1
|
Beerweiler CC, Salvermoser M, Theodorou J, Böck A, Sattler F, Kulig P, Tosevski V, Schaub B. Farm-dust mediated protection of childhood asthma: Mass cytometry reveals novel cellular regulation. Allergy 2024. [PMID: 39400913 DOI: 10.1111/all.16347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Farm-dust mediated asthma protection in childhood was replicated in numerous epidemiological studies. Central immune mechanisms are not fully understood. This exploratory study aimed to disentangle underlying immunological regulation of farm-dust mediated protection in peripheral blood on a single-cell level. METHODS Single-cell protein expression of in vitro farm-dust stimulated and unstimulated cells from allergic asthmatics and healthy controls were measured using mass cytometry. Analysis of innate and adaptive cellular proportions (linear regression) and T-cell proliferation was performed. Functional marker intensity was investigated using Earth Mover's Distance and the Monte Carlo permutation test. RESULTS Farm-dust stimulation induced cell type-specific regulation: Key-features of farm-dust stimulation comprised opposing regulation of immune-cell frequencies (downregulated innate cell populations (monocytes/DCs (p < .001), NK-cells (p < .05)) and upregulated adaptive populations (B-cells, CD4+ T-cells (both p < .05)), reduced CD4+ CD25- T-cell proliferation, and differential cell type-specific functional marker expression. Following stimulation, functional marker analysis revealed induced activation (CD25) in T-cells and NK-T-cells in both phenotypes even after correction for multiple testing. Cytotoxicity (GZMB) and inflammation (pERK1/2, pp38) related markers were reduced in T-cells exclusively in asthmatic children. Asthma-associated markers (Gata3, RORγ, and HLA-DR) were reduced in T- and innate- cell populations of asthmatics following stimulation. B-cells displayed a phenotypically independent increase of diverse functional markers upon farm-dust stimulation. CONCLUSIONS This study mimicking in vivo environmental exposure identified a novel profile of immune-regulatory markers using mass cytometry demonstrating decreased asthma-associated markers following farm-dust stimulation. These findings may be key for further studies on asthma prevention in childhood.
Collapse
Affiliation(s)
- Claudia Carina Beerweiler
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Center for Lung Research - DZL, LMU Munich, Munich, Germany
| | - Michael Salvermoser
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| | - Johanna Theodorou
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Center for Lung Research - DZL, LMU Munich, Munich, Germany
| | - Andreas Böck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Center for Child and Adolescent Health-DZKJ, LMU, Munich, Germany
| | - Franziska Sattler
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
| | - Paulina Kulig
- Mass Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Vinko Tosevski
- Mass Cytometry Facility, University of Zurich, Zurich, Switzerland
| | - Bianca Schaub
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospital, LMU Munich, Munich, Germany
- Member of German Center for Lung Research - DZL, LMU Munich, Munich, Germany
- Member of German Center for Child and Adolescent Health-DZKJ, LMU, Munich, Germany
| |
Collapse
|
2
|
Tong S, Scott JC, Eyoh E, Werthmann DW, Stone AE, Murrell AE, Sabino-Santos G, Trinh IV, Chandra S, Elliott DH, Smira AR, Velazquez JV, Schieffelin J, Ning B, Hu T, Kolls JK, Landry SJ, Zwezdaryk KJ, Robinson JE, Gunn BM, Rabito FA, Norton EB. Altered COVID-19 immunity in children with asthma by atopic status. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100236. [PMID: 38590754 PMCID: PMC11000189 DOI: 10.1016/j.jacig.2024.100236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 04/10/2024]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection causes a spectrum of clinical outcomes that may be complicated by severe asthma. Antiviral immunity is often compromised in patients with asthma; however, whether this is true for SARS-CoV-2 immunity and children is unknown. Objective We aimed to evaluate SARS-CoV-2 immunity in children with asthma on the basis of infection or vaccination history and compared to respiratory syncytial viral or allergen (eg, cockroach, dust mite)-specific immunity. Methods Fifty-three children from an urban asthma study were evaluated for medical history, lung function, and virus- or allergen-specific immunity using antibody or T-cell assays. Results Polyclonal antibody responses to spike were observed in most children from infection and/or vaccination history. Children with atopic asthma or high allergen-specific IgE, particularly to dust mites, exhibited reduced seroconversion, antibody magnitude, and SARS-CoV-2 virus neutralization after SARS-CoV-2 infection or vaccination. TH1 responses to SARS-CoV-2 and respiratory syncytial virus correlated with antigen-respective IgG. Cockroach-specific T-cell activation as well as IL-17A and IL-21 cytokines negatively correlated with SARS-CoV-2 antibodies and effector functions, distinct from total and dust mite IgE. Allergen-specific IgE and lack of vaccination were associated with recent health care utilization. Reduced lung function (forced expiratory volume in 1 second ≤ 80%) was independently associated with (SARS-CoV-2) peptide-induced cytokines, including IL-31, whereas poor asthma control was associated with cockroach-specific cytokine responses. Conclusion Mechanisms underpinning atopic and nonatopic asthma may complicate the development of memory to SARS-CoV-2 infection or vaccination and lead to a higher risk of repeated infection in these children.
Collapse
Affiliation(s)
- Sherry Tong
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Jordan C. Scott
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Enwono Eyoh
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Derek W. Werthmann
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Addison E. Stone
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Amelie E. Murrell
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Gilberto Sabino-Santos
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Ivy V. Trinh
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - Sruti Chandra
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Debra H. Elliott
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Ashley R. Smira
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Jalene V. Velazquez
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - John Schieffelin
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bo Ning
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, La
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Tony Hu
- Center for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, La
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Jay K. Kolls
- Department of Medicine, Tulane University School of Medicine, New Orleans, La
| | - Samuel J. Landry
- Department of Biochemistry & Molecular Biology, Tulane University School of Medicine, New Orleans, La
| | - Kevin J. Zwezdaryk
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| | - James E. Robinson
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, La
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global Health, Washington State University, Pullman, Wash
| | - Felicia A. Rabito
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, La
| | - Elizabeth B. Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, La
| |
Collapse
|
3
|
Wang H, He Y, Dang D, Zhao Y, Zhao J, Lu W. Gut Microbiota-Derived Tryptophan Metabolites Alleviate Allergic Asthma Inflammation in Ovalbumin-Induced Mice. Foods 2024; 13:1336. [PMID: 38731707 PMCID: PMC11082989 DOI: 10.3390/foods13091336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Asthma is a prevalent respiratory disease. The present study is designed to determine whether gut microbiota-derived tryptophan metabolites alleviate allergic asthma inflammation in ovalbumin (OVA)-induced mice and explore the effect and potential mechanism therein. Asthma model mice were constructed by OVA treatment, and kynurenine (KYN), indole-3-lactic acid (ILA), in-dole-3-carbaldehyde (I3C), and indole acetic acid (IAA) were administered by intraperitoneal injection. The percent survival, weight and asthma symptom score of mice were recorded. The total immunoglobulin E and OVA-specific (s)IgE in the serum and the inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) were detected by the corresponding ELISA kits. The composition of the gut microbiota and tryptophan-targeted metabolism in mouse feces were analyzed using 16S rRNA gene sequencing and targeted metabolomics, respectively. The four tryptophan metabolites improved the percent survival, weight and asthma symptoms of mice, and reduced the inflammatory cells in lung tissues, especially I3C. I3C and IAA significantly (p < 0.05) downregulated the levels of OVA-IgE and inflammatory cytokines. KYN was observed to help restore gut microbiota diversity. Additionally, I3C, KYN, and ILA increased the relative abundance of Anaeroplasma, Akkermansia, and Ruminococcus_1, respectively, which were connected with tryptophan metabolic pathways. IAA also enhanced capability of tryptophan metabolism by the gut microbiota, restoring tryptophan metabolism and increasing production of other tryptophan metabolites. These findings suggest that tryptophan metabolites may modulate asthma through the gut microbiota, offering potential benefits for clinical asthma management.
Collapse
Affiliation(s)
- Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuan He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Danting Dang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yurong Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
4
|
Cen Y, Li F, Li Y, Zhang K, Riaz F, Zhao K, Wei P, Pan F. Dimethyl fumarate alleviates allergic asthma by strengthening the Nrf2 signaling pathway in regulatory T cells. Front Immunol 2024; 15:1375340. [PMID: 38711519 PMCID: PMC11070462 DOI: 10.3389/fimmu.2024.1375340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Allergic asthma is a widely prevalent inflammatory condition affecting people across the globe. T cells and their secretory cytokines are central to the pathogenesis of allergic asthma. Here, we have evaluated the anti-inflammatory impact of dimethyl fumarate (DMF) in allergic asthma with more focus on determining its effect on T cell responses in allergic asthma. By utilizing the ovalbumin (OVA)-induced allergic asthma model, we observed that DMF administration reduced the allergic asthma symptoms and IgE levels in the OVA-induced mice model. Histopathological analysis showed that DMF treatment in an OVA-induced animal model eased the inflammation in the nasal and bronchial tissues, with a particular decrease in the infiltration of immune cells. Additionally, RT-qPCR analysis exhibited that treatment of DMF in an OVA-induced model reduced the expression of inflammatory cytokine (IL4, IL13, and IL17) while augmenting anti-inflammatory IL10 and Foxp3 (forkhead box protein 3). Mechanistically, we found that DMF increased the expression of Foxp3 by exacerbating the expression of nuclear factor E2-related factor 2 (Nrf2), and the in-vitro activation of Foxp3+ Tregs leads to an escalated expression of Nrf2. Notably, CD4-specific Nrf2 deletion intensified the allergic asthma symptoms and reduced the in-vitro iTreg differentiation. Meanwhile, DMF failed to exert protective effects on OVA-induced allergic asthma in CD4-specific Nrf2 knock-out mice. Overall, our study illustrates that DMF enhances Nrf2 signaling in T cells to assist the differentiation of Tregs, which could improve the anti-inflammatory immune response in allergic asthma.
Collapse
Affiliation(s)
- Yanhong Cen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Otolaryngology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Fangfang Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yikui Li
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kaimin Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Farooq Riaz
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kuaile Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Ping Wei
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Otolaryngology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
5
|
Mowahedi M, Aramesh A, Khouzani MS, Khouzani MS, Daryanoush S, Samet M, Samadi M. Association of Interleukin-4 Receptor α Chain I50V Gene Variant (rs1805010) and Asthma in Iranian Population: A Case-control Study. Open Respir Med J 2024; 18:e18743064266613. [PMID: 38660682 PMCID: PMC11037512 DOI: 10.2174/0118743064266613231123103523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/09/2023] [Accepted: 10/03/2023] [Indexed: 04/26/2024] Open
Abstract
Background Asthma is one of the respiratory disorders caused by chronic airway inflammation. IL-4 has been identified as one of the participating interleukins in the severity of asthma. Objective A case-control study was conducted to determine the association of rs1805010, a single nucleotide polymorphism in the interleukin 4 receptor α chain, with asthma and immunoglobulin E and IL-17A serum levels in Iranian populations. Methods ELISA was used to investigate the relationship between three different varieties of SNP I50V and serum IL-17A levels, as well as total IgE levels. Based on GINA criteria, patients were classified into mild, moderate, and severe groups based on the association between SNP I50V, IL-17A, and total IgE. In order to analyze the data, the student-t-test and the one-way ANOVA were used. Results The SNP I50V was associated with asthma in a significant way (p = 0.001). IL-17A and total IgE levels were significantly higher in asthmatic patients than in control participants (p 0.05 and p 0.021, respectively), but neither showed any association with SNP I50V in the asthmatic patients. Conclusion Asthma patients have a higher prevalence of the I allele, reflecting the significance of Th2 cells. Although total IgE and IL-17A levels increased in both disease subgroups, total IgE level augmentation correlates directly with disease severity, while IL-17A level enhancement does not.
Collapse
Affiliation(s)
- Masouma Mowahedi
- Abortion Research Center, Research and Clinical Center for Infertility, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azam Aramesh
- Thalassemia and Hemophilia Research Center, Shahid Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Marjan Sorkhi Khouzani
- Cellular and Molecular Biology - Genetic Center, Falavarjan Azad University, Isfahan, Iran
| | - Saeed Daryanoush
- Thalassemia and Hemophilia Research Center, Shahid Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Samet
- Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Morteza Samadi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
6
|
Liu N, Wang D, Tian J, Wang X, Shi H, Wang C, Jiang Y, Pang M, Fan X, Zhao J, Liu L, Wu H, Guan L, Zheng H, Shi D, Zhang Z. PM 2.5-bound metals and blood metals are associated with pulmonary function and Th17/Treg imbalance: A panel study of asthmatic adults. CHEMOSPHERE 2023; 340:139869. [PMID: 37597628 DOI: 10.1016/j.chemosphere.2023.139869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023]
Abstract
Growing research has demonstrated that exposure to fine particulate matter (PM2.5) was associated with decreased pulmonary function and obvious inflammatory response. However, few pieces of research focus on the effects of PM2.5-bound metals on people with asthma. Here, we assessed whether PM2.5 and PM2.5-bound metals exposure could worsen pulmonary function in asthmatic patients and further elucidate the possible mechanisms. Thirty-four asthmatic patients were recruited to follow up for one year with eight visits in 2019-2020 in Taiyuan City, China. The index of pulmonary function was detected and blood and nasal epithelial lining fluid (ELF) samples were acquired for biomarkers measurement at each follow-up. Linear mixed-effect (LME) models were used to evaluate the relations between PM2.5, PM2.5-bound metals, and blood metals with lung function and biomarkers of Th17/Treg balance. The individual PM2.5 exposure concentration varied from 37 μg/m3 to 194 μg/m3 (mean: 59.63 μg/m3) in the present study. An interquartile range (IQR) increment of PM2.5 total mass was associated with a faster decline in maximal mid-expiratory flow (MMEF) and higher interleukin-23 (IL-23). PM2.5-bound metals [e.g. copper (Cu), nickel (Ni), manganese (Mn), titanium (Ti), and zinc (Zn)] were significantly associated with IL-23 (Cu: 5.1126%, 95% CI: 9.3708, 0.8544; Mn: 14.7212%, 95% CI: 27.926, 1.5164; Ni: 1.0269%, 95% CI: 2.0273, 0.0264; Ti: 16.7536%, 95% CI: 31.6203, 1.8869; Zn: 24.5806%, 95% CI: 46.609, 2.5522). Meanwhile, blood lead (Pb) and Cu were associated with significant declines of 0.382-3.895% in MMEF and maximum ventilatory volume (MVV). Blood Pb was associated with descending transforming growth factor β (TGF-β). In conclusion, exposure to PM2.5-bound metals and blood metals is a risk factor for decreased pulmonary function, especially in small airways. These alterations might be partially attributed to the imbalance of Th17/Treg.
Collapse
Affiliation(s)
- Nannan Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Dan Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Jiayu Tian
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Xin Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Hao Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Caihong Wang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Yi Jiang
- Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Min Pang
- Department of Respiratory and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaozhou Fan
- Shanxi Eco-Environmental Monitoring and Emergency Support Center (Shanxi Academy of Eco-Environmental Sciences), Taiyuan, Shanxi, China
| | - Jing Zhao
- Shanxi Eco-Environmental Monitoring and Emergency Support Center (Shanxi Academy of Eco-Environmental Sciences), Taiyuan, Shanxi, China
| | - Liangpo Liu
- Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of Sanitary Inspection, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongyan Wu
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Department of Epidemiology and Health Statistics, Fudan University, Shanghai, China
| | - Linlin Guan
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Huiqiu Zheng
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Dongxing Shi
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China; Yellow River Basin Ecological Public Health Security Center, Shanxi Medical University, Taiyuan, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China.
| |
Collapse
|
7
|
Allgire E, Ahlbrand RA, Nawreen N, Ajmani A, Hoover C, McAlees JW, Lewkowich IP, Sah R. Altered Fear Behavior in Aeroallergen House Dust Mite Exposed C57Bl/6 Mice: A Model of Th2-skewed Airway Inflammation. Neuroscience 2023; 528:75-88. [PMID: 37516435 PMCID: PMC10530159 DOI: 10.1016/j.neuroscience.2023.07.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/05/2023] [Accepted: 07/19/2023] [Indexed: 07/31/2023]
Abstract
There is a growing interest for studying the impact of chronic inflammation, particularly lung inflammation, on the brain and behavior. This includes asthma, a chronic inflammatory condition, that has been associated with psychiatric conditions such as posttraumatic stress disorder (PTSD). Although asthma is driven by elevated production of Th2 cytokines (IL-4, IL-5 and IL-13), which drive asthma symptomology, recent work demonstrates that concomitant Th1 or Th17 cytokine production can worsen asthma severity. We previously demonstrated a detrimental link between PTSD-relevant fear behavior and allergen-induced lung inflammation associated with a mixed Th2/Th17-inflammatory profile in mice. However, the behavioral effects of Th2-skewed airway inflammation, typical to mild/moderate asthma, are unknown. Therefore, we investigated fear conditioning/extinction in allergen house dust mite (HDM)-exposed C57Bl/6 mice, a model of Th2-skewed allergic asthma. Behaviors relevant to panic, anxiety, and depression were also assessed. Furthermore, we investigated the accumulation of Th2/Th17-cytokine-expressing cells in lung and brain, and the neuronal activation marker, ΔFosB, in fear regulatory brain areas. HDM-exposed mice elicited lower freezing during fear extinction with no effects on acquisition and conditioned fear. No HDM effect on panic, anxiety or depression-relevant behaviors was observed. While HDM evoked a Th2-skewed immune response in lung tissue, no significant alterations in brain Th cell subsets were observed. Significantly reduced ΔFosB+ cells in the basolateral amygdala of HDM mice were observed post extinction. Our data indicate that allergen-driven Th2-skewed responses may induce fear extinction promoting effects, highlighting beneficial interactions of Th2-associated immune mediators with fear regulatory circuits.
Collapse
Affiliation(s)
- E Allgire
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH 45220, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45220, United States
| | - R A Ahlbrand
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH 45220, United States
| | - N Nawreen
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH 45220, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45220, United States
| | - A Ajmani
- Neuroscience Undergraduate Program, University of Cincinnati, Cincinnati, OH 45220, United States
| | - C Hoover
- Neuroscience Undergraduate Program, University of Cincinnati, Cincinnati, OH 45220, United States
| | - J W McAlees
- Division of Immunobiology, Children's Hospital Medical Center, Cincinnati, OH 45220, United States
| | - I P Lewkowich
- Division of Immunobiology, Children's Hospital Medical Center, Cincinnati, OH 45220, United States; Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45220, United States
| | - R Sah
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH 45220, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45220, United States; VA Medical Center, Cincinnati, OH 45220, United States.
| |
Collapse
|
8
|
Li J, Bao T, Cao L, Ma M, Zhang Y, Tian Z. Effects of early postnatal hyperoxia exposure combined with early ovalbumin sensitization on lung inflammation and bacterial flora in a juvenile mouse model of asthma. Front Microbiol 2023; 14:1220042. [PMID: 37485534 PMCID: PMC10358775 DOI: 10.3389/fmicb.2023.1220042] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Objective The aim of this study is to explore the effects of early postnatal hyperoxia exposure combined with early ovalbumin (OVA) sensitization on lung inflammation and bacterial flora in neonatal mice on a juvenile mouse model of asthma. Methods Thirty-two newborn female C57BL/6 J mice were randomly divided into four groups, which including room air+phosphate-buffered saline (PBS) group, hyperoxia+PBS group, room air+OVA group, and hyperoxia+OVA group, according to the hyperoxia exposure and/or OVA induction. Mice were exposed to either 95% O2 or room air for 7 days after birth; after 7 days, they were exposed to air and received an intraperitoneal injection of OVA suspension or PBS solution on postnatal days 21 (P21) and 28 (P28). From P36 to P42, the mice were allowed to inhale of 1% OVA or 0.9% NaCl solution. The mice were observed after the last excitation. HE staining was performed to observe the pathological changes in lung tissues. Wright-Giemsa staining was used to perform bronchoalveolar lavage fluid (BALF) leukocyte sorting. Enzyme-linked immunosorbent assay was used to determined the cytokines levels of interleukin (IL)-2, IL-5, IL-13, IL-17A, and IL-10 and serum IgE levels in BALF. Additionally, 16S rRNA sequencing was used to analyze the characteristics of lung microbiota. Results Mice in the hyperoxia+OVA group showed asthma-like symptoms. HE staining results revealed a significant thickening of the airway wall and airway inflammation. BALF analysis of cellular components showed significant increases in total leukocyte and eosinophil counts and the levels of cytokines related to Th2 (IL-5 and IL-13) and Th17 (IL-17A); 16S rRNA sequencing revealed that the main members of the pulmonary microflora were Actinobacteriota, Proteobacteria, Firmicutes, and Bacteroidota at the phylum level. In addition, the bacteria with a major role were Acinetobacter and Moraxellaceae in the O2 + OVA group. Conclusion The mouse suffering from postnatal hyperoxia exposure and early OVA sensitization, changes in symptoms, pathology, leukocyte and eosinophil counts, and levels of different T-cell cytokines in BALF and lung microbiota, which may provide a basis for the establishment of a juvenile mouse model of asthma.
Collapse
|
9
|
Cheru N, Hafler DA, Sumida TS. Regulatory T cells in peripheral tissue tolerance and diseases. Front Immunol 2023; 14:1154575. [PMID: 37197653 PMCID: PMC10183596 DOI: 10.3389/fimmu.2023.1154575] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/19/2023] Open
Abstract
Maintenance of peripheral tolerance by CD4+Foxp3+ regulatory T cells (Tregs) is essential for regulating autoreactive T cells. The loss of function of Foxp3 leads to autoimmune disease in both animals and humans. An example is the rare, X-linked recessive disorder known as IPEX (Immune Dysregulation, Polyendocrinopathy, Enteropathy X-linked) syndrome. In more common human autoimmune diseases, defects in Treg function are accompanied with aberrant effector cytokines such as IFNγ. It has recently become appreciated that Tregs plays an important role in not only maintaining immune homeostasis but also in establishing the tissue microenvironment and homeostasis of non-lymphoid tissues. Tissue resident Tregs show profiles that are unique to their local environments which are composed of both immune and non-immune cells. Core tissue-residence gene signatures are shared across different tissue Tregs and are crucial to homeostatic regulation and maintaining the tissue Treg pool in a steady state. Through interaction with immunocytes and non-immunocytes, tissue Tregs exert a suppressive function via conventional ways involving contact dependent and independent processes. In addition, tissue resident Tregs communicate with other tissue resident cells which allows Tregs to adopt to their local microenvironment. These bidirectional interactions are dependent on the specific tissue environment. Here, we summarize the recent advancements of tissue Treg studies in both human and mice, and discuss the molecular mechanisms that maintain tissue homeostasis and prevent pathogenesis.
Collapse
Affiliation(s)
- Nardos Cheru
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - David A. Hafler
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Tomokazu S. Sumida
- Department of Neurology, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
10
|
Th17/Treg Imbalance: Implications in Lung Inflammatory Diseases. Int J Mol Sci 2023; 24:ijms24054865. [PMID: 36902294 PMCID: PMC10003150 DOI: 10.3390/ijms24054865] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Regulatory T cells (Tregs) and T helper 17 cells (Th17) are two CD4+ T cell subsets with antagonist effects. Th17 cells promote inflammation, whereas Tregs are crucial in maintaining immune homeostasis. Recent studies suggest that Th17 cells and Treg cells are the foremost players in several inflammatory diseases. In this review, we explore the present knowledge on the role of Th17 cells and Treg cells, focusing on lung inflammatory diseases, such as chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), sarcoidosis, asthma, and pulmonary infectious diseases.
Collapse
|
11
|
Eldosoky MA, Hammad R, Rushdi A, Ibrahim HF, Tawfeik AM, Mora A, Fahmy SF, El-Ashmawy H, Ali E, Hamed DH, Mohammed AR, Mashaal A, Mohsen H. MicroRNA-146a-5p and microRNA-210-3p Correlate with T Regulatory Cells Frequency and Predict Asthma Severity in Egyptian Pediatric Population. J Asthma Allergy 2023; 16:107-121. [PMID: 36714048 PMCID: PMC9880026 DOI: 10.2147/jaa.s398494] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Abstract
Background Severe bronchial asthma (BA) affects 5-10% of children, which imposes socioeconomic burden. Therefore, it is crucial to identify biomarkers for risk stratification in children with BA. T regulatory cells (Tregs) play a balancing role in allergic response regulation. We aimed to investigate the relationship between Treg, miR-210-3p, and miR-146a-5p in relation to asthma phenotypes in search of novel biomarkers of disease severity. Methods This study included 50 children with BA classified into Group 1 (n = 25) children with mild to moderate asthma and Group 2 (n = 25) children with severe asthma. In addition to 26 control subjects. Flow cytometry was used to detect Tregs. Plasma miR-210-3p and miR-146a levels were determined using quantitative real-time PCR. Patients' FEV1 (Forced Expiratory Volume in the first second) was measured. Results miR-210-3p level correlated negatively with Treg frequency (r = -0.828, P < 0.001) and FEV1 (r = -0.621, P < 0.001). The level of miR-146a-5p positively correlated positively with Treg% (r = 0.303, P = 0.032). ROC curve analysis revealed that miR-210-3p was the most sensitive biomarker of severity, with the area under curve (AUC) = 0.923, 96% sensitivity, and 60% specificity. According to multivariate analysis, miR-210-3p is an independent risk factor for BA severity [OR =3.119, P = 0.030], while miR-146a-5p is a protective factor [OR =0.811, P = 0.049]. Conclusion Treg frequency is linked to FEV1, miR-146a-5p and miR-210-3p in childhood BA. Upregulation of miR-210-3p is a sensitive biomarker and an independent risk factor for BA severity in Egyptian children.
Collapse
Affiliation(s)
- Mona A Eldosoky
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Reham Hammad
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Areej Rushdi
- Microbiology and Immunology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Hanan F Ibrahim
- Microbiology and Immunology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Amany M Tawfeik
- Microbiology and Immunology Department, Faculty of Medicine, Badr University in Cairo (BUC), Cairo, Egypt
| | - Ahmed Mora
- Chemistry Department, Faculty of Science (for Boys), Al-Azhar University, Cairo, Egypt
| | - Sarah F Fahmy
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hossam El-Ashmawy
- Clinical Pathology Department, Faculty of Medicine (for Boys), Al-Azhar University, Assuit, Egypt
| | - Elham Ali
- Molecular Biology, Zoology and Entomology Department, Faculty of Science (For Girls), Al-Azhar University, Cairo, Egypt
| | - Dina H Hamed
- Pediatric Department, Pediatric Allergy and Pulmonology Unit, Children’s Hospital, Cairo University, Cairo, Egypt,Correspondence: Dina H Hamed, Email
| | - Amena Rezk Mohammed
- Biochemistry Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Alya Mashaal
- Immunology, Zoology and Entomology Department, Faculty of Science (For Girls), Al-Azhar University, Cairo, Egypt
| | - Hanan Mohsen
- Pediatric Department, Pediatric Allergy and Pulmonology Unit, Children’s Hospital, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
El-Baz LM, Elaidy SM, Hafez HS, Shoukry NM. Vismodegib, a sonic hedgehog signalling blockade, ameliorates ovalbumin and ovalbumin/lipopolysaccharide-induced airway inflammation and asthma phenotypical models. Life Sci 2022; 310:121119. [DOI: 10.1016/j.lfs.2022.121119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
|
13
|
Liu H, Nie H, Lai W, Shi Y, Liu X, Li K, Tian L, Xi Z, Lin B. Different exposure modes of PM 2.5 induces bronchial asthma and fibrosis in male rats through macrophage activation and immune imbalance induced by TIPE2 methylation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 247:114200. [PMID: 36274320 DOI: 10.1016/j.ecoenv.2022.114200] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/02/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Exposure to PM2.5 can aggravate the occurrence and development of bronchial asthma and fibrosis. Here, we investigated the differences in bronchial injury caused by different exposure modes of PM2.5 (high concentration intermittent exposure and low concentration continuous exposure), and the mechanism of macrophage activation and respiratory immune imbalance induced by PM2.5, leading to bronchial asthma and airway fibrosis using animal and cell models. A "PM2.5 real-time online concentrated animal whole-body exposure system" was used to conduct PM2.5 respiratory exposure of Wistar rats for 12 weeks, which can enhance oxidative stress in rat bronchus, activate epithelial cells and macrophages, release chemokines, recruit inflammatory cells, release inflammatory factors and extracellular matrix, promote bronchial mucus hypersecretion, inhibit the expression of epithelial cytoskeletal proteins, destroy airway barrier, and induce asthma. Furthermore, PM2.5 induced M2 polarization in lung bronchial macrophages through JAK/STAT and PI3K/Akt signaling pathways, and compared with low concentration continuous exposure, high concentration intermittent exposure of PM2.5 could regulate significantly higher expression of TIPE2 protein through promoter methylation of TIPE2 DNA, thereby activating PI3K/Akt signaling pathway and more effectively inducing M2 polarization of macrophages. Additionally, activated macrophages release IL-23, and activated epithelial cells and macrophages released TGF-β1, which promoted the differentiation of Th17 cells, triggered the Th17 dominant immune response, and activated the TGF-β1/Smad2 signaling pathway, finally causing bronchial fibrosis. Moreover, when the total amount of PM2.5 exposure was equal, high concentration-intermittent exposure was more serious than low concentration-continuous exposure. In vitro experiments, the co-culture models of PM2.5 with BEAS-2B, WL-38 and rat primary alveolar macrophages further confirmed that PM2.5 could induce the macrophage activation through oxidative stress and TIPE2 DNA methylation, and activate the TGF-β1/Smad2 signaling pathway, leading to the occurrence of bronchial fibrosis.
Collapse
Affiliation(s)
- Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Huipeng Nie
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Wenqing Lai
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Yue Shi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Xuan Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Lei Tian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China.
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment & Food Safety, Tianjin 300050, China.
| |
Collapse
|
14
|
Investigational Treatments in Phase I and II Clinical Trials: A Systematic Review in Asthma. Biomedicines 2022; 10:biomedicines10092330. [PMID: 36140430 PMCID: PMC9496184 DOI: 10.3390/biomedicines10092330] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Inhaled corticosteroids (ICS) remain the mainstay of asthma treatment, along with bronchodilators serving as control agents in combination with ICS or reliever therapy. Although current pharmacological treatments improve symptom control, health status, and the frequency and severity of exacerbations, they do not really change the natural course of asthma, including disease remission. Considering the highly heterogeneous nature of asthma, there is a strong need for innovative medications that selectively target components of the inflammatory cascade. The aim of this review was to systematically assess current investigational agents in Phase I and II randomised controlled trials (RCTs) over the last five years. Sixteen classes of novel therapeutic options were identified from 19 RCTs. Drugs belonging to different classes, such as the anti-interleukin (IL)-4Rα inhibitors, anti-IL-5 monoclonal antibodies (mAbs), anti-IL-17A mAbs, anti-thymic stromal lymphopoietin (TSLP) mAbs, epithelial sodium channel (ENaC) inhibitors, bifunctional M3 receptor muscarinic antagonists/β2-adrenoceptor agonists (MABAs), and anti-Fel d 1 mAbs, were found to be effective in the treatment of asthma, with lung function being the main assessed outcome across the RCTs. Several novel investigational molecules, particularly biologics, seem promising as future disease-modifying agents; nevertheless, further larger studies are required to confirm positive results from Phase I and II RCTs.
Collapse
|
15
|
Dong H, Hao Y, Li W, Yang W, Gao P. IL-36 Cytokines: Their Roles in Asthma and Potential as a Therapeutic. Front Immunol 2022; 13:921275. [PMID: 35903102 PMCID: PMC9314646 DOI: 10.3389/fimmu.2022.921275] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin (IL)-36 cytokines are members of the IL-1 superfamily, which consists of three agonists (IL-36α, IL-36β and IL-36γ) and an IL-36 receptor antagonist (IL-36Ra). IL-36 cytokines are crucial for immune and inflammatory responses. Abnormal levels of IL-36 cytokine expression are involved in the pathogenesis of inflammation, autoimmunity, allergy and cancer. The present study provides a summary of recent reports on IL-36 cytokines that participate in the pathogenesis of inflammatory diseases, and the potential mechanisms underlying their roles in asthma. Abnormal levels of IL-36 cytokines are associated with the pathogenesis of different types of asthma through the regulation of the functions of different types of cells. Considering the important role of IL-36 cytokines in asthma, these may become a potential therapeutic target for asthma treatment. However, existing evidence is insufficient to fully elucidate the specific mechanism underlying the action of IL-36 cytokines during the pathological process of asthma. The possible mechanisms and functions of IL-36 cytokines in different types of asthma require further studies.
Collapse
Affiliation(s)
- Hongna Dong
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yuqiu Hao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Peng Gao,
| |
Collapse
|
16
|
Ritzmann F, Lunding LP, Bals R, Wegmann M, Beisswenger C. IL-17 Cytokines and Chronic Lung Diseases. Cells 2022; 11:2132. [PMID: 35883573 PMCID: PMC9318387 DOI: 10.3390/cells11142132] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
IL-17 cytokines are expressed by numerous cells (e.g., gamma delta (γδ) T, innate lymphoid (ILC), Th17, epithelial cells). They contribute to the elimination of bacteria through the induction of cytokines and chemokines which mediate the recruitment of inflammatory cells to the site of infection. However, IL-17-driven inflammation also likely promotes the progression of chronic lung diseases, such as chronic obstructive pulmonary disease (COPD), lung cancer, cystic fibrosis, and asthma. In this review, we highlight the role of IL-17 cytokines in chronic lung diseases.
Collapse
Affiliation(s)
- Felix Ritzmann
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Lars Peter Lunding
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
- Helmholtz Institute for Pharmaceutical Research, 66123 Saarbrücken, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area Asthma and Allergy, Research Center Borstel—Leibniz Lung Center, 23845 Borstel, Germany; (L.P.L.); (M.W.)
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V—Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, 66421 Homburg, Germany; (F.R.); (R.B.)
| |
Collapse
|
17
|
Rahmawati SF, Vos R, Bos IST, Kerstjens HAM, Kistemaker LEM, Gosens R. Function-specific IL-17A and dexamethasone interactions in primary human airway epithelial cells. Sci Rep 2022; 12:11110. [PMID: 35773318 PMCID: PMC9247091 DOI: 10.1038/s41598-022-15393-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/23/2022] [Indexed: 11/09/2022] Open
Abstract
Asthmatics have elevated levels of IL-17A compared to healthy controls. IL-17A is likely to contribute to reduced corticosteroid sensitivity of human airway epithelium. Here, we aimed to investigate the mechanistic underpinnings of this reduced sensitivity in more detail. Differentiated primary human airway epithelial cells (hAECs) were exposed to IL-17A in the absence or presence of dexamethasone. Cells were then collected for RNA sequencing analysis or used for barrier function experiments. Mucus was collected for volume measurement and basal medium for cytokine analysis. 2861 genes were differentially expressed by IL-17A (Padj < 0.05), of which the majority was not sensitive to dexamethasone (< 50% inhibition). IL-17A did inhibit canonical corticosteroid genes, such as HSD11B2 and FKBP5 (p < 0.05). Inflammatory and goblet cell metaplasia markers, cytokine secretion and mucus production were all induced by IL-17A, and these effects were not prevented by dexamethasone. Dexamethasone did reverse IL-17A-stimulated epithelial barrier disruption, and this was associated with gene expression changes related to cilia function and development. We conclude that IL-17A induces function-specific corticosteroid-insensitivity. Whereas inflammatory response genes and mucus production in primary hAECs in response to IL-17A were corticosteroid-insensitive, corticosteroids were able to reverse IL-17A-induced epithelial barrier disruption.
Collapse
Affiliation(s)
- Siti Farah Rahmawati
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Department of Pharmacology and Clinical Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Rémon Vos
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - I Sophie T Bos
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Huib A M Kerstjens
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Aquilo Contract Research, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen (UMCG), Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands.
| |
Collapse
|
18
|
Dai H, Zheng R, Wang L, Wan J, Tong Y, Zhao W, Zhang W. ICS/LABA Combined With Subcutaneous Immunotherapy Modulates the Th17/Treg Imbalance in Asthmatic Children. Front Immunol 2022; 13:779072. [PMID: 35355985 PMCID: PMC8960042 DOI: 10.3389/fimmu.2022.779072] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Rationale The imbalance of T helper (Th17) cell and regulatory T (Treg) cell are involved in allergic asthma pathogenesis. We hypothesized that ICS/LABA could modulate the Th17/Treg imbalance and that subcutaneous immunotherapy (SCIT) could coordinate with ICS/LABA to rebalance the dysfunction of Th17/Treg. Methods Thirty house dust mites (HDM) allergic asthmatic children and fifteen healthy control subjects were enrolled in this study. Fifteen asthmatic children were treated by ICS/LABA powder inhalation, while the other fifteen asthmatic children were treated by ICS/LABA powder inhalation combined with HDM-SCIT. Asthmatic subjects were followed up for 6 months, but 2 asthmatics treated with ICS/LABA were lost to follow-up. Flow cytometry was used to determine the proportions of Th17 and Treg in CD4+ T cells from peripheral blood mononuclear cells (PBMCs). Serum levels of IL-17A and IL-10 were assessed by ELISA. Result ICS/LABA treatment significantly reduced the percentage of Th17 cells (1.252 ± 0.134% vs. 2.567 ± 0.386%), serum IL-17A (49.42 ± 2.643 pg/ml vs. 66.75 ± 3.442 pg/ml) and Th17/Treg ratio (0.194 ± 0.025 vs. 0.439 ± 0.072) compared to baseline (P<0.01). The ICS/LABA+HDM-SCIT treatment group showed similar reduction in the percentage of Th17 cells (1.11 ± 0.114% vs. 2.654 ± 0.276%), serum IL-17A (49.23 ± 2.131 pg/ml vs. 66.41 ± 2.616 pg/ml) and the Th17/Treg ratio (0.133 ± 0.015 vs. 0.4193 ± 0.050) (P<0.01). ICS/LABA+HDM-SCIT treatment group demonstrated elevated Treg percentages (8.483 ± 0.408% vs. 6.549 ± 0.299%) and serum IL-10 levels (127.4 ± 4.423 pg/ml vs. 93.15 ± 4.046 pg/ml), resulting in a lower Th17/Treg ratio than the ICS/LABA group. Conclusion ICS/LABA treatment regulates Th17/Treg imbalance mainly by mitigating Th17-induced inflammation in asthma patients. The addition of SCIT further enhanced such effect by upregulating Treg cells.
Collapse
Affiliation(s)
- Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongying Zheng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Like Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinyi Wan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Tong
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Zhao
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
19
|
Rahmawati SF, te Velde M, Kerstjens HAM, Dömling ASS, Groves MR, Gosens R. Pharmacological Rationale for Targeting IL-17 in Asthma. FRONTIERS IN ALLERGY 2021; 2:694514. [PMID: 35387016 PMCID: PMC8974835 DOI: 10.3389/falgy.2021.694514] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/07/2021] [Indexed: 01/09/2023] Open
Abstract
Asthma is a respiratory disease that currently affects around 300 million people worldwide and is defined by coughing, shortness of breath, wheezing, mucus overproduction, chest tightness, and expiratory airflow limitation. Increased levels of interleukin 17 (IL-17) have been observed in sputum, nasal and bronchial biopsies, and serum of patients with asthma compared to healthy controls. Patients with higher levels of IL-17 have a more severe asthma phenotype. Biologics are available for T helper 2 (Th2)-high asthmatics, but the Th17-high subpopulation has a relatively low response to these treatments, rendering it a rather severe asthma phenotype to treat. Several experimental models suggest that targeting the IL-17 pathway may be beneficial in asthma. Moreover, as increased activation of the Th17/IL-17 axis is correlated with reduced inhaled corticosteroids (ICS) sensitivity, targeting the IL-17 pathway might reverse ICS unresponsiveness. In this review, we present and discuss the current knowledge on the role of IL-17 in asthma and its interaction with the Th2 pathway, focusing on the rationale for therapeutic targeting of the IL-17 pathway.
Collapse
Affiliation(s)
- Siti Farah Rahmawati
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Department of Pharmacology and Clinical Pharmacy, Institut Teknologi Bandung, Bandung, Indonesia
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Maurice te Velde
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
| | - Huib A. M. Kerstjens
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- Department of Pulmonary Medicine, University of Groningen and University Medical Center Groningen (UMCG), Groningen, Netherlands
| | | | | | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Centre Groningen (UMCG), Groningen, Netherlands
- *Correspondence: Reinoud Gosens
| |
Collapse
|
20
|
Affiliation(s)
- Ricardo Wesley Alberca
- Laboratorio de Dermatologia e Imunodeficiencias (LIM-56), Departamento de Dermatologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
21
|
Li G, Zhang L, Han N, Zhang K, Li H. Increased Th17 and Th22 Cell Percentages Predict Acute Lung Injury in Patients with Sepsis. Lung 2020; 198:687-693. [PMID: 32462370 DOI: 10.1007/s00408-020-00362-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/08/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE This study was conducted to investigate the percentages of Th22 and Th17 cells in the peripheral blood of septic patients with and without acute lung injury (ALI) and their clinical significance. METHODS A total of 479 patients were divided into non-ALI and ALI groups. The percentages of Th22 and Th17 cells and the levels of interleukin 22 (IL-22), 6 (IL-6), and 17 (IL-17) were determined. Receiver operating characteristic curve analysis was performed to assess the diagnostic value of Th22 and Th17 cells to predict sepsis-induced ALI. RESULTS The lung injury prediction score (LIPS), IL-6, IL-17, and IL-22 levels and the percentages of Th17 and Th22 cells were significantly higher in the ALI group (P < 0.05). They were significant factors affecting sepsis-induced ALI (P < 0.05). Multivariate logistic regression analysis showed that the LIPS (OR = 1.130), IL-17 (OR = 1.982), IL-22 (OR = 2.612) and the percentages of Th17 (OR = 2.211) and Th22 (OR = 3.230) cells were independent risk factors for ALI. The area under the curve of Th22 cells was 0.844 to predict ALI with a cutoff value of 6.81%. The sensitivity and specificity for early diagnosis of sepsis-induced ALI by the Th22 cell percentage were 78.72% and 89.13%, respectively. CONCLUSIONS Th22 and Th17 cells in peripheral blood are significantly increased in septic patients with ALI and they may be used as biomarkers for early diagnosis of sepsis-induced ALI.
Collapse
Affiliation(s)
- Gang Li
- Department of Emergency Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, China.
| | - Liangtian Zhang
- Department of Critical Care Medicine, Chunan First People's Hospital, Zhejiang Provincial People's Hospital Chunan Branch, Chunan, China
| | - Nannan Han
- Department of Emergency Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, China
| | - Ke Zhang
- Department of Emergency Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, China
| | - Hengjie Li
- Department of Emergency Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310014, China
| |
Collapse
|