenicity of S. aureus as well as innate and acquired immunity. In-depth knowledge about them may help to discover the whole pathomechanism of the disease and to develop effective therapeutic options. In this review, we focus on the S. aureus-host immune interactions in the pathogenesis of recurrent furunculosis according to the most recent experimental and clinical findings.
Collapse
|
11
|
Guttman-Yassky E, Bissonnette R, Ungar B, Suárez-Fariñas M, Ardeleanu M, Esaki H, Suprun M, Estrada Y, Xu H, Peng X, Silverberg JI, Menter A, Krueger JG, Zhang R, Chaudhry U, Swanson B, Graham NMH, Pirozzi G, Yancopoulos GD, D Hamilton JD. Dupilumab progressively improves systemic and cutaneous abnormalities in patients with atopic dermatitis. J Allergy Clin Immunol 2018; 143:155-172. [PMID: 30194992 DOI: 10.1016/j.jaci.2018.08.022] [Citation(s) in RCA: 442] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/19/2018] [Accepted: 08/14/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Dupilumab is an IL-4 receptor α mAb inhibiting signaling of IL-4 and IL-13, key drivers of type 2-driven inflammation, as demonstrated by its efficacy in patients with atopic/allergic diseases. OBJECTIVE This placebo-controlled, double-blind trial (NCT01979016) evaluated the efficacy, safety, and effects of dupilumab on molecular/cellular lesional and nonlesional skin phenotypes and systemic type 2 biomarkers of patients with moderate-to-severe atopic dermatitis (AD). METHODS Skin biopsy specimens and blood were evaluated from 54 patients randomized 1:1 to weekly subcutaneous doses of 200 mg of dupilumab or placebo for 16 weeks. RESULTS Dupilumab (vs placebo) significantly improved clinical signs and symptoms of AD, was well tolerated, and progressively shifted the lesional transcriptome toward a nonlesional phenotype (weeks 4-16). Mean improvements in a meta-analysis-derived AD transcriptome (genes differentially expressed between lesional and nonlesional skin) were 68.8% and 110.8% with dupilumab and -10.5% and 55.0% with placebo (weeks 4 and 16, respectively; P < .001). Dupilumab significantly reduced expression of genes involved in type 2 inflammation (IL13, IL31, CCL17, CCL18, and CCL26), epidermal hyperplasia (keratin 16 [K16] and MKi67), T cells, dendritic cells (ICOS, CD11c, and CTLA4), and TH17/TH22 activity (IL17A, IL-22, and S100As) and concurrently increased expression of epidermal differentiation, barrier, and lipid metabolism genes (filaggrin [FLG], loricrin [LOR], claudins, and ELOVL3). Dupilumab reduced lesional epidermal thickness versus placebo (week 4, P = .001; week 16, P = .0002). Improvements in clinical and histologic measures correlated significantly with modulation of gene expression. Dupilumab also significantly suppressed type 2 serum biomarkers, including CCL17, CCL18, periostin, and total and allergen-specific IgEs. CONCLUSION Dupilumab-mediated inhibition of IL-4/IL-13 signaling through IL-4 receptor α blockade significantly and progressively improved disease activity, suppressed cellular/molecular cutaneous markers of inflammation and systemic measures of type 2 inflammation, and reversed AD-associated epidermal abnormalities.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY.
| | | | - Benjamin Ungar
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Mayte Suárez-Fariñas
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Hitokazu Esaki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Maria Suprun
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yeriel Estrada
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hui Xu
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiangyu Peng
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jonathan I Silverberg
- Department of Dermatology, Preventive Medicine and Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Alan Menter
- Department of Dermatology, Baylor University Medical Center, Dallas, Tex
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Rick Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY
| | | | | | | | | | | | | |
Collapse
|
12
|
Tuffs SW, Haeryfar SMM, McCormick JK. Manipulation of Innate and Adaptive Immunity by Staphylococcal Superantigens. Pathogens 2018; 7:pathogens7020053. [PMID: 29843476 PMCID: PMC6027230 DOI: 10.3390/pathogens7020053] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Staphylococcal superantigens (SAgs) constitute a family of potent exotoxins secreted by Staphylococcus aureus and other select staphylococcal species. SAgs function to cross-link major histocompatibility complex (MHC) class II molecules with T cell receptors (TCRs) to stimulate the uncontrolled activation of T lymphocytes, potentially leading to severe human illnesses such as toxic shock syndrome. The ubiquity of SAgs in clinical S. aureus isolates suggests that they likely make an important contribution to the evolutionary fitness of S. aureus. Although the apparent redundancy of SAgs in S. aureus has not been explained, the high level of sequence diversity within this toxin family may allow for SAgs to recognize an assorted range of TCR and MHC class II molecules, as well as aid in the avoidance of humoral immunity. Herein, we outline the major diseases associated with the staphylococcal SAgs and how a dysregulated immune system may contribute to pathology. We then highlight recent research that considers the importance of SAgs in the pathogenesis of S. aureus infections, demonstrating that SAgs are more than simply an immunological diversion. We suggest that SAgs can act as targeted modulators that drive the immune response away from an effective response, and thus aid in S. aureus persistence.
Collapse
Affiliation(s)
- Stephen W Tuffs
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, ON N6A 3K7, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| | - John K McCormick
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, Western University, London, ON N6A 3K7, Canada.
- Lawson Health Research Institute, London, ON N6C 2R5, Canada.
| |
Collapse
|
13
|
Rasmussen G, Cajander S, Bäckman A, Källman J, Söderquist B, Strålin K. Expression of HLA-DRA and CD74 mRNA in whole blood during the course of complicated and uncomplicated Staphylococcus aureus bacteremia. Microbiol Immunol 2018; 61:442-451. [PMID: 28862321 DOI: 10.1111/1348-0421.12533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 01/16/2023]
Abstract
To improve management of Staphylococcus aureus bacteremia (SAB), better understanding of host-pathogen interactions is needed. In vitro studies have shown that S. aureus bacteria induce dose-dependent immunosuppression that is evidenced by reduced expression of major histocompatibility complex (MHC) class II on antigen presenting cells. Thus, the aim of this study was to determine whether expression of the MHC class II-related genes HLA-DRA and CD74 is more greatly reduced in complicated SAB, with its probable higher loads of S. aureus, than in uncomplicated SAB. Adult patients with SAB were prospectively included and blood samples taken on the day of confirmation of SAB (Day 1) and on Days 2, 3, 5 and 7. HLA-DRA and CD74 mRNA expression was determined by quantitative reverse transcription PCR. Sepsis was defined according to the Sepsis-3 classification and SAB was categorized as complicated in patients with deep-seated infection and/or hematogenous seeding. Twenty patients with SAB were enrolled and samples obtained on all assessment days. HLA-DRA and CD74 expression did not differ significantly between patients with SAB and sepsis (n = 13) and those without sepsis (n = 7) on any assessment day. However, patients with complicated SAB (n = 14) had significantly weaker HLA-DRA expression on all five assessment days than patients with uncomplicated SAB (n = 6). Additionally, they tended to have weaker CD74 expressions. Neutrophil, monocyte and leukocyte counts did not differ significantly between complicated and uncomplicated SAB. In conclusion, patients with complicated SAB show weaker HLA-DRA expression than those with uncomplicated SAB during the first week of bacteremia.
Collapse
Affiliation(s)
- Gunlög Rasmussen
- Department of Infectious Diseases, Örebro University Hospital, Örebro University, Örebro.,Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro
| | - Sara Cajander
- Department of Infectious Diseases, Örebro University Hospital, Örebro University, Örebro.,Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro
| | - Anders Bäckman
- Faculty of Medicine and Health, Department of Clinical Research Laboratory, Örebro University, Örebro
| | - Jan Källman
- Department of Infectious Diseases, Örebro University Hospital, Örebro University, Örebro.,Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro
| | - Bo Söderquist
- Department of Infectious Diseases, Örebro University Hospital, Örebro University, Örebro.,Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro
| | - Kristoffer Strålin
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Chamoun MN, Blumenthal A, Sullivan MJ, Schembri MA, Ulett GC. Bacterial pathogenesis and interleukin-17: interconnecting mechanisms of immune regulation, host genetics, and microbial virulence that influence severity of infection. Crit Rev Microbiol 2018; 44:465-486. [PMID: 29345518 DOI: 10.1080/1040841x.2018.1426556] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Interleukin-17 (IL-17) is a pro-inflammatory cytokine involved in the control of many different disorders, including autoimmune, oncogenic, and diverse infectious diseases. In the context of infectious diseases, IL-17 protects the host against various classes of microorganisms but, intriguingly, can also exacerbate the severity of some infections. The regulation of IL-17 expression stems, in part, from the activity of Interleukin-23 (IL-23), which drives the maturation of different classes of IL-17-producing cells that can alter the course of infection. In this review, we analyze IL-17/IL-23 signalling in bacterial infection, and examine the interconnecting mechanisms that link immune regulation, host genetics, and microbial virulence in the context of bacterial pathogenesis. We consider the roles of IL-17 in both acute and chronic bacterial infections, with a focus on mouse models of human bacterial disease that involve infection of mucosal surfaces in the lungs, urogenital, and gastrointestinal tracts. Polymorphisms in IL-17-encoding genes in humans, which have been associated with heightened host susceptibility to some bacterial pathogens, are discussed. Finally, we examine the implications of IL-17 biology in infectious diseases for the development of novel therapeutic strategies targeted at preventing bacterial infection.
Collapse
Affiliation(s)
- Michelle N Chamoun
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Antje Blumenthal
- b The University of Queensland Diamantina Institute, Translational Research Institute , Brisbane , Australia
| | - Matthew J Sullivan
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| | - Mark A Schembri
- c School of Chemistry and Molecular Biosciences, and Australian Infectious Disease Research Centre , The University of Queensland , Brisbane , Australia
| | - Glen C Ulett
- a School of Medical Science, and Menzies Health Institute Queensland , Griffith University , Southport , Australia
| |
Collapse
|
15
|
Abstract
Safe and efficacious vaccines are arguably the most successful medical interventions of all time. Yet the ongoing discovery of new pathogens, along with emergence of antibiotic-resistant pathogens and a burgeoning population at risk of such infections, imposes unprecedented public health challenges. To meet these challenges, innovative strategies to discover and develop new or improved anti-infective vaccines are necessary. These approaches must intersect the most meaningful insights into protective immunity and advanced technologies with capabilities to deliver immunogens for optimal immune protection. This goal is considered through several recent advances in host-pathogen relationships, conceptual strides in vaccinology, and emerging technologies. Given a clear and growing risk of pandemic disease should the threat of infection go unmet, developing vaccines that optimize protective immunity against high-priority and antibiotic-resistant pathogens represents an urgent and unifying imperative.
Collapse
Affiliation(s)
- Michael R Yeaman
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California 90024.,Division of Molecular Medicine, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509; .,Division of Infectious Diseases, Department of Medicine, Harbor-UCLA Medical Center, Torrance, California 90509.,Los Angeles Biomedical Research Institute, Torrance, California 90502
| | | |
Collapse
|
16
|
Goldmann O, Medina E. Staphylococcus aureus strategies to evade the host acquired immune response. Int J Med Microbiol 2017; 308:625-630. [PMID: 28939437 DOI: 10.1016/j.ijmm.2017.09.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/01/2017] [Accepted: 09/13/2017] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus poses a significant public-health problem. Infection caused by S. aureus can manifest as acute or long-lasting persistent diseases that are often refractory to antibiotic and are associated with significant morbidity and mortality. To develop more effective strategies for preventing or treating these infections, it is crucial to understand why the immune response is incapable to eradicate the bacterium. When S. aureus first infect the host, there is a robust activation of the host innate immune responses. Generally, S. aureus can survive this initial interaction due to the expression of a wide array of virulence factors that interfere with the host innate immune defenses. After this initial interaction the acquired immune response is the arm of the host defenses that will try to clear the pathogen. However, S. aureus is capable of maintaining infection in the host even in the presence of a robust antigen-specific immune response. Thus, understanding the mechanisms underlying the ability of S. aureus to escape immune surveillance by the acquired immune response will help uncover potentially important targets for the development of immune-based adjunctive therapies and more efficient vaccines. There are several lines of evidence that lead us to believe that S. aureus can directly or indirectly disable the acquired immune response. This review will discuss the different immune evasion strategies used by S. aureus to modulate the different components of the acquired immune defenses.
Collapse
Affiliation(s)
- Oliver Goldmann
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| | - Eva Medina
- Infection Immunology Research Group, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany.
| |
Collapse
|
17
|
Szabo PA, Goswami A, Mazzuca DM, Kim K, O'Gorman DB, Hess DA, Welch ID, Young HA, Singh B, McCormick JK, Haeryfar SMM. Rapid and Rigorous IL-17A Production by a Distinct Subpopulation of Effector Memory T Lymphocytes Constitutes a Novel Mechanism of Toxic Shock Syndrome Immunopathology. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:2805-2818. [PMID: 28219889 PMCID: PMC6635948 DOI: 10.4049/jimmunol.1601366] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/25/2017] [Indexed: 01/13/2023]
Abstract
Toxic shock syndrome (TSS) is caused by staphylococcal and streptococcal superantigens (SAgs) that provoke a swift hyperinflammatory response typified by a cytokine storm. The precipitous decline in the host's clinical status and the lack of targeted therapies for TSS emphasize the need to identify key players of the storm's initial wave. Using a humanized mouse model of TSS and human cells, we herein demonstrate that SAgs elicit in vitro and in vivo IL-17A responses within hours. SAg-triggered human IL-17A production was characterized by remarkably high mRNA stability for this cytokine. A distinct subpopulation of CD4+ effector memory T (TEM) cells that secrete IL-17A, but not IFN-γ, was responsible for early IL-17A production. We found mouse "TEM-17" cells to be enriched within the intestinal epithelium and among lamina propria lymphocytes. Furthermore, interfering with IL-17A receptor signaling in human PBMCs attenuated the expression of numerous inflammatory mediators implicated in the TSS-associated cytokine storm. IL-17A receptor blockade also abrogated the secondary effect of SAg-stimulated PBMCs on human dermal fibroblasts as judged by C/EBP δ expression. Finally, the early IL-17A response to SAgs was pathogenic because in vivo neutralization of IL-17A in humanized mice ameliorated hepatic and intestinal damage and reduced mortality. Together, our findings identify CD4+ TEM cells as a key effector of TSS and reveal a novel role for IL-17A in TSS immunopathogenesis. Our work thus elucidates a pathogenic, as opposed to protective, role for IL-17A during Gram-positive bacterial infections. Accordingly, the IL-17-IL-17R axis may provide an attractive target for the management of SAg-mediated illnesses.
Collapse
Affiliation(s)
- Peter A Szabo
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - Ankur Goswami
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - Delfina M Mazzuca
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - Kyoungok Kim
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
| | - David B O'Gorman
- Cell and Molecular Biology Laboratory, Roth | McFarlane Hand and Upper Limb Centre, Western University, London, Ontario N6A 4V2, Canada
- Department of Biochemistry, Western University, London, Ontario N6A 5C1, Canada
- Lawson Health Research Institute, London, Ontario N6C 2R5, Canada
- Department of Surgery, Western University, London, Ontario N6A 4V2, Canada
| | - David A Hess
- Department of Physiology and Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Group, Robarts Research Institute, London, Ontario N6A 5B7, Canada
| | - Ian D Welch
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 2B5, Canada
| | - Howard A Young
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702
| | - Bhagirath Singh
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
- Lawson Health Research Institute, London, Ontario N6C 2R5, Canada
- Centre for Human Immunology, Western University, London, Ontario N6A 5C1, Canada; and
| | - John K McCormick
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada
- Lawson Health Research Institute, London, Ontario N6C 2R5, Canada
- Centre for Human Immunology, Western University, London, Ontario N6A 5C1, Canada; and
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario N6A 5C1, Canada;
- Lawson Health Research Institute, London, Ontario N6C 2R5, Canada
- Centre for Human Immunology, Western University, London, Ontario N6A 5C1, Canada; and
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario N6A 5A5, Canada
| |
Collapse
|
18
|
Li Z, Levast B, Madrenas J. Staphylococcus aureusDownregulates IP-10 Production and Prevents Th1 Cell Recruitment. THE JOURNAL OF IMMUNOLOGY 2017; 198:1865-1874. [DOI: 10.4049/jimmunol.1601336] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022]
|
19
|
Björkander S, Johansson MA, Hell L, Lasaviciute G, Nilsson C, Holmlund U, Sverremark-Ekström E. FOXP3 + CD4 T-cell maturity and responses to microbial stimulation alter with age and associate with early-life gut colonization. J Allergy Clin Immunol 2016; 138:905-908.e4. [DOI: 10.1016/j.jaci.2016.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/05/2016] [Accepted: 04/29/2016] [Indexed: 01/09/2023]
|
20
|
Haileselassie Y, Navis M, Vu N, Qazi KR, Rethi B, Sverremark-Ekström E. Lactobacillus reuteri and Staphylococcus aureus differentially influence the generation of monocyte-derived dendritic cells and subsequent autologous T cell responses. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:315-26. [PMID: 27621814 PMCID: PMC5004286 DOI: 10.1002/iid3.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/20/2016] [Accepted: 06/20/2016] [Indexed: 12/23/2022]
Abstract
Introduction In early‐life, the immature mucosal barrier allows contact between the gut microbiota and the developing immune system. Due to their strategic location and their ability to sample luminal antigen, dendritic cells (DC) play a central role in the interaction of microbes and immune cells in the gut. Here, we investigated how two bacteria associated with opposite immune profiles in children, that is, Lactobacillus (L.) reuteri and Staphylococcus (S.) aureus, influenced the differentiation of monocytes in vitro as well how the generated DC impacted T cell responses. Methods We exposed monocyte cultures to cell‐free supernatants (CFS) from these bacteria during their differentiation to DC. Results The presence of L. reuteri‐CFS during DC differentiation resulted in DC with a more mature phenotype, in terms of up‐regulated surface markers (HLA‐DR, CD86, CD83, CCR7) and enhanced cytokine production (IL6, IL10, and IL23), but had a reduced phagocytic capacity compared with non‐treated monocyte‐derived DC (Mo‐DC). However, upon LPS activation, L. reuteri‐CFS‐generated DC displayed a more regulated phenotype than control Mo‐DC with notable reduction of cytokine responses both at mRNA and protein levels. In contrast, S. aureus‐CFS‐generated DC were more similar to control Mo‐DC both without and after LPS stimulation, but they were still able to induce responses in autologous T cells, in the absence of further T cell stimulation. Conclusions We show that bacterial signals during DC differentiation have a profound impact on DC function and possibly also for shaping the T cell pool.
Collapse
Affiliation(s)
- Yeneneh Haileselassie
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Marit Navis
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Nam Vu
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Khaleda Rahman Qazi
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| | - Bence Rethi
- Department of Medicine Karolinska University Hospital Stockholm Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, The Wenner-Gren Institute Stockholm University Stockholm Sweden
| |
Collapse
|
21
|
Johansson MA, Björkander S, Mata Forsberg M, Qazi KR, Salvany Celades M, Bittmann J, Eberl M, Sverremark-Ekström E. Probiotic Lactobacilli Modulate Staphylococcus aureus-Induced Activation of Conventional and Unconventional T cells and NK Cells. Front Immunol 2016; 7:273. [PMID: 27462316 PMCID: PMC4939411 DOI: 10.3389/fimmu.2016.00273] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022] Open
Abstract
Lactobacilli are probiotic commensal bacteria and potent modulators of immunity. When present in the gut or supplemented as probiotics, they beneficially modulate ex vivo immune responsiveness. Further, factors derived from several lactobacilli strains act immune regulatory in vitro. In contrast, Staphylococcus aureus (S. aureus) is known to induce excessive T cell activation. In this study, we aimed to investigate S. aureus-induced activation of human mucosal-associated invariant T cells (MAIT cells), γδ T cells, NK cells, as well as of conventional CD4+ and CD8+ T cells in vitro. Further, we investigated if lactobacilli-derived factors could modulate their activation. PBMC were cultured with S. aureus 161:2 cell-free supernatants (CFS), staphylococcal enterotoxin A or CD3/CD28-beads alone, or in combination with Lactobacillus rhamnosus GG-CFS or Lactobacillus reuteri DSM 17938-CFS and activation of T and NK cells was evaluated. S. aureus-CFS induced IFN-γ and CD107a expression as well as proliferation. Costimulation with lactobacilli-CFS dampened lymphocyte-activation in all cell types analyzed. Preincubation with lactobacilli-CFS was enough to reduce subsequent activation, and the absence of APC or APC-derived IL-10 did not prevent lactobacilli-mediated dampening. Finally, lactate selectively dampened activation of unconventional T cells and NK cells. In summary, we show that molecules present in the lactobacilli-CFS are able to directly dampen in vitro activation of conventional and unconventional T cells and of NK cells. This study provides novel insights on the immune-modulatory nature of probiotic lactobacilli and suggests a role for lactobacilli in the modulation of induced T and NK cell activation.
Collapse
Affiliation(s)
- Maria A Johansson
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Sophia Björkander
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Manuel Mata Forsberg
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Khaleda Rahman Qazi
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Maria Salvany Celades
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Julia Bittmann
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK; Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Eva Sverremark-Ekström
- Arrhenius Laboratories for Natural Sciences, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|
22
|
Carvalho-Queiroz C, Johansson MA, Persson JO, Jörtsö E, Kjerstadius T, Nilsson C, Saghafian-Hedengren S, Sverremark-Ekström E. Associations between EBV and CMV Seropositivity, Early Exposures, and Gut Microbiota in a Prospective Birth Cohort: A 10-Year Follow-up. Front Pediatr 2016; 4:93. [PMID: 27630978 PMCID: PMC5006634 DOI: 10.3389/fped.2016.00093] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/23/2016] [Indexed: 01/05/2023] Open
Abstract
Early-life infections with persistent Epstein-Barr virus (EBV) and cytomegalovirus (CMV) are delayed in affluent countries, probably due to alterations in early environmental exposures, such as maternal age, siblings, and day-care attendance. We have previously reported that the timing of EBV and CMV contraction is related both to allergic sensitization and changes in functional competence of immune cells, while the presence/absence of lactobacilli [Lactobacillus (L.) casei, L. paracasei, and L. rhamnosus] or Staphylococcus (S.) aureus in feces is related to the risk for allergy. Here, we used the same prospective longitudinal birth cohort of children to investigate early-life environmental exposures and their influence on EBV and CMV contraction over time. Since gut microbes also belong to this category of early exposures, we investigated their association with herpesvirus contraction. Our results show that these two viruses are acquired with different kinetics and that EBV and CMV seroprevalence at 10 years of age was 47 and 57%, respectively. We also observed that a delayed EBV or CMV infection was associated with older maternal age [time ratio (TR) 1.14, 95% confidence interval (CI) 1.07-1.21, P adj < 0.001 and TR 1.09, CI 1.03-1.16, P adj = 0.008, respectively]. Further, we present the novel finding that S. aureus colonization reduced the time to CMV acquisition (TR 0.21, CI 0.06-0.78, P adj = 0.02). Together, these findings suggest that there is a relationship between timing of herpesvirus acquisition and early-life immune modulating exposures, which interestingly also includes the early infant gut microbiota.
Collapse
Affiliation(s)
- Claudia Carvalho-Queiroz
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Maria A Johansson
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| | - Jan-Olov Persson
- Department of Mathematics, Stockholm University , Stockholm , Sweden
| | - Evelina Jörtsö
- Department of Clinical Science and Education, Karolinska Institute, Stockholm, Sweden; Sachs' Children's and Youth Hospital, Stockholm South General Hospital, Stockholm, Sweden
| | - Torbjörn Kjerstadius
- Department of Clinical Virology and Microbiology, Karolinska University Laboratory, Solna, Sweden; Department of Clinical Microbiology, Central Hospital, Karlstad, Sweden
| | - Caroline Nilsson
- Department of Clinical Science and Education, Karolinska Institute, Stockholm, Sweden; Sachs' Children's and Youth Hospital, Stockholm South General Hospital, Stockholm, Sweden
| | - Shanie Saghafian-Hedengren
- Department of Women's and Children's Health, Paediatric Oncology Unit, Astrid Lindgren Children's Hospital, Karolinska Institute , Sweden
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University , Stockholm , Sweden
| |
Collapse
|