1
|
Shi J, Chen L, Yuan X, Yang J, Xu Y, Shen L, Huang Y, Wang B, Yu F. A potential XGBoost Diagnostic Score for Staphylococcus aureus bloodstream infection. Front Immunol 2025; 16:1574003. [PMID: 40330459 PMCID: PMC12052945 DOI: 10.3389/fimmu.2025.1574003] [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: 02/10/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Staphylococcus aureus (S. aureus) bloodstream infection is often life-threatening, and increasing in incidence. We identified 63 differentially expressed genes (DEGs) in the GSE33341 S. aureus infection samples. Subsequently, intersecting the 63 DEGs with 950 genes from the blue module through weighted gene co-expression network analysis (WGCNA) yielded 38 genes. We leveraged Boruta and least absolute shrinkage and selection operator (LASSO) algorithms and identified5 diagnostic genes (DRAM1, UPP1, IL18RAP, CLEC4A, and PGLYRP1). Comparative analysis revealed that Extreme Gradient Boosting (XGBoost) surpassed SVM-RFE and Random Forest models, demonstrating superior diagnostic performance for S. aureus bloodstream infection (mean AUC for XGBoost =0.954; mean AUC for SVM-RFE =0.93275; mean AUC for Random Forest =0.94625). The XGBoost Diagnostic Score correlated with multiple immune cells to varying degrees, manifesting significant negative associations with CD8 T cells and CD4 naive T cells in both human and mouse samples. The diagnostic power of the Diagnostic Score was further validated by RT-qPCR results obtained from both mouse and patient samples, as well as RNA-Seq analysis conducted on mouse samples. XGBoost Diagnostic Score, consisting of DRAM1, UPP1, IL18RAP, CLEC4A, and PGLYRP1, may serve as a Diagnostic tool for S. aureus bloodstream infection.
Collapse
Affiliation(s)
- Junhong Shi
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lan Chen
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Xinru Yuan
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jinjin Yang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanlei Xu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Shen
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Huang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bingjie Wang
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fangyou Yu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
2
|
Saadawi A, Mair F, Rosenwald E, Hoces D, Slack E, Kopf M. Investigating Polyreactivity of CD4 + T Cells to the Intestinal Microbiota. Eur J Immunol 2025; 55:e202451484. [PMID: 40223653 DOI: 10.1002/eji.202451484] [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: 08/28/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025]
Abstract
Antigen-specific recognition of microbiota by T cells enforces tolerance at homeostasis. Conversely, dysbiosis leads to imbalanced T-cell responses, triggering inflammatory and autoimmune diseases. Despite their significance, the identities of immunogenic microbial antigens remain largely enigmatic. Here, we leveraged a sensitive, unbiased, genome-wide screening platform to identify peptides from Akkermansia muciniphila (AKK) and Bacteroides thetaiotaomicron (BT) recognized by CD4+ T cells. The platform is based on screening peptide libraries using an NFAT-fluorescence reporter cell line transduced with a retrovirus encoding an MHC-TCR (MCR) hybrid molecule. We discovered several novel epitopes from AKK and BT. T-cell hybridomas reactive to AKK and BT bacteria demonstrated polyreactivity to microbiota-derived peptides in co-cultures with MCR reporter cells. Steady-state T cells recognized these epitopes in an MHC-restricted fashion. Intriguingly, most of the identified epitopes are broadly conserved within the given phylum and originate from membrane and intracellular proteins. Ex vivo stimulation of CD4+ T cells from mice vaccinated with the identified peptides revealed mono-specific IFN-γ and IL-17 responses. Our work showcases the potential of the MCR system for identifying immunogenic microbial epitopes, providing a valuable resource. Our study facilitates decoding antigen specificity in immune system-bacterial interactions, with applications in understanding microbiome and pathogenic bacterial immunity.
Collapse
Affiliation(s)
- Ahmed Saadawi
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Florian Mair
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Esther Rosenwald
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Daniel Hoces
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
3
|
Zhang H, Monk IR, Braverman J, Jones CM, Brooks AG, Stinear TP, Wakim LM. Staphylococcal superantigens evoke temporary and reversible T cell anergy, but fail to block the development of a bacterium specific cellular immune response. Nat Commun 2024; 15:9872. [PMID: 39543088 PMCID: PMC11564628 DOI: 10.1038/s41467-024-54074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024] Open
Abstract
Superantigens (sAgs) are bacterial virulence factors that induce a state of immune hyperactivation by forming a bridge between certain subsets of T cell receptor (TCR) β chains on T lymphocytes, and class II major histocompatibility complex (MHC-II) molecules; this cross-linking leads to indiscriminate T cell activation, cytokine storm and toxic shock. Here we show that sAg exposure drives the preferential expansion of naive and central memory T cell subsets, but not effector or resident memory T cells, which instead, hyper release pro-inflammatory cytokines. A targeted therapeutic approach to minimise cytokine release by effector memory T cells attenuated sAg-induced cytokine release. Irrespective of antigen experience, sAg activation does not render mature T cells permanently dysfunctional, and full restoration of effector function is observed following a transient and reversible anergy. Moreover, we show that in the face of sAg induced immune hyperactivation, an intact bacterium-specific CD4+ T cell response can be mounted.
Collapse
Affiliation(s)
- Heran Zhang
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Ian R Monk
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Jessica Braverman
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Claerwen M Jones
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Andrew G Brooks
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Timothy P Stinear
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia
| | - Linda M Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
4
|
Kaempfer R. Subduing the Inflammatory Cytokine Storm. Int J Mol Sci 2024; 25:11194. [PMID: 39456976 PMCID: PMC11508503 DOI: 10.3390/ijms252011194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
The inflammatory cytokine response is essential for protective immunity, yet bacterial and viral pathogens often elicit an exaggerated response ("cytokine storm") harmful to the host that can cause multi-organ damage and lethality. Much has been published recently on the cytokine storm within the context of the coronavirus pandemic, yet bacterial sepsis, severe wound infections and toxic shock provide other prominent examples. The problem of the cytokine storm is compounded by the increasing incidence of multidrug-resistant bacterial strains. We created an incisive molecular tool for analyzing the role of the B7/CD28 costimulatory axis in the human inflammatory response. To attenuate the cytokine storm underlying infection pathology, yet preserve host defenses, we uniquely targeted the engagement of CD28 with its B7 co-ligands by means of short peptide mimetics of the human CD28 and B7 receptor homodimer interfaces. These peptides are not only effective tools for dissecting mechanism but also serve to attenuate the inflammatory response as a broad host-oriented therapeutic strategy against the cytokine storm. Indeed, such peptides protect mice from lethal Gram-positive bacterial superantigen-induced toxic shock even when dosed in molar amounts well below that of the superantigen and show promise in protecting humans from the severe inflammatory disease necrotizing soft tissue infections ('flesh-eating' bacterial sepsis) following traumatic wound injuries.
Collapse
Affiliation(s)
- Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| |
Collapse
|
5
|
Kashani B, Zandi Z, Pourbagheri-Sigaroodi A, Yousefi AM, Ghaffari SH, Bashash D. The PI3K signaling pathway; from normal lymphopoiesis to lymphoid malignancies. Expert Rev Anticancer Ther 2024; 24:493-512. [PMID: 38690706 DOI: 10.1080/14737140.2024.2350629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/29/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION As a vital mechanism of survival, lymphopoiesis requires the collaboration of different signaling molecules to orchestrate each step of cell development and maturation. The PI3K pathway is considerably involved in the maturation of lymphatic cells and therefore, its dysregulation can immensely affect human well-being and cause some of the most prevalent malignancies. As a result, studies that investigate this pathway could pave the way for a better understanding of the lymphopoiesis mechanisms, the undesired changes that lead to cancer progression, and how to design drugs to solve this issue. AREAS COVERED The present review addresses the aforementioned aspects of the PI3K pathway and helps pave the way for future therapeutic approaches. In order to access the articles, databases such as Medicine Medline/PubMed, Scopus, Google Scholar, and Science Direct were utilized. The search formula was established by identifying main keywords including PI3K/Akt/mTOR pathway, Lymphopoiesis, Lymphoid malignancies, and inhibitors. EXPERT OPINION The PI3K pathway is crucial for lymphocyte development and differentiation, making it a potential target for therapeutic intervention in lymphoid cancers. Studies are focused on developing PI3K inhibitors to impede the progression of hematologic malignancies, highlighting the pathway's significance in lymphoma and lymphoid leukemia.
Collapse
Affiliation(s)
- Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Zhao JB, Fan MZ, Shi YX, Zhu YT, Gao SX, Li GL, Guan JC, Zhou P. Staphylococcal enterotoxin B exposed to pregnant rats inhibits the hedgehog signaling pathway in thymic T lymphocytes of the offspring. Microb Pathog 2024; 192:106723. [PMID: 38823465 DOI: 10.1016/j.micpath.2024.106723] [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: 10/30/2023] [Revised: 05/17/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
The Hedgehog (Hh) signaling pathway is involved in T cell differentiation and development and plays a major regulatory part in different stages of T cell development. A previous study by us suggested that prenatal exposure to staphylococcal enterotoxin B (SEB) changed the percentages of T cell subpopulation in the offspring thymus. However, it is unclear whether prenatal SEB exposure impacts the Hh signaling pathway in thymic T cells. In the present study, pregnant rats at gestational day 16 were intravenously injected once with 15 μg SEB, and the thymi of both neonatal and adult offspring rats were aseptically acquired to scrutinize the effects of SEB on the Hh signaling pathway. It firstly found that prenatal SEB exposure clearly caused the increased expression of Shh and Dhh ligands of the Hh signaling pathway in thymus tissue of both neonatal and adult offspring rats, but significantly decreased the expression levels of membrane receptors of Ptch1 and Smo, transcription factor Gli1, as well as target genes of CyclinD1, C-myc, and N-myc in Hh signaling pathway of thymic T cells. These data suggest that prenatal SEB exposure inhibits the Hh signaling pathway in thymic T lymphocytes of the neonatal offspring, and this effect can be maintained in adult offspring via the imprinting effect.
Collapse
Affiliation(s)
- Jia-Bao Zhao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Meng-Zhu Fan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Yin-Xing Shi
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Yu-Ting Zhu
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Shu-Xian Gao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Guang-Lin Li
- Majored in Biological Science, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jun-Chang Guan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| | - Ping Zhou
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| |
Collapse
|
7
|
Rampersadh K, Salie MT, Engel KC, Moodley C, Zühlke LJ, Engel ME. Presence of Group A streptococcus frequently assayed virulence genes in invasive disease: a systematic review and meta-analysis. Front Cell Infect Microbiol 2024; 14:1337861. [PMID: 39055978 PMCID: PMC11270091 DOI: 10.3389/fcimb.2024.1337861] [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: 11/13/2023] [Accepted: 01/18/2024] [Indexed: 07/28/2024] Open
Abstract
Introduction It is currently unclear what the role of Group A streptococcus (GAS) virulence factors (VFs) is in contributing to the invasive potential of GAS. This work investigated the evidence for the association of GAS VFs with invasive disease. Methods We employed a broad search strategy for studies reporting the presence of GAS VFs in invasive and non-invasive GAS disease. Data were independently extracted by two reviewers, quality assessed, and meta-analyzed using Stata®. Results A total of 32 studies reported on 45 putative virulence factors [invasive (n = 3,236); non-invasive (n = 5,218)], characterized by polymerase chain reaction (PCR) (n = 30) and whole-genome sequencing (WGS) (n = 2). The risk of bias was rated as low and moderate, in 23 and 9 studies, respectively. Meta-,analyses of high-quality studies (n = 23) revealed a significant association of speM [OR, 1.64 (95%CI, 1.06; 2.52)] with invasive infection. Meta-analysis of WGS studies demonstrated a significant association of hasA [OR, 1.91 (95%CI, 1.36; 2.67)] and speG [OR, 2.83 (95%CI, 1.63; 4.92)] with invasive GAS (iGAS). Meta-analysis of PCR studies indicated a significant association of speA [OR, 1.59 (95%CI, 1.10; 2.30)] and speK [OR, 2.95 (95%CI, 1.81; 4.80)] with invasive infection. A significant inverse association was observed between prtf1 [OR, 0.42 (95%CI, 0.20; 0.87)] and invasive infection. Conclusion This systematic review and genomic meta-analysis provides evidence of a statistically significant association with invasive infection for the hasA gene, while smeZ, ssa, pnga3, sda1, sic, and NaDase show statistically significantly inverse associations with invasive infection. SpeA, speK, and speG are associated with GAS virulence; however, it is unclear if they are markers of invasive infection. This work could possibly aid in developing preventative strategies.
Collapse
Affiliation(s)
- Kimona Rampersadh
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - M. Taariq Salie
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Kelin C. Engel
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Clinton Moodley
- Department of Pathology, Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- The National Health Laboratory Service, Microbiology, Groote Schuur Hospital, Cape Town, South Africa
| | - Liesl J. Zühlke
- Division of Paediatric Cardiology, Department of Paediatrics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Parrow Valley, Cape Town, South Africa
| | - Mark E. Engel
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Parrow Valley, Cape Town, South Africa
| |
Collapse
|
8
|
Rodríguez-Rodríguez N, Rosetti F, Crispín JC. CD8 is down(regulated) for tolerance. Trends Immunol 2024; 45:442-453. [PMID: 38782625 DOI: 10.1016/j.it.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024]
Abstract
Activated CD8+ T cells directly kill target cells. Therefore, the regulation of their function is central to avoiding immunopathology. Mechanisms that curb effector functions in CD4+ and CD8+ T cells are mostly shared, yet important differences occur. Here, we focus on the control of CD8+ T cell activity and discuss the importance of a poorly understood aspect of tolerance that directly impairs engagement of target cells: the downregulation of CD8. We contextualize this process and propose that it represents a key element during CD8+ T cell modulation.
Collapse
Affiliation(s)
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C Crispín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, Mexico.
| |
Collapse
|
9
|
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
|
10
|
Tsoukas P, Yeung RSM. Kawasaki Disease-Associated Cytokine Storm Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:365-383. [PMID: 39117827 DOI: 10.1007/978-3-031-59815-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Kawasaki disease (KD) is a hyperinflammatory syndrome manifesting as an acute systemic vasculitis characterized by fever, nonsuppurative conjunctival injection, rash, oral mucositis, extremity changes, and cervical lymphadenopathy. KD predominantly affects young children and shares clinical features and immunobiology with other hyperinflammation syndromes including systemic juvenile idiopathic arthritis (sJIA) and multisystem inflammatory syndrome in children (MIS-C). Cytokine storm syndrome (CSS) is an acute complication in ~2% of KD patients; however, the incidence is likely underestimated as many clinical and laboratory features of both diseases overlap. CSS should be entertained when a child with KD is unresponsive to IVIG therapy with recalcitrant fever. Early recognition and prompt institution of immunomodulatory treatment can substantially reduce the mortality and morbidity of CSS in KD. Given the known pathogenetic role of IL-1β in both syndromes, the early use of IL-1 blockers in refractory KD with CSS deserves consideration.
Collapse
Affiliation(s)
- Paul Tsoukas
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Rae S M Yeung
- Division of Rheumatology, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Paediatrics, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Pallesen EMH, Gluud M, Vadivel CK, Buus TB, de Rooij B, Zeng Z, Ahmad S, Willerslev-Olsen A, Röhrig C, Kamstrup MR, Bay L, Lindahl L, Krejsgaard T, Geisler C, Bonefeld CM, Iversen L, Woetmann A, Koralov SB, Bjarnsholt T, Frieling J, Schmelcher M, Ødum N. Endolysin Inhibits Skin Colonization by Patient-Derived Staphylococcus Aureus and Malignant T-Cell Activation in Cutaneous T-Cell Lymphoma. J Invest Dermatol 2023; 143:1757-1768.e3. [PMID: 36889662 DOI: 10.1016/j.jid.2023.01.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 12/15/2022] [Accepted: 01/10/2023] [Indexed: 03/08/2023]
Abstract
Staphylococcus aureus is suspected to fuel disease activity in cutaneous T-cell lymphomas. In this study, we investigate the effect of a recombinant, antibacterial protein, endolysin (XZ.700), on S. aureus skin colonization and malignant T-cell activation. We show that endolysin strongly inhibits the proliferation of S. aureus isolated from cutaneous T-cell lymphoma skin and significantly decreases S. aureus bacterial cell counts in a dose-dependent manner. Likewise, ex vivo colonization of both healthy and lesional skin by S. aureus is profoundly inhibited by endolysin. Moreover, endolysin inhibits the patient-derived S. aureus induction of IFNγ and the IFNγ-inducible chemokine CXCL10 in healthy skin. Whereas patient-derived S. aureus stimulates activation and proliferation of malignant T cells in vitro through an indirect mechanism involving nonmalignant T cells, endolysin strongly inhibits the effects of S. aureus on activation (reduced CD25 and signal transducer and activator of transcription 5 phosphorylation) and proliferation (reduced Ki-67) of malignant T cells and cell lines in the presence of nonmalignant T cells. Taken together, we provide evidence that endolysin XZ.700 inhibits skin colonization, chemokine expression, and proliferation of pathogenic S. aureus and blocks their potential tumor-promoting effects on malignant T cells.
Collapse
Affiliation(s)
- Emil M H Pallesen
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Maria Gluud
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Chella Krishna Vadivel
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Terkild B Buus
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Bob de Rooij
- Micreos Human Health B.V., Bilthoven, the Netherlands
| | - Ziao Zeng
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sana Ahmad
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Willerslev-Olsen
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Maria R Kamstrup
- Department of Dermatology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Lene Bay
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lise Lindahl
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Thorbjørn Krejsgaard
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Geisler
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte M Bonefeld
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Woetmann
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sergei B Koralov
- Department of Pathology, NYU School of Medicine, New York, New York, USA
| | - Thomas Bjarnsholt
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Tanaka A, Maeda S, Nomura T, Llamas-Covarrubias MA, Tanaka S, Jin L, Lim EL, Morikawa H, Kitagawa Y, Akizuki S, Ito Y, Fujimori C, Hirota K, Murase T, Hashimoto M, Higo J, Zamoyska R, Ueda R, Standley DM, Sakaguchi N, Sakaguchi S. Construction of a T cell receptor signaling range for spontaneous development of autoimmune disease. J Exp Med 2023; 220:213728. [PMID: 36454183 PMCID: PMC9718937 DOI: 10.1084/jem.20220386] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 10/06/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
Thymic selection and peripheral activation of conventional T (Tconv) and regulatory T (Treg) cells depend on TCR signaling, whose anomalies are causative of autoimmunity. Here, we expressed in normal mice mutated ZAP-70 molecules with different affinities for the CD3 chains, or wild type ZAP-70 at graded expression levels under tetracycline-inducible control. Both manipulations reduced TCR signaling intensity to various extents and thereby rendered those normally deleted self-reactive thymocytes to become positively selected and form a highly autoimmune TCR repertoire. The signal reduction more profoundly affected Treg development and function because their TCR signaling was further attenuated by Foxp3 that physiologically repressed the expression of TCR-proximal signaling molecules, including ZAP-70, upon TCR stimulation. Consequently, the TCR signaling intensity reduced to a critical range generated pathogenic autoimmune Tconv cells and concurrently impaired Treg development/function, leading to spontaneous occurrence of autoimmune/inflammatory diseases, such as autoimmune arthritis and inflammatory bowel disease. These results provide a general model of how altered TCR signaling evokes autoimmune disease.
Collapse
Affiliation(s)
- Atsushi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Department of Frontier Research in Tumor Immunology, Center of Medical Innovation and Translational Research, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shinji Maeda
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nomura
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mara Anais Llamas-Covarrubias
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Institute of Research in Biomedical Sciences, University Center of Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Satoshi Tanaka
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Lin Jin
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ee Lyn Lim
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Hiromasa Morikawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Yohko Kitagawa
- Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shuji Akizuki
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshinaga Ito
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Chihiro Fujimori
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Tosei Murase
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Motomu Hashimoto
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Junichi Higo
- Institute for Protein Research, Osaka University, Suita, Japan
| | - Rose Zamoyska
- Institute for Immunology and Infection Research, The University of Edinburgh, Edinburgh, UK
| | - Ryuzo Ueda
- Department of Tumor Immunology, Aichi Medical University School of Medicine, Aichi, Japan
| | - Daron M Standley
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Noriko Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.,Laboratory of Experimental Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
13
|
Medetgul-Ernar K, Davis MM. Standing on the shoulders of mice. Immunity 2022; 55:1343-1353. [PMID: 35947979 PMCID: PMC10035762 DOI: 10.1016/j.immuni.2022.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/13/2023]
Abstract
While inbred mice have informed most of what we know about the immune system in the modern era, they have clear limitations with respect to their ability to be informative regarding genetic heterogeneity or microbial influences. They have also not been very predictive as models of human disease or vaccination results. Although there are concerted attempts to compensate for these flaws, the rapid rise of human studies, driven by both technical and conceptual advances, promises to fill in these gaps, as well as provide direct information about human diseases and vaccination responses. Work on human immunity has already provided important additional perspectives on basic immunology such as the importance of clonal deletion to self-tolerance, and while many challenges remain, it seems inevitable that "the human model" will continue to inform us about the immune system and even allow for the discovery of new mechanisms.
Collapse
Affiliation(s)
- Kwat Medetgul-Ernar
- Immunology Program, Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Mark M Davis
- Howard Hughes Medical Institute, Institute for Immunity, Transplantation and Infection, Department of Microbiology and Immunology, Stanford University School of Medicine, Palo Alto, CA 94304, USA.
| |
Collapse
|
14
|
Noorbakhsh S, Ashouri S, Moradkhani M. Role of Superantigens In Various Childhood Inflamatory Diseases. Infect Disord Drug Targets 2022; 22:IDDT-EPUB-124062. [PMID: 35638540 DOI: 10.2174/1871526522666220530141031] [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: 10/20/2022] [Revised: 01/14/2022] [Accepted: 01/28/2022] [Indexed: 06/15/2023]
Abstract
----:: Super antigens (Sags) are some part of virus or bacteria proteins which stimulate T cells and antigen-presenting cells leading to systemic immune repose and inflammation. ---SAgs might have possible role in in various inflammatory childhood diseases (eg Kawasaki disease, atopic dermatitis, and chronic rhinosinusitis). ----Worldwide studies had done to determine the role of staphylococcal SAgs (TSST-1 ) in various inflammatory diseases. The SAgs (TSST-1) not only induce sepsis and septic shock (even in negative blood culture for S.aureus) ,but also might have significant role in various childhood inflammatory diseases ( eg KD, OMS, Polyp, dermatitis, psoriasis ) . In proven SAgs induced inflammatory diseases, inhibition the cell-destructive process by SAgs suppressants might be helpful. ----In toxic shock or sepsis like presentation even in cases with negative blood cultures immediate use pf anti staphylococcal drugs is required. ---Occasionaly, clinical presentation of some human viruses (eg coronavirus and adenovirus) mimic KD. ----- In addition ,coinfection with adenovirus, coronavirus, and para-influenza virus type 3 were observed with KD. -- Bacterial Sags induced increasement of acute-phase reactants and number of white blood cell, and neutrophil counts In developed KD. ----Multisystem inflammatory syndrome in children (MISC) and KS observed during the recent COVID-19 pandemia. This study summarized the relation between viral and bacterial SAgs and childhood inflammatory diseases.
Collapse
Affiliation(s)
- Samileh Noorbakhsh
- Pediatric Infectious Diseases department, Iran University of Medical Sciences, Tehran, Iran
| | - Sarvenaz Ashouri
- ENT and Head and Neck Research center and department, Iran university of medical sciences, Tehran, Iran
| | - Masoumeh Moradkhani
- Pediatric Resident ,Rasoul Hospital ,Iran university of medical sciences, Tehran, Iran
| |
Collapse
|
15
|
Shams H, Hollenbach JA, Matsunaga A, Mofrad MRK, Oksenberg JR, Didonna A. A short HLA-DRA isoform binds the HLA-DR2 heterodimer on the outer domain of the peptide-binding site. Arch Biochem Biophys 2022; 719:109156. [PMID: 35218721 PMCID: PMC9007275 DOI: 10.1016/j.abb.2022.109156] [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: 10/08/2021] [Revised: 02/06/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022]
Abstract
The human leukocyte antigen (HLA) locus encodes a large group of proteins governing adaptive and innate immune responses. Among them, HLA class II proteins form α/β heterodimers on the membrane of professional antigen-presenting cells (APCs), where they display both, self and pathogen-derived exogenous antigens to CD4+ T lymphocytes. We have previously shown that a shorter HLA-DRA isoform (sHLA-DRA) lacking 25 amino acids can be presented onto the cell membrane via binding to canonical HLA-DR2 heterodimers. Here, we employed atomistic molecular dynamics simulations to decipher the binding position of sHLA-DRA and its structural impact on functional regions of the HLA-DR2 molecule. We show that a loop region exposed only in the short isoform (residues R69 to G83) is responsible for binding HLA-DR2 on the outer domain of the peptide-binding site, and experimentally validated the critical role of F76 in mediating such interaction. Additionally, sHLA-DRA allosterically modifies the peptide-binding pocket conformation. In summary, this study unravels key molecular mechanisms underlying sHLA-DRA function, providing important insights into the role of full-length proteins in structural modulation of HLA class II receptors.
Collapse
Affiliation(s)
- Hengameh Shams
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Jill A Hollenbach
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, 94158, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, 94158, USA
| | - Atsuko Matsunaga
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Mohammad R K Mofrad
- Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Alessandro Didonna
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, 94158, USA; Department of Anatomy and Cell Biology, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
16
|
Nicotinamide Breaks Effector CD8 T cell Responses by Targeting mTOR Signaling. iScience 2022; 25:103932. [PMID: 35243268 PMCID: PMC8886054 DOI: 10.1016/j.isci.2022.103932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 11/22/2022] Open
|
17
|
Ahmad Z, Somanath PR. AKT Isoforms in the Immune Response in Cancer. Curr Top Microbiol Immunol 2022; 436:349-366. [PMID: 36243852 DOI: 10.1007/978-3-031-06566-8_15] [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: 06/16/2023]
Abstract
AKT is a protein kinase that exists in three isoforms: AKT1, AKT2, and AKT3. Though similar in structure, these isoforms display different effects. AKT is activated downstream of PI3K, and together, this signaling pathway helps regulate cellular processes including cell growth, proliferation, metabolism, survival, and apoptosis. Disruption in these pathways has been associated with disorders including cardiovascular diseases, developmental disorders, inflammatory responses, autoimmune diseases, neurologic disorders, type 2 diabetes, and several cancers. In cancer, deregulation in the PI3K/AKT pathway can be manifested as tumorigenesis, pathological angiogenesis, and metastasis. Increased activity has been correlated with tumor progression and resistance to cancer treatments. Recent studies have suggested that inhibition of the PI3K/AKT pathway plays a significant role in the development, expansion, and proliferation of cells of the immune system. Additionally, AKT has been found to play an important role in differentiating regulatory T cells, activating B cells, and augmenting tumor immunosurveillance. This emphasizes AKT as a potential target for inhibition in cancer therapy. This chapter reviews AKT structure and regulation, its different isoforms, its role in immune cells, and its modulation in oncotherapy.
Collapse
Affiliation(s)
- Zayd Ahmad
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Georgia Cancer Center, Vascular Biology Center and Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
18
|
Planeta Kepp K. Bioinorganic Chemistry of Zinc in Relation to the Immune System. Chembiochem 2021; 23:e202100554. [PMID: 34889510 DOI: 10.1002/cbic.202100554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/09/2021] [Indexed: 01/18/2023]
Abstract
Zinc is well-known to have a central role in human inflammation and immunity and is itself an anti-inflammatory and antiviral agent. Despite its massively documented role in such processes, the underlying chemistry of zinc in relation to specific proteins and pathways of the immune system has not received much focus. This short review provides an overview of this topic, with emphasis on the structures of key proteins, zinc coordination chemistry, and probable mechanisms involved in zinc-based immunity, with some focus points for future chemical and biological research.
Collapse
Affiliation(s)
- Kasper Planeta Kepp
- DTU Chemistry, Technical University of Denmark, Building 206, 2800, Kongens Lyngby, Denmark
| |
Collapse
|
19
|
Deacy AM, Gan SKE, Derrick JP. Superantigen Recognition and Interactions: Functions, Mechanisms and Applications. Front Immunol 2021; 12:731845. [PMID: 34616400 PMCID: PMC8488440 DOI: 10.3389/fimmu.2021.731845] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.
Collapse
Affiliation(s)
- Anthony M. Deacy
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, Experimental Drug Development Centre – Bioinformatics Institute (EDDC-BII), Agency for Science Technology and Research (ASTAR), 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
| |
Collapse
|
20
|
Banerjee S, Mohammed A, Wong HR, Palaniyar N, Kamaleswaran R. Machine Learning Identifies Complicated Sepsis Course and Subsequent Mortality Based on 20 Genes in Peripheral Blood Immune Cells at 24 H Post-ICU Admission. Front Immunol 2021; 12:592303. [PMID: 33692779 PMCID: PMC7937924 DOI: 10.3389/fimmu.2021.592303] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/28/2021] [Indexed: 01/08/2023] Open
Abstract
A complicated clinical course for critically ill patients admitted to the intensive care unit (ICU) usually includes multiorgan dysfunction and subsequent death. Owing to the heterogeneity, complexity, and unpredictability of the disease progression, ICU patient care is challenging. Identifying the predictors of complicated courses and subsequent mortality at the early stages of the disease and recognizing the trajectory of the disease from the vast array of longitudinal quantitative clinical data is difficult. Therefore, we attempted to perform a meta-analysis of previously published gene expression datasets to identify novel early biomarkers and train the artificial intelligence systems to recognize the disease trajectories and subsequent clinical outcomes. Using the gene expression profile of peripheral blood cells obtained within 24 h of pediatric ICU (PICU) admission and numerous clinical data from 228 septic patients from pediatric ICU, we identified 20 differentially expressed genes predictive of complicated course outcomes and developed a new machine learning model. After 5-fold cross-validation with 10 iterations, the overall mean area under the curve reached 0.82. Using a subset of the same set of genes, we further achieved an overall area under the curve of 0.72, 0.96, 0.83, and 0.82, respectively, on four independent external validation sets. This model was highly effective in identifying the clinical trajectories of the patients and mortality. Artificial intelligence systems identified eight out of twenty novel genetic markers (SDC4, CLEC5A, TCN1, MS4A3, HCAR3, OLAH, PLCB1, and NLRP1) that help predict sepsis severity or mortality. While these genes have been previously associated with sepsis mortality, in this work, we show that these genes are also implicated in complex disease courses, even among survivors. The discovery of eight novel genetic biomarkers related to the overactive innate immune system, including neutrophil function, and a new predictive machine learning method provides options to effectively recognize sepsis trajectories, modify real-time treatment options, improve prognosis, and patient survival.
Collapse
Affiliation(s)
- Shayantan Banerjee
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, India
| | - Akram Mohammed
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Hector R. Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
21
|
Chiu ES, VandeWoude S. Endogenous Retroviruses Drive Resistance and Promotion of Exogenous Retroviral Homologs. Annu Rev Anim Biosci 2020; 9:225-248. [PMID: 33290087 DOI: 10.1146/annurev-animal-050620-101416] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Endogenous retroviruses (ERVs) serve as markers of ancient viral infections and provide invaluable insight into host and viral evolution. ERVs have been exapted to assist in performing basic biological functions, including placentation, immune modulation, and oncogenesis. A subset of ERVs share high nucleotide similarity to circulating horizontally transmitted exogenous retrovirus (XRV) progenitors. In these cases, ERV-XRV interactions have been documented and include (a) recombination to result in ERV-XRV chimeras, (b) ERV induction of immune self-tolerance to XRV antigens, (c) ERV antigen interference with XRV receptor binding, and (d) interactions resulting in both enhancement and restriction of XRV infections. Whereas the mechanisms governing recombination and immune self-tolerance have been partially determined, enhancement and restriction of XRV infection are virus specific and only partially understood. This review summarizes interactions between six unique ERV-XRV pairs, highlighting important ERV biological functions and potential evolutionary histories in vertebrate hosts.
Collapse
Affiliation(s)
- Elliott S Chiu
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA; ,
| | - Sue VandeWoude
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado 80523, USA; ,
| |
Collapse
|
22
|
Bhattacharya M, Sharma AR, Sharma G, Patra P, Mondal N, Patra BC, Lee SS, Chakraborty C. Computer aided novel antigenic epitopes selection from the outer membrane protein sequences of Aeromonas hydrophila and its analyses. INFECTION GENETICS AND EVOLUTION 2020; 82:104320. [PMID: 32298854 DOI: 10.1016/j.meegid.2020.104320] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Gram-negative bacteria are among the causative microorganisms for zoonotic diseases in humans and teleosts. Outer membrane proteins (Omps) of Aeromonas hydrophila, a gram-negative bacterium, are critical for the subcellular integration to eukaryotic cell that can modulate the functions of macrophages. Hence Omps are recognized as immune markers for the vaccine development. METHODS In the present study, a 3-D model of Omps was identified using in silico technique and recognized through the Swiss model web-server and confirmed with Procheck and ProSA server.. The B-cell binding sites of the protein were selected from sequence alignment.. Further, the identification of B-cell epitope was carried out using modules of BCpred server (i.e., BCPred and Amino Acid Pairs). The identified antigenic amino acid sequences for B-cells were used to determine the T-cell epitope (both MHC I & II allelic binding sequences) using ProPred 1 and ProPred servers. RESULTS The epitopic regions (9 mer: LAGKTTNES and GFDGSQYGK) in the Omps that are bound together with the MHC molecules (MHC-I & II), and have maximum possible numbers of MHC alleles are recognized. It was observed that Omps of A. hydrophila are conserved across the serotypes and are immunogenic. These epitopes can stimulate significant immune responses and can be advantageous while preparing peptide-based vaccines against A. hydrophila infections. Thus, suggesting the use of Omps in the development of vaccines and immunotherapeutics against the bacterial diseases in humans and teleosts.
Collapse
Affiliation(s)
- Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore 756020, Odisha, India; Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Garima Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chuncheon-si 24341, Gangwon-do, Republic of Korea
| | - Prasanta Patra
- Centre For Aquaculture Research, Extension & Livelihood, Department of Aquaculture Management & Technology, Vidyasagar University, Midnapore 721 102, West Bengal, India
| | - Niladri Mondal
- Centre For Aquaculture Research, Extension & Livelihood, Department of Aquaculture Management & Technology, Vidyasagar University, Midnapore 721 102, West Bengal, India
| | - Bidhan Chandra Patra
- Centre For Aquaculture Research, Extension & Livelihood, Department of Aquaculture Management & Technology, Vidyasagar University, Midnapore 721 102, West Bengal, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea.
| | - Chiranjib Chakraborty
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea; Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Rd, Kolkata, West Bengal 700126, India.
| |
Collapse
|
23
|
Lieberman JA, Stansbury LG, Kufera JA, Chiu WC, Punch LJ, Hess JR, Scalea TM, Henry SM. Red Blood Cell Transfusions and Anemia on Admission Are Associated with Poor Outcomes in Necrotizing Soft Tissue Infections. J Appl Lab Med 2019; 3:250-260. [PMID: 33636946 DOI: 10.1373/jalm.2017.025643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/27/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND Necrotizing soft tissue infections (NSTIs) are highly morbid infections often requiring critical care and transfusion support. We explored a large 2-year experience from a regional trauma center with a dedicated soft tissue service (STS) in an attempt to identify factors in current care with potential for improving outcomes for these critically ill patients. METHODS New adult (>17 years) STS admissions, 2008-2009, were identified from the Trauma Registry. Patient records were extracted and assessed via descriptive statistics, univariate analysis, and multivariable logistic regression models. RESULTS Mortality among 253 eligible primary admissions was 8.3% overall and 10.3% for those with an admission diagnosis of NSTI. No significant differences in wound characteristics, use of VAC (vacuum-assisted closure) dressing or hyperbaric oxygen, or wound microbiology emerged between survivors and nonsurvivors. Median time to first debridement was 5 h (interquartile range, 2-21 h). Multivariable modeling indicated association of worse outcome (death or discharge to chronic/rehab care) with age >60 years [odds ratio (OR), 3.82; P < 0.001], anemia (OR, 0.98; P = 0.03), increasing number of transfusions (OR, 1.09; P < 0.001), NSTI diagnosis (OR, 2.47; P = 0.005), preexisting diabetes mellitus (OR, 3.20; P = 0.001), and low admission hemoglobin (OR, 0.80; P = 0.004). CONCLUSIONS Mortality was less than previously reported. Number of transfusions and anemia at admission emerged as risk factors for poor outcomes. Future research should focus on the effects of transfusion on NSTI outcomes, on potentially confounding factors, and on whether a restrictive transfusion strategy reduces mortality.
Collapse
Affiliation(s)
- Joshua A Lieberman
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Lynn G Stansbury
- Shock Trauma and Anesthesia Research Center, R. Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD
| | - Joseph A Kufera
- Shock Trauma and Anesthesia Research Center, R. Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD
| | - William C Chiu
- R. Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD
| | - Laurie J Punch
- Acute and Critical Care Surgery, Washington University School of Medicine, St Louis, MO
| | - John R Hess
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | - Thomas M Scalea
- R. Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD
| | - Sharon M Henry
- R. Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD
| |
Collapse
|
24
|
Bottasso E. Toward the Existence of a Sympathetic Neuroplasticity Adaptive Mechanism Influencing the Immune Response. A Hypothetical View-Part II. Front Endocrinol (Lausanne) 2019; 10:633. [PMID: 31620088 PMCID: PMC6760024 DOI: 10.3389/fendo.2019.00633] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/30/2019] [Indexed: 01/16/2023] Open
Abstract
In the preceding work, a hypothesis on the existence of a specific neural plasticity program from sympathetic fibers innervating secondary lymphoid organs was introduced. This proposed adaptive mechanism would involve segmental retraction and degeneration of noradrenergic terminals during the immune system (IS) activation followed by regeneration once the IS returns to the steady-state. Starting from such view, this second part presents clinical and experimental evidence allowing to envision that this sympathetic neural plasticity mechanism is also operative on inflamed non-lymphoid peripheral tissues. Importantly, the sympathetic nervous system regulates most of the physiological bodily functions, ranging from cardiovascular, respiratory and gastro-intestinal functions to endocrine and metabolic ones, among others. Thus, it seems sensible to think that compensatory programs should be put into place during inflammation in non-lymphoid tissues as well, to avoid the possible detrimental consequences of a sympathetic blockade. Nevertheless, in many pathological scenarios like severe sepsis, chronic inflammatory diseases, or maladaptive immune responses, such compensatory programs against noradrenergic transmission impairment would fail to develop. This would lead to a manifest sympathetic dysfunction in the above-mentioned settings, partly accounting for their underlying pathophysiological basis; which is also discussed. The physiological/teleological significance for the whole neural plasticity process is postulated, as well.
Collapse
Affiliation(s)
- Emanuel Bottasso
- Departments of Pathology and Physiology, Faculty of Medicine, Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Rosario, Argentina
| |
Collapse
|
25
|
Minasyan H. Sepsis: mechanisms of bacterial injury to the patient. Scand J Trauma Resusc Emerg Med 2019; 27:19. [PMID: 30764843 PMCID: PMC6376788 DOI: 10.1186/s13049-019-0596-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 12/17/2022] Open
Abstract
In bacteremia the majority of bacterial species are killed by oxidation on the surface of erythrocytes and digested by local phagocytes in the liver and the spleen. Sepsis-causing bacteria overcome this mechanism of human innate immunity by versatile respiration, production of antioxidant enzymes, hemolysins, exo- and endotoxins, exopolymers and other factors that suppress host defense and provide bacterial survival. Entering the bloodstream in different forms (planktonic, encapsulated, L-form, biofilm fragments), they cause different types of sepsis (fulminant, acute, subacute, chronic, etc.). Sepsis treatment includes antibacterial therapy, support of host vital functions and restore of homeostasis. A bacterium killing is only one of numerous aspects of antibacterial therapy. The latter should inhibit the production of bacterial antioxidant enzymes and hemolysins, neutralize bacterial toxins, modulate bacterial respiration, increase host tolerance to bacterial products, facilitate host bactericidal mechanism and disperse bacterial capsule and biofilm.
Collapse
|
26
|
Heterogeneity in FoxP3- and GARP/LAP-Expressing T Regulatory Cells in an HLA Class II Transgenic Murine Model of Necrotizing Soft Tissue Infections by Group A Streptococcus. Infect Immun 2018; 86:IAI.00432-18. [PMID: 30224551 DOI: 10.1128/iai.00432-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/03/2018] [Indexed: 11/20/2022] Open
Abstract
Invasive group A streptococcus (GAS) infections include necrotizing soft tissue infections (NSTI) and streptococcal toxic shock syndrome (STSS). We have previously shown that host HLA class II allelic variations determine the risk for necrotizing fasciitis (NF), a dominant subgroup of NSTI, and STSS by modulating responses to GAS superantigens (SAgs). SAgs are pivotal mediators of uncontrolled T-cell activation, triggering a proinflammatory cytokine storm in the host. FoxP3-expressing CD4+ CD25+ T regulatory cells (Tregs) comprise phenotypically and functionally heterogeneous subsets with a profound ability to suppress inflammatory responses. Specifically, activated Tregs, which express glycoprotein A repetitions predominant (GARP) and display latent transforming growth factor β1 (TGF-β1) complexes (latency-associated peptide [LAP]), exhibit strong immunosuppressive functions. The significance of Tregs that may participate in suppressing inflammatory responses during NSTI is unknown. Here, we phenotypically characterized FoxP3/GARP/LAP-expressing Tregs in GAS-infected or SAg (SmeZ)-stimulated splenocytes from transgenic (tg) mice expressing human HLA-II DRB1*15 (DR15 allele associated with nonsevere NF/STSS-protective responses) or DRB1*0402/DQB1*0302 (DR4/DQ8 alleles associated with neutral risk for combined NF/STSS). We demonstrated both in vivo and in vitro that the neutral-risk allele upregulates expression of CD4+ CD25+ activated effector T cells, with a significantly lower frequency of Foxp3+/GARP+ LAP- but higher frequency of Foxp3- LAP+ Tregs than seen with the protective allele. Additional in vitro studies revealed that the presentation of SmeZ by the neutral-risk allele significantly increases proliferation and expression of effector cytokines gamma interferon (IFN-γ) and interleukin-2 (IL-2) and upregulates CD4+ CD25+ T cell receptors (TCRs) carrying specific Vβ 11 chain (TCRVβ11+) T cells and Th1 transcription factor Tbx21 mRNA levels. Our data suggest that neutral-risk alleles may drive Th1 differentiation while attenuating the induction of Tregs associated with suppressive function.
Collapse
|
27
|
Pierson JD, Hansmann MA, Davis CC, Forney LJ. The effect of vaginal microbial communities on colonization by Staphylococcus aureus with the gene for toxic shock syndrome toxin 1 (TSST-1): a case-control study. Pathog Dis 2018; 76:4919727. [PMID: 29762733 DOI: 10.1093/femspd/fty015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
Menstrual toxic shock syndrome is associated with vaginal colonization by Staphylococcus aureus strains that encode toxic shock syndrome toxin 1 (tst+). Interestingly, a small proportion of women are colonized by S. aureus tst+ but do not have symptoms of toxic shock syndrome. Here we sought to determine if differences in the species composition of vaginal bacterial communities reflect a differential risk of colonization by S. aureus capable of producing toxic shock syndrome toxin 1 (TSST-1). The composition of vaginal communities of women that were or were not colonized with S. aureus tst+ were compared based on terminal restriction fragment length polymorphism (T-RFLP) profiles and sequences of cloned 16S rRNA genes. There were no detectable differences in community composition or species rank abundance between communities of women vaginally colonized with S. aureus tst+ as compared to those that were not. Phylogenetic analysis of cloned 16S rRNA gene sequences showed that the predominant members of communities of women colonized with S. aureus tst+ were indistinguishable from those of other healthy women. The data suggest that the numerically dominant members of vaginal communities do not preclude colonization and proliferation of S. aureus tst+ within indigenous microbial communities of the vagina.
Collapse
Affiliation(s)
- Jacob D Pierson
- Institute for Bioinformatics and Evolutionary Studies and Department of Biological Sciences University of Idaho, Moscow, ID 83844-3051, USA
| | - Melanie A Hansmann
- FemCare Product Safety and Regulatory Affairs, The Procter and Gamble Company, Cincinnati, OH 45224, USA
| | - Catherine C Davis
- FemCare Product Safety and Regulatory Affairs, The Procter and Gamble Company, Cincinnati, OH 45224, USA
| | - Larry J Forney
- Institute for Bioinformatics and Evolutionary Studies and Department of Biological Sciences University of Idaho, Moscow, ID 83844-3051, USA
| |
Collapse
|
28
|
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
29
|
Muluk NB, Altın F, Cingi C. Role of Superantigens in Allergic Inflammation: Their Relationship to Allergic Rhinitis, Chronic Rhinosinusitis, Asthma, and Atopic Dermatitis. Am J Rhinol Allergy 2018; 32:502-517. [PMID: 30253652 DOI: 10.1177/1945892418801083] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Our intention was to review all material published to date regarding superantigens (SAgs) and allergy from an otorhinolaryngological viewpoint to understand this association more clearly. METHODS We identified all materials published mentioning both SAg and allergic rhinitis (AR), chronic sinusitis, asthma, and atopic dermatitis (AD) that are indexed on PubMed, Google, or the ProQuest Central databases. RESULTS Staphylococcus aureus is a significant bacterial pathogen in humans and has the ability to produce enterotoxins with superantigenic features. The inflammatory response in allergy seen in both B cell and T cell may be attributed to SAgs. Sufferers of both allergic asthma with rhinitis and AR alone produce serological evidence of immunoglobulin E formation to SAgs produced by S. aureus. Perennial AR sufferers carry S. aureus more frequently and the presence of the organism within the nasal cavity may exacerbate perennial AR. SAg produced by S. aureus potentially worsens the asthmatic inflammatory response within the airway and may lead to the airways becoming hyperresponsive, as well as possibly activating T cells if asthmatic control is poor. Staphylococcal SAgs potentially increase the risk of developing chronic rhinosinusitis with nasal polyposis, additionally being a marker for more severe disease. If SAgs bring about chronic inflammatory responses in the nose and sinuses, then T cells excreting interferon-gamma may be a crucial mediator. In allergic dermatitis, S. aureus could be a key player in exacerbation of the condition. Even in younger pediatric patients with allergic dermatitis, allergic hypersensitivity to SAgs is frequent and may be a factor explaining how severe the condition becomes. CONCLUSION Just as SAgs are known to feature in many allergic conditions, they play their part in AR, chronic rhinosinusitis, asthma, and AD. Further research is required before the relationship between SAgs and allergy can be adequately explained.
Collapse
Affiliation(s)
- Nuray Bayar Muluk
- 1 Department of Otorhinolaryngology, Medical Faculty, Kirikkale University, Kirikkale, Turkey
| | - Fazilet Altın
- 2 ENT Clinics, Haseki Training and Research Hospital, Istanbul, Turkey
| | - Cemal Cingi
- 3 Department of Otorhinolaryngology, Medical Faculty, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
30
|
Schmitz M, Roux X, Huttner B, Pugin J. Streptococcal toxic shock syndrome in the intensive care unit. Ann Intensive Care 2018; 8:88. [PMID: 30225523 PMCID: PMC6141408 DOI: 10.1186/s13613-018-0438-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 09/11/2018] [Indexed: 11/10/2022] Open
Abstract
The streptococcal toxic shock syndrome is a severe complication associated with invasive infections by group A streptococci. In spite of medical progresses in the care of patients with septic shock during the last decades, this condition has remained associated with a high mortality. Early recognition and multidisciplinary management are key to the care of patients with streptococcal toxic shock syndrome, with intensive and appropriate intensive support of failing organs, rapid diagnosis of infectious source(s), and surgical management. The epidemiology and risk factors for streptococcal toxic shock syndrome remain to be better studied, including the possible causal role of exposure to nonsteroidal anti-inflammatory drugs. In this review article, the authors review the current knowledge of streptococcal toxic shock syndrome and discuss the pathophysiology as well as its supportive and specific treatment.
Collapse
Affiliation(s)
- Marylin Schmitz
- Division of Intensive Care, Faculty of Medicine Geneva, University Hospitals of Geneva, University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| | - Xavier Roux
- Division of Intensive Care, Faculty of Medicine Geneva, University Hospitals of Geneva, University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| | - Benedikt Huttner
- Division of Infectious Diseases, Faculty of Medicine Geneva, University Hospitals of Geneva, University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| | - Jérôme Pugin
- Division of Intensive Care, Faculty of Medicine Geneva, University Hospitals of Geneva, University of Geneva, Rue Gabrielle-Perret-Gentil 4, 1211 Geneva 14, Switzerland
| |
Collapse
|
31
|
Fähnrich A, Klein S, Sergé A, Nyhoegen C, Kombrink S, Möller S, Keller K, Westermann J, Kalies K. CD154 Costimulation Shifts the Local T-Cell Receptor Repertoire Not Only During Thymic Selection but Also During Peripheral T-Dependent Humoral Immune Responses. Front Immunol 2018; 9:1019. [PMID: 29867987 PMCID: PMC5966529 DOI: 10.3389/fimmu.2018.01019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/24/2018] [Indexed: 12/20/2022] Open
Abstract
CD154 is a transmembrane cytokine expressed transiently on activated CD4 T cells upon T-cell receptor (TCR) stimulation that interacts with CD40 on antigen-presenting cells. The signaling via CD154:CD40 is essential for B-cell maturation and germinal center formation and also for the final differentiation of CD4 T cells during T-dependent humoral immune responses. Recent data demonstrate that CD154 is critically involved in the selection of T-cell clones during the negative selection process in the thymus. Whether CD154 signaling influences the TCR repertoire during peripheral T-dependent humoral immune responses has not yet been elucidated. To find out, we used CD154-deficient mice and assessed the global TCRβ repertoire in T-cell zones (TCZ) of spleens by high-throughput sequencing after induction of a Th2 response to the multiepitopic antigen sheep red blood cells. Qualitative and quantitative comparison of the splenic TCZ-specific TCRβ repertoires revealed that CD154 deficiency shifts the distribution of Vβ-Jβ genes after antigen exposure. This data led to the conclusion that costimulation via CD154:CD40 during the interaction of T cells with CD40-matured B cells contributes to the recruitment of T-cell clones into the immune response and thereby shapes the peripheral TCR repertoire.
Collapse
Affiliation(s)
- Anke Fähnrich
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Sebastian Klein
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| | - Arnauld Sergé
- Centre de Recherche en Cancérologie de Marseille (CRCM) U1068 INSERM - UMR7258 CNRS - Institut Paoli Calmette, Aix-Marseille University, UM105, Marseille, France
| | | | - Sabrina Kombrink
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Karsten Keller
- Institute of Mathematics, University of Luebeck, Luebeck, Germany
| | | | - Kathrin Kalies
- Institute of Anatomy, University of Luebeck, Luebeck, Germany
| |
Collapse
|
32
|
Bernstein JM, Ballow M, Schlievert PM, Rich G, Allen C, Dryja D. A Superantigen Hypothesis for the Pathogenesis of Chronic Hyperplastic Sinusitis with Massive Nasal Polyposis. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/194589240301700601] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The pathogenesis of chronic hyperplastic sinusitis with massive nasal polyposis is still an enigma; however, the molecular biology of this disease is beginning to become unraveled and the proinflammatory cytokines and the message and the product of these cytokines have all been identified in nasal polyps. However, the initial trigger that causes inflammation of the lateral wall of the nose to up-regulate lymphocytes and eosinophils is still unknown. Methods Thirteen patients with massive polyposis were studied. The mucus of the nasal cavities surrounding the nasal polyps was studied for both bacterial and fungal species. The lymphocytes of the nasal polyps were extracted and evaluated for the T-cell receptor, particularly, the variable β region of this receptor. Enterotoxins (superantigens) of the bacteria were studied. Finally, the histopathology of nasal polyps was studied. Results Fifty-five percent of the patients had toxin-producing Staphylococcus aureus in the nasal mucus adjacent to the polyps. Three different enterotoxins were isolated, including Staphylococcus enterotoxin A, Staphylococcus enterotoxin B, and toxic shock syndrome toxin 1. The variable B specificity for these superantigens was identified also in the polyp lymphocyte T-cell receptor. Conclusion A superantigen hypothesis for massive polyposis is suggested because the most common bacterial species found in the nasal mucus is Staphylococcus aureus. These bacteria produce enterotoxins in all of the cases studied and the corresponding variable β region of the T-cell receptor also was up-regulated in the polyp lymphocytes in cases studied thus far. These data taken together suggest that the initial injury to the lateral wall of the nose may be the result of toxin-producing Staphylococci. Superantigens (enterotoxins) may up-regulate lymphocytes to produce cytokines that are responsible for the massive up-regulation of lymphocytes, eosinophils, and macrophages, the three most common inflammatory cells found in massive nasal polyposis.
Collapse
Affiliation(s)
- Joel M. Bernstein
- Departments of Otolaryngology and Pediatrics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, and the Division of Infectious Diseases, Children's Hospital of Buffalo, Buffalo, New York
| | - Mark Ballow
- Department of Pediatrics, State University of New York at Buffalo and Chief, Division of Allergy and Immunology, and Immunobiology Laboratory, Children's Hospital of Buffalo, Buffalo, New York
| | | | - Gary Rich
- Department of Pediatrics, State University of New York at Buffalo
| | - Cheryl Allen
- Department of Pediatrics, State University of New York at Buffalo
| | - Diane Dryja
- Department of Microbiology, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
33
|
Calvo V, Izquierdo M. Imaging Polarized Secretory Traffic at the Immune Synapse in Living T Lymphocytes. Front Immunol 2018; 9:684. [PMID: 29681902 PMCID: PMC5897431 DOI: 10.3389/fimmu.2018.00684] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/20/2018] [Indexed: 12/20/2022] Open
Abstract
Immune synapse (IS) formation by T lymphocytes constitutes a crucial event involved in antigen-specific, cellular and humoral immune responses. After IS formation by T lymphocytes and antigen-presenting cells, the convergence of secretory vesicles toward the microtubule-organizing center (MTOC) and MTOC polarization to the IS are involved in polarized secretion at the synaptic cleft. This specialized mechanism appears to specifically provide the immune system with a fine strategy to increase the efficiency of crucial secretory effector functions of T lymphocytes, while minimizing non-specific, cytokine-mediated stimulation of bystander cells, target cell killing and activation-induced cell death. The molecular bases involved in the polarized secretory traffic toward the IS in T lymphocytes have been the focus of interest, thus different models and several imaging strategies have been developed to gain insights into the mechanisms governing directional secretory traffic. In this review, we deal with the most widely used, state-of-the-art approaches to address the molecular mechanisms underlying this crucial, immune secretory response.
Collapse
Affiliation(s)
- Víctor Calvo
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
| | - Manuel Izquierdo
- Departamento de Bioquímica, Instituto de Investigaciones Biomédicas Alberto Sols CSIC-UAM, Madrid, Spain
| |
Collapse
|
34
|
Abstract
Recent progress in both conceptual and technological approaches to human immunology have rejuvenated a field that has long been in the shadow of the inbred mouse model. This is a healthy development both for the clinical relevance of immunology and for the fact that it is a way to gain access to the wealth of phenomenology in the many human diseases that involve the immune system. This is where we are likely to discover new immunological mechanisms and principals, especially those involving genetic heterogeneity or environmental influences that are difficult to model effectively in inbred mice. We also suggest that there are likely to be novel immunological mechanisms in long-lived, less fecund mammals such as human beings since they must remain healthy far longer than short-lived rodents in order for the species to survive.
Collapse
Affiliation(s)
- Mark M Davis
- Department of Microbiology and Immunology, The Howard Hughes Medical Institute, and the Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California 94305, USA;
| | - Petter Brodin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, 17121 Solna, Sweden.,Department of Neonatology, Karolinska University Hospital, 17176 Solna, Sweden
| |
Collapse
|
35
|
Pompura SL, Dominguez-Villar M. The PI3K/AKT signaling pathway in regulatory T-cell development, stability, and function. J Leukoc Biol 2018; 103:1065-1076. [PMID: 29357116 DOI: 10.1002/jlb.2mir0817-349r] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
The PI3K/AKT signaling pathway is an essential node in mammalian cells that controls cell growth, migration, proliferation, and metabolism. During the last decade, a number of works have demonstrated an important role for the PI3K/AKT pathway in regulatory T cell development, function, and stability. This review summarizes our current knowledge of how the PI3K/AKT pathway regulates thymic and peripheral Treg generation and function, with an emphasis on translation of these observations to therapies targeting Tregs in several pathologies.
Collapse
Affiliation(s)
- Saige L Pompura
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurology, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut, USA
| | - Margarita Dominguez-Villar
- Department of Neurology, Human and Translational Immunology Program, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Strains of bacterial species induce a greatly varied acute adaptive immune response: The contribution of the accessory genome. PLoS Pathog 2018; 14:e1006726. [PMID: 29324905 PMCID: PMC5764401 DOI: 10.1371/journal.ppat.1006726] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/02/2017] [Indexed: 11/20/2022] Open
Abstract
A fundamental question in human susceptibility to bacterial infections is to what extent variability is a function of differences in the pathogen species or in individual humans. To focus on the pathogen species, we compared in the same individual the human adaptive T and B cell immune response to multiple strains of two major human pathogens, Staphylococcus aureus and Streptococcus pyogenes. We found wide variability in the acute adaptive immune response induced by various strains of a species, with a unique combination of activation within the two arms of the adaptive response. Further, this was also accompanied by a dramatic difference in the intensity of the specific protective T helper (Th) response. Importantly, the same immune response differences induced by the individual strains were maintained across multiple healthy human donors. A comparison of isogenic phage KO strains, demonstrated that of the pangenome, prophages were the major contributor to inter-strain immune heterogeneity, as the T cell response to the remaining “core genome” was noticeably blunted. Therefore, these findings extend and modify the notion of an adaptive response to a pathogenic bacterium, by implying that the adaptive immune response signature of a bacterial species should be defined either per strain or alternatively to the species’ ‘core genome’, common to all of its strains. Further, our results demonstrate that the acquired immune response variation is as wide among different strains within a single pathogenic species as it is among different humans, and therefore may explain in part the clinical heterogeneity observed in patients infected with the same species. We address a fundamental question regarding the variability in human susceptibility to bacterial infection. Recent attention has been directed to human immune variation as the major element in infection susceptibility, however, our study focuses on the contribution of the pathogenic bacteria. To separate the contribution of bacterial versus human variability, we compared the acute adaptive immune response (both T and B cells) induced by several strains within two human disease-causing bacterial species in the same individual, and verified in 10 other humans. Our results clearly demonstrate that different strains of a species vary widely in the acute adaptive immune response they induce. Most importantly, using mutant strains, we show that the observed variability is a function of the differences found in the bacterial accessory genome, i.e., its lysogen. Previous publications generally base their results on a single strain as the representative of a species. Our findings extend and modify this notion by implying that the adaptive immune response signature to a pathogenic species should be defined either per strain or alternatively to the species’ ‘core genome, common to all of its strains. The results, while novel and unexpected, may set the stage to ultimately better predict patient disease outcome.
Collapse
|
37
|
Langley RJ, Ting YT, Clow F, Young PG, Radcliff FJ, Choi JM, Sequeira RP, Holtfreter S, Baker H, Fraser JD. Staphylococcal enterotoxin-like X (SElX) is a unique superantigen with functional features of two major families of staphylococcal virulence factors. PLoS Pathog 2017; 13:e1006549. [PMID: 28880913 PMCID: PMC5589262 DOI: 10.1371/journal.ppat.1006549] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 07/24/2017] [Indexed: 11/23/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen that produces many virulence factors. Two major families of which are the staphylococcal superantigens (SAgs) and the Staphylococcal Superantigen-Like (SSL) exoproteins. The former are immunomodulatory toxins that induce a Vβ-specific activation of T cells, while the latter are immune evasion molecules that interfere with a wide range of innate immune defences. The superantigenic properties of Staphylococcal enterotoxin-like X (SElX) have recently been established. We now reveal that SElX also possesses functional characteristics of the SSLs. A region of SElX displays high homology to the sialyl-lactosamine (sLacNac)-specific binding site present in a sub-family of SSLs. By analysing the interaction of SElX with sLacNac-containing glycans we show that SElX has an equivalent specificity and host cell binding range to the SSLs. Mutation of key amino acids in this conserved region affects the ability of SElX to bind to cells of myeloid origin and significantly reduces its ability to protect S. aureus from destruction in a whole blood killing (WBK) assay. Like the SSLs, SElX is up-regulated early during infection and is under the control of the S. aureus exotoxin expression (Sae) two component gene regulatory system. Additionally, the structure of SElX in complex with the sLacNac-containing tetrasaccharide sialyl Lewis X (sLeX) reveals that SElX is a unique single-domain SAg. In summary, SElX is an ‘SSL-like’ SAg. The ability of Staphylococcus aureus to cause disease can be attributed to the wide range of toxins and immune evasion molecules it produces. The 25-member superantigen (SAg) family of toxins disrupts adaptive immunity by activating large proportions of T cells. In contrast, the structurally-related 14-member Staphylococcal Superantigen-Like (SSL) family inhibits a wide range of innate immune functions. We have discovered that the SAg staphylococcal enterotoxin-like X (SElX) has the sialylated-glycan-dependent active site found in a sub-family of SSLs. Through this site it possesses the ability to affect host innate immunity defences. By solving the X-ray crystal structure of SElX we have also discovered that SElX is a unique single-domain SAg. While it retains a typical β-grasp domain, it lacks the OB-fold domain that is present in all other staphylococcal SAgs.
Collapse
Affiliation(s)
- Ries J. Langley
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
- * E-mail:
| | - Yi Tian Ting
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Fiona Clow
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Paul G. Young
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Fiona J. Radcliff
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Jeong Min Choi
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Richard P. Sequeira
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Silva Holtfreter
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - Heather Baker
- School of Biological Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| | - John D. Fraser
- School of Medical Sciences, and The Maurice Wilkins Centre for Molecular Biodiscovery, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
38
|
Anania C, Olivero F, Spagnolo A, Chiesa C, Pacifico L. Immune response to vaccines in children with celiac disease. World J Gastroenterol 2017; 23:3205-3213. [PMID: 28566880 PMCID: PMC5434426 DOI: 10.3748/wjg.v23.i18.3205] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/07/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated systemic condition evoked by ingestion of gluten and related prolamines in genetically susceptible subjects. The disease is featured by a variable combination of clinical signs, specific antibodies, HLA-DQ2 and HLA-DQ8 haplotypes, and enteropathy. Vaccination is the most potent intervention for infectious disease prevention. Several factors including age, gender, ethnicity, quality and quantity of vaccine antigen, doses, and route of administration can influence immune response to vaccination, although the main cause of variation in the responsiveness among vaccine recipients is host genetic variability. The HLA system has a fundamental role in identifying the antigens introduced into the host with the vaccines and in the development of specific antibodies, and some HLA phenotypes have been associated with a less effective immunological response. The available literature indicates that the immunological response to vaccines in CD children does not differ markedly from that of general population and antibody titres are high enough to provide long-term protection, except for hepatitis B virus vaccine. In this article, we review and discuss the scarce literature in this field in order to provide clinical practice guidelines to achieve the most efficient monitoring of the response to vaccines in pediatric CD patients.
Collapse
|
39
|
Svedova J, Ménoret A, Mittal P, Ryan JM, Buturla JA, Vella AT. Therapeutic blockade of CD54 attenuates pulmonary barrier damage in T cell-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2017; 313:L177-L191. [PMID: 28473322 DOI: 10.1152/ajplung.00050.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/27/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious, often fatal condition without available pharmacotherapy. Although the role of innate cells in ARDS has been studied extensively, emerging evidence suggests that T cells may be involved in disease etiology. Staphylococcus aureus enterotoxins are potent T-cell mitogens capable of triggering life-threatening shock. We demonstrate that 2 days after inhalation of S. aureus enterotoxin A, mice developed T cell-mediated increases in vascular permeability, as well as expression of injury markers and caspases in the lung. Pulmonary endothelial cells underwent sequential phenotypic changes marked by rapid activation coinciding with inflammatory events secondary to T-cell priming, followed by reductions in endothelial cell number juxtaposing simultaneous T-cell expansion and cytotoxic differentiation. Although initial T-cell activation influenced the extent of lung injury, CD54 (ICAM-1) blocking antibody administered well after enterotoxin exposure substantially attenuated pulmonary barrier damage. Thus CD54-targeted therapy may be a promising candidate for further exploration into its potential utility in treating ARDS patients.
Collapse
Affiliation(s)
- Julia Svedova
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - Antoine Ménoret
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut.,Institute for Systems Genomics, UConn Health, Farmington, Connecticut; and
| | - Payal Mittal
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - Joseph M Ryan
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut
| | - James A Buturla
- Department of Internal Medicine, UConn Health, Farmington, Connecticut
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, Connecticut;
| |
Collapse
|
40
|
Benvenuti M, An T, Amaro E, Lovejoy S, Mencio G, Martus J, Mignemi M, Schoenecker JG. Double-Edged Sword: Musculoskeletal Infection Provoked Acute Phase Response in Children. Orthop Clin North Am 2017; 48:181-197. [PMID: 28336041 DOI: 10.1016/j.ocl.2016.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The acute phase response has a crucial role in mounting the body's response to tissue injury. Excessive activation of the acute phase response is responsible for many complications that occur in orthopedic patients. Given that infection may be considered continuous tissue injury that persistently activates the acute phase response, children with musculoskeletal infections are at markedly increased risk for serious complications. Future strategies that modulate the acute phase response have the potential to improve treatment and prevent complications associated with musculoskeletal infection.
Collapse
Affiliation(s)
| | - Thomas An
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Emilie Amaro
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Steven Lovejoy
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gregory Mencio
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Martus
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Megan Mignemi
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan G Schoenecker
- Department of Orthopaedics, Vanderbilt University Medical Center, Nashville, TN, USA; Departments of Orthopaedics, Pharmacology, and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
41
|
Abstract
γδ T cells constitute the third arm of a tripartite adaptive immune system in jawed vertebrates, besides αβ T cells and B cells. Like the other two lymphocyte-types, they express diverse antigen receptors, capable of specific ligand recognition. Functionally, γδ T cells represent a system of differentiated subsets, sometimes engaged in cross-regulation, which ultimately determines their effect on other components of the immune system, including B cells and antibodies. γδ T cells are capable of providing help to B cells in antibody production. More recently it became clear that γδ T cells influence B cell differentiation during the peripheral stages of B cell development, control levels of circulating immunoglobulin (all subclasses), and affect production of autoantibodies. Because of this relationship between γδ T cells and B cells, the extensive variation of γδ T cells among human individuals might be expected to modulate their humoral responsiveness.
Collapse
Affiliation(s)
- Willi K Born
- National Jewish Health, Denver, CO, United States; University of Colorado Health Sciences Center, Aurora, CO, United States.
| | - Yafei Huang
- Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; Joint Laboratory for Stem Cell Engineering and Technology Transfer, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - R Lee Reinhardt
- National Jewish Health, Denver, CO, United States; University of Colorado Health Sciences Center, Aurora, CO, United States
| | - Hua Huang
- National Jewish Health, Denver, CO, United States; University of Colorado Health Sciences Center, Aurora, CO, United States
| | - Deming Sun
- Doheny Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rebecca L O'Brien
- National Jewish Health, Denver, CO, United States; University of Colorado Health Sciences Center, Aurora, CO, United States
| |
Collapse
|
42
|
Chen JB, James LK, Davies AM, Wu YCB, Rimmer J, Lund VJ, Chen JH, McDonnell JM, Chan YC, Hutchins GH, Chang TW, Sutton BJ, Kariyawasam HH, Gould HJ. Antibodies and superantibodies in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2016; 139:1195-1204.e11. [PMID: 27658758 PMCID: PMC5380656 DOI: 10.1016/j.jaci.2016.06.066] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 05/07/2016] [Accepted: 06/13/2016] [Indexed: 01/19/2023]
Abstract
Background Chronic rhinosinusitis with nasal polyps is associated with local immunoglobulin hyperproduction and the presence of IgE antibodies against Staphylococcus aureus enterotoxins (SAEs). Aspirin-exacerbated respiratory disease is a severe form of chronic rhinosinusitis with nasal polyps in which nearly all patients express anti-SAEs. Objectives We aimed to understand antibodies reactive to SAEs and determine whether they recognize SAEs through their complementarity-determining regions (CDRs) or framework regions. Methods Labeled staphylococcal enterotoxin (SE) A, SED, and SEE were used to isolate single SAE-specific B cells from the nasal polyps of 3 patients with aspirin-exacerbated respiratory disease by using fluorescence-activated cell sorting. Recombinant antibodies with “matched” heavy and light chains were cloned as IgG1, and those of high affinity for specific SAEs, assayed by means of ELISA and surface plasmon resonance, were recloned as IgE and antigen-binding fragments. IgE activities were tested in basophil degranulation assays. Results Thirty-seven SAE-specific, IgG- or IgA-expressing B cells were isolated and yielded 6 anti-SAE clones, 2 each for SEA, SED, and SEE. Competition binding assays revealed that the anti-SEE antibodies recognize nonoverlapping epitopes in SEE. Unexpectedly, each anti-SEE mediated SEE-induced basophil degranulation, and IgG1 or antigen-binding fragments of each anti-SEE enhanced degranulation by the other anti-SEE. Conclusions SEEs can activate basophils by simultaneously binding as antigens in the conventional manner to CDRs and as superantigens to framework regions of anti-SEE IgE in anti-SEE IgE-FcεRI complexes. Anti-SEE IgG1s can enhance the activity of anti-SEE IgEs as conventional antibodies through CDRs or simultaneously as conventional antibodies and as “superantibodies” through CDRs and framework regions to SEEs in SEE–anti-SEE IgE-FcεRI complexes.
Collapse
Affiliation(s)
- Jiun-Bo Chen
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Louisa K James
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - Anna M Davies
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - Yu-Chang Bryan Wu
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - Joanne Rimmer
- Allergy and Rhinology, Royal National Throat Nose Ear Hospital, London, United Kingdom
| | - Valerie J Lund
- Allergy and Rhinology, Royal National Throat Nose Ear Hospital, London, United Kingdom
| | - Jou-Han Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - James M McDonnell
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - Yih-Chih Chan
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - George H Hutchins
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Tse Wen Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Brian J Sutton
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom
| | - Harsha H Kariyawasam
- Allergy and Rhinology, Royal National Throat Nose Ear Hospital, London, United Kingdom
| | - Hannah J Gould
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, King's College London, Guy's Campus, London, United Kingdom.
| |
Collapse
|
43
|
Chapes S, Beharka A. Lipopolysaccharide is required for the lethal effects of enterotoxin B after D-galactosamine sensitization. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199500200406] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We tested the D-galactosamine sensitization model with staphylococcal enterotoxin B (SEB). LPS was required for the lethal effects of SEB in D-galactosamine sensitized mice. Only two (2/62) among the C3HeB/FeJ (H-2k), Balb/c (H-2d) and C57BL/6J (H-2b) mice died in response to SEB in the absence of LPS whereas injection of SEB and minimally lethal concentrations of LPS became highly toxic. Similar to LPS, the lethal effect of SEB was dependent on the mouse strain used. Mouse strains more sensitive to the effects of LPS (Balb/c and C57BL/6J) were also more sensitive to the effects of SEB in comparison to C3H mice when equivalent doses of LPS and SEB were used. Among Balb/c and C3HeB/FeJ but not C57BL/6J mice, SEB (20 μg) potentiated the lethal effects of LPS at low doses (0.1 μg LPS), but had an apparent protective effect at high doses (1 μg LPS). Lastly, there was an inverse correlation between pathogenesis and serum IL-2 concentrations and splenic T cell activation in C3H mice. However, macrophage mobilization did correlate with lethality. Therefore, some questions remain about the mechanisms involved in the D-galactosamine/SEB pathogenesis model. We conclude that when sensitizing mice with D-galactosamine and assessing the lethal effects of SEB, endotoxin contamination must be assessed.
Collapse
Affiliation(s)
- S.K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - A.A. Beharka
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
44
|
Chinnadurai R, Copland IB, Garcia MA, Petersen CT, Lewis CN, Waller EK, Kirk AD, Galipeau J. Cryopreserved Mesenchymal Stromal Cells Are Susceptible to T-Cell Mediated Apoptosis Which Is Partly Rescued by IFNγ Licensing. Stem Cells 2016; 34:2429-42. [PMID: 27299362 DOI: 10.1002/stem.2415] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/18/2016] [Indexed: 12/16/2022]
Abstract
We have previously demonstrated that cryopreservation and thawing lead to altered Mesenchymal stromal cells (MSC) functionalities. Here, we further analyzed MSC's fitness post freeze-thaw. We have observed that thawed MSC can suppress T-cell proliferation when separated from them by transwell membrane and the effect is lost in a MSC:T-cell coculture system. Unlike actively growing MSCs, thawed MSCs were lysed upon coculture with activated autologous Peripheral Blood Mononuclear Cells (PBMCs) and the lysing effect was further enhanced with allogeneic PBMCs. The use of DMSO-free cryoprotectants or substitution of Human Serum Albumin (HSA) with human platelet lysate in freezing media and use of autophagy or caspase inhibitors did not prevent thaw defects. We tested the hypothesis that IFNγ prelicensing before cryobanking can enhance MSC fitness post thaw. Post thawing, IFNγ licensed MSCs inhibit T cell proliferation as well as fresh MSCs and this effect can be blocked by 1-methyl Tryptophan, an Indoleamine 2,3-dioxygenase (IDO) inhibitor. In addition, IFNγ prelicensed thawed MSCs inhibit the degranulation of cytotoxic T cells while IFNγ unlicensed thawed MSCs failed to do so. However, IFNγ prelicensed thawed MSCs do not deploy lung tropism in vivo following intravenous injection as well as fresh MSCs suggesting that IFNγ prelicensing does not fully rescue thaw-induced lung homing defect. We identified reversible and irreversible cryoinjury mechanisms that result in susceptibility to host T-cell cytolysis and affect MSC's cell survival and tissue distribution. The susceptibility of MSC to negative effects of cryopreservation and the potential to mitigate the effects with IFNγ prelicensing may inform strategies to enhance the therapeutic efficacy of MSC in clinical use. Stem Cells 2016;34:2429-2442.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Ian B Copland
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA.,Emory Personalized Immunotherapy Center, Emory Healthcare, Atlanta, Georgia, USA
| | - Marco A Garcia
- Emory Personalized Immunotherapy Center, Emory Healthcare, Atlanta, Georgia, USA
| | - Christopher T Petersen
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Christopher N Lewis
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Edmund K Waller
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA.,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA.,Emory Personalized Immunotherapy Center, Emory Healthcare, Atlanta, Georgia, USA
| | - Allan D Kirk
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Jacques Galipeau
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, Georgia, USA. .,Winship Cancer Institute, Emory University, Atlanta, Georgia, USA. .,Department of Pediatrics, Emory University, Atlanta, Georgia, USA. .,Emory Personalized Immunotherapy Center, Emory Healthcare, Atlanta, Georgia, USA.
| |
Collapse
|
45
|
Svedova J, Tsurutani N, Liu W, Khanna KM, Vella AT. TNF and CD28 Signaling Play Unique but Complementary Roles in the Systemic Recruitment of Innate Immune Cells after Staphylococcus aureus Enterotoxin A Inhalation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4510-21. [PMID: 27183621 PMCID: PMC4875807 DOI: 10.4049/jimmunol.1600113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/27/2016] [Indexed: 01/12/2023]
Abstract
Staphylococcus aureus enterotoxins cause debilitating systemic inflammatory responses, but how they spread systemically and trigger inflammatory cascade is unclear. In this study, we showed in mice that after inhalation, Staphylococcus aureus enterotoxin A rapidly entered the bloodstream and induced T cells to orchestrate systemic recruitment of inflammatory monocytes and neutrophils. To study the mechanism used by specific T cells that mediate this process, a systems approach revealed inducible and noninducible pathways as potential targets. It was found that TNF caused neutrophil entry into the peripheral blood, whereas CD28 signaling, but not TNF, was needed for chemotaxis of inflammatory monocytes into blood and lymphoid tissue. However, both pathways triggered local recruitment of neutrophils into lymph nodes. Thus, our findings revealed a dual mechanism of monocyte and neutrophil recruitment by T cells relying on overlapping and nonoverlapping roles for the noninducible costimulatory receptor CD28 and the inflammatory cytokine TNF. During sepsis, there might be clinical value in inhibiting CD28 signaling to decrease T cell-mediated inflammation and recruitment of innate cells while retaining bioactive TNF to foster neutrophil circulation.
Collapse
Affiliation(s)
- Julia Svedova
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030
| | - Naomi Tsurutani
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030
| | - Wenhai Liu
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030
| | - Kamal M Khanna
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030
| | - Anthony T Vella
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT 06030
| |
Collapse
|
46
|
Elliott DM, Nagarkatti M, Nagarkatti PS. 3,39-Diindolylmethane Ameliorates Staphylococcal Enterotoxin B–Induced Acute Lung Injury through Alterations in the Expression of MicroRNA that Target Apoptosis and Cell-Cycle Arrest in Activated T Cells. J Pharmacol Exp Ther 2016; 357:177-87. [PMID: 26818958 PMCID: PMC4809322 DOI: 10.1124/jpet.115.226563] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 01/26/2016] [Indexed: 12/20/2022] Open
Abstract
3,39-Diindolylmethane (DIM), a natural indole found in cruciferous vegetables, has significant anti-cancer and anti-inflammatory properties. In this current study, we investigated the effects of DIM on acute lung injury (ALI) induced by exposure to staphylococcal enterotoxin B (SEB). We found that pretreatment of mice with DIM led to attenuation of SEB-induced inflammation in the lungs, vascular leak, and IFN-g secretion. Additionally, DIM could induce cell-cycle arrest and cell death in SEB-activated T cells in a concentration-dependent manner. Interestingly, microRNA (miRNA) microarray analysis uncovered an altered miRNA profile in lung-infiltrating mononuclear cells after DIM treatment of SEB-exposed mice. Moreover, computational analysis of miRNA gene targets and regulation networks indicated that DIM alters miRNA in the cell death and cell-cycle progression pathways. Specifically, DIM treatment significantly downregulated several miRNA and a correlative increase associated gene targets. Furthermore, overexpression and inhibition studies demonstrated that DIM-induced cell death, at least in part, used miR-222. Collectively, these studies demonstrate for the first time that DIM treatment attenuates SEB-induced ALI and may do so through the induction of microRNAs that promote apoptosis and cell-cycle arrest in SEB-activated T cells.
Collapse
|
47
|
Heming N, Lamothe L, Ambrosi X, Annane D. Emerging drugs for the treatment of sepsis. Expert Opin Emerg Drugs 2016; 21:27-37. [DOI: 10.1517/14728214.2016.1132700] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
48
|
Yu W, Jiang N, Ebert PJR, Kidd BA, Müller S, Lund PJ, Juang J, Adachi K, Tse T, Birnbaum ME, Newell EW, Wilson DM, Grotenbreg GM, Valitutti S, Quake SR, Davis MM. Clonal Deletion Prunes but Does Not Eliminate Self-Specific αβ CD8(+) T Lymphocytes. Immunity 2015; 42:929-41. [PMID: 25992863 DOI: 10.1016/j.immuni.2015.05.001] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 02/27/2015] [Accepted: 03/23/2015] [Indexed: 02/06/2023]
Abstract
It has long been thought that clonal deletion efficiently removes almost all self-specific T cells from the peripheral repertoire. We found that self-peptide MHC-specific CD8(+) T cells in the blood of healthy humans were present in frequencies similar to those specific for non-self antigens. For the Y chromosome-encoded SMCY antigen, self-specific T cells exhibited only a 3-fold lower average frequency in males versus females and were anergic with respect to peptide activation, although this inhibition could be overcome by a stronger stimulus. We conclude that clonal deletion prunes but does not eliminate self-specific T cells and suggest that to do so would create holes in the repertoire that pathogens could readily exploit. In support of this hypothesis, we detected T cells specific for all 20 amino acid variants at the p5 position of a hepatitis C virus epitope in a random group of blood donors.
Collapse
Affiliation(s)
- Wong Yu
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Hematology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ning Jiang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter J R Ebert
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brian A Kidd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sabina Müller
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France and Université Toulouse III Paul-Sabatier, 31024 Toulouse, France
| | - Peder J Lund
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jeremy Juang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Keishi Adachi
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tiffany Tse
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael E Birnbaum
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Evan W Newell
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Darrell M Wilson
- Department of Pediatric Endocrinology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Salvatore Valitutti
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France and Université Toulouse III Paul-Sabatier, 31024 Toulouse, France
| | - Stephen R Quake
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
Singh S, Barr H, Liu YC, Robins A, Heeb S, Williams P, Fogarty A, Cámara M, Martínez-Pomares L. Granulocyte-macrophage colony stimulatory factor enhances the pro-inflammatory response of interferon-γ-treated macrophages to Pseudomonas aeruginosa infection. PLoS One 2015; 10:e0117447. [PMID: 25706389 PMCID: PMC4338139 DOI: 10.1371/journal.pone.0117447] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 12/23/2014] [Indexed: 01/07/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can cause severe infections at compromised epithelial surfaces, such those found in burns, wounds, and in lungs damaged by mechanical ventilation or recurrent infections, particularly in cystic fibrosis (CF) patients. CF patients have been proposed to have a Th2 and Th17-biased immune response suggesting that the lack of Th1 and/or over exuberant Th17 responses could contribute to the establishment of chronic P. aeruginosa infection and deterioration of lung function. Accordingly, we have observed that interferon (IFN)-γ production by peripheral blood mononuclear cells from CF patients positively correlated with lung function, particularly in patients chronically infected with P. aeruginosa. In contrast, IL-17A levels tended to correlate negatively with lung function with this trend becoming significant in patients chronically infected with P. aeruginosa. These results are in agreement with IFN-γ and IL-17A playing protective and detrimental roles, respectively, in CF. In order to explore the protective effect of IFN-γ in CF, the effect of IFN-γ alone or in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF), on the ability of human macrophages to control P. aeruginosa growth, resist the cytotoxicity induced by this bacterium or promote inflammation was investigated. Treatment of macrophages with IFN-γ, in the presence and absence of GM-CSF, failed to alter bacterial growth or macrophage survival upon P. aeruginosa infection, but changed the inflammatory potential of macrophages. IFN-γ caused up-regulation of monocyte chemoattractant protein-1 (MCP-1) and TNF-α and down-regulation of IL-10 expression by infected macrophages. GM-CSF in combination with IFN-γ promoted IL-6 production and further reduction of IL-10 synthesis. Comparison of TNF-α vs. IL-10 and IL-6 vs. IL-10 ratios revealed the following hierarchy in regard to the pro-inflammatory potential of human macrophages infected with P. aeruginosa: untreated < treated with GM-CSF < treated with IFN-γ < treated with GM-CSF and IFN-γ.
Collapse
Affiliation(s)
- Sonali Singh
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Helen Barr
- School of Medicine, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Yi-Chia Liu
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Adrian Robins
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Stephan Heeb
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Paul Williams
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Andrew Fogarty
- School of Community Health Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
| | - Miguel Cámara
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
- * E-mail: (LMP); (MC)
| | - Luisa Martínez-Pomares
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2RD, United Kingdom
- * E-mail: (LMP); (MC)
| |
Collapse
|
50
|
Khalifian S, Raimondi G, Lee WA, Brandacher G. Taming inflammation by targeting cytokine signaling: new perspectives in the induction of transplantation tolerance. Immunotherapy 2015; 6:637-53. [PMID: 24896631 DOI: 10.2217/imt.14.25] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transplantation tolerance remains an elusive goal, partly due to limitations in our understanding of the interplay between inflammatory mediators and their role in the activation and regulation of T lymphocytes. Although multiple mechanisms acting both centrally and peripherally are responsible for tolerance induction, the signaling pathways leading to activation or regulation of adaptive immunity are often complex, branched, redundant and modulated by the microenvironment's inflammatory milieu. Accumulating evidence clearly indicates that inflammatory cytokines limit the tolerogenic potential of immunomodulatory protocols by supporting priming of the immune system and counteracting regulatory mechanisms, ultimately promoting rejection. In this review, we summarize recent progress in the development of novel therapeutics to manipulate this inflammatory environment and achievements in targeted inhibition of inflammatory cytokine signaling. Ultimately, robust transplant tolerance induction will probably require a multifaceted, holistic approach that integrates the various mechanisms of tolerance induction, incorporates the dynamic alterations in costimulatory requirements of alloreactive T cells, while maintaining endogenous mechanisms of immune regulation.
Collapse
Affiliation(s)
- Saami Khalifian
- Department of Plastic and Reconstructive Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | | |
Collapse
|