1
|
Latayan J, Akkenapally SV, Madala SK. Emerging Concepts in Cytokine Regulation of Airway Remodeling in Asthma. Immunol Rev 2025; 330:e70020. [PMID: 40116139 PMCID: PMC11926778 DOI: 10.1111/imr.70020] [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: 08/24/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Asthma, a chronic respiratory condition that has seen a dramatic rise in prevalence over the past few decades, now affects more than 300 million people globally and imposes a significant burden on healthcare systems. The key pathological features of asthma include inflammation, airway hyperresponsiveness, mucus cell metaplasia, smooth muscle hypertrophy, and subepithelial fibrosis. Cytokines released by lung epithelial cells, stromal cells, and immune cells during asthma are critical to pathological tissue remodeling in asthma. Over the past few decades, researchers have made great strides in understanding key cells involved in asthma and the cytokines that they produce. Epithelial cells as well as many adaptive and innate immune cells are activated by environmental signals to produce cytokines, namely, type 2 cytokines (IL-4, IL-5, IL-13), IFN-γ, IL-17, TGF-β, and multiple IL-6 family members. However, the precise mechanisms through which these cytokines contribute to airway remodeling remain elusive. Additionally, multiple cell types can produce the same cytokines, making it challenging to decipher how specific cell types and cytokines uniquely contribute to asthma pathogenesis. This review highlights recent advances and provides a comprehensive overview of the key cells involved in the production of cytokines and how these cytokines modulate airway remodeling in asthma.
Collapse
Affiliation(s)
- Jana Latayan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
- Immunology Graduate ProgramUniversity of CincinnatiCincinnatiOhioUSA
| | - Santhoshi V. Akkenapally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| | - Satish K. Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
2
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
3
|
Kuo CY, Huang CY, Chen HM, Chen LC, Kuo ML. Antagonism of CD28 blocks allergic responses in the ovalbumin-induced asthmatic model mice. Int Immunopharmacol 2025; 148:114071. [PMID: 39842142 DOI: 10.1016/j.intimp.2025.114071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
Allergen-reactive T helper (Th) 2 cells play a pivotal role in initiating asthma pathogenesis. The absence or interruption of CD28 signaling causes significant consequences for T-cell activation, leading to reduced cell proliferation and interleukin (IL)-2 production. A novel compound, Cyn-1324, exhibits a higher binding affinity to CD28 than CD80. Thus, targeting the CD28-CD80 interaction emerges as a promising therapeutic approach for allergic asthma. However, the impact of CD28 antagonists on allergen-induced asthma remains unreported. In this study, we explored the effects of intranasally administered Cyn-1324 on airway inflammation in the ovalbumin (OVA)-induced murine allergic model. The results revealed a significant reduction in airway hyper-responsiveness (AHR), eosinophil recruitment, and cell infiltration in lung tissues, as well as decreased OVA-specific IgE in serum and Th2 cytokine levels in OVA-stimulated lymphocyte cultures. Additionally, we demonstrated the immunosuppressive effects of Cyn-1324 in vitro, including decreased T-cell proliferation and IL-2 secretion, together with increased p27kip1 expression via inhibiting the PI3K signaling pathway. Notably, Cyn-1324 not only inhibited the NF-κB pathway, but also appeared to suppress p38 activation, which is downstream of CD3 signaling, and reduced calcium-induced NFAT protein expression. These findings suggest that Cyn-1324 alleviates allergic responses by inhibiting the CD28-CD80 interaction and holds promise as an immunosuppressive agent for allergic patients.
Collapse
Affiliation(s)
- Chieh-Ying Kuo
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
| | - Chih-Yu Huang
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan
| | - Hueih-Min Chen
- Taiwan Semiconductor Research Institute National Applied Research Laboratories Hsinchu Taiwan
| | - Li-Chen Chen
- Division of Allergy Asthma and Rheumatology Department of Pediatrics Chang Gung Memorial Hospital at Linkou Taoyuan Taiwan; Department of Pediatrics New Taipei Municipal TuCheng Hospital New Taipei Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan; Division of Allergy Asthma and Rheumatology Department of Pediatrics Chang Gung Memorial Hospital at Linkou Taoyuan Taiwan; Department of Pediatrics New Taipei Municipal TuCheng Hospital New Taipei Taiwan; Research Center for Chinese Herbal Medicine College of Human Ecology Chang Gung University of Science and Technology Taoyuan Taiwan.
| |
Collapse
|
4
|
Yang Z, Krammer S, Mitländer H, Grund JC, Zirlik S, Wirtz S, Rauh M, Shermeh AS, Finotto S. NFATc1 in CD4 + T cells and CD11c + dendritic cells drives T H2-mediated eosinophilic inflammation in allergic asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100355. [PMID: 39629220 PMCID: PMC11613943 DOI: 10.1016/j.jacig.2024.100355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 12/07/2024]
Abstract
Background Asthma, a chronic lung disease, is a significant public health problem worldwide. It is marked by increased TH2 response resulting in eosinophil accumulation. The pathophysiology of asthma involves various cell types, including epithelial cells, dendritic cells (DCs), innate lymphoid cells, B cells, and effector cells. Nuclear factor of activated T cells, cytoplasmic 1 (NFATc1), a critical transcription factor for immune regulation, is known for its role in T cells and, more recently, in myeloid cells. However, the specific contributions of NFATc1 in T cells and DCs in the context of asthma are not well understood. Objective We explored NFATc1's role in T cells and DCs in modulating TH2 immune responses within the pathophysiology of allergic asthma. Methods We induced asthma in mice lacking Nfatc1 in CD4+ T cells or CD11c+ DCs using house dust mite, thereby enabling investigation into NFATc1's role in both cell types in experimental allergic asthma. Additionally, we examined NFATc1 expression in these cell types and its correlation with blood eosinophil levels in an adult asthma cohort. Results In a house dust mite-induced asthma model, we found that Nfatc1 deficiency either in CD4+ T cells or CD11c+ DCs resulted in reduced TH2-driven eosinophilic inflammation, IgE levels, and mast cell presence in the lung of asthmatic mice. Nfatc1's absence in CD4+ T cells directly hampered TH2 cell polarization and functionality, whereas in CD11c+ DCs, it affected DC differentiation and maturation, thereby weakening T-cell priming, proliferation, and subsequent TH2 differentiation. Correspondingly, translational research indicated significant correlations between CD4+NFATc1+ and CD11c+NFATc1+ cell populations and eosinophil levels in asthmatic patients, but not in healthy controls. Conclusion NFATc1 in T cells and DCs modulates TH2-mediated eosinophilic inflammation in allergic asthma, thus offering insight into asthma pathogenesis and identifying NFATc1 as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hannah Mitländer
- Department of Molecular Pneumology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Janina C. Grund
- Department of Molecular Pneumology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- Department of Internal Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Atefeh Sadeghi Shermeh
- Department of Immune Modulation, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Bavarian Cancer Research Center, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
5
|
García-Moguel I, Martínez-Mesa Á, Andújar-Espinosa R, Díaz-Campos R, Velasco-Garrido JL, Sanchez-Trincado JL, Luzon E, Nuevo J, Alconada C, Gutiérrez MÁ, Niza G, Padilla-Galo A. The impact of blood eosinophil count and FeNO on benralizumab effectiveness in clinical practice: An ORBE II subanalysis. Respir Med 2025; 237:107940. [PMID: 39814271 DOI: 10.1016/j.rmed.2025.107940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND The ORBE II study showed the real-world effectiveness of benralizumab in severe eosinophilic asthma (SEA). This subgroup analysis aimed to characterize patients and outcomes based on baseline blood eosinophil count (BEC) and/or fractional exhaled nitric oxide (FeNO) levels. METHODS In this analysis of the ORBE II retrospective study, SEA patients receiving benralizumab were categorized into subgroups based on individual or combined BEC/FeNO levels, according to the following thresholds: high BEC (hiBEC): ≥300 cells/μL; low BEC (loBEC): <300 cells/μL; high FeNO (hiFeNO): ≥50 ppb; low FeNO (loFeNO): <50 ppb. Baseline and up to 1 year of follow-up data are described. RESULTS Most patients with available data were classified as hiBEC (72.6 %) and 38.3 % as hiFeNO. The distribution according to combined baseline BEC and FeNO levels revealed a heterogeneous patient population. Although common SEA features were shared among subgroups, some distinct characteristics were observed, including elevated allergic asthma prevalence in hiBEC/loFeNO patients, high obesity prevalence and fewer non-smokers in loBEC/loFeNO patients, remarkable severe exacerbation rates in loBEC/hiFeNO patients [5.5 SD (6.0)], and more severe symptoms in the hiBEC/loBEC subgroup. All subgroups showed benefits following benralizumab treatment, with super-responder rates ranging from 68.2 % to 83.3 % and clinical remission rates reaching 70.0 %. Particularly good responses were noted in hiBEC/hiFeNO patients. CONCLUSIONS This study shows the variability of T2 biomarkers in ORBE II SEA patients, emphasizing the prevalence of high BEC values. While benralizumab benefits were important regardless of BEC, high BEC predicted good outcomes and FeNO had less influence on treatment effectiveness.
Collapse
Affiliation(s)
- Ismael García-Moguel
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, Spain
| | | | | | - Rocío Díaz-Campos
- Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Madrid, Spain
| | | | | | - Elisa Luzon
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | - Javier Nuevo
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | - Carlos Alconada
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | | | - Gabriel Niza
- Departamento Médico, AstraZeneca Farmacéutica S.A., Madrid, Spain
| | | |
Collapse
|
6
|
Szefler S, Corren J, Silverberg JI, Okragly A, Sun Z, Natalie CR, Zitnik R, Siu K, Blauvelt A. Lebrikizumab decreases type 2 inflammatory biomarker levels in patients with asthma: data from randomized phase 3 trials (LAVOLTA I and II). Immunotherapy 2025; 16:1211-1216. [PMID: 39781908 PMCID: PMC11759530 DOI: 10.1080/1750743x.2024.2439777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
AIM Lebrikizumab is an interleukin (IL)-13 inhibitor that specifically blocks IL-13 signaling. Here, we report the effects of lebrikizumab on asthma serum biomarkers in 2 phase 3 clinical studies. METHODS LAVOLTA I and LAVOLTA II are replicate, double-blind, placebo-controlled trials with 52-week placebo-controlled treatment periods that evaluated lebrikizumab 37.5- and 125-mg doses every 4 weeks. Patients were aged 18-75 years with uncontrolled asthma on stable background therapy. Biomarkers assessed included immunoglobulin E (IgE), periostin, CC motif chemokine ligand (CCL)13, and CCL17. Statistical significance was assessed for difference in fold-change for lebrikizumab versus placebo using a mixed-effects model for repeated measures. RESULTS At early time points in LAVOLTA I and II (weeks 1 and 4), decreases in periostin and CCL13 were statistically significant versus placebo (all p < 0.001) for both lebrikizumab doses. For the 125-mg lebrikizumab dose at week 1 in LAVOLTA I, the decrease in CCL17 was statistically significant (p = 0.001). Reductions in periostin, CCL13, and CCL17 were maintained throughout the trial duration. Significant decreases versus placebo (p ≤ 0.001) were seen in IgE by weeks 12 and 24 in LAVOLTA I and LAVOLTA II, respectively. CONCLUSION Significant reductions in relevant inflammatory biomarkers were observed in the LAVOLTA I and LAVOLTA II studies.
Collapse
Affiliation(s)
- Stanley Szefler
- The Breathing Institute and Pediatric Pulmonary and Sleep Medicine Section, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO, USA
| | - Jonathan Corren
- Division of Allergy and Clinical Immunology, Department of Medicine and Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jonathan I. Silverberg
- Department of Dermatology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | | | - Zhe Sun
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | | | | | | |
Collapse
|
7
|
Ramonell RP, Gauthier MC, Ray A, Wenzel SE. Biologic Medications for Severe Asthma: Implications for Understanding Pathogenic Heterogeneity and Endotypes. Annu Rev Med 2025; 76:339-355. [PMID: 39586024 DOI: 10.1146/annurev-med-070323-103158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Asthma is a chronic inflammatory disease of the airways long known for phenotypic heterogeneity. Phenotyping studies in asthma have led to a better characterization of disease pathogenesis, yet further work is needed to pair available treatments with disease endotypes. In this review, the biology of targeted pathways is discussed along with the efficacy of biologic therapies targeting those pathways. Results of asthma clinical trials are included, as well as results of trials in related diseases. This review then analyzes how biologics help to inform the complex immunobiology of asthma and further guide their use while identifying areas for future research.
Collapse
Affiliation(s)
- Richard P Ramonell
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Marc C Gauthier
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
8
|
Zhang H, He K, Li Z, Tsoi LC, Zhou X. FABIO: TWAS fine-mapping to prioritize causal genes for binary traits. PLoS Genet 2024; 20:e1011503. [PMID: 39621803 DOI: 10.1371/journal.pgen.1011503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 12/16/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Transcriptome-wide association studies (TWAS) have emerged as a powerful tool for identifying gene-trait associations by integrating gene expression mapping studies with genome-wide association studies (GWAS). While most existing TWAS approaches focus on marginal analyses through examining one gene at a time, recent developments in TWAS fine-mapping methods enable the joint modeling of multiple genes to refine the identification of potentially causal ones. However, these fine-mapping methods have primarily focused on modeling quantitative traits and examining local genomic regions, leading to potentially suboptimal performance. Here, we present FABIO, a TWAS fine-mapping method specifically designed for binary traits that is capable of modeling all genes jointly on an entire chromosome. FABIO employs a probit model to directly link the genetically regulated expression (GReX) of genes to binary outcomes while taking into account the GReX correlation among all genes residing on a chromosome. As a result, FABIO effectively controls false discoveries while offering substantial power gains over existing TWAS fine-mapping approaches. We performed extensive simulations to evaluate the performance of FABIO and applied it for in-depth analyses of six binary disease traits in the UK Biobank. In the real datasets, FABIO significantly reduced the size of the causal gene sets by 27.9%-36.9% over existing approaches across traits. Leveraging its improved power, FABIO successfully prioritized multiple potentially causal genes associated with the diseases, including GATA3 for asthma, ABCG2 for gout, and SH2B3 for hypertension. Overall, FABIO represents an effective tool for TWAS fine-mapping of disease traits.
Collapse
Affiliation(s)
- Haihan Zhang
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kevin He
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Zheng Li
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Lam C Tsoi
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
9
|
Ma N, Wang H, Li Q, Chang M, Zhu J, Nan S, Zhang Q, Li Q, Yang D, Ming K, Zhuang S, Guo P, Yin R, Sun J, Wang H, Lei Q, Liu Z, Ding M, Zhou X, Ding Y. Gut-derived IL-13 contributes to growth via promoting hepatic IGF-1 production. MICROBIOME 2024; 12:248. [PMID: 39580435 PMCID: PMC11585157 DOI: 10.1186/s40168-024-01929-3] [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: 01/09/2024] [Accepted: 09/10/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The gut microbiota has a profound effect on immunity and metabolic status of the host, which has increasingly attracted research communities. However, the intrinsic mechanism underlying the interplay among these three aspects remains unclear. RESULTS Different immune states were established via shaping the population structure of gut microbiota with antibacterial agents. The gut microbiota population structures altered with the subtherapeutic level of antibacterial agents facilitated growth phenotype in both piglets and infant mice. Notably, increased colonization of Prevotella copri was observed in the intestinal microbiota, which shifted the immune balance from a CD4+ T cell-dominated population toward a T helper 2 cell (Th2) phenotype, accompanied by a significant elevation of interleukin-13 (IL-13) levels in the portal vein, which was found to display a strong positive correlation with hepatic insulin-like growth factor-1 (IGF-1) levels. Subsequent investigations unveiled that gut-derived IL-13 stimulated the production of hepatic IGF-1 by activating the IL-13R/Jak2/Stat6 pathway in vitro. The IGF-1 levels were increased in the muscles, leading to an upregulation of and resulted the increased genes associated with related to myofibrillar synthesis and differentiation, which ultimately improving the growth phenotype. CONCLUSIONS Our findings highlight the modification of gut immunity states as a central strategy for increasing anabolism of the host, which has significant implications for addressing human undernutrition/stunting, sarcopenia, obesity and related comorbidities. Video Abstract.
Collapse
Affiliation(s)
- Ning Ma
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Haolong Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qiuhua Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mengyu Chang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiandi Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Sha Nan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qiulin Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qiao Li
- College of Veterinary Medicine, Nanjing Agricultural University, Jiangsu, 210095, People's Republic of China
| | - Diqi Yang
- School of Animal Science and Technology, Hainan University, Haikou, 570228, People's Republic of China
| | - Ke Ming
- School of Life Sciences, Hubei University, Wuhan, 430070, People's Republic of China
| | - Shen Zhuang
- College of Animal Science and Technology, Northwest A&F University, Shanxi, 712100, People's Republic of China
| | - Panpan Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ruiling Yin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jinrui Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Huikang Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Qianghui Lei
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhenli Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mingxing Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaoshu Zhou
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| | - Yi Ding
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
10
|
Lee GM, Mun BG, Hussain A, Kim E, Lee DS, Kim MO, Yun BW. Brown garlic: A nutritionally improved garlic with therapeutic value in asthma treatment via modulation of S-nitrosothiols. Heliyon 2024; 10:e36976. [PMID: 39286149 PMCID: PMC11402760 DOI: 10.1016/j.heliyon.2024.e36976] [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: 02/12/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Nitric Oxide (NO) regulates important physiological functions. Garlic (Allium sativum) is an important food component consumed fresh and processed for thousands of years. It has high L-arginine, which contributes to the NO system in the body. Both garlic and NO impact important physiological processes. Here we produced brown garlic, with significantly higher nutritional and therapeutic value compared to fresh and black garlic. Lower exhaled NO was recorded in asthmatic mice fed with brown garlic but with higher blood SNOs and no change in eNOS and iNOS expression. Lung biopsy showed reduced eosinophil accumulation in asthmatic mice fed with brown garlic. Real-time PCR and Western blot analyses indicated high expression of antioxidant genes but reduced interleukin genes, IL-4, IL-5, IL-6, IL-13, IL1β, and TNF-α brown garlic-fed asthmatic mice as compared to that in fresh and black garlic-fed asthmatic mice. This study provides the first comprehensive and conclusive insight into the nutritional benefits of brown garlic and its therapeutic value for the treatment of asthma in animals.
Collapse
Affiliation(s)
- Geun-Mo Lee
- Department of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Bong-Gyu Mun
- Department of Environmental and Biological Chemistry, Chungbuk National University, Cheongju, Republic of Korea
| | - Adil Hussain
- Department of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
- Department of Agriculture, Abdul Wali Khan University Mardan, 23200 Khyber Pakhtunkhwa, Pakistan
| | - Eungyung Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Republic of Korea
| | - Da-Sol Lee
- Department of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju, Republic of Korea
| | - Byung-Wook Yun
- Department of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
11
|
Xiao H, Tang AZ, Xu ML, Chen HL, Wang F, Li CQ. Mycobacterium vaccae attenuates airway inflammation by inhibiting autophagy and activating PI3K/Akt signaling pathway in OVA-induced allergic airway inflammation mouse model. Mol Immunol 2024; 173:30-39. [PMID: 39018745 DOI: 10.1016/j.molimm.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/11/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
PURPOSE The etiology of asthma remains elusive, with no known cure. Based on accumulating evidence, autophagy, a self-degradation process that maintains cellular metabolism and homeostasis, participates in the development of asthma. Mycobacterium vaccae vaccine (M. vaccae), an immunomodulatory agent, has previously been shown to effectively alleviate airway inflammation and airway remodeling. However, its therapeutic effect on asthma via the regulation of autophagy remains unknown. Therefore, this study aimed to investigate the impact of M. vaccae in attenuating asthma airway inflammation via autophagy-mediated pathways. METHODS Balb/c mice were used to generate an ovalbumin (OVA)-immunized allergic airway model and were subsequently administered either M. vaccae or M. vaccae + rapamycin (an autophagy activator) prior to each challenge. Next, airway inflammation, mucus secretion, and airway remodeling in mouse lung tissue were assessed via histological analyses. Lastly, the expression level of autophagy proteins LC3B, Beclin1, p62, and autolysosome was determined both in vivo and in vitro, along with the expression level of p-PI3K, PI3K, p-Akt, and Akt in mouse lung tissue. RESULTS The findings indicated that aerosol inhalation of M. vaccae in an asthma mouse model has the potential to decrease eosinophil counts, alleviate airway inflammation, mucus secretion, and airway remodeling through the inhibition of autophagy. Likewise, M. vaccae could reduce the levels of OVA-specific lgE, IL-5, IL-13, and TNF-α in asthma mouse models by inhibiting autophagy. Furthermore, this study revealed that M. vaccae also suppressed autophagy in IL-13-stimulated BEAS-2B cells. Moreover, M. vaccae may activate the PI3K/Akt signaling pathway in the lung tissue of asthmatic mice. CONCLUSION In summary, the present study suggests that M. vaccae may contribute to alleviating airway inflammation and remodeling in allergic asthma by potentially modulating autophagy and the PI3K/Akt signaling pathway. These discoveries offer a promising avenue for the development of therapeutic interventions targeting allergic airway inflammation.
Collapse
Affiliation(s)
- Huan Xiao
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China
| | - An-Zhou Tang
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China
| | - Mei-Li Xu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China
| | - Hong-Liu Chen
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China
| | - Fan Wang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China
| | - Chao-Qian Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning 530021, China.
| |
Collapse
|
12
|
Tóth G, Golubova A, Falk A, Lind SB, Nicholas M, Lanekoff I. Interleukin-13 Treatment of Living Lung Tissue Model Alters the Metabolome and Proteome-A Nano-DESI MS Metabolomics and Shotgun Proteomics Study. Int J Mol Sci 2024; 25:5034. [PMID: 38732251 PMCID: PMC11084154 DOI: 10.3390/ijms25095034] [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: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Asthma is a chronic respiratory disease with one of the largest numbers of cases in the world; thus, constant investigation and technical development are needed to unravel the underlying biochemical mechanisms. In this study, we aimed to develop a nano-DESI MS method for the in vivo characterization of the cellular metabolome. Using air-liquid interface (ALI) cell layers, we studied the role of Interleukin-13 (IL-13) on differentiated lung epithelial cells acting as a lung tissue model. We demonstrate the feasibility of nano-DESI MS for the in vivo monitoring of basal-apical molecular transport, and the subsequent endogenous metabolic response, for the first time. Conserving the integrity of the ALI lung-cell layer enabled us to perform temporally resolved metabolomic characterization followed by "bottom-up" proteomics on the same population of cells. Metabolic remodeling was observed upon histamine and corticosteroid treatment of the IL-13-exposed lung cell monolayers, in correlation with alterations in the proteomic profile. This proof of principle study demonstrates the utility of in vivo nano-DESI MS for characterizing ALI tissue layers, and the new markers identified in our study provide a good starting point for future, larger-scale studies.
Collapse
Affiliation(s)
- Gábor Tóth
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | - Alexander Falk
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| | | | | | - Ingela Lanekoff
- Department of Chemistry—BMC, Uppsala University, 75237 Uppsala, Sweden
| |
Collapse
|
13
|
Kliem CV, Schaub B. The role of regulatory B cells in immune regulation and childhood allergic asthma. Mol Cell Pediatr 2024; 11:1. [PMID: 38172451 PMCID: PMC10764675 DOI: 10.1186/s40348-023-00174-2] [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: 09/16/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND As the most common chronic disease in childhood, asthma displays a major public health problem worldwide with the incidence of those affected rising. As there is currently no cure for allergic asthma, it is mandatory to get a better understanding of the underlying molecular mechanism. MAIN BODY By producing IgE antibodies upon allergen contact, B cells play a pivotal role in allergic asthma. Besides that, IL-10-secreting B cell subsets, namely regulatory B cells (Bregs), are reported in mice and humans to play a role in allergic asthma. In humans, several Breg subsets with distinct phenotypic and functional properties are identified among B cells at different maturational and differentiation stages that exert anti-inflammatory functions by expressing several suppressor molecules. Emerging research has focused on the role of Bregs in allergic asthma as well as their role for future diagnostic and preventive strategies. CONCLUSION Knowledge about the exact function of human Bregs in allergic asthma is still very limited. This review aims to summarize the current knowledge on Bregs. We discuss different human Breg subsets, several ways of Breg induction as well as the mechanisms through which they exert immunoregulatory functions, and their role in (childhood) allergic asthma.
Collapse
Affiliation(s)
- Caroline Vanessa Kliem
- Pediatric Allergology, Department of Pediatrics, Dr. Von Hauner Children´S Hospital, University Hospital, Lindwurmstraße 4, 80337, LMU, Munich, Germany
| | - Bianca Schaub
- Pediatric Allergology, Department of Pediatrics, Dr. Von Hauner Children´S Hospital, University Hospital, Lindwurmstraße 4, 80337, LMU, Munich, Germany.
- Member of German Center for Lung Research - DZL, LMU, Munich, Germany.
- Member of German Center for Child and Adolescent Health-DZKJ, LMU, Munich, Germany.
| |
Collapse
|
14
|
Dydak P, Sozańska B. Exposure to farm environment and its correlations with total IgE, IL-13, and IL-33 serum levels in patients with atopy and asthma. Allergol Immunopathol (Madr) 2023; 51:33-40. [PMID: 37695228 DOI: 10.15586/aei.v51i5.823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/17/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND The aim of the study was to evaluate total immunoglobulin E (IgE), IL-13, and IL-33 serum level in people with bronchial asthma and atopy, and in healthy control group depending on their exposure to farm animals currently and in the first year of life. METHODS The study included 174 individuals living in rural areas and in a small town. Standardized questions from the International Study of Asthma and Allergy in Childhood and The European Community Respiratory Health Survey (ECRHS) questionnaires were used to define asthma. Atopic status was verified by skin prick tests. Rural exposure including contact with livestock was verified by adequate questionnaire. Total serum IgE, IL-13, and IL-33 levels were assessed by ELISA (enzyme-linked immunosorbent assay) tests. RESULTS Participants with atopy and bronchial asthma were characterized by high level of immunoglobulin E. Tendency to lower serum IgE level was observed among people reporting present contact with farm animals. Also, among those having contact with livestock in their first year of life, the analogous tendency was noticed. No difference in serum IL-13 levels in participants with asthma and atopy, and controls was observed, and there was no effect of exposure on farm animals on the concentration of IL-13. The highest IL-33 level was found in the atopic group, and the lowest in the control group. Participants currently exposed to farm animals were predisposed to have lower IL-33 serum level. CONCLUSION Exposure of farm animals currently and in first year of life may result in a lower level of total IgE. Correlation between IL-13 and IL-33 serum levels and contact with livestock was not confirmed.
Collapse
Affiliation(s)
- Paulina Dydak
- 1st Department of Pediatrics, Allergology and Cardiology, Wrocław Medical University, Wrocław, Poland;
| | - Barbara Sozańska
- 1st Department of Pediatrics, Allergology and Cardiology, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
15
|
Zhou Y, Duan Q, Yang D. In vitro human cell-based models to study airway remodeling in asthma. Biomed Pharmacother 2023; 159:114218. [PMID: 36638596 DOI: 10.1016/j.biopha.2023.114218] [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: 07/20/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Airway remodeling, as a predominant characteristic of asthma, refers to the structural changes that occurred both in the large and small airways. These pathological changes not only contribute to airway hyperresponsiveness and airway obstruction, but also predict poor outcomes of patients. In vitro models are the alternatives to animal models that facilitate airway remodeling research. Current approaches to mimic airway remodeling in vitro include mono cultures of cell lines and primary cells that are derived from the respiratory tract, and co-culture systems that consist of different cell subpopulations. Moreover, recent advances in microfluid chips and organoids show promise in simulating the complex architecture and functionality of native organs. According, they enable highly physiological-relevant investigations of human diseases in vitro. Here we aim to detail the current human cell-based models regarding their key pros and cons, and to discuss how they may be used to facilitate our understanding of airway remodeling in asthma.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Qirui Duan
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China
| | - Dong Yang
- Department of Anesthesiology, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shijingshan District, Beijing 100144, China.
| |
Collapse
|
16
|
Esnault S, Jarjour NN. Development of Adaptive Immunity and Its Role in Lung Remodeling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:287-351. [PMID: 37464127 DOI: 10.1007/978-3-031-32259-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is characterized by airflow limitations resulting from bronchial closure, which can be either reversible or fixed due to changes in airway tissue composition and structure, also known as remodeling. Airway remodeling is defined as increased presence of mucins-producing epithelial cells, increased thickness of airway smooth muscle cells, angiogenesis, increased number and activation state of fibroblasts, and extracellular matrix (ECM) deposition. Airway inflammation is believed to be the main cause of the development of airway remodeling in asthma. In this chapter, we will review the development of the adaptive immune response and the impact of its mediators and cells on the elements defining airway remodeling in asthma.
Collapse
|
17
|
Jaén M, Martín-Regalado Á, Bartolomé RA, Robles J, Casal JI. Interleukin 13 receptor alpha 2 (IL13Rα2): Expression, signaling pathways and therapeutic applications in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188802. [PMID: 36152905 DOI: 10.1016/j.bbcan.2022.188802] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 10/14/2022]
Abstract
Interleukin 13 receptor alpha 2 (IL13Rα2) is increasingly recognized as a relevant player in cancer invasion and metastasis. Despite being initially considered a decoy receptor for dampening the levels of interleukin 13 (IL-13) in diverse inflammatory conditions, accumulating evidences in the last decades indicate the capacity of IL13Rα2 for mediating IL-13 signaling in cancer cells. The biological reasons behind the expression of this receptor with such extremely high affinity for IL-13 in cancer cells remain unclear. Elevated expression of IL13Rα2 is commonly associated with invasion, late stage and cancer metastasis that results in poor prognosis for glioblastoma, colorectal or breast cancer, among others. The discovery of new mediators and effectors of IL13Rα2 signaling has been critical for deciphering its underlying molecular mechanisms in cancer progression. Still, many questions about the effects of inflammation, the cancer type and the tumor degree in the expression of IL13Rα2 remain largely uncharacterized. Here, we review and discuss the current status of the IL13Rα2 biology in cancer, with particular emphasis in the role of inflammation-driven expression and the regulation of different signaling pathways. As IL13Rα2 implications in cancer continue to grow exponentially, we highlight new targeted therapies recently developed for glioblastoma, colorectal cancer and other IL13Rα2-positive tumors.
Collapse
Affiliation(s)
- Marta Jaén
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ángela Martín-Regalado
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Javier Robles
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain; Protein Alternatives SL, Tres Cantos, Madrid, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
18
|
Current Understanding of Asthma Pathogenesis and Biomarkers. Cells 2022; 11:cells11172764. [PMID: 36078171 PMCID: PMC9454904 DOI: 10.3390/cells11172764] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Asthma is a heterogeneous lung disease with variable phenotypes (clinical presentations) and distinctive endotypes (mechanisms). Over the last decade, considerable efforts have been made to dissect the cellular and molecular mechanisms of asthma. Aberrant T helper type 2 (Th2) inflammation is the most important pathological process for asthma, which is mediated by Th2 cytokines, such as interleukin (IL)-5, IL-4, and IL-13. Approximately 50% of mild-to-moderate asthma and a large portion of severe asthma is induced by Th2-dependent inflammation. Th2-low asthma can be mediated by non-Th2 cytokines, including IL-17 and tumor necrosis factor-α. There is emerging evidence to demonstrate that inflammation-independent processes also contribute to asthma pathogenesis. Protein kinases, adapter protein, microRNAs, ORMDL3, and gasdermin B are newly identified molecules that drive asthma progression, independent of inflammation. Eosinophils, IgE, fractional exhaled nitric oxide, and periostin are practical biomarkers for Th2-high asthma. Sputum neutrophils are easily used to diagnose Th2-low asthma. Despite progress, more studies are needed to delineate complex endotypes of asthma and to identify new and practical biomarkers for better diagnosis, classification, and treatment.
Collapse
|
19
|
Asadi Jozani K, Kouthouridis S, Hirota JA, Zhang B. Next generation preclinical models of lung development, physiology and disease. CAN J CHEM ENG 2022. [DOI: 10.1002/cjce.24581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Kimia Asadi Jozani
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
| | - Sonya Kouthouridis
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| | - Jeremy Alexander Hirota
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Medicine, Division of Respirology McMaster University Hamilton Ontario Canada
- Firestone Institute for Respiratory Health St. Joseph’s Hospital, Hamilton Ontario Canada
| | - Boyang Zhang
- School of Biomedical Engineering, McMaster University 1280 Main Street West, Hamilton Ontario Canada
- Department of Chemical Engineering McMaster University Hamilton Ontario Canada
| |
Collapse
|
20
|
Farmanzadeh A, Qujeq D, Yousefi T. The Interaction Network of MicroRNAs with Cytokines and Signaling Pathways in Allergic Asthma. Microrna 2022; 11:104-117. [PMID: 35507792 DOI: 10.2174/2211536611666220428134324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/15/2022] [Accepted: 03/10/2022] [Indexed: 01/01/2023]
Abstract
Allergic asthma is a complicated disease that is affected by many factors. Numerous cytokines and signaling pathways are attributed to the cause of asthma symptoms. MicroRNAs (miRNAs) are a group of small non-coding single-stranded RNA molecules that are involved in gene silencing and posttranscriptional regulation of gene expression by targeting mRNAs. In pathological conditions, altered expression of microRNAs differentially regulates cytokines and signaling pathways and therefore, can be the underlying reason for the pathogenesis of allergic asthma. Indeed, microRNAs participate in airway inflammation via inducing airway structural cells and activating immune responses by targeting cytokines and signaling pathways. Thus, to make a complete understanding of allergic asthma, it is necessary to investigate the communication network of microRNAs with cytokines and signaling pathways which is contributed to the pathogenesis of allergic asthma. Here, we shed light on this aspect of asthma pathology by Summarizing our current knowledge of this topic.
Collapse
Affiliation(s)
- Ali Farmanzadeh
- Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
21
|
Kishida S, Kato-Mori Y, Okamoto M, Hagiwara K. Anti-inflammatory effect a specific Lactiplantibacillus plantarum in an ovalbumin-induced asthma model. Microbiol Immunol 2022; 66:442-452. [PMID: 35674213 DOI: 10.1111/1348-0421.13014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/13/2022] [Accepted: 06/02/2022] [Indexed: 11/29/2022]
Abstract
Autoimmune, allergic, and respiratory inflammatory diseases are some of the most important health issues worldwide. Disorders of the gut microbiota have been associated with the induction of allergic and inflammatory diseases, and probiotics are being tested for disease prevention. We examined functional Lactiplantibacillus plantarum RGU (Lp-1) to mice with ovalbumin (OVA)-induced asthma model to elucidate the inhibitory effect on pathological progression in asthma model. Prior to the experiments, the intestinal lactic acid bacteria were reduced by administering multiple antibiotics (MAB) to evaluate the administration effect of lactic acid bacteria. Mice were administered with Lp-1 or comparative control lactic acid bacteria in each group. After that, OVA-induced asthma was induced, and cytokine gene expression and histological findings were compared. Exacerbation of lung lesions was confirmed in the MAB group. The Lp-1 group mice had alleviated lung lesions with a decrease in IL-1β, IL-13, IL-17 and an increase in IL-10 of the splenocytes and bronchial lymph nodes compared with the MAB group, but not in the other groups. In OVA-induced asthma, administration of specific Lactiplantibacillus was confirmed to induce anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Shigefumi Kishida
- Department of Pathobiology, Graduate School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Yuko Kato-Mori
- Graduate School of Science, Technology and Innovation, Kobe University, Kobe, Japan
| | - Minoru Okamoto
- Department of Pathobiology, Graduate School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Katsuro Hagiwara
- Department of Pathobiology, Graduate School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| |
Collapse
|
22
|
Zeng Z, Xu X, Zhu Y, Wang Q, Zhang Y, Huo X. Pb and Cd exposure linked with Il-10 and Il-13 gene polymorphisms in asthma risk relevant immunomodulation in children. CHEMOSPHERE 2022; 294:133656. [PMID: 35051511 DOI: 10.1016/j.chemosphere.2022.133656] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/05/2023]
Abstract
Genetic variants and environmental factor of heavy metal exposure accelerate the risk of immune-mediated respiratory diseases, including asthma in children. This study aims to investigate the effects and interaction of Pb, Cd exposure from e-waste and interleukin (IL)-10, IL-13 gene polymorphisms on interleukin expressions in children. Pb, Cd levels, Il-10 (rs1800871, rs1800872, rs1800896) and Il-13 (rs20541, rs1800925) polymorphisms were determined in blood or urine of 155 children (75 from e-waste-exposed area, Guiyu and 80 from reference area, Haojiang). Blood Pb and urine Cd levels of e-waste-exposed children were both higher (mean: 5.89 vs. 3.35 μg/dL; 6.04 vs. 1.82 μg/g, both P < 0.05). Exposed children had a larger proportion of high blood Pb level (> 5.00 μg/dL) (66.67% vs. 4.11%, P < 0.001), but they had no statistically different proportion of high urine Cd level (> 1.00 μg/g creatinine). Median levels of IL-13 decreased (3.674 vs. 4.410 ng/L, P < 0.01), but of IL-10 did not vary in serum of exposed children. The analyses revealed no significant associations of Pb or Cd with Il-10 or Il-13 gene polymorphisms. The high blood Pb and urine Cd level were respectively associated with the low IL-13 ( < 3.696 ng/L) and low IL-10 ( < 0.361 ng/L) level, after adjustment for children's age, gender, and BMI (both P ≤ 0.05). Children homozygous carrying major allele for Il-13 (rs20541 and rs1800925) had additive interactions with high blood Pb level on low IL-13 expression (OR = 5.37, 95% CI: 1.96, 14.73 and OR = 8.45, 95% CI: 2.61, 27.32; both P ≤ 0.001). In contrast, no interaction was observed amongst Pb or Cd with Il-10 gene polymorphisms on its expression. Our findings suggest that Pb exposure interacting with Il-13 gene polymorphisms negatively regulates IL-13 expression, which may pose a risk to abnormal asthma-relevant immunomodulation in preschool children.
Collapse
Affiliation(s)
- Zhijun Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yongzhan Zhu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
23
|
Li B, Xiao Q, Liu J, Mu X, Zhang J, Qi Y, Zhang B, Xiao P, Liu H. Chemical Characterization and Potential Mechanism of the Anti-Asthmatic Activity of a Subfraction from Schisandra chinensis Fruit Extract. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:5015-5025. [PMID: 35416657 DOI: 10.1021/acs.jafc.2c01034] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Schisandra chinensis fruit is a widely edible and medicinal resource, whose extract had a good inhibitory effect on airway inflammation in asthmatic mice. However, the main active components remain unknown. In this work, we found that PET2, a subfraction of its ethanolic extract petroleum ether, displayed significant anti-inflammatory effects in interleukin (IL)-4/tumor necrosis factor (TNF)-α-stimulated BEAS-2B cells. Meanwhile, in the ovalbumin (OVA)-induced allergic asthma mice model, PET2 (200 and 400 mg/kg) had significant effects on attenuating airway inflammatory cell infiltration and reducing serum Th2-related cytokines. Further studies led to the isolation and identification of 14 compounds, guided by ultraperformance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS)-based rapid characterization of chemical constituents. Combining network pharmacology analysis and in vitro experiments, we found that six compounds from PET2 had good anti-inflammatory properties. The potential mechanism may be involved in Fc epsilon RI, T cell receptor, and Jak-STAT signaling pathways. This study clarified the anti-inflammatory properties of the main active fraction and active compounds of S. chinensis fruit and provided a theoretical basis for its anti-asthma scientific utilization.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Qi Xiao
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Jiushi Liu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Xinlu Mu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Jianuo Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Yaodong Qi
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Bengang Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Peigen Xiao
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Haitao Liu
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
24
|
Qian L, Mehrabi Nasab E, Athari SM, Athari SS. Mitochondria signaling pathways in allergic asthma. J Investig Med 2022; 70:863-882. [PMID: 35168999 PMCID: PMC9016245 DOI: 10.1136/jim-2021-002098] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2021] [Indexed: 12/23/2022]
Abstract
Mitochondria, as the powerhouse organelle of cells, are greatly involved in regulating cell signaling pathways, including those related to the innate and acquired immune systems, cellular differentiation, growth, death, apoptosis, and autophagy as well as hypoxic stress responses in various diseases. Asthma is a chronic complicated airway disease characterized by airway hyperresponsiveness, eosinophilic inflammation, mucus hypersecretion, and remodeling of airway. The asthma mortality and morbidity rates have increased worldwide, so understanding the molecular mechanisms underlying asthma progression is necessary for new anti-asthma drug development. The lung is an oxygen-rich organ, and mitochondria, by sensing and processing O2, contribute to the generation of ROS and activation of pro-inflammatory signaling pathways. Asthma pathophysiology has been tightly associated with mitochondrial dysfunction leading to reduced ATP synthase activity, increased oxidative stress, apoptosis induction, and abnormal calcium homeostasis. Defects of the mitochondrial play an essential role in the pro-remodeling mechanisms of lung fibrosis and airway cells' apoptosis. Identification of mitochondrial therapeutic targets can help repair mitochondrial biogenesis and dysfunction and reverse related pathological changes and lung structural remodeling in asthma. Therefore, we here overviewed the relationship between mitochondrial signaling pathways and asthma pathogenic mechanisms.
Collapse
Affiliation(s)
- Ling Qian
- Department of Pulmonary and Critical Care Medicine, Shanghai Fifth People's Hospital, Shanghai, China
| | - Entezar Mehrabi Nasab
- Department of Cardiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran (the Islamic Republic of)
| | | | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran (the Islamic Republic of)
| |
Collapse
|
25
|
Wang R, Wang Y, Liao G, Chen B, Panettieri RA, Penn RB, Tang DD. Abi1 mediates airway smooth muscle cell proliferation and airway remodeling via Jak2/STAT3 signaling. iScience 2022; 25:103833. [PMID: 35198891 PMCID: PMC8851273 DOI: 10.1016/j.isci.2022.103833] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/10/2021] [Accepted: 01/21/2022] [Indexed: 11/05/2022] Open
Abstract
Asthma is a complex pulmonary disorder with multiple pathological mechanisms. A key pathological feature of chronic asthma is airway remodeling, which is largely attributed to airway smooth muscle (ASM) hyperplasia that contributes to thickening of the airway wall and further drives asthma pathology. The cellular processes that mediate ASM cell proliferation are not completely elucidated. Using multiple approaches, we demonstrate that the adapter protein Abi1 (Abelson interactor 1) is upregulated in ∼50% of ASM cell cultures derived from patients with asthma. Loss-of-function studies demonstrate that Abi1 regulates the activation of Jak2 (Janus kinase 2) and STAT3 (signal transducers and activators of transcription 3) as well as the proliferation of both nonasthmatic and asthmatic human ASM cell cultures. These findings identify Abi1 as a molecular switch that activates Jak2 kinase and STAT3 in ASM cells and demonstrate that a dysfunctional Abi1-associated pathway contributes to the progression of asthma.
Collapse
Affiliation(s)
- Ruping Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Yinna Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Guoning Liao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Bohao Chen
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Reynold A. Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Robert Wood Johnson School of Medicine, New Brunswick, NJ 08901, USA
| | - Raymond B. Penn
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Dale D. Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
26
|
Xiao H, Zhang QN, Sun QX, Li LD, Xu SY, Li CQ. Transcriptomic Analysis Reveals a Link Between Hippo Signaling Pathway and Macrophages in Lungs of Mice with OVA-Induced Allergic Asthma. J Inflamm Res 2022; 15:423-437. [PMID: 35082511 PMCID: PMC8784274 DOI: 10.2147/jir.s346505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Huan Xiao
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Qian-nan Zhang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Qi-xiang Sun
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Lao-dong Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Si-yue Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
| | - Chao-qian Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Province, People’s Republic of China
- Correspondence: Chao-qian Li, Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Province, People’s Republic of China, Tel +86 13807887867, Fax +86 771-5350031, Email
| |
Collapse
|
27
|
Ip S, Ms S, Av K, Aa N, Ed B, Vi K, Li V, Vn T, Kv Y, Mm K, Ve B, I S, A M, DA K, O P, M R K. The mixture of siRNAs targeted to IL-4 and IL-13 genes effectively reduces the airway hyperreactivity and allergic inflammation in a mouse model of asthma. Int Immunopharmacol 2021; 103:108432. [PMID: 34923422 DOI: 10.1016/j.intimp.2021.108432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
Bronchial asthma (BA) is one of the most common chronic inflammatory disease of airways. There are huge experimental data indicating that Th2-cytokines IL-4 and IL-13 play a key role in BA pathogenesis. They are implicated in the IgE synthesis, eosinophil infiltration to the lungs and in the development of airway hyperreactivity (AHR), that makes these cytokines the promising targets. Neutralization of IL-4 and IL-13 or its common receptor chain (IL-4Rα) by monoclonal antibodies substantially reduce asthma symptoms. RNA interference provides a novel method for regulation of gene expression by siRNA molecules. In this study we evaluated whether the siRNA targeted to IL-4 and IL-13 reduce BA symptoms in mice model. Experimental BA was induced in BALB/c mice by sensitization to ovalbumin allergen followed by intranasal challenge. The intranasal delivery of siRNAs targeted to IL-4 and IL-13 inhibited the lung expression of these cytokines by more than 50% that led to the attenuation of AHR and pulmonary inflammation; the quantity of eosinophils in lungs which are one of the major inflammatory cells involved in allergic asthma pathogenesis decreased by more than 50% after siRNA treatment. These data support the possibility of a dual IL-4 and IL-13 inhibition by locally delivered siRNAs which in turn leads to the suppression of allergen-induced pulmonary inflammation and AHR.
Collapse
Affiliation(s)
- Shilovskiy Ip
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation.
| | - Sundukova Ms
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Korneev Av
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Nikolskii Aa
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Barvinskaya Ed
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Kovchina Vi
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Vishniakova Li
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Turenko Vn
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Yumashev Kv
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Kaganova Mm
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Brylina Ve
- Federal State Budgetary Educational Institution of Higher Education «Moscow state Academy of Veterinary Medicine and Biotechnology - MVA by K.I. Skryabin» of the Ministry of Agriculture of the Russian Federation, 109472, Moscow, Russian Federation
| | - Sergeev I
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Maerle A
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Kudlay DA
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation; Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenovskiy University), 119991, Moscow, Russian Federation
| | - Petukhova O
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation
| | - Khaitov M R
- National Research Center - Institute of Immunology of Federal Medico-Biological Agency. 115478, 24, Kashirskoye shosse, Moscow, Russian Federation; Federal State Autonomous Educational Institution of Higher Education «N.I. Pirogov Russian National Research Medical University» of the Ministry of Health of the Russian Federation, 117997, Moscow, Russian Federation
| |
Collapse
|
28
|
Azman S, Sekar M, Wahidin S, Gan SH, Vaijanathappa J, Bonam SR, Alvala M, Lum PT, Thakur V, Beladiya JV, Mehta AA. Embelin Alleviates Severe Airway Inflammation in OVA-LPS-Induced Rat Model of Allergic Asthma. J Asthma Allergy 2021; 14:1511-1525. [PMID: 34938083 PMCID: PMC8685448 DOI: 10.2147/jaa.s298613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Asthma is a chronic lung disease, which causes wheezing, tightness in the chest, shortness of breath and coughing. In the wake of coronavirus disease 2019 (COVID-19), which affect the lungs, asthma patients are at high risk. Embelin, a natural benzoquinone obtained mainly from Embelia ribes Burm, has excellent biological properties, including protection against acute asthma. However, since asthma is a chronic and multi-factorial inflammatory disease, asthma conferred by a single allergen in an animal may not be clinically significant. Therefore, the purpose of the current study was to evaluate the effectiveness of embelin against ovalbumin (OVA)-lipopolysaccharide (LPS)-induced severe airway inflammation in experimental animals and to investigate the plausible mechanism of action. METHODS Rats (n=36) were divided into six groups. Group I served as a normal control. Groups II-VI were sensitised with severe allergens (OVA and LPS) on day 7, 14 and 21, followed by OVA and LPS challenge for 30 min three times/week for 3 weeks. Group II acted as an asthmatic disease control and received only vehicle. On the other hand, groups III-V received embelin (12.5, 25 and 50 mg/kg, P.O. respectively) while group VI received a standard dexamethasone (2.5 mg/kg, P.O.) for 15 days from day 27. Lung function parameters, including the respiratory rate, tidal volume and airflow rate were measured at the end of the experiment (day 42). The total and differential counts of leukocytes in the blood and bronchoalveolar fluid (BALF) were calculated. Th2-mediated serum pro-inflammatory cytokines such as interleukin (IL)-4, IL-5 and IL-13 levels were analyzed. At the end of the study protocol, the lung tissues were removed for a histopathology study. Additionally, a molecular docking simulation on embelin and standard dexamethasone was applied to support the in vivo findings. RESULTS Significant inhibition of eosinophils, neutrophils, lymphocytes and monocytes in the blood and the BALF was seen in the groups, which received embelin (25 and 50 mg/kg) and dexamethasone (2.5 mg/kg). Moreover, the lung function parameters were normalised by embelin (25 and 50 mg/kg) treatment significantly. The lung histopathological changes confirmed the protective effect of embelin against severe airway inflammation. The docking findings indicated good binding efficacy of embelin to IL-13. CONCLUSION Overall, our findings indicate that embelin can alleviate severe airway inflammation in OVA-LPS-induced model of allergic asthma occurring by suppression of Th2-mediated immune response. Due to its promising anti-asthmatic effect, it is recommended that embelin should be investigated in clinical trials against asthma. It should also be further explored against COVID-19 or COVID-like diseases due to its ameliorative effects on cytokines and immune cell infiltration.
Collapse
Affiliation(s)
- Shazalyana Azman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
- Bioengineering and Technology Section, Universiti Kuala Lumpur Malaysian Institute of Chemical & Bioengineering Technology, Alor Gajah, Melaka, 78000, Malaysia
| | - Mahendran Sekar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | - Suzana Wahidin
- Bioengineering and Technology Section, Universiti Kuala Lumpur Malaysian Institute of Chemical & Bioengineering Technology, Alor Gajah, Melaka, 78000, Malaysia
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Jaishree Vaijanathappa
- Faculty of Life Sciences, JSS Academy of Higher Education and Research Mauritius, Republic of Mauritius
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale; Centre de Recherche des Cordeliers, Equipe-Immunopathologie et Immunointervention Thérapeutique, Sorbonne Université, Université De Paris, Paris, F-75006, France
| | | | - Pei Teng Lum
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Health Sciences, Universiti Kuala Lumpur Royal College of Medicine Perak, Ipoh, Perak, 30450, Malaysia
| | - Vandana Thakur
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Jayesh V Beladiya
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Anita A Mehta
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| |
Collapse
|
29
|
Subramanian H, Hashem T, Bahal D, Kammala AK, Thaxton K, Das R. Ruxolitinib Ameliorates Airway Hyperresponsiveness and Lung Inflammation in a Corticosteroid-Resistant Murine Model of Severe Asthma. Front Immunol 2021; 12:786238. [PMID: 34777398 PMCID: PMC8586657 DOI: 10.3389/fimmu.2021.786238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022] Open
Abstract
Asthma prevalence has increased considerably over the decades and it is now considered as one of the most common chronic disorders in the world. While the current anti-asthmatic therapies are effective for most asthma patients, there are 5-10% subjects whose disease is not controlled by such agents and they account for about 50% of the asthma-associated healthcare costs. Such patients develop severe asthma (SA), a condition characterized by a dominant Th1/Th17 cytokine response that is accompanied by Type 2 (T2)-low endotype. As JAK (Janus Kinase) signaling is very important for the activation of several cytokine pathways, we examined whether inhibition of JAKs might lessen the clinical and laboratory manifestations of SA. To that end, we employed a recently described murine model that recapitulates the complex immune response identified in the airways of human SA patients. To induce SA, mice were sensitized with house dust mite extract (HDME) and cyclic (c)-di-GMP and then subsequently challenged with HDME and a lower dose of c-di-GMP. In this model, treatment with the JAK inhibitor, Ruxolitinib, significantly ameliorated all the features of SA, including airway hyperresponsiveness and lung inflammation as well as total IgE antibody titers. Thus, these studies highlight JAKs as critical targets for mitigating the hyper-inflammation that occurs in SA and provide the framework for their incorporation into future clinical trials for patients that have severe or difficult-to manage asthma.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Tanwir Hashem
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Devika Bahal
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Ananth K Kammala
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Kanedra Thaxton
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Rupali Das
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
30
|
Huang J, Chen X, Xie A. Formononetin ameliorates IL‑13‑induced inflammation and mucus formation in human nasal epithelial cells by activating the SIRT1/Nrf2 signaling pathway. Mol Med Rep 2021; 24:832. [PMID: 34590155 PMCID: PMC8503736 DOI: 10.3892/mmr.2021.12472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023] Open
Abstract
Formononetin has proven to be anti‑inflammatory and able to alleviate symptoms of certain allergic diseases. The present study aimed to determine and elucidate the potential effects of formononetin in allergic rhinitis. JME/CF15 cells were pretreated with formononetin at different doses, followed by stimulation with IL‑13. Cell Counting Kit‑8 assay was performed to determine the cytotoxicity of formononetin. The expression levels of inflammation‑related proteins, histamine, IgE, TNF‑α, IL‑1β, IL‑6, granulocyte‑macrophage colony‑stimulating factor and eotaxin in IL‑13‑stimulated JME/CF15 cells were detected using ELISAs. The expression levels of phosphorylated‑NF‑κB p65, NF‑κB p65 and cyclooxygenase‑2 (Cox‑2) were analyzed using western blotting. Reverse transcription‑quantitative PCR, western blotting and immunofluorescence were performed to measure the levels of mucin 5AC oligomeric mucus/gel‑forming. Expression levels of sirtuin 1 (SIRT1) and nuclear erythroid factor 2‑related factor 2 (Nrf2) proteins were also measured using western blotting. The results of the present study revealed that formononetin exerted no cytotoxic effect on the viability of JME/CF15 cells. Following stimulation of JME/CF15 cells with IL‑13, formononetin suppressed the upregulated expression levels of proinflammatory cytokines. IL‑13‑induced formation of mucus was also attenuated by formononetin treatment. Furthermore, it was found that the SIRT1/Nrf2 signaling pathway was activated in formononetin‑treated JME/CF15 cells, whereas treatment with the SIRT1 inhibitor, EX527, reversed the effects of formononetin on IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. In conclusion, the findings of the current study indicated that formononetin may activate the SIRT1/Nrf2 signaling pathway, thereby inhibiting IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. These results suggested that formononetin may represent a promising agent for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Juanjuan Huang
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xianfeng Chen
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Aihua Xie
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
31
|
Lu Y, Zhou Y, Lin Y, Li W, Tian S, Hao X, Guo H. Preventive effects of donkey milk powder on the ovalbumin-induced asthmatic mice. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
32
|
Nakayama T, Hirahara K, Kimura MY, Iwamura C, Kiuchi M, Kokubo K, Onodera A, Hashimoto K, Motohashi S. CD4 + T cells in inflammatory diseases : pathogenic T-helper cells and the CD69-Myl9 system. Int Immunol 2021; 33:699-704. [PMID: 34427648 DOI: 10.1093/intimm/dxab053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
CD4 + T cells direct immune responses against infectious microorganisms but are also involved in the pathogenesis of inflammatory diseases. In the last two to three decades, various researchers have identified and characterized several functional CD4 + T cell subsets, including T-helper 1 (Th1), Th2, Th9 and Th17 cells and regulatory T (Treg) cells. In this mini-review, we introduce the concept of pathogenic Th cells that induce inflammatory diseases with a model of disease induction by a population of pathogenic Th cells; "pathogenic Th population disease-induction model". We will focus on Th2 cells that induce allergic airway inflammation-pathogenic Th2 cells (Tpath2 cells)-and discuss the nature of Tpath2 cells that shape the pathology of chronic inflammatory diseases. Various Tpath2 cell subsets have been identified and their unique features are summarized in mouse and human systems. Second, we will discuss how Th cells migrate and are maintained in chronic inflammatory lesions. We propose a model known as the "CD69-Myl9 system". CD69 is a cell surface molecule expressed on activated T cells and interaction with its ligand myosin light chain 9 (Myl9) is required for the induction of inflammatory diseases. Myl9 molecules in the small vessels of inflamed lungs may play a crucial role in the migration of activated T cells into inflammatory lesions. Emerging evidence may provide new insight into the pathogenesis of chronic inflammatory diseases and contribute to the development of new therapeutic strategies for intractable inflammatory disorders.
Collapse
Affiliation(s)
- Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan.,AMED-CREST, AMED, Inohana Chuo-ku, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Motoko Y Kimura
- Department of Experimental Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Chiaki Iwamura
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| | - Kahoko Hashimoto
- Department of Life Science, Faculty of Advanced Engineering, Chiba Institute of Technology, Tsudanuma, Narashino-city, Chiba, Japan
| | - Shinichiro Motohashi
- Department of Medical Immunology, Graduate School of Medicine, Chiba University, Inohana Chuo-ku, Chiba, Japan
| |
Collapse
|
33
|
Shastri MD, Allam VSRR, Shukla SD, Jha NK, Paudel KR, Peterson GM, Patel RP, Hansbro PM, Chellappan DK, Dua K. Interleukin-13: A pivotal target against influenza-induced exacerbation of chronic lung diseases. Life Sci 2021; 283:119871. [PMID: 34352260 DOI: 10.1016/j.lfs.2021.119871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/18/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022]
Abstract
Non-communicable, chronic respiratory diseases (CRDs) affect millions of individuals worldwide. The course of these CRDs (asthma, chronic obstructive pulmonary disease, and cystic fibrosis) are often punctuated by microbial infections that may result in hospitalization and are associated with increased risk of morbidity and mortality, as well as reduced quality of life. Interleukin-13 (IL-13) is a key protein that regulates airway inflammation and mucus hypersecretion. There has been much interest in IL-13 from the last two decades. This cytokine is believed to play a decisive role in the exacerbation of inflammation during the course of viral infections, especially, in those with pre-existing CRDs. Here, we discuss the common viral infections in CRDs, as well as the potential role that IL-13 plays in the virus-induced disease pathogenesis of CRDs. We also discuss, in detail, the immune-modulation potential of IL-13 that could be translated to in-depth studies to develop IL-13-based therapeutic entities.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia.
| | | | - Shakti D Shukla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, Greater Noida, UP, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart 7005, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University (IMU), Kuala Lumpur, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia; Centre for Inflammation, Centenary Institute, Sydney, NSW 2050, Australia; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia.
| |
Collapse
|
34
|
Gao W, Jin Z, Zheng Y, Xu Y. Psoralen inhibits the inflammatory response and mucus production in allergic rhinitis by inhibiting the activator protein 1 pathway and the downstream expression of cystatin‑SN. Mol Med Rep 2021; 24:652. [PMID: 34278468 PMCID: PMC8299190 DOI: 10.3892/mmr.2021.12291] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/22/2021] [Indexed: 12/14/2022] Open
Abstract
Psoralen (PSO) exerts anti-inflammatory pharmacological effects and plays an important role in a variety of inflammatory diseases. However, the effects of PSO with allergic rhinitis (AR) are yet to be reported. In the present study, an in vitro AR model was generated by inducing JME/CF15 human nasal epithelial cells with IL-13, after which MTT was used to assess the cytotoxicity of PSO. The expression levels of inflammatory cytokines (granulocyte-macrophage colony-stimulating factor and Eotaxin) were determined by ELISA. Furthermore, the expression of inflammatory IL-6 and −8, as well as mucin 5AC, was assessed by reverse transcription-quantitative PCR and western blotting, and cellular reactive oxygen species were detected using a 2′,7′-dichlorodihydrofluorescein diacetate fluorescent probe. Western blotting was also used to detect the expression and phosphorylation of c-Fos and c-Jun in the activator protein 1 (AP-1) pathway, as well as the expression of cystatin-SN (CST1). PSO inhibited the inflammatory response and mucus production in IL-13-induced JME/CF15 cells. Furthermore, the levels of c-Fos and c-Jun phosphorylation in the AP-1 pathway were decreased in IL-13-induced JME/CF15 cells following PSO treatment. The expression of pathway proteins was activated by the addition of PMA, an AP-1 pathway activator, which concurrently reversed the inhibitory effects of PSO on the inflammatory response and mucus formation. The addition of an AP-1 inhibitor (SP600125) further inhibited pathway activity, and IL-13-induced inflammation and mucus formation was restored. In conclusion, PSO regulates the expression of CST1 by inhibiting the AP-1 pathway, thus suppressing the IL-13-induced inflammatory response and mucus production in nasal mucosal epithelial cells.
Collapse
Affiliation(s)
- Wenying Gao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Zhenglong Jin
- Department of Neurology, Jiangmen Wuyi Hospital of TCM Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen, Guangdong 529000, P.R. China
| | - Yanxia Zheng
- Department of TCM Pediatrics, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Youjia Xu
- Department of TCM Pediatrics, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| |
Collapse
|
35
|
Carlier FM, de Fays C, Pilette C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front Physiol 2021; 12:691227. [PMID: 34248677 PMCID: PMC8264588 DOI: 10.3389/fphys.2021.691227] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells, which provide a complex and adaptive module that ensures first-line defense against external toxics, irritants, antigens, and pathogens. The underlying mechanisms of host protection encompass multiple physical, chemical, and immune pathways. In the lung, inhaled agents continually challenge the airway epithelial barrier, which is altered in chronic diseases such as chronic obstructive pulmonary disease, asthma, cystic fibrosis, or pulmonary fibrosis. In this review, we describe the epithelial barrier abnormalities that are observed in such disorders and summarize current knowledge on the mechanisms driving impaired barrier function, which could represent targets of future therapeutic approaches.
Collapse
Affiliation(s)
- François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology and Lung Transplant, Centre Hospitalier Universitaire UCL Namur, Yvoir, Belgium
| | - Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| |
Collapse
|
36
|
Kammala AK, Bahal D, Yang C, Panettieri RA, Das R, Subramanian H. Na +/H + Exchanger Regulatory Factor 1 Mediates the Pathogenesis of Airway Inflammation in a Murine Model of House Dust Mite-Induced Asthma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2301-2311. [PMID: 33952618 PMCID: PMC8113128 DOI: 10.4049/jimmunol.2001199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/16/2021] [Indexed: 11/19/2022]
Abstract
Na+/H+ exchanger regulatory factor 1 (NHERF1), a class I PDZ-binding protein, regulates G protein-coupled receptor signaling in some cell types. NHERF1 also functions as a scaffolding protein and activates non-G protein-coupled receptor signaling pathways, thereby contributing to the pathogenesis of various diseases. Although we have previously shown that NHERF1 regulates mast cell functions, there is little information regarding the role of NHERF1 in other immune cells. How NHERF1 regulates the pathogenesis of allergic disease such as asthma also remains unknown. In the current study, we show that NHERF1 promotes allergic airway inflammation in a house dust mite extract (HDME)-induced mouse model of asthma. Specifically, HDME-specific serum IgE levels, airway leukocyte numbers, and goblet cell hyperplasia were reduced in NHERF1+/- mice as compared with NHERF1+/+ mice. Interestingly, the gene expression of inflammatory (IL-17a, IL-25, and IL-33) as well as T helper 2 (Th2) cytokines (IL-4, IL-5, and IL-13) and several chemokines that recruit eosinophils, neutrophils, and lymphocytes were also decreased in the lungs of NHERF1+/- mice exposed to HDME. Consistent with these observations, microRNAs regulating mucus production, inflammation, Th2 effector functions, and IL-13 expression were increased in the lungs of HDME-treated NHERF1+/- mice. Overall, our studies reveal a unique role for NHERF1 in regulating asthma pathogenesis, and further elucidation of the mechanisms through which NHERF1 modulates allergic inflammation will lead to the development of new therapeutic strategies for asthma.
Collapse
Affiliation(s)
- Ananth K Kammala
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Devika Bahal
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Canchai Yang
- Department of Physiology, Michigan State University, East Lansing, MI
| | | | - Rupali Das
- Department of Physiology, Michigan State University, East Lansing, MI
| | | |
Collapse
|
37
|
Zou B, Fu Y, Cao C, Pan D, Wang W, Kong L. Gentiopicroside ameliorates ovalbumin-induced airway inflammation in a mouse model of allergic asthma via regulating SIRT1/NF-κB signaling pathway. Pulm Pharmacol Ther 2021; 68:102034. [PMID: 33887491 DOI: 10.1016/j.pupt.2021.102034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 11/28/2022]
Abstract
Allergic asthma is a common airway inflammatory disorder with increasing morbidity and mortality worldwide. Gentiopicroside (GPS) is a secoiridoid glycoside compound that exhibits anti-inflammatory property. However, the effect of GPS on allergic asthma has not been reported yet. In this study, we investigated the role of GPS in a mouse model of ovalbumin (OVA)-induced allergic asthma and explored its potential mechanism. Mice were sensitized with OVA and gavaged with 20, 40, or 80 mg/kg GPS. Administration of GPS decreased lung wet-to-dry weight ratio. Histological analysis of H&E and PAS staining showed that GPS treatment alleviated inflammatory cell infiltration and goblet cell hyperplasia in lung tissue of OVA-sensitized mice. Moreover, GPS inhibited the recruitment of inflammatory cells including total cells, macrophages, eosinophils, lymphocytes and neutrophils and the secretion of T helper type 2 (Th2) cytokines (interleukin (IL)-4, IL-5 and IL-13) in bronchoalveolar lavage fluid (BALF) of OVA-sensitized mice in a dose dependent manner. The levels of OVA-specific immunoglobulin E (IgE) and pro-inflammatory tumor necrosis factor (TNF)-α were also attenuated by GPS treatment. Interestingly, GPS upregulated the expression of silent information regulator 1 (SIRT1) while downregulated the expression of acetyl-nuclear factor kappa B (NF-κB) p65 in lung tissue of OVA-sensitized mice. Furthermore, treatment with an SIRT1 inhibitor (EX-527) partially abolished the inhibitory effect of GPS on OVA-induced airway inflammation, suggesting that the anti-inflammation of GPS might be achieved through regulating SIRT1/NF-κB p65 signaling pathway. These findings indicate that GPS might be a novel drug candidate in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Bo Zou
- Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China; Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Yue Fu
- Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Chaofan Cao
- Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China; Department of Respiratory Medicine, The Second Affiliated Hospital of Shenyang Medical College, Shenyang City, Liaoning Province, People's Republic of China
| | - Dianzhu Pan
- Department of Respiratory Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, Liaoning Province, People's Republic of China
| | - Wei Wang
- Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China
| | - Lingfei Kong
- Institute of Respiratory Diseases, The First Hospital of China Medical University, Shenyang City, Liaoning Province, People's Republic of China.
| |
Collapse
|
38
|
Oliveria JP, Agayby R, Gauvreau GM. Regulatory and IgE + B Cells in Allergic Asthma. Methods Mol Biol 2021; 2270:375-418. [PMID: 33479910 DOI: 10.1007/978-1-0716-1237-8_21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Allergic asthma is triggered by inhalation of environmental allergens resulting in bronchial constriction and inflammation, which leads to clinical symptoms such as wheezing, coughing, and difficulty breathing. Asthmatic airway inflammation is initiated by inflammatory mediators released by granulocytic cells. However, the immunoglobulin E (IgE) antibody is necessary for the initiation of the allergic cascade, and IgE is produced and released exclusively by memory B cells and plasma cells. Acute allergen exposure has also been shown to increase IgE levels in the airways of patients diagnosed with allergic asthma; however, more studies are needed to understand local airway inflammation. Additionally, regulatory B cells (Bregs) have been shown to modulate IgE-mediated inflammatory processes in allergic asthma pathogenesis, particularly in mouse models of allergic airway disease. However, the levels and function of these IgE+ B cells and Bregs remain to be elucidated in human models of asthma. The overall objective for this chapter is to provide detailed methodological, and insightful technological advances to study the biology of B cells in allergic asthma pathogenesis. Specifically, we will describe how to investigate the frequency and function of IgE+ B cells and Bregs in allergic asthma, and the kinetics of these cells after allergen exposure in a human asthma model.
Collapse
Affiliation(s)
- John Paul Oliveria
- School of Medicine, Department of Pathology, Stanford University, Stanford, CA, USA.,Department of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Rita Agayby
- Department of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Gail M Gauvreau
- Department of Medicine, Division of Respirology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
39
|
MicroRNA Targets for Asthma Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:89-105. [PMID: 33788189 DOI: 10.1007/978-3-030-63046-1_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic inflammatory obstructive lung disease that is stratified into endotypes. Th2 high asthma is due to an imbalance of Th1/Th2 signaling leading to abnormally high levels of Th2 cytokines, IL-4, IL-5, and IL-13 and in some cases a reduction in type I interferons. Some asthmatics express Th2 low, Th1/Th17 high phenotypes with or without eosinophilia. Most asthmatics with Th2 high phenotype respond to beta-adrenergic agonists, muscarinic antagonists, and inhaled corticosteroids. However, 5-10% of asthmatics are not well controlled by these therapies despite significant advances in lung immunology and the pathogenesis of severe asthma. This problem is being addressed by developing novel classes of anti-inflammatory agents. Numerous studies have established efficacy of targeting pro-inflammatory microRNAs in mouse models of mild/moderate and severe asthma. Current approaches employ microRNA mimics and antagonists designed for use in vivo. Chemically modified oligonucleotides have enhanced stability in blood, increased cell permeability, and optimized target specificity. Delivery to lung tissue limits clinical applications, but it is a tractable problem. Future studies need to define the most effective microRNA targets and effective delivery systems. Successful oligonucleotide drug candidates must have adequate lung cell uptake, high target specificity, and efficacy with tolerable off-target effects.
Collapse
|
40
|
Zhan JB, Zheng J, Zeng LY, Fu Z, Huang QJ, Wei X, Zeng M. Downregulation of miR-96-5p Inhibits mTOR/NF-κb Signaling Pathway via DEPTOR in Allergic Rhinitis. Int Arch Allergy Immunol 2021; 182:210-219. [PMID: 33477144 DOI: 10.1159/000509403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/14/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This study aims to investigate the regulatory effect of microRNA-96-5p (miR-96-5p) in the pathophysiological process of allergic rhinitis (AR). METHODS Nasal mucosal tissue samples were collected from AR patients and healthy controls. An in vitro AR model was established by stimulating human nasal epithelial cells (HNECs) with interleukin (IL)-13. The expressions of target genes and proteins were measured by qPCR, Western blot, or ELISA. Dual-luciferase reporter assay and pull-down assay were performed to confirm the interaction between miR-96-5p and DEP domain-containing mammalian target of rapamycin-interacting protein (DEPTOR). RESULTS The level of miR-96-5p was increased while the expression of DEPTOR was decreased in AR patients. The expressions of proinflammatory cytokines were markedly increased and the mammalian target of rapamycin (mTOR)/NF-κB pathway was activated in HNECs following IL-13 stimulation. miR-96-5p downregulation alleviated the stimulated function by IL-13. DEPTOR was the target of miR-96-5p. Knockdown of DEPTOR reversed the function of miR-96-5p inhibitor on IL-13-stimulated HNECs. CONCLUSIONS The current study showed that miR-96-5p and DEPTOR were aberrantly expressed in AR nasal mucosa. miR-96-5p knockdown inhibited the production of inflammatory cytokines and the activation of mTOR/NF-κB pathway via targeting DEPTOR. These findings suggested that miR-96-5p might be used as a diagnostic marker and therapeutic target for the treatment of AR.
Collapse
Affiliation(s)
- Jia-Bin Zhan
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jing Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Lian-Ya Zeng
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Zhi Fu
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Qiu-Ju Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Xin Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China,
| | - Min Zeng
- Medical Center, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| |
Collapse
|
41
|
Sohn KH, Baek MG, Choi SM, Bae B, Kim RY, Kim YC, Kim HY, Yi H, Kang HR. Alteration of Lung and Gut Microbiota in IL-13-Transgenic Mice Simulating Chronic Asthma. J Microbiol Biotechnol 2020; 30:1819-1826. [PMID: 33046682 PMCID: PMC9728179 DOI: 10.4014/jmb.2009.09019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests a potential role of microbial colonization in the inception of chronic airway diseases. However, it is not clear whether the lung and gut microbiome dysbiosis is coincidental or a result of mutual interaction. In this study, we investigated the airway microbiome in interleukin 13 (IL-13)-rich lung environment and related alterations of the gut microbiome. IL-13- overexpressing transgenic (TG) mice presented enhanced eosinophilic inflammatory responses and mucus production, together with airway hyperresponsiveness and subepithelial fibrosis. While bronchoalveolar lavage fluid and cecum samples obtained from 10-week-old IL-13 TG mice and their C57BL/6 wild-type (WT) littermates showed no significant differences in alpha diversity of lung and gut microbiome, they presented altered beta diversity in both lung and gut microbiota in the IL-13 TG mice compared to the WT mice. Lung-specific IL-13 overexpression also altered the composition of the gut as well as the lung microbiome. In particular, IL-13 TG mice showed an increased proportion of Proteobacteria and Cyanobacteria and a decreased amount of Bacteroidetes in the lungs, and depletion of Firmicutes and Proteobacteria in the gut. The patterns of polymicrobial interaction within the lung microbiota were different between WT and IL-13 TG mice. For instance, in IL-13 TG mice, lung Mesorhizobium significantly affected the alpha diversity of both lung and gut microbiomes. In summary, chronic asthma-like pathologic changes can alter the lung microbiota and affect the gut microbiome. These findings suggest that the lung-gut microbial axis might actually work in asthma.
Collapse
Affiliation(s)
- Kyoung-Hee Sohn
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Pulmonology, Allergy and Critical Care, Department of Internal Medicine, Kyung Hee University Medical Center, Seoul 0447, Republic of Korea
| | - Min-gyung Baek
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sung-Mi Choi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Boram Bae
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Ruth Yuldam Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Young-Chan Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hye-Young Kim
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 08826, Republic of Korea
| | - Hana Yi
- Department of Public Health Sciences, Graduate School, Korea University, Seoul 02841, Republic of Korea,School of Biosystems and Biomedical Sciences, Korea University, Seoul 02841, Republic of Korea,Corresponding authors H.Yi Phone: +82-2-3290-5644 Fax: +82-2-940-2849 E-mail:
| | - Hye-Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul National University College of Medicine, Seoul 08826, Republic of Korea,Division of Allergy and Clinical Immunology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul 0882, Republic of Korea,H-R.Kang Phone: 82-2-2072-0820 Fax: 82-2-742-3291 E-mail:
| |
Collapse
|
42
|
Tereschenko SY, Smolnikova MV, Kasparov EV, Shakhtshneider EV, Malinchik MA, Konopleva OS, Smirnova SV. Role of IL13 genetic polymorphism in the development of bronchial asthma in children. MEDICAL IMMUNOLOGY (RUSSIA) 2020. [DOI: 10.15789/1563-0625-roi-1986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Bronchial asthma is a multifactorial disease, with both environmental factors and genetic predisposal affecting its development. A number of gene associations have been obtained between polymorphisms of cytokine genes produced by different types of immune cells and asthma development. Interleukin-13 is involved in allergic inflammation, increased bronchial hypersensitivity, regulation of eosinophil levels and IgE production by B cells, thus making it promising for studying IL13 gene polymorphisms in bronchial asthma coupled to development of the disease. The aim of this study was to investigate possible association between asthma and IL13 rs1800925 polymorphism in the children of Caucasian origin in Eastern Siberia. Four groups of patients with asthma were examined (mean age 12.8±1.2 years): with a controlled (n = 95) and uncontrolled course (n = 107), with severe (n = 71) and moderate severity (n = 131) diseases. The control group consisted of healthy individuals: children (n = 33) and adults (n = 102). DNA was isolated with sorbent method; genotyping was carried out using RT-PCR using specific oligonucleotide primers and fluorescent TaqMan probes. The allele and genotype frequencies were compared by the χ-square test using an online calculator. The odds ratio (OR) with a 95% confidence interval (CI) was performed to link genetic markers with pathological phenotypes. The CT IL13 rs1800925 genotype was shown to be associated with moderate asthma and cases of uncontrollable clinical course, whereas the TT genotype was associated with severe asthma. Thus, rs1800925 polymorphism of IL13 gene (the T* variant is known to be associated with increased IL-13 expression) may be associated with bronchial asthma in children. Our data are consistent with results of other authors. E.g., Liu Z. et al. revealed an association between rs1800925 IL13 and the risk of developing asthma in children, with CT and TT genotypes being more common in the patient group. Radhakrishnan A. et al., was studied rs1800925 IL13 in adult population of Malaysia and found that the T* allele frequency in the group of patients significantly exceeds the frequency of this allele in the control group. Thus, the results of our study showed that IL13 rs1800925 polymorphism is associated with bronchial asthma in children, especially, with level of its control and severity of the disease.
Collapse
Affiliation(s)
- S. Yu. Tereschenko
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - M. V. Smolnikova
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - E. V. Kasparov
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - E. V. Shakhtshneider
- Institute of Internal and Preventive Medicine, Branch of Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences
| | - M. A. Malinchik
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| | - O. S. Konopleva
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk; Krasnoyarsk State V. Voino-Yasenetsky Medical University
| | - S. V. Smirnova
- Research Institute of Medical Problems of the North, Krasnoyarsk Scientific Center, Siberian Branch, Russian Academy of Sciences, Krasnoyarsk
| |
Collapse
|
43
|
Role of interleukins 12B and 17A genetic variation in house dust mites allergy. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2020. [DOI: 10.1186/s43042-020-00098-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Abstract
Background
The house dust mites (HDM) constitute a major cause of allergic diseases all over the world. Genes encoding interleukins 12B and 17A which determine the course of T cell-mediated immune response are prime candidates as allergic disease susceptibility. The purpose of this study was to evaluate whether a single-nucleotide polymorphisms (SNP) of interleukins 12B + 1188A/C (rs3212227) and 17A −197G/A (rs2275913) confers susceptibility to HDM allergic diseases. Through a case-control study, 120 subjects served as 60 dust mites' allergic patients and 60 healthy non-allergic controls. Total immunoglobulin (Ig) E level, eosinophilic count, serum interleukins 4, 10, 12B, and 17A levels for the studied subjects were measured. Then, genotyping of single-nucleotide polymorphisms (SNPs) at +1188A/C for IL12B and −197G/A for IL17A gene were conducted using restriction fragment length polymorphisms (RFLP-PCR).
Results
The present study showed that in HDMs' allergic subjects there was a significant increase in IL12B (+1188 A/C) and IL17A (−197 G/A) genotype variants compared to that of the controls. There was a significant increase in total IgE levels, eosinophil counts, and the levels of both IL-4 and IL-17A, while IL12B was significantly lower in patients compared to that of the controls. There was no significant difference in IL-10 levels between patients and controls.
Conclusion
Our findings indicate that IL12B (+1188 A/C) and IL17A (−197G/A) might be associated with an increase in the susceptibility to dust mites’ allergic patients.
Collapse
|
44
|
Chiba Y, Ueda C, Kohno N, Yamashita M, Miyakawa Y, Ando Y, Suto W, Hirabayashi T, Takenoya F, Takasaki I, Kamei J, Sakai H, Shioda S. Attenuation of relaxing response induced by pituitary adenylate cyclase-activating polypeptide in bronchial smooth muscle of experimental asthma. Am J Physiol Lung Cell Mol Physiol 2020; 319:L786-L793. [PMID: 32877227 DOI: 10.1152/ajplung.00315.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Bronchomotor tone is regulated by contraction and relaxation of airway smooth muscle (ASM). A weakened ASM relaxation might be a cause of airway hyperresponsiveness (AHR), a characteristic feature of bronchial asthma. Pituitary adenylyl cyclase-activating polypeptide (PACAP) is known as a mediator that causes ASM relaxation. To date, whether or not the PACAP responsiveness is changed in asthmatic ASM is unknown. The current study examined the hypothesis that relaxation induced by PACAP is reduced in bronchial smooth muscle (BSM) of allergic asthma. The ovalbumin (OA)-sensitized mice were repeatedly challenged with aerosolized OA to induce asthmatic reaction. Twenty-four hours after the last antigen challenge, the main bronchial smooth muscle (BSM) tissues were isolated. Tension study showed a BSM hyperresponsiveness to acetylcholine in the OA-challenged mice. Both quantitative RT-PCR and immunoblot analyses revealed a significant decrease in PAC1 receptor expression in BSMs of the diseased mice. Accordingly, in the antigen-challenged group, the PACAP-induced PAC1 receptor-mediated BSM relaxation was significantly attenuated, whereas the relaxation induced by vasoactive intestinal polypeptide was not changed. These findings suggest that the relaxation induced by PACAP is impaired in BSMs of experimental asthma due to a downregulation of its binding partner PAC1 receptor. Impaired BSM responsiveness to PACAP might contribute to the AHR in asthma.
Collapse
Affiliation(s)
- Yoshihiko Chiba
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Chihiro Ueda
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Naoko Kohno
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Michio Yamashita
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Yui Miyakawa
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Yusuke Ando
- Department of Biomolecular Pharmacology, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Wataru Suto
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Takahiro Hirabayashi
- Peptide Drug Innovation Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Fumiko Takenoya
- Department of Physiology and Molecular Sciences, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Ichiro Takasaki
- Department of Pharmacology, Graduate School of Science and Engineering, University of Toyama, Toyama, Japan
| | - Junzo Kamei
- Department of Biomolecular Pharmacology, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Hiroyasu Sakai
- Department of Biomolecular Pharmacology, Hoshi University School of Pharmacy, Tokyo, Japan
| | - Seiji Shioda
- Peptide Drug Innovation Global Research Center for Innovative Life Science, Hoshi University School of Pharmacy, Tokyo, Japan
| |
Collapse
|
45
|
Long A, Bunning B, Sampath V, DeKruyff RH, Nadeau KC. Epigenetics and the Environment in Airway Disease: Asthma and Allergic Rhinitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1253:153-181. [PMID: 32445095 DOI: 10.1007/978-981-15-3449-2_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Asthma and rhinitis are complex, heterogeneous diseases characterized by chronic inflammation of the upper and lower airways. While genome-wide association studies (GWAS) have identified a number of susceptible loci and candidate genes associated with the pathogenesis of asthma and allergic rhinitis (AR), the risk-associated alleles account for only a very small percent of the genetic risk. In allergic airway and other complex diseases, it is thought that epigenetic modifications, including DNA methylation, histone modifications, and non-coding microRNAs, caused by complex interactions between the underlying genome and the environment may account for some of this "missing heritability" and may explain the high degree of plasticity in immune responses. In this chapter, we will focus on the current knowledge of classical epigenetic modifications, DNA methylation and histone modifications, and their potential role in asthma and AR. In particular, we will review epigenetic variations associated with maternal airway disease, demographics, environment, and non-specific associations. The role of specific genetic haplotypes in environmentally induced epigenetic changes are also discussed. A major limitation of many of the current studies of asthma epigenetics is that they evaluate epigenetic modifications in both allergic and non-allergic asthma, making it difficult to distinguish those epigenetic modifications that mediate allergic asthma from those that mediate non-allergic asthma. Additionally, most DNA methylation studies in asthma use peripheral or cord blood due to poor accessibility of airway cells or tissue. Unlike DNA sequences, epigenetic alterations are quite cell- and tissue-specific, and epigenetic changes found in airway tissue or cells may be discordant from that of circulating blood. These two confounding factors should be considered when reviewing epigenetic studies in allergic airway disease.
Collapse
Affiliation(s)
- Andrew Long
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, 94305, USA.,Department of Pharmacy, Lucile Packard Children's Hospital, Stanford, CA, 94304, USA
| | - Bryan Bunning
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, 94305, USA
| | - Vanitha Sampath
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, 94305, USA
| | - Rosemarie H DeKruyff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, 94305, USA
| | - Kari C Nadeau
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
46
|
Wang X, Lu X, Ma C, Ma L, Han S. Combination of TLR agonist and miR146a mimics attenuates ovalbumin-induced asthma. Mol Med 2020; 26:65. [PMID: 32600285 PMCID: PMC7325265 DOI: 10.1186/s10020-020-00191-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Background microRNA-146a has been reported to be a regulator in the process of attenuating asthma by inhibiting Toll-like receptor 2 (TLR2) pathway. This study aimed to investigate how miR146a-inhibitor affect the symptom of asthma and the underlying mechanisms. Methods Ovalbumin (OVA)-induced allergic asthma mice model was established by intraperitoneal injection with 20 μg of OVA. Total cells and differential inflammatory cells in bronchoalveolar lavage fluid were counted by flow cytometry. The expression levels of molecules and cytokines in TLR2 signaling pathway were detected by Q-PCR and ELISA. Results miR146a-inhibitor attenuated OVA-induced allergic asthma by increasing Th1 cytokines in OVA-induced allergic asthma model, and the treatment of miR146a-inhibitor can reduce the inflammation caused by asthma, followed by the down-regulation of IL-5 and IL-13 in sorted ILC2. The inhibition of miR-146a significantly reduced symptoms of asthma model with TLR2-related molecules being up-regulated. Conclusion It was found that miR-146a is an important regulator in OVA-induced allergic asthma model, which can relieve symptoms of asthma through regulating TLR2 pathway. These findings provide a theoretical basis for solving asthma in clinical treatment.
Collapse
Affiliation(s)
- Xinhua Wang
- Department of Respiratory Medicine, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, No.68 Zhongshan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Xiaoxian Lu
- Department of Critical Care Medicine, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, No.68 Zhongshan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Chenhui Ma
- Department of Respiratory Medicine, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, No.68 Zhongshan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Lihong Ma
- Department of Respiratory Medicine, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, No.68 Zhongshan Road, Liangxi District, Wuxi, 214002, Jiangsu, China
| | - Shuguang Han
- Department of Respiratory Medicine, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, No.68 Zhongshan Road, Liangxi District, Wuxi, 214002, Jiangsu, China.
| |
Collapse
|
47
|
Komatsu‐Fujii T, Nonoyama S, Ogawa M, Fukumoto T, Tanabe H. Rapid effects of dupilumab treatment on papuloerythroderma of Ofuji. J Eur Acad Dermatol Venereol 2020; 34:e739-e741. [DOI: 10.1111/jdv.16581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 11/30/2022]
Affiliation(s)
- T. Komatsu‐Fujii
- Department of Dermatology Tenri Hospital Tenri Japan
- Department of Dermatology Kyoto University Graduate School of Medicine Kyoto Japan
| | - S. Nonoyama
- Department of Dermatology Tenri Hospital Tenri Japan
| | - M. Ogawa
- Department of Dermatology Tenri Hospital Tenri Japan
| | - T. Fukumoto
- Department of Dermatology Tenri Hospital Tenri Japan
| | - H. Tanabe
- Department of Dermatology Tenri Hospital Tenri Japan
| |
Collapse
|
48
|
Zataria multiflora affects clinical symptoms, oxidative stress and cytokines in asthmatic patient: A randomized, double blind, placebo-controlled, phase II clinical trial. Cytokine 2020; 133:155169. [PMID: 32585581 DOI: 10.1016/j.cyto.2020.155169] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Z. multiflora effect on clinical symptoms, pulmonary function tests (PFT), oxidative stress and cytokine levels in asthmatic patients were evaluated. METHODS 36 asthmatic patients were divided to; placebo group (P), two groups treated with Z. multiflora extract (5 and 10 mg/kg/day, as Z5 and Z10, respectively), (n = 12 in each group). Medications were administered three times a day for two months and several parameters were evaluated before treatment (step 0), one (step 1) and two months (step 2) after treatment. RESULTS Clinical symptoms and PFTs were significantly improved in Z5 and Z10 groups in steps 1 and 2 compared to step 0 (p < 0.05 to p < 0.001). Improvement of oxidative stress, cytokines levels and their gene expression after treatment with both doses of extract were observed in step 2 compared to step 0 (p < 0.05 to p < 0.001). CONCLUSION These results indicated therapeutic value of Z. multiflora for the management of asthma.
Collapse
|
49
|
Cui J, Dong M, Yi L, Wei Y, Tang W, Zhu X, Dong J, Wang W. Acupuncture inhibited airway inflammation and group 2 innate lymphoid cells in the lung in an ovalbumin-induced murine asthma model. Acupunct Med 2020; 39:217-225. [PMID: 32539427 DOI: 10.1177/0964528420924033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are known to serve important functions in the pathogenesis of allergic airway inflammation. Studies have shown that acupuncture has an anti-inflammatory effect in the airways. However, how acupuncture treatment affects innate immunity, especially with regard to the function of ILC2s in ovalbumin (OVA)-induced allergic airway inflammation, is poorly understood. METHODS BALB/c mice were injected and subsequently challenged with OVA ± treated with manual acupuncture. At the end of the experimental course, lung function was assessed by measurement of airway resistance (RL) and lung dynamic compliance (Cdyn). Cytokine levels were detected by enzyme-linked immunosorbent assay (ELISA). ILC2 proportions in the lung were analyzed by flow cytometry. RESULTS The results showed that airway inflammation and mucus secretion were significantly suppressed by acupuncture treatment. RL decreased while Cdyn increased after acupuncture treatment. There was an apparent decrease in the bronchoalveolar lavage fluid (BALF) concentrations of interleukin (IL)-5, IL-13, IL-9, IL-25 and IL-33 and an increase in soluble IL-33 receptor (sST2) levels compared with untreated asthmatic mice. Acupuncture also reduced the lin-CD45+KLRG1+ST2+ cell proportion in the lung. CONCLUSION In conclusion, this study has demonstrated that acupuncture treatment alleviates allergic airway inflammation and inhibits pulmonary ILC2 influx and IL-5, IL-9 and IL-13 production. The inhibition of ILC2s by acupuncture may be associated with the IL-33/ST2-signaling pathway and IL-25 levels, thereby offering protection from the respiratory inflammation associated with asthma.
Collapse
Affiliation(s)
- Jie Cui
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ming Dong
- Gumei Community Health Center of Minhang District of Shanghai, Shanghai, China
| | - La Yi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Wenqian Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Maruthamuthu V, Henry LJK, Ramar MK, Kandasamy R. Myxopyrum serratulum ameliorates airway inflammation in LPS-stimulated RAW 264.7 macrophages and OVA-induced murine model of allergic asthma. JOURNAL OF ETHNOPHARMACOLOGY 2020; 255:112369. [PMID: 31683035 DOI: 10.1016/j.jep.2019.112369] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/12/2019] [Accepted: 10/29/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myxopyrum serratulum A. W. Hill. (Oleaceae) is a traditionally used Indian medicinal plant for the treatment of cough, asthma and many other inflammatory diseases. AIM OF THE STUDY In this study, the protective effects of M. serratulum on airway inflammation was investigated in ovalbumin (OVA)-induced murine model of allergic asthma and lipopolysaccharide (LPS)-stimulated inflammation in RAW 264.7 murine macrophages, and the possible mechanisms were elucidated. MATERIALS AND METHODS The phytochemicals present in the methanolic leaf extract of M. serratulum (MEMS) were identified by reverse phase high performance liquid chromatography (RP-HPLC) analysis. In vitro anti-inflammatory activity of MEMS were evaluated by estimating the levels of nitric oxide (NO), reactive oxygen species (ROS) and cytokines (IL-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12, IL-17A, IFN-γ, TNF-α, G-CSF and GM-CSF) in LPS-stimulated RAW 264.7 macrophages. In vivo anti-asthmatic activity of MEMS was studied using OVA-induced murine model. Airway hyperresponsiveness (AHR), was measured; total and differential cell counts, eosinophil peroxidase (EPO), prostaglandin E2 (PGE2), NO, ROS, and cytokines (IL-4, IL-5 and IL-13), were estimated in bronchoalveolar lavage fluid (BALF). Serum total IgE level was measured; and the histopathological changes of lung tissues were observed. The expressions of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in lung tissue homogenates were detected by Western blot. RESULTS The chromatographic analysis of MEMS identified the presence of gallic acid, protocatechuic acid, catechin, ellagic acid, rutin, p-coumaric acid, quercetin, naringenin and apigenin. MEMS (125 and 250 μg/mL) dose-dependently reduced the levels of NO, ROS and pro-inflammatory cytokines in LPS-stimulated RAW 264.7 macrophages. MEMS (200 and 400 mg/kg, p.o.) significantly (p < 0.05) alleviated AHR; number of inflammatory cells, EPO, PGE2, NO, ROS, and cytokines (IL-4, IL-5 and IL-13) in BALF; serum total IgE and the histopathological changes associated with lung inflammation. Western blot studies showed that MEMS substantially suppressed COX-2 and iNOS protein expressions in the lung tissues of OVA-sensitized/challenged mice. CONCLUSIONS The present study corroborates for the first time the ameliorative effects of MEMS on airway inflammation by reducing the levels of oxidative stress, pro-inflammatory cytokines and inhibiting COX-2, iNOS protein expressions, thereby validating the ethnopharmacological uses of M. serratulum.
Collapse
Affiliation(s)
- Vijayalakshmi Maruthamuthu
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Linda Jeeva Kumari Henry
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India; National Facility for Drug Development (NFDD) for Academia, Pharmaceutical and Allied Industries, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Mohan Kumar Ramar
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India; National Facility for Drug Development (NFDD) for Academia, Pharmaceutical and Allied Industries, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India
| | - Ruckmani Kandasamy
- Department of Pharmaceutical Technology, Centre for Excellence in Nanobio Translational Research, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India; National Facility for Drug Development (NFDD) for Academia, Pharmaceutical and Allied Industries, Bharathidasan Institute of Technology, Anna University, Tiruchirappalli, 620024, Tamil Nadu, India.
| |
Collapse
|