1
|
Jamali MC, Mohamed AH, Jamal A, Kamal MA, Al Abdulmonem W, Saeed BA, Mansuri N, Ahmad F, Mudhafar M, Shafie A, Hattiwale HM. Biological mechanisms and therapeutic prospects of interleukin-33 in pathogenesis and treatment of allergic disease. J Inflamm (Lond) 2025; 22:17. [PMID: 40355878 PMCID: PMC12070619 DOI: 10.1186/s12950-025-00438-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 02/15/2025] [Indexed: 05/15/2025] Open
Abstract
Allergic diseases significantly impact the quality of life of people around the world. Cytokines play a crucial role in regulating the immune system. Due to their importance in pro-inflammatory mechanisms, cytokines are used to understand pathogenesis and serve as biomarkers in many diseases. One such cytokine is interleukin-33, a member of the IL-1 family, including IL- 1α, IL-1β, and IL-18. The IL-33 receptor is a heterodimer of IL-1 receptor-like 1 and IL-1 receptor accessory protein. IL-33 plays a critical role in regulating innate and adaptive immune responses. The primary targets of IL-33 in vivo are tissue-resident immune cells, including mast cells, group 2 innate lymphoid cells, regulatory T cells, T helper 2 cells, eosinophils, basophils, dendritic cells, Th1 cells, CD8 + T cells, NK cells, iNKT cells, B cells, neutrophils, and macrophages. However, IL-33 appears to act as an alarm signal that is promptly released by producing cells under cellular damage or stress conditions. IL-33 regulates signaling and various biological functions, including induction of pro-inflammatory cytokines, regulation of cell proliferation, and involvement in tissue remodeling. IL-33 is fundamental in immune-related diseases and plays a critical role in the control of inflammation. Recently, IL-33 has been shown to significantly impact allergic diseases, primarily by inducing Th2 immune responses. IL-33 is a key regulator of mast cell function and a promising therapeutic target for treating allergic diseases. This review provides an overview of the current understanding of the role of IL-33 in allergy pathogenesis and potential clinical approaches.
Collapse
Affiliation(s)
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi, Majmaah University, Al-Majmaah 11952,, Saudi Arabia
- Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952 , Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Bashar Abdullah Saeed
- Department of Medical Laboratory Technics, Al-Noor University College, Nineveh, Iraq
| | - Nasrin Mansuri
- Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713 , Saudi Arabia
| | - Mustafa Mudhafar
- Department of Medical Physics, Faculty of Medical Applied Sciences, University of Kerbala, 56001, Karbala, Iraq
- Department of Anesthesia Techniques and Intensive Care, Al-Taff university college, 56001, Kerbala, Iraq
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif, 21944, Saudi Arabia
| | - Haroonrashid M Hattiwale
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al-Majmaah 11952 , Saudi Arabia.
| |
Collapse
|
2
|
Chakraborty R, Chronopoulos J, Sun R, Morozan A, Joy S, Divangahi M, Lauzon AM, Martin JG. Anti-ST2 antibody reduces airway hyperresponsiveness mediated by monocyte-derived macrophages during influenza A infection. Mucosal Immunol 2025:S1933-0219(25)00046-7. [PMID: 40319941 DOI: 10.1016/j.mucimm.2025.04.008] [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: 10/29/2024] [Revised: 03/23/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Influenza A virus (IAV) infections trigger asthma attacks and cause airway hyperresponsiveness (AHR) in murine models. However, the mechanism by which AHR is induced remains to be fully elucidated. Here, we show that targeting the interleukin (IL)-33 suppression of tumorigenicity 2 (ST2) receptor complex with an anti-ST2 antibody during acute IAV infection of C57BL/6 mice reduced AHR, without affecting expansion of ILC2s and independently of IL-13. Among the lung inflammatory cells, the anti-ST2 antibody selectively reduced the monocyte-derived macrophages (MMs). Furthermore, AHR was reduced in C-C chemokine receptor 2 (CCR2)-knockout mice that have deficient MM recruitment. Depletion of MMs achieved by anti-Ly6C antibody administration also reduced AHR. The treatment of airway smooth muscle (ASM) with conditioned medium from IL-33-treated human THP-1-derived macrophages enhanced potassium chloride-induced ASM contraction. These findings suggest that MMs contribute to acute AHR following IAV infection in an IL-33-dependent manner, but independent of the ILC2/IL-13 axis. Additionally, IL-33 stimulates the release of macrophage-derived mediators that enhance airway smooth muscle contraction, offering a potential mechanistic basis for IAV-induced AHR.
Collapse
Affiliation(s)
- Rohin Chakraborty
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Julia Chronopoulos
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Arina Morozan
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Sydney Joy
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - James G Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Su P, Jiang C, Zhang Y. The implication of infection with respiratory syncytial virus in pediatric recurrent wheezing and asthma: knowledge expanded post-COVID-19 era. Eur J Clin Microbiol Infect Dis 2024; 43:403-416. [PMID: 38153660 DOI: 10.1007/s10096-023-04744-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection has been identified to serve as the primary cause of acute lower respiratory infectious diseases in children under the age of one and a significant risk factor for the emergence and development of pediatric recurrent wheezing and asthma, though the exact mechanism is still unknown. METHODS AND RESULTS In this study, we discuss the key routes that lead to recurrent wheezing and bronchial asthma following RSV infection. It is interesting to note that following the coronavirus disease 2019 (COVID-19) epidemic, the prevalence of RSV changes significantly. This presents us with a rare opportunity to better understand the associated mechanism for RSV infection, its effects on the respiratory system, and the immunological response to RSV following the COVID-19 epidemic. To better understand the associated mechanisms in the occurrence and progression of pediatric asthma, we thoroughly described how the RSV infection directly destroys the physical barrier of airway epithelial tissue, promotes inflammatory responses, enhances airway hyper-responsiveness, and ultimately causes the airway remodeling. More critically, extensive discussion was also conducted regarding the potential impact of RSV infection on host pulmonary immune response. CONCLUSION In conclusion, this study offers a comprehensive perspective to better understand how the RSV infection interacts in the control of the host's pulmonary immune system, causing recurrent wheezing and the development of asthma, and it sheds fresh light on potential avenues for pharmaceutical therapy in the future.
Collapse
Affiliation(s)
- Peipei Su
- Xi'an Medical University, Xi'an, 710068, Shaanxi, China
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Congshan Jiang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China
| | - Yanmin Zhang
- Key Laboratory of Precision Medicine to Pediatric Diseases of Shaanxi Province, National Regional Children's Medical Centre (Northwest), Xi'an Key Laboratory of Children's Health and Diseases, Shaanxi Institute for Pediatric Diseases, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China.
- Department of Cardiology, Xi'an Children's Hospital, Affiliated Children's Hospital of Xi'an Jiaotong University, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
4
|
Alvarez F, Istomine R, Da Silva Lira Filho A, Al-Aubodah TA, Huang D, Okde R, Olivier M, Fritz JH, Piccirillo CA. IL-18 is required for the T H1-adaptation of T REG cells and the selective suppression of T H17 responses in acute and chronic infections. Mucosal Immunol 2023; 16:462-475. [PMID: 37182738 DOI: 10.1016/j.mucimm.2023.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/24/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Interleukin (IL)-18, a member of the IL-1 family of alarmins, is abundantly released in the lungs following influenza A (IAV) infections yet its role in orchestrating the local adaptive immune response remains ill defined. Through genetic disruption of the IL-18 receptor, we demonstrate that IL-18 not only promotes pulmonary TH1 responses but also influences regulatory T cells (TREG) function in the infected lungs. As the response unfolds, TREG cells accumulating in the lungs express Helios, T-bet, CXCR3, and IL-18R1 and produce interferon γ in the presence of IL-12. During IAV, IL-18R1 is required for TREG cells to control TH17, but not TH1, responses and promote a return to lung homeostasis, revealing a novel mechanism of selective suppression. Moreover, this observation was not limited to the lungs, as skin-localized TREG cells require an IL-18 signal to specifically suppress IL-17A production by TH17 and γδ T cells in a model of chronic cutaneous Leishmania major infection. Overall, these results uncover how IL-18 orchestrates the tissue adaptation of TREG cells to selectively favor TH1 over TH17 responses during TH1-driven immune responses and provide a novel perspective into how IL-18 dictates the immune response during viral and parasitic infections.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada
| | - Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada
| | | | - Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada
| | - Daniel Huang
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada
| | - Rakan Okde
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, Canada
| | - Jörg H Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada; McGill University Research Centre on Complex Traits (MRCCT), Montréal, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, Canada; Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Canada; Centre of Excellence in Translational Immunology (CETI), Montréal, Canada; McGill University Research Centre on Complex Traits (MRCCT), Montréal, Canada.
| |
Collapse
|
5
|
Macchia I, La Sorsa V, Urbani F, Moretti S, Antonucci C, Afferni C, Schiavoni G. Eosinophils as potential biomarkers in respiratory viral infections. Front Immunol 2023; 14:1170035. [PMID: 37483591 PMCID: PMC10358847 DOI: 10.3389/fimmu.2023.1170035] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/30/2023] [Indexed: 07/25/2023] Open
Abstract
Eosinophils are bone marrow-derived granulocytes that, under homeostatic conditions, account for as much as 1-3% of peripheral blood leukocytes. During inflammation, eosinophils can rapidly expand and infiltrate inflamed tissues, guided by cytokines and alarmins (such as IL-33), adhesion molecules and chemokines. Eosinophils play a prominent role in allergic asthma and parasitic infections. Nonetheless, they participate in the immune response against respiratory viruses such as respiratory syncytial virus and influenza. Notably, respiratory viruses are associated with asthma exacerbation. Eosinophils release several molecules endowed with antiviral activity, including cationic proteins, RNases and reactive oxygen and nitrogen species. On the other hand, eosinophils release several cytokines involved in homeostasis maintenance and Th2-related inflammation. In the context of SARS-CoV-2 infection, emerging evidence indicates that eosinophils can represent possible blood-based biomarkers for diagnosis, prognosis, and severity prediction of disease. In particular, eosinopenia seems to be an indicator of severity among patients with COVID-19, whereas an increased eosinophil count is associated with a better prognosis, including a lower incidence of complications and mortality. In the present review, we provide an overview of the role and plasticity of eosinophils focusing on various respiratory viral infections and in the context of viral and allergic disease comorbidities. We will discuss the potential utility of eosinophils as prognostic/predictive immune biomarkers in emerging respiratory viral diseases, particularly COVID-19. Finally, we will revisit some of the relevant methods and tools that have contributed to the advances in the dissection of various eosinophil subsets in different pathological settings for future biomarker definition.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina La Sorsa
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Rome, Italy
| | - Caterina Antonucci
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Afferni
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
6
|
Liu W, Wang S, Wang J, Zheng R, Wang D, Yu R, Liu B. Neuromedin U Induces Pulmonary ILC2 Activation via the NMUR1 Pathway during Acute Respiratory Syncytial Virus Infection. Am J Respir Cell Mol Biol 2023; 68:256-266. [PMID: 36227802 DOI: 10.1165/rcmb.2022-0123oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Activated group 2 innate lymphoid cells (ILC2s) play a crucial role in respiratory syncytial virus (RSV)-induced airway inflammation and allergy-like symptoms because of their ability to secrete large quantities of type 2 cytokines. Cytokines such as IL-33, IL-25, and thymic stromal lymphopoietin are activators of ILC2s. Besides, a regulatory effect of neurotransmitters on ILC2 activation has been reported recently. However, whether and how RSV infection induces neurotransmitter production in the lungs and regulates pulmonary ILC2 activation remains unclear. In this study, using a murine model established by intranasal infection with RSV, we found that acute RSV infection induced the production of a neurotransmitter, neuromedin U (NMU), in the lungs of RSV-infected mice and upregulated the expression of NMUR1 (neuromedin U receptor 1) on ILC2s. Moreover, in vivo administration of NMU exacerbated RSV-induced airway inflammation by promoting the proliferation and activation of pulmonary ILC2s via the NMUR1 pathway, which involved PI3K, mitogen-activated protein kinase kinase, and NFAT signaling proteins. Furthermore, pulmonary neurons responded to the stimulation of RSV infection and secreted NMU in a Toll-like receptor 4- and Toll-like receptor 7-dependent manner. Collectively, our data suggest that NMU is a powerful neuropeptide to activate ILC2s, highlighting the critical regulatory effects of neurotransmitters on antiviral, inflammatory, and tissue homeostasis at the mucosal barrier during a viral respiratory infection.
Collapse
Affiliation(s)
- Weiwei Liu
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang, China.,Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China; and
| | - Si Wang
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Jia Wang
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Rui Zheng
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang, China
| | | | - Rui Yu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Pathogenic Microbiology, School of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
7
|
Murdaca G, Paladin F, Tonacci A, Borro M, Greco M, Gerosa A, Isola S, Allegra A, Gangemi S. Involvement of Il-33 in the Pathogenesis and Prognosis of Major Respiratory Viral Infections: Future Perspectives for Personalized Therapy. Biomedicines 2022; 10:715. [PMID: 35327516 PMCID: PMC8944994 DOI: 10.3390/biomedicines10030715] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
Interleukin (IL)-33 is a key cytokine involved in type-2 immunity and allergic airway disease. At the level of lung epithelial cells, where it is clearly expressed, IL-33 plays an important role in both innate and adaptive immune responses in mucosal organs. It has been widely demonstrated that in the course of respiratory virus infections, the release of IL-33 increases, with consequent pro-inflammatory effects and consequent exacerbation of the clinical symptoms of chronic respiratory diseases. In our work, we analyzed the pathogenetic and prognostic involvement of IL-33 during the main respiratory viral infections, with particular interest in the recent SARS-CoV-2virus pandemic and the aim of determining a possible connection point on which to act with a targeted therapy that is able to improve the clinical outcome of patients.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy; (F.P.); (A.G.)
| | - Francesca Paladin
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy; (F.P.); (A.G.)
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy;
| | - Matteo Borro
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy; (M.B.); (M.G.)
| | - Monica Greco
- Internal Medicine Department, San Paolo Hospital, 17100 Savona, Italy; (M.B.); (M.G.)
| | - Alessandra Gerosa
- Department of Internal Medicine, Ospedale Policlinico San Martino, 16132 Genoa, Italy; (F.P.); (A.G.)
| | - Stefania Isola
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (S.I.); (S.G.)
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Hematology, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (S.I.); (S.G.)
| |
Collapse
|
8
|
Inhibition of NF- κB/IL-33/ST2 Axis Ameliorates Acute Bronchiolitis Induced by Respiratory Syncytial Virus. J Immunol Res 2021; 2021:6625551. [PMID: 34395633 PMCID: PMC8357524 DOI: 10.1155/2021/6625551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/04/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022] Open
Abstract
Background/Aim Bronchiolitis is a common acute lower respiratory tract infectious disease in infants. Respiratory syncytial virus (RSV) infection is one of the main causes. Bronchiolitis can lead to a significant increase in the incidence of asthma in young children, but the mechanism of bronchiolitis transforming into asthma is still unclear. The study was aimed at investigating the role of NF-κB/IL-33/ST2 axis on RSV-induced acute bronchiolitis. Methods A total of 40 infants diagnosed with acute bronchiolitis infected by RSV, and 20 normal infants were included in this study. BALB/c mice (6-8 weeks old, 20 ± 1.1 g) were used as study models. Enzyme-linked immunosorbent assay (ELISA), quantitative real time PCR, western blot analysis, immunohistochemical staining, and flow cytometry analysis were performed to examine relevant indicators. Results IL-33 level was significantly elevated, and Th1/Th2 ratio is imbalance after in infants with acute bronchiolitis. In vivo study, we found that NF-κB/IL-33/ST2 axis is mediated the Th2 cytokine levels and BAL cell number induced by RSV. Acute bronchiolitis induced by RSV in a mouse model is attenuated after inhibition of NF-κB/IL-33/ST2 pathway. Moreover, we also confirmed that macrophages are important sources of IL-33 and are regulated by NF-κB pathway in RSV-induced mice. Conclusion We confirmed that inhibition of NF-κB/IL-33/ST2 axis could attenuate acute bronchiolitis by RSV infected. Our findings not only demonstrate the potential role of IL-33 antibody in attenuating RSV-induced lung damage but also provide a new insight into better prevention of RSV-induced asthma by mediating NF-κB/IL-33/ST2 axis.
Collapse
|
9
|
Warren KJ, Poole JA, Sweeter JM, DeVasure JM, Dickinson JD, Peebles RS, Wyatt TA. Neutralization of IL-33 modifies the type 2 and type 3 inflammatory signature of viral induced asthma exacerbation. Respir Res 2021; 22:206. [PMID: 34266437 PMCID: PMC8281667 DOI: 10.1186/s12931-021-01799-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
Background Respiratory viral infections are one of the leading causes of need for emergency care and hospitalizations in asthmatic individuals, and airway-secreted cytokines are released within hours of viral infection to initiate these exacerbations. IL-33, specifically, contributes to these allergic exacerbations by amplifying type 2 inflammation. We hypothesized that blocking IL-33 in RSV-induced exacerbation would significantly reduce allergic inflammation. Methods Sensitized BALB/c mice were challenged with aerosolized ovalbumin (OVA) to establish allergic inflammation, followed by RSV-A2 infection to yield four treatment groups: saline only (Saline), RSV-infected alone (RSV), OVA alone (OVA), and OVA-treated with RSV infection (OVA-RSV). Lung outcomes included lung mRNA and protein markers of allergic inflammation, histology for mucus cell metaplasia and lung immune cell influx by cytospin and flow cytometry. Results While thymic stromal lymphopoietin (TSLP) and IL-33 were detected 6 h after RSV infection in the OVA-RSV mice, IL-23 protein was uniquely upregulated in RSV-infected mice alone. OVA-RSV animals varied from RSV- or OVA-treated mice as they had increased lung eosinophils, neutrophils, group 2 innate lymphoid cells (ILC2) and group 3 innate lymphoid cells (ILC3) detectable as early as 6 h after RSV infection. Neutralized IL-33 significantly reduced ILC2 and eosinophils, and the prototypical allergic proteins, IL-5, IL-13, CCL17 and CCL22 in OVA-RSV mice. Numbers of neutrophils and ILC3 were also reduced with anti-IL-33 treatment in both RSV and OVA-RSV treated animals as well. Conclusions Taken together, our findings indicate a broad reduction in allergic-proinflammatory events mediated by IL-33 neutralization in RSV-induced asthma exacerbation.
Collapse
Affiliation(s)
- Kristi J Warren
- Division of Pulmonary Medicine, Department of Internal Medicine, University of Utah Health, 26 N 1900 E, Salt Lake City, UT, 84132, USA. .,VA Salt Lake City Health Care System, Salt Lake City, UT, 84148, USA.
| | - Jill A Poole
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - Jenea M Sweeter
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - Jane M DeVasure
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - John D Dickinson
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA
| | - R Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, USA
| | - Todd A Wyatt
- Critical Care and Sleep Division, Department of Internal Medicine, Pulmonary, University of Nebraska Medical Center, Omaha, USA.,Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, 68198-5910, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| |
Collapse
|
10
|
Vu LD, Saravia J, Jaligama S, Baboeram Panday RV, Sullivan RD, Mancarella S, Cormier SA, Kimura D. Deficiency in ST2 signaling ameliorates RSV-associated pulmonary hypertension. Am J Physiol Heart Circ Physiol 2021; 321:H309-H317. [PMID: 34170196 DOI: 10.1152/ajpheart.00018.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulmonary hypertension (PH) observed during respiratory syncytial virus (RSV) bronchiolitis is associated with morbidity and mortality, especially in children with congenital heart disease. Yet, the pathophysiological mechanisms of RSV-associated PH remain unclear. Therefore, this study aimed to investigate the pathophysiological mechanism of RSV-associated PH. We used a translational mouse model of RSV-associated PH, in which wild-type (WT) and suppression of tumorigenicity 2 (ST2) knockout neonatal mice were infected with RSV at 5 days old and reinfected 4 wk later. The development of PH in WT mice following RSV reinfection was evidenced by elevated right ventricle systolic pressure, shortened pulmonary artery acceleration time (PAT), and decreased PAT/ejection time (ET) ratio. It coincided with the augmentation of periostin and IL-13 expression and increased arginase bioactivity by both arginase 1 and 2 as well as induction of nitric oxide synthase (NOS) uncoupling. Absence of ST2 signaling prevented RSV-reinfected mice from developing PH by suppressing NOS uncoupling. In summary, ST2 signaling was involved in the development of RSV-associated PH. ST2 signaling inhibition may be a novel therapeutic target for RSV-associated PH.NEW & NOTEWORTHY We report that the pathogenic role of ST2-mediated type 2 immunity and mechanisms contribute to RSV-associated pulmonary hypertension. Inhibiting ST2 signaling may be a novel therapeutic target for this condition.
Collapse
Affiliation(s)
- Luan D Vu
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana
| | - Jordy Saravia
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Le Bonheur Children's Hospital, Memphis, Tennessee.,Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Sridhar Jaligama
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Le Bonheur Children's Hospital, Memphis, Tennessee.,IIT Research Institute, Chicago, Illinois
| | | | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Internal Medicine, University of Arizona College of Medicine-Phoenix, Phoenix, Arizona
| | - Salvatore Mancarella
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Stephania A Cormier
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, Louisiana.,Le Bonheur Children's Hospital, Memphis, Tennessee
| | - Dai Kimura
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee.,Le Bonheur Children's Hospital, Memphis, Tennessee
| |
Collapse
|
11
|
Wirtz S, Schulz-Kuhnt A, Neurath MF, Atreya I. Functional Contribution and Targeted Migration of Group-2 Innate Lymphoid Cells in Inflammatory Lung Diseases: Being at the Right Place at the Right Time. Front Immunol 2021; 12:688879. [PMID: 34177944 PMCID: PMC8222800 DOI: 10.3389/fimmu.2021.688879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
During the last decade, group-2 innate lymphoid cells (ILC2s) have been discovered and successfully established as crucial mediators of lung allergy, airway inflammation and fibrosis, thus affecting the pathogenesis and clinical course of many respiratory diseases, like for instance asthma, cystic fibrosis and chronic rhinosinusitis. As an important regulatory component in this context, the local pulmonary milieu at inflammatory tissue sites does not only determine the activation status of lung-infiltrating ILC2s, but also influences their motility and migratory behavior. In general, many data collected in recent murine and human studies argued against the former concept of a very strict tissue residency of innate lymphoid cells (ILCs) and instead pointed to a context-dependent homing capacity of peripheral blood ILC precursors and the inflammation-dependent capacity of specific ILC subsets for interorgan trafficking. In this review article, we provide a comprehensive overview of the so far described molecular mechanisms underlying the pulmonary migration of ILC2s and thereby the numeric regulation of local ILC2 pools at inflamed or fibrotic pulmonary tissue sites and discuss their potential to serve as innovative therapeutic targets in the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Stefan Wirtz
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Anja Schulz-Kuhnt
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
12
|
Rey-Jurado E, Bohmwald K, Gálvez NMS, Becerra D, Porcelli SA, Carreño LJ, Kalergis AM. Contribution of NKT cells to the immune response and pathogenesis triggered by respiratory viruses. Virulence 2021; 11:580-593. [PMID: 32463330 PMCID: PMC7549913 DOI: 10.1080/21505594.2020.1770492] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human respiratory syncytial virus (hRSV) and human metapneumovirus (hMPV) cause acute respiratory tract infections in children worldwide. Natural killer T (NKT) cells are unconventional T lymphocytes, and their TCRs recognize glycolipids bound to the MHC-I-like molecule, CD1d. These cells modulate the inflammatory response in viral infections. Here, we evaluated the contribution of NKT cells in both hRSV and hMPV infections. A significant decrease in the number of neutrophils, eosinophils, and CD103+DCs infiltrating to the lungs, as well as an increased production of IFN-γ, were observed upon hRSV-infection in CD1d-deficient BALB/c mice, as compared to wild-type control mice. However, this effect was not observed in the CD1d-deficient BALB/c group, upon infection with hMPV. Importantly, reduced expression of CD1d in CD11b+ DCs and epithelial cells was found in hRSV -but not hMPV-infected mice. Besides, a reduction in the expression of CD1d in alveolar macrophages of lungs from hRSV- and hMPV-infected mice was found. Such reduction of CD1d expression interfered with NKT cells activation, and consequently IL-2 secretion, as characterized by in vitro experiments for both hRSV and hMPV infections. Furthermore, increased numbers of NKT cells recruited to the lungs in response to hRSV- but not hMPV-infection was detected, resulting in a reduction in the expression of IFN-γ and IL-2 by these cells. In conclusion, both hRSV and hMPV might be differently impairing NKT cells function and contributing to the immune response triggered by these viruses.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Daniela Becerra
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile
| | - Steven A Porcelli
- Department of Microbiology and Immunology, and Department of Medicine, Albert Einstein College of Medicine , Bronx, NY, USA
| | - Leandro J Carreño
- Millennium Institute on Immunology and Immunotherapy, Programa de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile , Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile , Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile , Santiago, Chile
| |
Collapse
|
13
|
Zeng Z, Hong XY, Li Y, Chen W, Ye G, Li Y, Luo Y. Serum-soluble ST2 as a novel biomarker reflecting inflammatory status and illness severity in patients with COVID-19. Biomark Med 2020; 14:1619-1629. [PMID: 33336592 DOI: 10.2217/bmm-2020-0410] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: The authors studied the role of soluble ST2 (sST2) in COVID-19 and its relationship with inflammatory status and disease severity. Materials & methods: Serum levels of sST2 and interleukin (IL)-33, C-reactive protein (CRP), serum amyloid protein (SAA), IL-6 and procalcitonin (PCT), and T lymphocyte subsets from 80 subjects diagnosed with COVID-19 including 36 mild, 41 severe and three asymptomatic cases were tested. Results: Serum sST2 levels were significantly increased in COVID-19 patients, which were positively correlated with CRP, but negatively correlated with CD4+ and CD8+ T lymphocyte counts. Serum sST2 levels in nonsurviving severe cases were persistently high during disease progression. Conclusion: Serum sST2 level test is helpful for reflecting inflammatory status and illness severity of COVID-19.
Collapse
Affiliation(s)
- Zhikun Zeng
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| | - Xiao-Yue Hong
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| | - Yunhui Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Wei Chen
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| | - Guangming Ye
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| | - Yi Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Hubei, Wuhan, 430071, China
| |
Collapse
|
14
|
Jiang Y, Liao H, Zhang X, Cao S, Hu X, Yang Z, Fang Y, Wang H. IL-33 synergistically promotes the proliferation of lung cancer cells in vitro by inducing antibacterial peptide LL-37 and proinflammatory cytokines in macrophages. Immunobiology 2020; 225:152025. [PMID: 33190003 DOI: 10.1016/j.imbio.2020.152025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/30/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Lung cancer is the primary cause of cancer-related deaths, and the persistent inflammation is inextricably linked with the lung cancer tumorigenesis. Pro-inflammatory cytokine interleukin-33 (IL-33) is able to serve as a potent modulator of cancer. Mounting evidence indicates IL-33 has significant effect on lung cancer progression by regulating host immune response, but the current opinions about the function and mechanism of IL-33 in lung cancer are still controversial. Meanwhile, antibacterial peptide LL-37 also exerts a momentous effect on immune responses to lung cancer. LL-37 is regarded as versatile, including antimicrobial activities, chemotaxis and immunoregulation. However, the immunomodulatory mechanism of IL-33 and LL-37 in lung cancer remains thoroughly not defined. Here, we determined the secretion of LL-37 was up-regulated in lung cancer serum samples. Similarly, the expression of CRAMP was enhancive in macrophages after co-cultured with lung cancer cells. Moreover, we expounded that IL-33 could up-regulate LL-37 secretion in macrophages, resulting in the massive releases of IL-6 and IL-1β. Additionally, LL-37 cooperated with IL-33 to increase the phosphorylation of p38 MAPK and NF-κB p65 pathways, and augmented IL-6 and IL-1β secretion, which resulting in the proliferation of lung cancer cells in vitro. In conclusion, our study identified that IL-33 aggravated the inflammation of lung cancer by increasing LL-37 expression in macrophages, thereby promoting lung cancer cell proliferation in vitro. It is contributed to our present understanding of the immunomodulatory relationship between pro-inflammatory cytokines and antibacterial peptides in the tumor immune response, and offer a novel perspective for controlling the progress of lung cancer.
Collapse
Affiliation(s)
- Yinting Jiang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hongyi Liao
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; Department of Clinical Laboratory Medicine, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Sijia Cao
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xuexue Hu
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zihan Yang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuting Fang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hong Wang
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing 400016, China; School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
15
|
Respiratory syncytial virus upregulates IL-33 expression in mouse model of virus-induced inflammation exacerbation in OVA-sensitized mice and in asthmatic subjects. Cytokine 2020; 138:155349. [PMID: 33132030 DOI: 10.1016/j.cyto.2020.155349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Bronchial asthma (BA) is a chronic disease of the airways. The great majority of BA exacerbations are associated with respiratory viral infections. Recent findings point out a possible role of proinflammatory cytokine interleukin-33 (IL-33) in the development of atopic diseases. Although, little is known about the role of IL-33 in virus-induced BA exacerbations. METHODS We used mouse models of RSV (respiratory syncytial virus)-induced inflammation exacerbation in OVA-sensitized mice and RSV infection alone in adult animals to characterize expression of il33 in the mouse lungs. Moreover, we studied the influence of il33 knockdown with intranasally administrated siRNA on the development of RSV-induced inflammation exacerbation. In addition, we evaluated the expression of IL33 in the ex vivo stimulated PBMCs from allergic asthma patients and healthy subjects with and without confirmed acute respiratory viral infection. RESULTS Using mouse models, we found that infection with RSV drives enhanced il33 mRNA expression in the mouse lung. Treatment with anti-il33 siRNA diminishes airway inflammation in the lungs (we found a decrease in the number of inflammatory cells in the lungs and in the severity of histopathological alterations) of mice with RSV-induced inflammation exacerbation, but do not influence viral load. Elevated level of the IL33 mRNA was detected in ex vivo stimulated blood lymphocytes of allergic asthmatics infected with respiratory viruses. RSV and rhinovirus were the most detected viruses in volunteers with symptoms of respiratory infection. CONCLUSION The present study provides additional evidence of the crucial role of the IL-33 in pathogenesis of RSV infection and virus-induced allergic bronchial asthma exacerbations.
Collapse
|
16
|
Valeff N, Juriol L, Quadrana F, Muzzio DO, Zygmunt M, Quiroga MF, Ventimiglia MS, Jensen F. Expression of IL-33 Receptor Is Significantly Up-Regulated in B Cells During Pregnancy and in the Acute Phase of Preterm Birth in Mice. Front Immunol 2020; 11:446. [PMID: 32292403 PMCID: PMC7118206 DOI: 10.3389/fimmu.2020.00446] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 02/26/2020] [Indexed: 12/22/2022] Open
Abstract
Interleukin-33 (IL-33) is a mucosal alarmin belonging to the IL-1 cytokine family and is now recognized to have a key role in innate and adaptive immunity, contributing to tissue homeostasis and response to environmental stresses. In addition, IL-33 has also been shown to work as a positive regulator that initiates and maintains a Th2 immune response. In the context of pregnancy, it has been recently demonstrated that upon certain stress conditions, such as an infection induced inflammation, IL-33 is released from the uterine mucosa and triggers decidual B cells to produce anti-inflammatory molecules, which in turn restore immune homeostasis and prevents the development of preterm birth. In this study we therefore performed a detailed characterization of IL-33 receptor (Il1rl1 or ST2) expression in B cells during normal pregnancy, as well as in a mouse model of preterm birth. We observed that splenic B cells significantly up-regulate the expression of Il1rl1 during pregnancy and identified the B1 B cell population as the main ST2-expressing B cell subset. A further kinetic analysis showed that percentages of ST2-expressing B1 B cells are significantly augmented on days 12 and 14 of pregnancy, both in the spleen and peritoneal cavity of pregnant mice, and then drop toward the end of pregnancy to the levels observed in non-pregnant animals. Furthermore, using a mouse model of LPS-induced preterm birth, we demonstrated that not only are the percentages of ST2-expressing B1 B cells significantly enlarged in the spleen during the acute phase of preterm birth, but decidual B cells also significantly up-regulate ST2 expression as compared to term-pregnant mice. Overall, our results suggest a functional role of ST2 expression in B cells during pregnancy and reinforce the importance of the IL-33/ST2 axis in B cells as a critical mechanism to control inflammation-induced preterm birth.
Collapse
Affiliation(s)
- Natalin Valeff
- Laboratory for Immunology of Pregnancy, Center for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), Buenos Aires, Argentina
| | - Lorena Juriol
- Laboratory for Immunology of Pregnancy, Center for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), Buenos Aires, Argentina
| | - Florencia Quadrana
- Laboratory for Immunology of Pregnancy, Center for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), Buenos Aires, Argentina
| | - Damián Oscar Muzzio
- Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Marek Zygmunt
- Research Laboratory, Department of Obstetrics and Gynecology, University of Greifswald, Greifswald, Germany
| | - Maria Florencia Quiroga
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Facultad de Medicina, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Silvia Ventimiglia
- Laboratory for Immunology of Pregnancy, Center for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), Buenos Aires, Argentina
| | - Federico Jensen
- Laboratory for Immunology of Pregnancy, Center for Pharmacological and Botanical Studies (CEFYBO-CONICET-UBA), Buenos Aires, Argentina.,Institute of Health Sciences, National University Arturo Jauretche, Buenos Aires, Argentina
| |
Collapse
|
17
|
Wu Y, Lai AC, Chi P, Thio CL, Chen W, Tsai C, Lee YL, Lukacs NW, Chang Y. Pulmonary IL-33 orchestrates innate immune cells to mediate respiratory syncytial virus-evoked airway hyperreactivity and eosinophilia. Allergy 2020; 75:818-830. [PMID: 31622507 DOI: 10.1111/all.14091] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is epidemiologically linked to asthma. During RSV infection, IL-33 is elevated and promotes immune cell activation, leading to the development of asthma. However, which immune cells are responsible for triggering airway hyperreactivity (AHR), inflammation and eosinophilia remained to be clarified. We aimed to elucidate the individual roles of IL-33-activated innate immune cells, including ILC2s and ST2+ myeloid cells, in RSV infection-triggered pathophysiology. METHODS The role of IL-33/ILC2 axis in RSV-induced AHR inflammation and eosinophilia were evaluated in the IL-33-deficient and YetCre-13 Rosa-DTA mice. Myeloid-specific, IL-33-deficient or ST2-deficient mice were employed to examine the role of IL-33 and ST2 signaling in myeloid cells. RESULTS We found that IL-33-activated ILC2s were crucial for the development of AHR and airway inflammation, during RSV infection. ILC2-derived IL-13 was sufficient for RSV-driven AHR, since reconstitution of wild-type ILC2 rescued RSV-driven AHR in IL-13-deficient mice. Meanwhile, myeloid cell-derived IL-33 was required for airway inflammation, ST2+ myeloid cells contributed to exacerbation of airway inflammation, suggesting the importance of IL-33 signaling in these cells. Local and peripheral eosinophilia is linked to both ILC2 and myeloid IL-33 signaling. CONCLUSIONS This study highlights the importance of IL-33-activated ILC2s in mediating RSV-triggered AHR and eosinophilia. In addition, IL-33 signaling in myeloid cells is crucial for airway inflammation.
Collapse
Affiliation(s)
- Yi‐Hsiu Wu
- Taiwan International Graduate Program in Molecular Medicine National Yang‐Ming University and Academia Sinica Taipei Taiwan
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | | | - Po‐Yu Chi
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | | | - Wei‐Yu Chen
- Institute for Translational Research in Biomedicine Chang Gung Memorial Hospital Kaohsiung Taiwan
| | - Ching‐Hui Tsai
- Institute of Epidemiology and Preventive Medicine National Taiwan University Taipei Taiwan
| | - Yungling Leo Lee
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
- Institute of Epidemiology and Preventive Medicine National Taiwan University Taipei Taiwan
| | | | - Ya‐Jen Chang
- Taiwan International Graduate Program in Molecular Medicine National Yang‐Ming University and Academia Sinica Taipei Taiwan
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| |
Collapse
|
18
|
Portugal CAA, de Araújo Castro Í, Prates MCM, Gagliardi TB, Martins RB, de Jesus BLS, de Souza Cardoso R, da Silva MVG, Aragon DC, Arruda Neto E, Alves Filho JCF, Cunha FDQ, Carlotti APDCP. IL-33 and ST2 as predictors of disease severity in children with viral acute lower respiratory infection. Cytokine 2020; 127:154965. [PMID: 31901762 PMCID: PMC7129023 DOI: 10.1016/j.cyto.2019.154965] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/09/2019] [Accepted: 12/25/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Mechanisms influencing severity of acute lower respiratory infection (ALRI) in children are not established. We aimed to assess the role of inflammatory markers and respiratory viruses in ALRI severity. METHODS Concentrations of interleukin(IL)-33, soluble suppression of tumorigenicity (sST)2, IL-1ß, tumor necrosis factor α, IL-4, IL-6 and IL- 8 and types of respiratory viruses were evaluated in children at the first and fifth days after hospital admission. Disease severity was defined as need for mechanical ventilation. RESULTS Seventy-nine children <5 years-old were included; 33(41.8%) received mechanical ventilation. No associations between virus type, viral load or co-detections and severity of disease were observed. Detection of IL-33 and sST2 in nasopharyngeal aspirates (NPA) on admission were associated with higher risk for mechanical ventilation (RR = 2.89 and RR = 4.57, respectively). IL-6 and IL-8 concentrations were higher on Day 5 in mechanically ventilated children. IL-6 NPA concentrations decreased from Day 1 to Day 5 in children who did not receive mechanical ventilation. Increase in sST2 NPA concentrations from Day 1 to Day 5 was associated with longer hospital length of stay (p < 0.01). CONCLUSIONS An exacerbated local activation of the IL-33/ST2 axis and persistently high sST2 concentrations over time were associated with severity of viral ALRI in children.
Collapse
Affiliation(s)
| | - Ítalo de Araújo Castro
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mirela Cristina Moreira Prates
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Talita Bianca Gagliardi
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ronaldo Bragança Martins
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruna Laís Santos de Jesus
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ricardo de Souza Cardoso
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marcus Vinícius Gomes da Silva
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Davi Casale Aragon
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eurico Arruda Neto
- Department of Cell Biology and Virology Research Center, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Fernando de Queiroz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | |
Collapse
|
19
|
Johansson K, McSorley HJ. Interleukin-33 in the developing lung-Roles in asthma and infection. Pediatr Allergy Immunol 2019; 30:503-510. [PMID: 30734382 DOI: 10.1111/pai.13040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 02/07/2023]
Abstract
It has become increasingly clear that interleukin-33 (IL-33) plays a crucial role in initiation of type 2 immunity. The last decade of intense research has uncovered multiple mechanisms through which IL-33 targets key effector cells of the allergic immune response. Recently, IL-33 has been implicated in shaping the immune system of the lungs early in life, at a time which is crucial in the subsequent development of allergic asthma. In this review, we will address the current literature describing the role of IL-33 in the healthy and diseased lung. In particular, we will focus on the evidence for IL-33 in the development of immune responses in the lung, including the role of IL-33-responsive immune cells that may explain susceptibility to allergic sensitization at a young age and the association between genetic variants of IL-33 and asthma in humans. Finally, we will indicate areas for potential therapeutic modulation of the IL-33 pathway.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California, San Francisco, California.,Department of Medicine, University of California, San Francisco, California
| | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
20
|
Interleukin 1 Receptor-Like 1 (IL1RL1) Promotes Airway Bacterial and Viral Infection and Inflammation. Infect Immun 2019; 87:IAI.00340-19. [PMID: 31061143 DOI: 10.1128/iai.00340-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 01/14/2023] Open
Abstract
Interleukin 1 receptor-like 1 (IL1RL1), also known as suppression of tumorigenicity 2 (ST2), is the receptor for interleukin 33 (IL-33) and has been increasingly studied in type 2 inflammation. An increase in airway IL-33/ST2 signaling in asthma has been associated with eosinophilic inflammation, but little is known about the role of ST2 in neutrophilic inflammation. Airway Mycoplasma pneumoniae and human rhinovirus (HRV) infections are linked to neutrophilic inflammation during acute exacerbations of asthma. However, whether ST2 contributes to M. pneumoniae- and HRV-mediated airway inflammation is poorly understood. The current study sought to determine the functions of ST2 during airway M. pneumoniae or HRV infection. In cultured normal human primary airway epithelial cells, ST2 overexpression (OE) increased the production of neutrophilic chemoattractant IL-8 in the absence or presence of M. pneumoniae or HRV1B infection. ST2 OE also enhanced HRV1B-induced IP-10, a chemokine involved in asthma exacerbations. In the M. pneumoniae-infected mouse model, ST2 deficiency, in contrast to sufficiency, significantly reduced the levels of neutrophils following acute (≤24 h) infection, while in the HRV1B-infected mouse model, ST2 deficiency significantly reduced the levels of proinflammatory cytokines KC, IP-10, and IL-33 in bronchoalveolar lavage (BAL) fluid. Overall, ST2 overexpression in human epithelial cells and ST2 sufficiency in mice increased the M. pneumoniae and HRV loads in cell supernatants and BAL fluid. After pathogen infection, ST2-deficient mice showed a higher level of the host defense protein lactotransferrin in BAL fluid. Our data suggest that ST2 promotes proinflammatory responses (e.g., neutrophils) to airway bacterial and viral infection and that blocking ST2 signaling may broadly attenuate airway infection and inflammation.
Collapse
|
21
|
Zhang Y, Li S, Huang S, Cao L, Liu T, Zhao J, Wu J, Wang J, Cao L, Xu J, Dong L. IL33/ST2 contributes to airway remodeling via p-JNK MAPK/STAT3 signaling pathway in OVA-induced allergic airway inflammation in mice. Exp Lung Res 2019; 45:65-75. [PMID: 31112061 DOI: 10.1080/01902148.2019.1611972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aim of this study: Airway remodeling, which encompasses structural changes in airway is a main feature of asthma. Interleukin-33 (IL-33) has been reported to be a vital cytokine in airway remodeling in asthma, but the underlying mechanisms are not clear yet. This study focused on discussing the role of IL-33 in airway remodeling in asthma. Material and methods: Female BALB/c mice were divided into a control group, an OVA induced allergic airway disease group and an anti-ST2 antibody intervention group. Immunohistochemistry and western blot were performed to detect IL-33, ST2 expression in addition to airway remodeling markers a-smooth muscle actin (a-SMA) and type 1 collagen in OVA-induced mice model. Levels of p-JNK and p-STAT3 activation in mice were detected by western blot. Human lung fibroblast (HLF) were stimulated with rhIL-33, anti-ST2 antibody and JNK inhibitor sp600125 and levels of JNK and STAT3 activation were determined via western blot and immunofluorescence staining. Results: Anti-ST2 treatment inhibited JNK/STAT3 phosphorylation and airway remodeling in OVA-induced mouse model. IL-33 induced a-SMA and collagen 1 expression was inhibited by anti-ST2 antibody and sp600125 treatment via decreased JNK/STAT3 phosphorylation in human lung fibroblast. Conclusions: IL-33 promoted airway remodeling by interacting with ST2 to activate the JNK/STAT3 signaling pathway in asthma.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Shuo Li
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Siyuan Huang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Liuzhao Cao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Tian Liu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jiping Zhao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jinxiang Wu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Junfei Wang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Lili Cao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jiawei Xu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Liang Dong
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| |
Collapse
|
22
|
Alvarez F, Fritz JH, Piccirillo CA. Pleiotropic Effects of IL-33 on CD4 + T Cell Differentiation and Effector Functions. Front Immunol 2019; 10:522. [PMID: 30949175 PMCID: PMC6435597 DOI: 10.3389/fimmu.2019.00522] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/26/2019] [Indexed: 12/16/2022] Open
Abstract
IL-33, a member of the IL-1 family of cytokines, was originally described in 2005 as a promoter of type 2 immune responses. However, recent evidence reveals a more complex picture. This cytokine is released locally as an alarmin upon cellular damage where innate cell types respond to IL-33 by modulating their differentiation and influencing the polarizing signals they provide to T cells at the time of antigen presentation. Moreover, the prominent expression of the IL-33 receptor, ST2, on GATA3+ T helper 2 cells (TH2) demonstrated that IL-33 could have a direct impact on T cells. Recent observations reveal that T-bet+ TH1 cells and Foxp3+ regulatory T (TREG) cells can also express the ST2 receptor, either transiently or permanently. As such, IL-33 can have a direct effect on the dynamics of T cell populations. As IL-33 release was shown to play both an inflammatory and a suppressive role, understanding the complex effect of this cytokine on T cell homeostasis is paramount. In this review, we will focus on the factors that modulate ST2 expression on T cells, the effect of IL-33 on helper T cell responses and the role of IL-33 on TREG cell function.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Center, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
| | - Jörg H. Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
- McGill University Research Center on Complex Traits, McGill University, Montréal, QC, Canada
| | - Ciriaco A. Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, The Research Institute of the McGill University Health Center, Montréal, QC, Canada
- Centre of Excellence in Translational Immunology, Montréal, QC, Canada
- McGill University Research Center on Complex Traits, McGill University, Montréal, QC, Canada
| |
Collapse
|
23
|
Sheshadri A, Chemaly RF, Alousi AM, Shah PK, Rondon G, Bashoura L, Kmeid J, Azzi J, Blanco DW, Kaous M, Dickey BF, Champlin RE, Shah DP. Pulmonary Impairment after Respiratory Viral Infections Is Associated with High Mortality in Allogeneic Hematopoietic Cell Transplant Recipients. Biol Blood Marrow Transplant 2018; 25:800-809. [PMID: 30521974 DOI: 10.1016/j.bbmt.2018.11.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/19/2018] [Indexed: 11/28/2022]
Abstract
Pulmonary impairment predicts increased mortality in many settings, and respiratory viral infection (RVI) causes considerable morbidity and mortality in allogeneic hematopoietic cell transplant recipients (allo-HCT). We hypothesized that pulmonary impairment after RVI, defined as a decline of forced expiratory volume in 1 second values by ≥10%, may identify allo-HCT recipients at high risk for mortality. We studied all allo-HCT recipients at our institution who had RVI with respiratory syncytial virus, parainfluenza virus, or influenza from 2004 to 2013 and had pre-RVI and post-RVI pulmonary function tests. We used competing risk regression models to identify risk factors for 2-year nonrelapse mortality (NRM) as the primary outcome after RVI and relapse-related mortality as a competing risk. From 223 eligible patients, pulmonary impairment after RVI was associated with over a 3-fold increase in 2-year NRM (pulmonary impairment, 25.3%; no impairment, 7.4%; univariate subhazard ratio [SHR], 3.9; 95% confidence interval [CI], 1.9 to 8.1; P < .001). After adjusting for age and systemic steroid use, pulmonary impairment after RVI was still associated with increased 2-year NRM (SHR, 3.3 [95% CI, 1.6 to 6.9]; P = .002). After adjustment for race and graft-versus-host disease (GVHD) prophylaxis, chronic GVHD at the time of RVI (odds ratio [OR], 2.8 [95% CI, 1.4 to 5.4]; p = .003) and lymphopenia (OR, 2.2 [95% CI, 1.1 to 4.2]; P = .02) were associated with increased odds of pulmonary impairment, whereas use of nonmyeloablative conditioning was associated with reduced odds of pulmonary impairment (OR, .4 [95% CI, .2 to .8]; P = .006). In allo-HCT recipients with RVIs, pulmonary impairment after RVI is associated with high NRM at 2years.
Collapse
Affiliation(s)
- Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roy F Chemaly
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Amin M Alousi
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pankil K Shah
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriela Rondon
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lara Bashoura
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joumana Kmeid
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jacques Azzi
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David W Blanco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maryam Kaous
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Burton F Dickey
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard E Champlin
- Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dimpy P Shah
- Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
24
|
Abukabda AB, McBride CR, Batchelor TP, Goldsmith WT, Bowdridge EC, Garner KL, Friend S, Nurkiewicz TR. Group II innate lymphoid cells and microvascular dysfunction from pulmonary titanium dioxide nanoparticle exposure. Part Fibre Toxicol 2018; 15:43. [PMID: 30413212 PMCID: PMC6230229 DOI: 10.1186/s12989-018-0280-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Background The cardiovascular effects of pulmonary exposure to engineered nanomaterials (ENM) are poorly understood, and the reproductive consequences are even less understood. Inflammation remains the most frequently explored mechanism of ENM toxicity. However, the key mediators and steps between lung exposure and uterine health remain to be fully defined. The purpose of this study was to determine the uterine inflammatory and vascular effects of pulmonary exposure to titanium dioxide nanoparticles (nano-TiO2). We hypothesized that pulmonary nano-TiO2 exposure initiates a Th2 inflammatory response mediated by Group II innate lymphoid cells (ILC2), which may be associated with an impairment in uterine microvascular reactivity. Methods Female, virgin, Sprague-Dawley rats (8–12 weeks) were exposed to 100 μg of nano-TiO2 via intratracheal instillation 24 h prior to microvascular assessments. Serial blood samples were obtained at 0, 1, 2 and 4 h post-exposure for multiplex cytokine analysis. ILC2 numbers in the lungs were determined. ILC2s were isolated and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) levels were measured. Pressure myography was used to assess vascular reactivity of isolated radial arterioles. Results Pulmonary nano-TiO2 exposure was associated with an increase in IL-1ß, 4, 5 and 13 and TNF- α 4 h post-exposure, indicative of an innate Th2 inflammatory response. ILC2 numbers were significantly increased in lungs from exposed animals (1.66 ± 0.19%) compared to controls (0.19 ± 0.22%). Phosphorylation of the transactivation domain (Ser-468) of NF-κB in isolated ILC2 and IL-33 in lung epithelial cells were significantly increased (126.8 ± 4.3% and 137 ± 11% of controls respectively) by nano-TiO2 exposure. Lastly, radial endothelium-dependent arteriolar reactivity was significantly impaired (27 ± 12%), while endothelium-independent dilation (7 ± 14%) and α-adrenergic sensitivity (8 ± 2%) were not altered compared to control levels. Treatment with an anti- IL-33 antibody (1 mg/kg) 30 min prior to nano-TiO2 exposure resulted in a significant improvement in endothelium-dependent dilation and a decreased level of IL-33 in both plasma and bronchoalveolar lavage fluid. Conclusions These results provide evidence that the uterine microvascular dysfunction that follows pulmonary ENM exposure may be initiated via activation of lung-resident ILC2 and subsequent systemic Th2-dependent inflammation. Electronic supplementary material The online version of this article (10.1186/s12989-018-0280-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alaeddin Bashir Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll Rolland McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas Paul Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William Travis Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Compton Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Lee Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Robert Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
25
|
Boudaud M, Turcotte S, Stankova J, Rola-Pleszczynski M. IL-33 Upregulates Cysteinyl Leukotriene Receptor Type 1 Expression in Human Peripheral Blood CD4 + T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2018; 201:2787-2798. [PMID: 30242072 DOI: 10.4049/jimmunol.1701463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 08/21/2018] [Indexed: 12/25/2022]
Abstract
IL-33 and cysteinyl leukotrienes (cysLTs) are key components of asthma pathogenesis, and both contribute to the initiation and maintenance of the type 2 inflammatory environment. However, little is known about the potential interactions between the two mediators. In this work, we aimed at studying the regulation of expression of the cysLT receptors CysLT1 and CysLT2 by IL-33 in human PBLs. Our results show that the IL-33/ST2L axis increases CysLT1 but not CysLT2 expression in a concentration- and time-dependent manner in PBLs. IL-33-induced CysLT1 upregulation was observed at the protein but not at the mRNA level and was accompanied by an increase in LTD4-induced calcium mobilization and migration of CD4+ T lymphocytes. We also show that purified naive CD4+ T lymphocytes expressed ST2L and responded to IL-33 in the absence of Ag or TCR stimulation, suggesting a mechanism independent of Ag presentation. These results contribute to expanding our knowledge in the field of IL-33 by proposing a new mode of action of the cytokine on T cells and by extending its role to the regulation of naive T cell trafficking, therefore reinforcing its interest as a potential therapeutic target for the treatment of asthma.
Collapse
Affiliation(s)
- Marie Boudaud
- Service d'Immunologie et Allergologie, Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Sylvie Turcotte
- Service d'Immunologie et Allergologie, Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Jana Stankova
- Service d'Immunologie et Allergologie, Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| | - Marek Rola-Pleszczynski
- Service d'Immunologie et Allergologie, Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, Canada
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Recent studies have highlighted the role of alarmins in asthma pathophysiology and tested the roles of these cytokines in asthmatic patients. This review will discuss the recent advances in the role of alarmins in asthma and the potential of future targeted therapies in asthma. RECENT FINDINGS Epithelial-derived cytokines can be released upon exposure to external stimuli, causing damage to the epithelial barrier and resulting in tissue inflammation. Of these cytokines, IL-25, IL-33 and thymic stromal lymphopoeitin (TSLP), have been associated with asthma. These alarmins are all not only overexpressed in asthmatic airways, particularly in airway epithelial cells, but also in other structural and immune cells. Furthermore, all three alarmins drive type-2 pro-inflammatory responses in several immune cells that have been identified as key players in the pathogenesis of asthma, including innate lymphoid type-2 cells. Clinical trials testing therapeutics that block pathways of the alarmins are in progress. SUMMARY To-date, only TSLP blockade has been reported in human clinical trials, and this approach has shown efficacy in asthmatic patients. Current body of evidence suggests that alarmins are useful upstream targets for treatment of asthma.
Collapse
|
27
|
Restori KH, Srinivasa BT, Ward BJ, Fixman ED. Neonatal Immunity, Respiratory Virus Infections, and the Development of Asthma. Front Immunol 2018; 9:1249. [PMID: 29915592 PMCID: PMC5994399 DOI: 10.3389/fimmu.2018.01249] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/18/2018] [Indexed: 12/27/2022] Open
Abstract
Infants are exposed to a wide range of potential pathogens in the first months of life. Although maternal antibodies acquired transplacentally protect full-term neonates from many systemic pathogens, infections at mucosal surfaces still occur with great frequency, causing significant morbidity and mortality. At least part of this elevated risk is attributable to the neonatal immune system that tends to favor T regulatory and Th2 type responses when microbes are first encountered. Early-life infection with respiratory viruses is of particular interest because such exposures can disrupt normal lung development and increase the risk of chronic respiratory conditions, such as asthma. The immunologic mechanisms that underlie neonatal host-virus interactions that contribute to the subsequent development of asthma have not yet been fully defined. The goals of this review are (1) to outline the differences between the neonatal and adult immune systems and (2) to present murine and human data that support the hypothesis that early-life interactions between the immune system and respiratory viruses can create a lung environment conducive to the development of asthma.
Collapse
Affiliation(s)
- Katherine H Restori
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Bharat T Srinivasa
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Brian J Ward
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Elizabeth D Fixman
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| |
Collapse
|
28
|
ILC2s in infectious diseases and organ-specific fibrosis. Semin Immunopathol 2018; 40:379-392. [PMID: 29623414 DOI: 10.1007/s00281-018-0677-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/12/2018] [Indexed: 12/13/2022]
Abstract
Type 2 immune responses evolved to provide host protection against parasitic infections and to support the repair of infection-induced tissue injury. However, persistent chronic organ damage can result in dysregulated production of critical type 2 cytokines supporting tissue remodeling and fibrosis development. Recently, group 2 innate lymphoid cells (ILC2s) were newly described as central innate mediators of type 2 responses. In particular, by secretion of the cytokines IL-5, IL-9, and IL-13 and the growth factor amphiregulin in response to the release of tissue-derived alarmins, ILC2s have been shown to substantially contribute to both the dismissal of metazoan parasites and the repair of infection-dependent or sterile tissue damage. Conversely, cytokine production by ILC2s emerged as a driving force for tissue remodeling and excessive fibrosis in several organ systems including the lung, liver, and skin. In this review, we discuss how ILC2s are specifically implicated in the body's immune response to different pathogenic infections and how dysregulated ILC2s may promote organ-specific fibrosis.
Collapse
|
29
|
Seo S, Yu J, Jenkins IC, Leisenring WM, Steven-Ayers T, Kuypers JM, Huang ML, Jerome KR, Boeckh M, Paczesny S. Diagnostic and Prognostic Plasma Biomarkers for Idiopathic Pneumonia Syndrome after Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2017; 24:678-686. [PMID: 29223372 DOI: 10.1016/j.bbmt.2017.11.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/28/2017] [Indexed: 11/16/2022]
Abstract
Idiopathic pneumonia syndrome (IPS) is a noninfectious pulmonary complication after hematopoietic cell transplantation (HCT) and is difficult to diagnose. In 41 patients with IPS, we evaluated 6 candidate proteins in plasma samples at day 7 post-HCT and at onset of IPS to identify potential diagnostic or prognostic biomarkers for IPS. Samples at similar times from 162 HCT recipients without documented infections and 37 HCT recipients with respiratory viral pneumonia served as controls. In multivariable models, a combination of Stimulation-2 (ST2; odds ratio [OR], 2.8; P < .001) and IL-6 (OR, 1.4; P = .025) was the best panel for distinguishing IPS at diagnosis from unaffected controls, whereas tumor necrosis factor receptor 1 (TNFR1; OR, 2.9; P = .002) was the best marker when comparing patients with IPS and viral pneumonia. The areas under the curve of the receiver operating characteristic (ROC) curves for discriminating between IPS and unaffected controls at day 7 post-HCT were .8 for ST2, .75 for IL-6, and .68 for TNFR1. Using estimated sensitivity and specificity values from cutoffs determined with the ROC analysis (cutoff level: ST2, 21 ng/mL; IL-6, 61 pg/mL; TNFR1, 3421 pg/mL), we calculated positive predictive values (PPV) for a range of estimated population prevalence values of IPS. Among the 3 markers, ST2 showed the highest PPV for IPS occurrence. Based on an assumed prevalence of 8%, a positive ST2 test increased likelihood of IPS to 50%. We conclude that a prospective validation study is warranted to determine whether a plasma biomarker panel can aid the noninvasive diagnosis and prognosis of IPS.
Collapse
Affiliation(s)
- Sachiko Seo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Hematology and Oncology, National Cancer Research Center East, Chiba, Japan
| | - Jeffrey Yu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Isaac C Jenkins
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wendy M Leisenring
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Terry Steven-Ayers
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jane M Kuypers
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Meei-Li Huang
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Michael Boeckh
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Medicine, University of Washington, Seattle, Washington.
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana.
| |
Collapse
|
30
|
Du LX, Wang YQ, Hua GQ, Mi WL. IL-33/ST2 Pathway as a Rational Therapeutic Target for CNS Diseases. Neuroscience 2017; 369:222-230. [PMID: 29175156 DOI: 10.1016/j.neuroscience.2017.11.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/13/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022]
Abstract
Interleukin (IL)-33 is a member of the interleukin-1 cytokine family that is produced by many different types of tissues including the central nervous system (CNS). IL-33 mediates its effects via its heterodimeric receptor complex, comprised of ST2 and the IL-1 receptor accessory protein (IL-1RAcp). As a pleiotropic nuclear cytokine, IL-33 is a crucial factor in the development of cardiovascular diseases, allergic diseases, infectious diseases, and autoimmune diseases. Recently, accumulated evidence shows that the IL-33/ST2 axis plays a crucial and diverse role in the pathogenesis of CNS diseases, including neurodegenerative diseases, cerebrovascular diseases, infectious diseases, traumatic CNS injury, chronic pain, etc. In this review, we discuss the recent findings in the cellular signaling of IL-33 and advancement of the role of IL-33 in several CNS diseases, as well as its therapeutic potential for the treatment of those diseases.
Collapse
Affiliation(s)
- Li-Xia Du
- Department of Integrative Medicine and Neurobiology, Academy of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, People's Republic of China
| | - Yan-Qing Wang
- Department of Integrative Medicine and Neurobiology, Academy of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, People's Republic of China
| | - Guo-Qiang Hua
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, People's Republic of China
| | - Wen-Li Mi
- Department of Integrative Medicine and Neurobiology, Academy of Integrative Medicine, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, People's Republic of China.
| |
Collapse
|
31
|
Han X, Chai R, Qi F, Bai S, Cui Y, Teng Y, Liu B. Natural helper cells mediate respiratory syncytial virus-induced airway inflammation by producing type 2 cytokines in an IL-33-dependent manner. Immunotherapy 2017; 9:715-722. [PMID: 28771101 DOI: 10.2217/imt-2017-0037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Type 2 cytokine production during respiratory virus infection is considered to be linked with asthma exacerbation. As potent Th2 cytokine producers, natural helper (NH) cells play a key role in influenza virus-induced airway hyper-responsiveness. However, it is unclear whether NH cells contribute to respiratory syncytial virus (RSV)-induced airway inflammation, and how the cytokine profile in NH cells is changed during RSV infection. METHODS BALB/c mice were infected intranasally with RSV. The number of NH cells in lungs was detected by flow cytometry. The expression of cytokine mRNAs was performed by real-time RT-PCR. Cytokines levels were determined by ELISA. RESULTS Following intranasal infection with RSV, BALB/c mice showed an increase in the expression of mRNAs for Th2-like cytokines in NH cells. Furthermore, adoptive transfer of pulmonary NH cells resulted in a massive infiltration of mononuclear cells, in particular eosinophils and neutrophils in lungs, in parallel with an augmented production of Th2-associated cytokines, such as IL-4, IL-5 and IL-10 in bronchoalveolar lavage fluids, providing convincing evidence that NH cells contribute to RSV-induced lung pathogenesis by producing type 2 cytokines. It should be noted that blocking IL-33 with antibody can diminish the absolute number of pulmonary NH cells and the relative expression of mRNAs for type 2 cytokines in pulmonary NH cells, suggesting that IL-33 is necessary for activating Th2-type NH cells. CONCLUSION These results reveal that pulmonary NH cells might participate in RSV-induced airway inflammation by producing large quality of type 2 cytokines in an IL-33-dependent manner.
Collapse
Affiliation(s)
- Xu Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, PR China
| | - Ruonan Chai
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Feifei Qi
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Song Bai
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Yulin Cui
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, PR China
| | - Beixing Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, PR China
| |
Collapse
|
32
|
Rey-Jurado E, Soto J, Gálvez N, Kalergis AM. A safe and efficient BCG vectored vaccine to prevent the disease caused by the human Respiratory Syncytial Virus. Hum Vaccin Immunother 2017; 13:2092-2097. [PMID: 28598702 DOI: 10.1080/21645515.2017.1334026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The human Respiratory Syncytial Virus (hRSV) causes lower respiratory tract infections including pneumonia and bronchiolitis. Such infections also cause a large number of hospitalizations and affects mainly newborns, young children and the elderly worldwide. Symptoms associated with hRSV infection are due to an exacerbated immune response characterized by low levels of IFN-γ, recruitment of neutrophils and eosinophils to the site of infection and lung damage. Although hRSV is a major health problem, no vaccines are currently available. Different immunization approaches have been developed to achieve a vaccine that activates the immune system, without triggering an unbalanced inflammation. These approaches include live attenuated vaccine, DNA or proteins technologies, and the use of vectors to express proteins of the virus. In this review, we discuss the host immune response to hRSV and the immunological mechanisms underlying an effective and safe BCG vectored vaccine against hRSV.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- a Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Jorge Soto
- a Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Nicolás Gálvez
- a Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile
| | - Alexis M Kalergis
- a Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas , Pontificia Universidad Católica de Chile , Santiago , Chile.,b Departamento de Endocrinología, Facultad de Medicina , Pontificia Universidad Católica de Chile , Santiago , Chile
| |
Collapse
|
33
|
Macrophages produce IL-33 by activating MAPK signaling pathway during RSV infection. Mol Immunol 2017; 87:284-292. [PMID: 28531812 DOI: 10.1016/j.molimm.2017.05.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/26/2017] [Accepted: 05/13/2017] [Indexed: 12/11/2022]
Abstract
It has been reported that RSV infection can enhance IL-33 production in lung macrophages. However, little is known about specific signaling pathways for activation of macrophages during RSV infection. In the present study, by using real-time RT-PCR as well as western blot assay, it became clear that RSV infection can enhance not only the expression of mRNAs for MAPK molecules (including p38, JNK1/2, and ERK1/2), but also the levels of MAPK proteins in lung macrophages as well as RAW264.7 cells. Furthermore, infection with RSV resulted in an increased level of phosphorylated MAPK proteins in RAW264.7 cells, suggesting that MAPK signaling pathway may participate in the process of RSV-induced IL-33 secretion by macrophages. In fact, the elevated production of IL-33 in RAW264.7 was attenuated significantly by pretreatment of the cells with special MAPK inhibitor before RSV infection, further confirming the function of MAPKs pathway in RSV-induced IL-33 production in macrophages. In contrast, the expression of NF-κB mRNA as well as the production of NF-κB protein in lung macrophages and RAW264.7 cells was not enhanced markedly after RSV infection. Moreover, RSV infection failed to induce the phosphorylation of NF-κB in RAW264.7 cells, suggesting that NF-κB signaling pathway may be not involved in RSV-induced IL-33 production in macrophages. Conclusion, these results indicate that RSV-induced production of IL-33 in macrophages is dependent on the activation of MAPK signaling pathway.
Collapse
|
34
|
Rey-Jurado E, Kalergis AM. Immunological Features of Respiratory Syncytial Virus-Caused Pneumonia-Implications for Vaccine Design. Int J Mol Sci 2017; 18:E556. [PMID: 28273842 PMCID: PMC5372572 DOI: 10.3390/ijms18030556] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/22/2017] [Accepted: 02/26/2017] [Indexed: 01/05/2023] Open
Abstract
The human respiratory syncytial virus (hRSV) is the causative agent for high rates of hospitalizations due to viral bronchiolitis and pneumonia worldwide. Such a disease is characterized by an infection of epithelial cells of the distal airways that leads to inflammation and subsequently to respiratory failure. Upon infection, different pattern recognition receptors recognize the virus and trigger the innate immune response against the hRSV. Further, T cell immunity plays an important role for virus clearance. Based on animal studies, it is thought that the host immune response to hRSV is based on a biased T helper (Th)-2 and Th17 T cell responses with the recruitment of T cells, neutrophils and eosinophils to the lung, causing inflammation and tissue damage. In contrast, human immunity against RSV has been shown to be more complex with no definitive T cell polarization profile. Nowadays, only a humanized monoclonal antibody, known as palivizumab, is available to protect against hRSV infection in high-risk infants. However, such treatment involves several injections at a significantly high cost. For these reasons, intense research has been focused on finding novel vaccines or therapies to prevent hRSV infection in the population. Here, we comprehensively review the recent literature relative to the immunological features during hRSV infection, as well as the new insights into preventing the disease caused by this virus.
Collapse
Affiliation(s)
- Emma Rey-Jurado
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330644, Chile.
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8330644, Chile.
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330644, Chile.
| |
Collapse
|
35
|
Christiaansen AF, Syed MA, Ten Eyck PP, Hartwig SM, Durairaj L, Kamath SS, Varga SM. Altered Treg and cytokine responses in RSV-infected infants. Pediatr Res 2016; 80:702-709. [PMID: 27486703 PMCID: PMC6215710 DOI: 10.1038/pr.2016.130] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 01/17/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in children under 1 y of age in the USA. The host immune response is believed to contribute to RSV-induced disease. We hypothesize that severe RSV infection in infants is mediated by insufficient regulation of the host immune response of regulatory T cells (Tregs) resulting in immunopathology. METHODS Blood and nasal aspirates from 23 RSV-infected and 17 control infants under 1 y of age were collected. Treg frequencies were determined by flow cytometry from peripheral blood mononuclear cells. Analysis of 24 cytokines was measured by multiplex assay on nasal aspirates. RESULTS We demonstrate that the frequency of activated Tregs is significantly reduced in the peripheral blood of RSV-infected infants compared with age-matched controls. Surprisingly, T helper (Th)17 related cytokines including interleukin (IL)-1β, IL-17A, and IL-23 were associated with a reduction in clinical symptoms of respiratory distress. In addition, the amount of IL-33 protein in nasal washes, a cytokine important in maintaining Treg homeostasis in mucosal tissues, was decreased in RSV-infected children. CONCLUSION These results suggest that decreased Treg numbers and an inability to properly control the host inflammatory response results in severe RSV infection.
Collapse
Affiliation(s)
| | | | - Patrick P. Ten Eyck
- Institute for Clinical and Translational Science, University of Iowa, Iowa City, IA
| | | | - Lakshmi Durairaj
- Department of Internal Medicine, University of Iowa, Iowa City, IA
| | | | - Steven M. Varga
- Department of Microbiology, University of Iowa, Iowa City, IA
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
- Department of Pathology, University of Iowa, Iowa City, IA
| |
Collapse
|
36
|
Lan F, Zhang N, Gevaert E, Zhang L, Bachert C. Viruses and bacteria in Th2-biased allergic airway disease. Allergy 2016; 71:1381-92. [PMID: 27188632 DOI: 10.1111/all.12934] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 01/24/2023]
Abstract
Allergic airway diseases are typically characterized by a type 2-biased inflammation. Multiple distinct viruses and bacteria have been detected in the airways. Recently, it has been confirmed that the microbiome of allergic individuals differs from that of healthy subjects, showing a close relationship with the type 2 response in allergic airway disease. In this review, we summarize the recent findings on the prevalence of viruses and bacteria in type 2-biased airway diseases and on the mechanisms employed by viruses and bacteria in propagating type 2 responses. The understanding of the microbial composition and postinfectious immune programming is critical for the reconstruction of the normal microflora and immune status in allergic airway diseases.
Collapse
Affiliation(s)
- F. Lan
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - N. Zhang
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - E. Gevaert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - L. Zhang
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - C. Bachert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Division of ENT Diseases; Clintec; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
37
|
Qi F, Wang D, Liu J, Zeng S, Xu L, Hu H, Liu B. Respiratory macrophages and dendritic cells mediate respiratory syncytial virus-induced IL-33 production in TLR3- or TLR7-dependent manner. Int Immunopharmacol 2016; 29:408-415. [PMID: 26603638 DOI: 10.1016/j.intimp.2015.10.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 09/30/2015] [Accepted: 10/19/2015] [Indexed: 12/24/2022]
Abstract
Respiratory syncytial virus (RSV) infection can increase the production of IL-33 in lungs of mice. However, little is known about cellular source of IL-33, particularly the types of IL-33-producing cells in innate immune cells during RSV infection. In this study, by using BALB/c mice that were infected intranasally with RSV, it became clear that RSV infection can enhance not only the number of IL-33(+)-alveolar macrophages (AMs) and dendritic cells (DCs), but also the expression of IL-33 mRNA in these cells, suggesting that innate immune cells participate in the production of IL-33. Indeed, in vitro experiments by using murine cell lines found that RSV infection results in more expression of IL-33 mRNA in AMs and DCs, further confirming that these cell types may be an important source of IL-33 during RSV infection. It should be noted that the expression of mRNA for TLR3 and TLR7 was up-regulated in pulmonary AMs during RSV infection. Blockade of TLRs by TLR3 or TLR7 antagonist significantly reduces the levels of IL-33 mRNA in AMs and DCs, suggesting that RSV-induced IL-33 production might be TLRs-dependent manner. Although the expression of TLRs mRNA in pulmonary interstitial macrophages (IMs) was enhanced after RSV infection, stimulation with agonists or inactivated RSV cannot alter the expression of IL-33 mRNA in IMs, suggesting that pulmonary IMs may not be a source of IL-33 during RSV infection. Thus, these results demonstrate that during RSV infection, respiratory macrophages and dendritic cells mediate the production of IL-33 in a TLR-dependent manner.
Collapse
Affiliation(s)
- Feifei Qi
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Dandan Wang
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Jing Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Sheng Zeng
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Lei Xu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Haiyan Hu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Beixing Liu
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
38
|
Bohmwald K, Espinoza JA, Rey-Jurado E, Gómez RS, González PA, Bueno SM, Riedel CA, Kalergis AM. Human Respiratory Syncytial Virus: Infection and Pathology. Semin Respir Crit Care Med 2016; 37:522-37. [PMID: 27486734 PMCID: PMC7171722 DOI: 10.1055/s-0036-1584799] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human respiratory syncytial virus (hRSV) is by far the major cause of acute lower respiratory tract infections (ALRTIs) worldwide in infants and children younger than 2 years. The overwhelming number of hospitalizations due to hRSV-induced ALRTI each year is due, at least in part, to the lack of licensed vaccines against this virus. Thus, hRSV infection is considered a major public health problem and economic burden in most countries. The lung pathology developed in hRSV-infected individuals is characterized by an exacerbated proinflammatory and unbalanced Th2-type immune response. In addition to the adverse effects in airway tissues, hRSV infection can also cause neurologic manifestations in the host, such as seizures and encephalopathy. Although the origins of these extrapulmonary symptoms remain unclear, studies with patients suffering from neurological alterations suggest an involvement of the inflammatory response against hRSV. Furthermore, hRSV has evolved numerous mechanisms to modulate and evade the immune response in the host. Several studies have focused on elucidating the interactions between hRSV virulence factors and the host immune system, to rationally design new vaccines and therapies against this virus. Here, we discuss about the infection, pathology, and immune response triggered by hRSV in the host.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janyra A Espinoza
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Emma Rey-Jurado
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roberto S Gómez
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M Bueno
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Departamento de Ciencias Biológicas y Facultad de Medicina, Millennium Institute on Immunology and Immunotherapy, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Departamento de Genética Molecular y Microbiología, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|