1
|
Zhang C, Luo Z, Ji L. Identification of potential diagnostic markers and molecular mechanisms of asthma and ulcerative colitis based on bioinformatics and machine learning. Front Mol Biosci 2025; 12:1554304. [PMID: 40443529 PMCID: PMC12119298 DOI: 10.3389/fmolb.2025.1554304] [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: 01/01/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Backgrounds Asthma and ulcerative colitis (UC) are chronic inflammatory diseases linked through the "gut-lung axis," but their shared mechanisms remain unclear. This study aims to identify common biomarkers and pathways between asthma and UC using bioinformatics. Methods Gene expression data for asthma and UC were retrieved from the GEO database, and differentially expressed genes (DEGs) were analyzed. Weighted Gene Coexpression Network Analysis (WGCNA) identified UC-associated gene modules. Shared genes between asthma and UC were derived by intersecting DEGs with UC-associated modules, followed by functional enrichment and protein-protein interaction (PPI) analysis. Machine learning identified hub genes, validated through external datasets using ROC curves, nomograms, and boxplots. Gene Set Enrichment Analysis (GSEA) explored pathway alterations, while immune infiltration patterns were analyzed using the CIBERSORT algorithm. Molecular docking (MD) was performed to predict therapeutic compounds, followed by molecular dynamics simulations on the top-ranked docked complex to assess its binding stability. Results A total of 41 shared genes were identified, linked to inflammatory and immune pathways, including TNF, IL-17, and chemokine signaling. Four key hub genes-NOS2, TCN1, CHI3L1, and TIMP1-were validated as diagnostic biomarkers. Immune infiltration analysis showed strong correlations with multiple immune cells. Molecular docking identified several potential therapeutic compounds, with PD 98059, beclomethasone, and isoproterenol validated as promising candidates. The stability of the TIMP1-Beclomethasone complex was determined through molecular dynamics simulations. Conclusion This study highlights NOS2, TCN1, CHI3L1, and TIMP1 as potential biomarkers and therapeutic targets for asthma and UC, providing insights into shared mechanisms and new strategies for diagnosis and treatment.
Collapse
Affiliation(s)
- Chenxuyu Zhang
- Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zheng Luo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liang Ji
- Mianyang Hospital of Traditional Chinese Medicine, Mianyang, China
| |
Collapse
|
2
|
Sun B, Lin J. Kv1.3 expression on CD4 (+) T cells promotes interleukin-17A-associated airway inflammation and airway remodeling in asthma. Transl Res 2025; 279:40-54. [PMID: 40221022 DOI: 10.1016/j.trsl.2025.04.001] [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: 10/28/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Different types of T helper cells play an important role in disease severity and treatment response in patients with asthma. The potassium channel Kv1.3 is a type of potentially therapeutic target in T-cell-mediated inflammatory diseases. OBJECTIVE This study aimed to explore the potential of Kv1.3 as a therapeutic target for asthma and to assess the efficacy of the Kv1.3 inhibitor PAP-1 in the treatment of asthma. METHODS Kv1.3 expression on CD4+T cells was determined using data from public databases. CD4+T cells were isolated from peripheral blood samples obtained from healthy individuals and patients with asthma. The mouse models of OVA-induced asthma and Kv1.3 knockout were established. The underlying mechanism was investigated using mouse splenic CD4+T cells and BEAS-2B cells. OVA-induced asthmatic mice were treated with the Kv1.3 selective blocker PAP-1. RESULTS Based on public data, we determined the distribution of Kv1.3 on CD4+T cells, its up-regulation in asthma, and its correlation with Th17/Treg balance. Upregulation of Kv1.3 in CD4+T cells was associated with enhanced activation of these cells and airway inflammation in patients and mice with asthma, accompanied by increased IL-17A levels in alveolar lavage fluid. Conversely, Kv1.3 deficiency significantly attenuated airway inflammation, lowered IL-17A levels in bronchoalveolar lavage fluid, and inhibited airway epithelial-mesenchymal transition in asthmatic mice. Furthermore, treatment with the Kv1.3 selective blocker PAP-1 attenuated inflammation in lung tissues and prevented airway remodeling in OVA-induced asthmatic mice. CONCLUSIONS Kv1.3 expression on CD4+ T cells was correlated with IL-17A-associated airway inflammation and remodeling in asthma, which may be regarded as a potential diagnostic marker and therapeutic target for asthma. TRANSLATIONAL SIGNIFICANCE Based on our study, Kv1.3 expression on CD4+T cells was correlated with IL-17A-associated airway inflammation and remodeling in asthma, which may be regarded as a potential diagnostic marker and therapeutic target for asthma. The treatment with the Kv1.3 selective blocker PAP-1 attenuated inflammation in lung tissues and prevented airway remodeling in OVA-induced asthmatic mice. Our discoveries offer novel perspectives for a better understanding of IL-17A-associated airway remodeling in asthma. The development of drugs targeting Kv1.3 holds application value for IL-17A-associated asthma.
Collapse
Affiliation(s)
- Bingqing Sun
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiangtao Lin
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
3
|
Chen Z, Shang Y, Yuan Y, Ji X, Gong S, Zeng Q, Xiang X. Aged mice-derived bronchial epithelial cells regulate Th17 cell differentiation in asthma via the MBD2-sICOSL axis. Cell Immunol 2025; 411-412:104954. [PMID: 40252480 DOI: 10.1016/j.cellimm.2025.104954] [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: 01/01/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Th17 cells are involved in the pathogenesis of elderly asthma. Bronchial epithelial cells (BECs) can act as antigen-presenting cells, and our previous studies have shown that methyl-CPG binding domain protein 2 (MBD2) in BECs can promote Th17 cell differentiation in asthma. However, the effect of BECs from different age groups (young and old) on Th17 cells remains unclear. In this study, BECs were co-cultured with CD4+ T cells, and it was found that BECs from young mice promoted the biased differentiation of Th2 cells, while BECs from older mice facilitated the biased differentiation of Th17 cells. Interestingly, MBD2 was highly expressed in BECs from older mice compared to BECs from young mice. MBD2 silencing induced inhibition of Th17 cell differentiation, while MBD2 overexpression reversed this change and promoted Th cell differentiation into Th17 cells. Soluble inducible T cell costimulator ligand (sICOSL) is mainly involved in the regulation of T cells after activation. In this study, we found that sICOSL levels were lower in BECs of old mice compared to BECs of young mice. Mechanistically, sICOSL levels increased with MBD2 silencing and decreased with MBD2 overexpression. As expected, the addition of anti-sICOSL antibodies significantly enhanced Th17 cell differentiation and suppressed Th2 cell differentiation, while exogenous sICOSL supplementation promoted Th2 cell differentiation and inhibited Th17 cell differentiation. However, neither anti-sICOSL nor exogenous sICOSL affected the expression of MBD2. Taken together, these results suggest that BECs from older mice regulate Th17 cell differentiation via the MBD2-sICOSL axis. These findings provide new insights into the pathogenesis of Th17-activated asthma in elderly patients.
Collapse
Affiliation(s)
- Zhifeng Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Yulin Shang
- Ophthalmology and Otorhinolaryngology, Zigui County Traditional Chinese Medicine Hospital, 30 Pinghu Avenue, Zigui, Hubei 443600, China
| | - Yu Yuan
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Xiaoying Ji
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guizhou Medical University, 28 Guiyi Street, Guiyang, Guizhou 550004, China
| | - Subo Gong
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China
| | - Qingping Zeng
- Department of Respiratory and Critical Care Medicine, Longshan County People's Hospital, 50 Yuelu Avenue, Longshan, Hunan 416800, China.
| | - Xudong Xiang
- Department of Emergency, The Second Xiangya Hospital, Central South University, 139 Middle Renmin Road, Changsha, Hunan 410011, China.
| |
Collapse
|
4
|
Xu C, Liu M, Xie X, Li Z, Zhu Y, Ye Y, Du M, Hu S, Liu T, Guo Y, Wen W, Liu H, Tu Z. Multifunctional Boron-based 2D Nanoplatforms Ameliorate Severe Respiratory Inflammation by Targeting Multiple Inflammatory Mediators. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412626. [PMID: 39950864 PMCID: PMC11967860 DOI: 10.1002/advs.202412626] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/04/2025] [Indexed: 04/05/2025]
Abstract
Effective management of serious respiratory diseases, such as asthma and recalcitrant rhinitis, remains a global challenge. Here, it is shown that induced sputum supernatants (ISS) from patients with asthma contain higher levels of cell-free DNA (cfDNA) compared to that of healthy volunteers. Although cfDNA scavenging strategies have been developed for inflammation modulation in previous studies, this fall short in clinical settings due to the excessive neutrophil extracellular trap (NET) formation, reactive oxygen and nitrogen species (RONS) and bacterial infections in injured airway tissues. Based on this, a multifunctional boron-based 2D nanoplatform B-PM is designed by coating boron nanosheets (B-NS) with polyamidoamine generation 1 (PG1) dendrimer, which can simultaneously target cfDNA, NETs, RONS, and bacteria. The effects of B-PM in promoting mucosal repair, reducing airway inflammation, and mucus production have been demonstrated in model mice, and the therapeutic effect is superior to dexamethasone. Furthermore, flow cytometry with clustering analysis and transcriptome analysis with RNA-sequencing are adopted to comprehensively evaluate the in vivo anti-inflammation therapeutic effects. These findings emphasize the significance of a multi-targeting strategy to modulate dysregulated inflammation and highlight multifunctional boron-based 2D nanoplatforms for the amelioration of respiratory inflammatory diseases.
Collapse
Affiliation(s)
- Changyi Xu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Ming Liu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Xinran Xie
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Zhixin Li
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Yuefei Zhu
- Department of Biomedical EngineeringColumbia UniversityNew York10027USA
| | - Yang Ye
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Mengya Du
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Suhua Hu
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Tianrun Liu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Yubiao Guo
- Department of Pulmonary and Critical Care MedicineThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510655China
| | - Weiping Wen
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Department of OtolaryngologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Huanliang Liu
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Zhaoxu Tu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| |
Collapse
|
5
|
Quach C, Li X, Shafiei-Jahani P, Li M, Shen S, Helou DG, Hurrell BP, Soroosh P, Akbari O. BTLA agonist attenuates Th17-driven inflammation in a mouse model of steroid-resistant asthma. Front Immunol 2025; 16:1552394. [PMID: 40226621 PMCID: PMC11986467 DOI: 10.3389/fimmu.2025.1552394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/05/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Steroid-resistant asthma does not respond adequately to corticosteroid treatment. The underlying mechanisms driving corticosteroid resistance remain poorly understood, partly due to the absence of suitable animal models. Identifying the immunomodulatory pathways and mechanisms driving steroid resistance is crucial for developing effective therapies. Methods In this study, we screened 58 murine strains exposed to house dust mite and identified that the BXD75 strain exhibited neutrophil-skewed, steroid-resistant asthma and elevated Th17 cells. RNA sequencing of lung CD4+ T cells from BXD75 was performed to identify immunomodulatory pathways involved in steroid-resistance. The effects of BTLA agonist treatment were assessed on airway hyperreactivity and lung inflammation. Results Transcriptomic analysis revealed increased HVEM expression and decreased BTLA expression, both critical immune regulators associated with stimulatory and inhibitory signaling, respectively. These T cells demonstrated enhanced inflammatory signaling through both canonical and non-canonical NF-κB pathways. BTLA agonist treatment in vivo reduced airway hyperreactivity and lung inflammation, while ex vivo treatment of Th17 cells induced inhibitory signaling via SHP-1, suppressed NF-κB signaling, reduced cell numbers, and lowered IL-17 levels. Discussion Our findings establish BXD75 mice as a model for steroid-resistant asthma and demonstrate that BTLA agonism attenuates airway hyperreactivity and lung inflammation, highlighting it as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Christine Quach
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Meng Li
- University of Southern California, Libraries Bioinformatics Service, University of Southern California, Los Angeles, CA, United States
| | - Stephen Shen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Benjamin P. Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Pejman Soroosh
- Janssen Research and Development, San Diego, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
6
|
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] [Download PDF] [Figures] [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
|
7
|
Thompson DA, Wabara YB, Duran S, Reichenbach A, Chen L, Collado K, Yon C, Greally DMed JM, Rastogi D. Single cell analysis identifies distinct CD4 + T cells associated with the pathobiology of pediatric obesity related asthma. Sci Rep 2025; 15:6844. [PMID: 40000680 PMCID: PMC11861978 DOI: 10.1038/s41598-025-88423-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Pediatric obesity-related asthma is characterized by non-atopic T helper 1 (Th1) inflammation and steroid resistance. CDC42 upregulation in CD4 + T cells underlies Th1 inflammation but the CD4 + T cell subtype(s) with CDC42 upregulation and their contribution to steroid resistance are not known. Compared to healthy-weight asthma, obesity-alone and healthy-weight controls, single-cell transcriptomics of obese asthma CD4 + T cells revealed CDC42 upregulation in 3 clusters comprised of naïve and central memory T cells, which differed from the cluster enriched for Th1 responses that was comprised of effector T cells. NR3C1, coding for the glucocorticoid receptor, was downregulated, while genes coding for NLRP3 inflammasome were upregulated, in clusters with CDC42 upregulation and Th1 responses. Conserved genes in these clusters correlated with pulmonary function deficits in obese asthma. These findings suggest that several distinct CD4 + T cell subtypes are programmed in obese asthma for CDC42 upregulation, Th1 inflammation, and steroid resistance, and together contribute to the obese asthma phenotype.
Collapse
Affiliation(s)
- David A Thompson
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yvonne B Wabara
- Children's National Hospital, George Washington University, Washington, DC, USA
| | - Sarai Duran
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Anna Reichenbach
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Laura Chen
- Department of Pediatrics, Yale University, New Haven, CT, USA
| | - Kayla Collado
- Montefiore Health System, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Changsuek Yon
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John M Greally DMed
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore Health System, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deepa Rastogi
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA.
- Children's Hospital at Montefiore Albert Einstein College of Medicine, 3415 Bainbridge Ave, Bronx, NY, 10467, USA.
| |
Collapse
|
8
|
Al-Beltagi M, Bediwy AS, Saeed NK, Bediwy HA, Elbeltagi R. Diabetes-inducing effects of bronchial asthma. World J Diabetes 2025; 16:97954. [PMID: 39817208 PMCID: PMC11718464 DOI: 10.4239/wjd.v16.i1.97954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/12/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The relationship between diabetes mellitus (DM) and asthma is complex and can impact disease trajectories. AIM To explore the bidirectional influences between the two conditions on clinical outcomes and disease control. METHODS We systematically reviewed the literature on the relationship between DM and asthma, focusing on their impacts, mechanisms, and therapeutic implications. Various studies were assessed, which investigated the effect of glycemic control on asthma outcomes, lung function, and exacerbations. The study highlighted the role of specific diabetes medications in managing asthma. RESULTS The results showed that poor glycemic control in diabetes can exacerbate asthma, increase hospitalizations, and reduce lung function. Conversely, severe asthma, especially in obese individuals, can complicate diabetes management and make glycemic control more difficult. The diabetes-associated mechanisms, such as inflammation, microangiopathy, and oxidative stress, can exacerbate asthma and decrease lung function. Some diabetes medications exhibit anti-inflammatory effects that show promise in mitigating asthma exacerbations. CONCLUSION The complex interrelationship between diabetes and asthma suggests bidirectional influences that affect disease course and outcomes. Inflammation and microvascular complications associated with diabetes may worsen asthma outcomes, while asthma severity, especially in obese individuals, complicates diabetes control. However, the current research has limitations, and more diverse longitudinal studies are required to establish causal relationships and identify effective treatment strategies for individuals with both conditions.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Manama, Bahrain
| | - Adel Salah Bediwy
- Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
- Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Manama, Bahrain
| | - Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Kingdom of Bahrain, Manama 26671, Manama, Bahrain
- Medical Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Busaiteen 15503, Muharraq, Bahrain
| | | | - Reem Elbeltagi
- Department of Medicine, The Royal College of Surgeons in Ireland-Bahrain, Busiateen 15503, Muharraq, Bahrain
| |
Collapse
|
9
|
Ju X, Fard NE, Bhalla A, Dvorkin-Gheva A, Xiao M, Radford K, Zhang K, Ditta R, Oliveria JP, Paré G, Mukherjee M, Nair P, Sehmi R. A population of c-kit + IL-17A + ILC2s in sputum from individuals with severe asthma supports ILC2 to ILC3 trans-differentiation. Sci Transl Med 2025; 17:eado6649. [PMID: 39813318 DOI: 10.1126/scitranslmed.ado6649] [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: 02/14/2024] [Revised: 07/29/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
In prednisone-dependent severe asthma, uncontrolled sputum eosinophilia is associated with increased numbers of group 2 innate lymphoid cells (ILC2s). These cells represent a relatively steroid-insensitive source of interleukin-5 (IL-5) and IL-13 and are considered critical drivers of asthma pathology. The abundance of ILC subgroups in severe asthma with neutrophilic or mixed granulocytic (both eosinophilic and neutrophilic) airway inflammation, prone to recurrent infective exacerbations, remains unclear. Here, we found by flow cytometry that sputum ILC3s are increased in severe asthma with intense airway neutrophilia, whereas equivalently raised sputum ILC2s and ILC3s were found in severe asthma with mixed granulocytic inflammation. Unbiased clustering analyses identified an "intermediate-ILC2" population displaying markers of both ILC2s (prostaglandin D2 receptor 2; CRTH2, IL-5, and IL-13) and ILC3s (c-kit and IL-17A) that were most abundant in severe asthma with mixed granulocytic airway inflammation. Intermediate ILC2s correlated with airway neutrophilia and were associated with increased amounts of IL-1β and IL-18 in sputum supernatants. Coculture of sort-purified canonical ILC2s with IL-1β and IL-18 in vitro up-regulated c-kit and IL-17A as well as gene expression profiles related to both type 2 and type 17 inflammatory pathways. Together, we have identified an intermediate-ILC2 phenotype in the airways of individuals with severe mixed granulocytic asthma, representing a candidate therapeutic target for controlling neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Xiaotian Ju
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Nahal Emami Fard
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Anurag Bhalla
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Anna Dvorkin-Gheva
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Maria Xiao
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Katherine Radford
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Kayla Zhang
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Reina Ditta
- Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Clinical Research Laboratory and Biobank and the Genetic and Molecular Epidemiology Laboratory (CRLB-GMEL), Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada
| | - John Paul Oliveria
- Department of Biomarker Development, Genentech Inc., South San Francisco, CA 94080, USA
| | - Guillaume Paré
- Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Clinical Research Laboratory and Biobank and the Genetic and Molecular Epidemiology Laboratory (CRLB-GMEL), Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, Hamilton, ON L8L 2X2, Canada
| | - Manali Mukherjee
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| | - Roma Sehmi
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON L8N 4A6, Canada
| |
Collapse
|
10
|
Dera N, Kosińska-Kaczyńska K, Żeber-Lubecka N, Brawura-Biskupski-Samaha R, Massalska D, Szymusik I, Dera K, Ciebiera M. Impact of Early-Life Microbiota on Immune System Development and Allergic Disorders. Biomedicines 2025; 13:121. [PMID: 39857705 PMCID: PMC11762082 DOI: 10.3390/biomedicines13010121] [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: 12/03/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Introduction: The shaping of the human intestinal microbiota starts during the intrauterine period and continues through the subsequent stages of extrauterine life. The microbiota plays a significant role in the predisposition and development of immune diseases, as well as various inflammatory processes. Importantly, the proper colonization of the fetal digestive system is influenced by maternal microbiota, the method of pregnancy completion and the further formation of the microbiota. In the subsequent stages of a child's life, breastfeeding, diet and the use of antibiotics influence the state of eubiosis, which determines proper growth and development from the neonatal period to adulthood. The literature data suggest that there is evidence to confirm that the intestinal microbiota of the infant plays an important role in regulating the immune response associated with the development of allergic diseases. However, the identification of specific bacterial species in relation to specific types of reactions in allergic diseases is the basic problem. Background: The main aim of the review was to demonstrate the influence of the microbiota of the mother, fetus and newborn on the functioning of the immune system in the context of allergies and asthma. Methods: We reviewed and thoroughly analyzed the content of over 1000 articles and abstracts between the beginning of June and the end of August 2024. Over 150 articles were selected for the detailed study. Results: The selection was based on the PubMed National Library of Medicine search engine, using selected keywords: "the impact of intestinal microbiota on the development of immune diseases and asthma", "intestinal microbiota and allergic diseases", "the impact of intrauterine microbiota on the development of asthma", "intrauterine microbiota and immune diseases", "intrauterine microbiota and atopic dermatitis", "intrauterine microbiota and food allergies", "maternal microbiota", "fetal microbiota" and "neonatal microbiota". The above relationships constituted the main criteria for including articles in the analysis. Conclusions: In the present review, we showed a relationship between the proper maternal microbiota and the normal functioning of the fetal and neonatal immune system. The state of eubiosis with an adequate amount and diversity of microbiota is essential in preventing the development of immune and allergic diseases. The way the microbiota is shaped, resulting from the health-promoting behavior of pregnant women, the rational conduct of the medical staff and the proper performance of the diagnostic and therapeutic process, is necessary to maintain the health of the mother and the child. Therefore, an appropriate lifestyle, rational antibiotic therapy as well as the way of completing the pregnancy are indispensable in the prevention of the above conditions. At the same time, considering the intestinal microbiota of the newborn in relation to the genera and phyla of bacteria that have a potentially protective effect, it is worth noting that the use of suitable probiotics and prebiotics seems to contribute to the protective effect.
Collapse
Affiliation(s)
- Norbert Dera
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
| | - Katarzyna Kosińska-Kaczyńska
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, 02-781 Warsaw, Poland;
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Robert Brawura-Biskupski-Samaha
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Diana Massalska
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| | - Iwona Szymusik
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, 01-809 Warsaw, Poland; (N.D.); (K.K.-K.); (R.B.-B.-S.); (I.S.)
| | - Kacper Dera
- Pediatric Ward, Department of Pediatrics, Center of Postgraduate Medical Education, Bielański Hospital, 01-809 Warsaw, Poland
| | - Michał Ciebiera
- Warsaw Institute of Women’s Health, 00-189 Warsaw, Poland; (D.M.); (M.C.)
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, 00-189 Warsaw, Poland
| |
Collapse
|
11
|
Berkinbayeva M, Gu W, Chen Z, Gao P. Group 3 Innate Lymphoid Cells: A Potential Therapeutic Target for Steroid Resistant Asthma. Clin Rev Allergy Immunol 2024; 68:1. [PMID: 39751959 PMCID: PMC11698894 DOI: 10.1007/s12016-024-09012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Asthma is a chronic airway inflammatory disease that affects millions globally. Although glucocorticoids are a mainstay of asthma treatment, a subset of patients show resistance to these therapies, resulting in poor disease control and increased morbidity. The complex mechanisms underlying steroid-resistant asthma (SRA) involve Th1 and Th17 lymphocyte activity, neutrophil recruitment, and NLRP3 inflammasome activation. Recent studies provided evidence that innate lymphoid cells type 3 (ILC3s) might be potential therapeutic targets for non-eosinophilic asthma (NEA) and SRA. Like Th17 cells, ILC3s play crucial roles in immune responses, inflammation, and tissue homeostasis, contributing to disease severity and corticosteroid resistance in NEA. Biologics targeting ILC3-related pathways have shown promise in managing Th2-low asthma, suggesting new avenues for SRA treatment. This review aims to explore the risk factors for SRA, discuss the challenges and mechanisms underlying SRA, consolidate current findings on innate lymphoid cells, and elucidate their role in respiratory conditions. We present the latest findings on the involvement of ILC3s in human diseases and explore their potential mechanisms in SRA development. Furthermore, we review emerging therapeutic biologics targeting ILC3-related pathways in managing NEA and SRA. This review highlights current challenges, and emerging therapeutic strategies, and addresses a significant gap in asthma research, with implications for improving the management of steroid-resistant asthma.
Collapse
Affiliation(s)
- Marzhan Berkinbayeva
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
| | - Wenjing Gu
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhifeng Chen
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA.
| |
Collapse
|
12
|
Panda ES, Gautam AS, Pandey SK, Singh RK. IL-17A-Induced Redox Imbalance and Inflammatory Responses in Mice Lung via Act1-TRAF6-IKBα Signaling Pathway: Implications for Lung Disease Pathogenesis. Inflammation 2024:10.1007/s10753-024-02199-9. [PMID: 39607627 DOI: 10.1007/s10753-024-02199-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
IL-17A is a potent proinflammatory cytokine that plays a crucial role in the pathogenesis of various lung diseases. This study focused on the evaluation of the role of IL-17 receptor signaling through one-week intranasal exposure of IL-17A in lung tissues of BALB/c mice. IL-17A triggered inflammatory responses in the mice lungs and led to changes in the morphological alveolar arrangements. Exposure of IL-17A induced redox imbalance by triggering an increase in the level of the pro-oxidants (reactive oxygen species, nitrite and malondialdehyde) and reduction of the levels of antioxidant proteins (glutathione, superoxide dismutase and catalase) in the lung tissue. IL-17A also caused a significant elevation in the levels of proinflammatory cytokines lines including TNF-α, IL-1β and IL-6, in lung tissue as well as in plasma. More interestingly, these changes were accompanied by the alterations in IL-17 receptor downstream signaling through activation of IL-17R-Act1-TRAF6-IKBα-mediated pathway. IL-17A exposure also caused lung tissue injury, recruitment and polarization of immune cells from anti-inflammatory to pro-inflammatory. This study clearly demonstrated the role of IL-17A-induced signaling in worsening lung inflammatory diseases, and hence points towards its emergence as an important therapeutic target to control lung inflammation.
Collapse
Affiliation(s)
- Ekta Swarnamayee Panda
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Shivam Kumar Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
13
|
Guan J, Yao W, Zhang L, Xie H, Li L, Wen Y, Chen H, Huang Y, Wen J, Ou C, Liang C, Wang J, Zhang Q, Tao A, Yan J. Contribution of Pseudomonas aeruginosa - mediated club cell necroptosis to the bias of type 17 inflammation and steroid insensitivity in asthma. J Adv Res 2024:S2090-1232(24)00475-2. [PMID: 39442871 DOI: 10.1016/j.jare.2024.10.020] [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: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
INTRODUCTION Opportunistic pathogen infection is one of the important inducements for asthma exacerbation. Pseudomonas aeruginosa (PA) is a kind of dominant pathogenic bacteria in the respiratory tract that is associated with severe asthma, but the underlying mechanisms still remains unclear. OBJECTIVES To examine the role of PA infection in the bias of the inflammatory endotype in asthma and its effect on the sensitivity to steroid therapy. METHODS An adjusted HDM (House Dust Mite) -induced asthma model with PA inoculation in the airway was utilized to mimic the process of opportunistic PA infection in asthma, focusing on the interaction between bacteria and epithelium. Dexamethasone administration in vivo was used to test the sensitivity to steroid therapy. RESULTS It was uncovered that PA could promote the loss of club cells in the necroptosis pattern through cellular CYP450 activation, leading to an imbalance of inflammatory response and steroid insensitivity. Club cell loss results in the activation of cellular E-cadherin/β-catenin axis in the rest of club cells for goblet metaplasia and mucus hypersecretion, as well as epithelial damage and GR downregulation for steroid resistance. For clinical applications, the necroptosis inhibitor Nec-1 can effectively relieve the pathological symptoms of asthma in vivo. Meanwhile, CCSP administration in the airway can regulate the pulmonary inflammation and restore the steroid sensitivity in asthma. CONCLUSION These experiments provide a novel mechanism of concurrent PA infection in asthma through club cell necroptosis and the pathological consequences. Nec-1 treatment and CCSP supplementation may be possible therapeutic strategies for asthma treatment.
Collapse
Affiliation(s)
- Jieying Guan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China; Department of Obstetrics and Gynecology, Center for Reproductive Medicine, The First People's Hospital of Zhaoqing, Guangdong province, China
| | - Wenruo Yao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Le Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Huancheng Xie
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Linmei Li
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yuhuan Wen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Honglv Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Yuyi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Junjie Wen
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changxing Ou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Canyang Liang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Jing Wang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.
| | - Jie Yan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
14
|
Li NC, Iannuzo N, Christenson SA, Langlais PR, Kraft M, Ledford JG, Li X. Investigation of lactotransferrin messenger RNA expression levels as an anti-type 2 asthma biomarker. J Allergy Clin Immunol 2024; 154:609-618. [PMID: 38797239 PMCID: PMC11380595 DOI: 10.1016/j.jaci.2024.05.013] [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: 09/12/2023] [Revised: 02/15/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Lactotransferrin (LTF) has an immunomodulatory function, and its expression levels are associated with asthma susceptibility. OBJECTIVES We sought to investigate LTF messenger RNA (mRNA) expression levels in human bronchial epithelial cells (BECs) as an anti-type 2 (T2) asthma biomarker. METHODS Association analyses between LTF mRNA expression levels in BECs and asthma-related phenotypes were performed in the Severe Asthma Research Program (SARP) cross-sectional (n = 155) and longitudinal (n = 156) cohorts using a generalized linear model. Correlation analyses of mRNA expression levels between LTF and all other genes were performed by Spearman correlation. RESULTS Low LTF mRNA expression levels were associated with asthma susceptibility and severity (P < .025), retrospective and prospective asthma exacerbations, and low lung function (P < 8.3 × 10-3). Low LTF mRNA expression levels were associated with high airway T2 inflammation biomarkers (sputum eosinophils and fractional exhaled nitric oxide; P < 8.3 × 10-3) but were not associated with blood eosinophils or total serum IgE. LTF mRNA expression levels were negatively correlated with expression levels of TH2 or asthma-associated genes (POSTN, NOS2, and MUC5AC) and eosinophil-related genes (IL1RL1, CCL26, and IKZF2) and positively correlated with expression levels of TH1 and inflammation genes (IL12A, MUC5B, and CC16) and TH17-driven cytokines or chemokines for neutrophils (CXCL1, CXCL6, and CSF3) (P < 3.5 × 10-6). CONCLUSIONS Low LTF mRNA expression levels in BECs are associated with asthma susceptibility, severity, and exacerbations through upregulation of airway T2 inflammation. LTF is a potential anti-T2 biomarker, and its expression levels may help determine the balance of eosinophilic and neutrophilic asthma.
Collapse
Affiliation(s)
- Nicholas C Li
- University of Arizona Internship, Basis Tucson North, Tucson, Ariz
| | - Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Ariz
| | - Stephanie A Christenson
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, Calif
| | - Paul R Langlais
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, Ariz
| | - Monica Kraft
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Julie G Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Ariz
| | - Xingnan Li
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Medicine, Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, Ariz.
| |
Collapse
|
15
|
Thompson DA, Wabara YB, Duran S, Reichenbach A, Chen L, Collado K, Yon C, Greally JM, Rastogi D. Single-cell analysis identifies distinct CD4+ T cells associated with the pathobiology of pediatric obesity-related asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607447. [PMID: 39211259 PMCID: PMC11361012 DOI: 10.1101/2024.08.13.607447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pediatric obesity-related asthma is characterized by non-atopic T helper 1 (Th1) inflammation and steroid resistance. CDC42 upregulation in CD4+T cells underliesTh1 inflammation but the CD4+T cell subtype(s) with CDC42 upregulation and their contribution to steroid resistance are not known. Compared to healthy-weight asthma, obesity-alone and healthy-weight controls, single-cell transcriptomics of obese asthma CD4+T cells revealed CDC42 upregulation in 3 clusters comprised of naïve and central memory T cells, which differed from the cluster enriched for Th1 responses that was comprised of effector T cells. NR3C1, coding for glucocorticoid receptor, was downregulated, while genes coding for NLRP3 inflammasome were upregulated, in clusters with CDC42 upregulation and Th1 responses. Conserved genes in these clusters correlated with pulmonary function deficits in obese asthma. These findings suggest that several distinct CD4+T cell subtypes are programmed in obese asthma for CDC42 upregulation, Th1 inflammation, and steroid resistance, and together contribute to obese asthma phenotype. Summary CD4+T cells from obese children with asthma are distinctly programmed for non-allergic immune responses, steroid resistance and inflammasome activation, that underlie the obese asthma phenotype.
Collapse
|
16
|
Qiao L, Li SM, Liu JN, Duan HL, Jiang XF. Revealing the regulation of allergic asthma airway epithelial cell inflammation by STEAP4 targeting MIF through machine learning algorithms and single-cell sequencing analysis. Front Mol Biosci 2024; 11:1427352. [PMID: 39176391 PMCID: PMC11338762 DOI: 10.3389/fmolb.2024.1427352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/24/2024] Open
Abstract
Asthma comprises one of the most common chronic inflammatory conditions, yet still lacks effective diagnostic markers and treatment targets. To gain deeper insights, we comprehensively analyzed microarray datasets of airway epithelial samples from asthmatic patients and healthy subjects in the Gene Expression Omnibus database using three machine learning algorithms. Our investigation identified a pivotal gene, STEAP4. The expression of STEAP4 in patients with allergic asthma was found to be reduced. Furthermore, it was found to negatively correlate with the severity of the disease and was subsequently validated in asthmatic mice in this study. A ROC analysis of STEAP4 showed the AUC value was greater than 0.75. Functional enrichment analysis of STEAP4 indicated a strong correlation with IL-17, steroid hormone biosynthesis, and ferroptosis signaling pathways. Subsequently, intercellular communication analysis was performed using single-cell RNA sequencing data obtained from airway epithelial cells. The results revealed that samples exhibiting low levels of STEAP4 expression had a richer MIF signaling pathway in comparison to samples with high STEAP4 expression. Through both in vitro and in vivo experiments, we further confirmed the overexpression of STEAP4 in airway epithelial cells resulted in decreased expression of MIF, which in turn caused a decrease in the levels of the cytokines IL-33, IL-25, and IL-4; In contrast, when the STEAP4 was suppressed in airway epithelial cells, there was an upregulation of MIF expression, resulting in elevated levels of the cytokines IL-33, IL-25, and IL-4. These findings suggest that STEAP4 in the airway epithelium reduces allergic asthma Th2-type inflammatory reactions by inhibiting the MIF signaling pathway.
Collapse
Affiliation(s)
- Lu Qiao
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shi-meng Li
- Department of Clinical Laboratory, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jun-nian Liu
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hong-lei Duan
- Department of Digestive, Weihai Municipal Hospital, Weihai, Shandong, China
| | - Xiao-feng Jiang
- Department of Clinical Laboratory, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
17
|
Luo J, Liu P, Luo Y. Genetic prediction of asthma increases multiple sepsis risks: A Mendelian randomization study. World Allergy Organ J 2024; 17:100937. [PMID: 39156599 PMCID: PMC11327466 DOI: 10.1016/j.waojou.2024.100937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024] Open
Abstract
Background Observational epidemiological studies have indicated a potential association between asthma and sepsis, although the causal relationship between these 2 conditions remains uncertain. To further investigate this relationship, the present study utilized Mendelian randomization (MR) analysis approach to explore the potential links between asthma and various types of sepsis. Methods In a large-scale genome-wide association study, single nucleotide polymorphisms (SNPs) associated with asthma were selected as instrumental variables. Three methods, including inverse-variance weighted (IVW), MR-Egger regression, and weighted median were used to assess the causal relationship between asthma and sepsis. The odds ratio (OR) and 95% confidence interval (CI) were used as the evaluation metrics for causal relationships, and sensitivity analysis was conducted to assess pleiotropy and instrument validity. Finally, a reverse MR analysis was conducted to investigate whether there is a causal relationship between sepsis and asthma. Results We found a positive association between asthma and an increased risk of sepsis (OR=1.18, P<0.05), streptococcal sepsis (OR=1.23, P=0.04), pneumonia-related sepsis (OR=1.57, P<0.05), pneumococcal sepsis (OR=1.58, P=0.01), other sepsis (OR=1.15, P<0.05), and sepsis in intensive care unit (ICU) settings (OR=1.23, P=0.02). Sensitivity analysis showed consistent results without heterogeneity or pleiotropy. The reverse MR analysis reveals no causal relationship between various types of sepsis and asthma. Conclusion Our study demonstrates a causal relationship between asthma and different types of sepsis. These findings suggest the importance of healthcare providers paying attention to the potential risk of sepsis in asthma patients and implementing appropriate preventive and intervention measures in a timely manner.
Collapse
Affiliation(s)
- Jihang Luo
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Puyu Liu
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yawen Luo
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
18
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Campen M, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances Th17 Responses and Promotes Airway Neutrophilia. Am J Respir Cell Mol Biol 2024; 71:169-181. [PMID: 38593442 PMCID: PMC11299091 DOI: 10.1165/rcmb.2023-0424oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024] Open
Abstract
Heightened unfolded protein responses (UPRs) are associated with the risk for asthma, including severe asthma. Treatment-refractory severe asthma manifests a neutrophilic phenotype with T helper (Th)17 responses. However, how UPRs participate in the deregulation of Th17 cells leading to neutrophilic asthma remains elusive. This study found that the UPR sensor IRE1 is induced in the murine lung with fungal asthma and is highly expressed in Th17 cells relative to naive CD4+ T cells. Cytokine (e.g., IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by both human and mouse Th17 cells. Ern1 (encoding IRE1) deficiency decreases the expression of endoplasmic reticulum stress factors and impairs the differentiation and cytokine secretion of Th17 cells. Genetic ablation of Ern1 leads to alleviated Th17 responses and airway neutrophilia in a fungal airway inflammation model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances Th17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPR-mediated secretory function of Th17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in Th17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology and
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | | - Ruoning Wang
- Department of Molecular Genetics and Microbiology and
| | | | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas; and
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology and
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | | | - Meilian Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, and
| | | |
Collapse
|
19
|
Kuramoto K, Morishima Y, Yoshida K, Ano S, Kawashima K, Yabuuchi Y, Sakai C, Matsumura S, Nishino K, Yazaki K, Matsuyama M, Kiwamoto T, Ishii Y, Hizawa N. Nrf2 Deficiency Accelerates IL-17-Dependent Neutrophilic Airway Inflammation in Asthmatic Mice. Antioxidants (Basel) 2024; 13:818. [PMID: 39061887 PMCID: PMC11274244 DOI: 10.3390/antiox13070818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Asthma is a heterogeneous disease that can be broadly classified into type 2, which is primarily steroid-sensitive and eosinophilic, and non-type 2, which is primarily steroid-resistant and neutrophilic. While the mechanisms leading to the development of molecular-targeted therapies for type 2 asthma are being elucidated, much remains to be learned about non-type 2 asthma. To investigate the role of oxidative stress in refractory allergic airway inflammation, we compared asthma models generated by immunizing wild-type and nuclear factor erythroid-2-related factor 2 (Nrf2)-deficient mice with the house dust mite antigen. Both asthma models had similar levels of airway inflammation and hyperresponsiveness, but the Nrf2-deficient mice had increased oxidative stress and exacerbated neutrophilic airway inflammation compared with the wild-type mice. Type 2 cytokines and the expression of GATA3, a transcription factor that is important for Th2 cell differentiation, had decreased in Nrf2-deficient mice compared with the wild-type mice, whereas helper T (Th) 17 cytokines and the expression of RORγt, which is important for Th17 cell differentiation, had increased. Furthermore, the neutrophilic airway inflammation caused by Nrf2 deficiency was ameliorated by interleukin (IL)-17 neutralization. We have concluded that the disruption of the Nrf2-mediated antioxidant defense system contributed to the induction of Th17 differentiation and exacerbated allergic neutrophilic airway inflammation.
Collapse
Affiliation(s)
| | - Yuko Morishima
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan (C.S.); (K.N.); (Y.I.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bowman WS, Schmidt RJ, Sanghar GK, Thompson GR, Ji H, Zeki AA, Haczku A. "Air That Once Was Breath" Part 1: Wildfire-Smoke-Induced Mechanisms of Airway Inflammation - "Climate Change, Allergy and Immunology" Special IAAI Article Collection: Collegium Internationale Allergologicum Update 2023. Int Arch Allergy Immunol 2024; 185:600-616. [PMID: 38452750 PMCID: PMC11487202 DOI: 10.1159/000536578] [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: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Wildfires are a global concern due to their wide-ranging environmental, economic, and public health impacts. Climate change contributes to an increase in the frequency and intensity of wildfires making smoke exposure a more significant and recurring health concern for individuals with airway diseases. Some of the most prominent effects of wildfire smoke exposure are asthma exacerbations and allergic airway sensitization. Likely due to the delayed recognition of its health impacts in comparison with cigarette smoke and industrial or traffic-related air pollution, research on the composition, the mechanisms of toxicity, and the cellular/molecular pathways involved is poor or non-existent. SUMMARY This review discusses potential underlying pathological mechanisms of wildfire-smoke-related allergic airway disease and asthma. We focused on major gaps in understanding the role of wildfire smoke composition in the development of airway disease and the known and potential mechanisms involving cellular and molecular players of oxidative injury at the epithelial barrier in airway inflammation. We examine how PM2.5, VOCs, O3, endotoxin, microbes, and toxic gases may affect oxidative stress and inflammation in the respiratory mucosal barrier. We discuss the role of AhR in mediating smoke's effects in alarmin release and IL-17A production and how glucocorticoid responsiveness may be impaired by IL-17A-induced signaling and epigenetic changes leading to steroid-resistant severe airway inflammation. KEY MESSAGE Effective mitigation of wildfire-smoke-related respiratory health effects would require comprehensive research efforts aimed at a better understanding of the immune regulatory effects of wildfire smoke in respiratory health and disease.
Collapse
Affiliation(s)
- Willis S. Bowman
- UC Davis Lung Center, University of California, Davis, CA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Sacramento, CA, USA
| | - Rebecca J. Schmidt
- Department of Public Health Sciences, School of Medicine, Sacramento, CA, USA
| | - Gursharan K. Sanghar
- UC Davis Lung Center, University of California, Davis, CA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Sacramento, CA, USA
| | - George R. Thompson
- UC Davis Lung Center, University of California, Davis, CA, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Sacramento, CA, USA
| | - Hong Ji
- UC Davis Lung Center, University of California, Davis, CA, USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, Davis, CA, USA
| | - Amir A. Zeki
- UC Davis Lung Center, University of California, Davis, CA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Sacramento, CA, USA
| | - Angela Haczku
- UC Davis Lung Center, University of California, Davis, CA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, Sacramento, CA, USA
| |
Collapse
|
21
|
Lezmi G, Poirault C, Grauso M, Dietrich C, Adel-Patient K, Leite-de-Moraes M. Identification of the major immune differences in severe asthmatic children according to their atopic dermatitis status. Cell Immunol 2024; 397-398:104815. [PMID: 38428350 DOI: 10.1016/j.cellimm.2024.104815] [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: 12/15/2023] [Revised: 02/02/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
Severe asthma (SA) affects 2% to 5% of asthmatic children. Atopic dermatitis can affect up to 34% of children with SA (cwSA). Atopic dermatitis and asthma share common genetic and immunological features. However, not all children with SA suffer from AD, and it remains unclear whether the overall immune profiles of these children are similar. In this study, seventeen cwSA (9.8 [7.1-13.2] years; seven with and ten without AD) were enrolled. Bronchoalveolar lavage (BAL) and blood samples were collected from these patients. Seventy-three cytokines/chemokines and distinct immune T cell populations were evaluated in blood and BAL. We found that BAL and blood immune profiles of cwSA with and without AD were globally similar. However, specific differences were observed, namely lower frequency of Tc2, Th17 and IL-17-producing mucosal associated invariant T (MAIT-17) cells and higher CD8/CD4 ratio and IL-22 concentrations in BAL and of CCL19 concentrations in plasma from cwSA with AD. Further, in contrast with cwSA without AD, we found a positive correlation between a set of plasma cytokines and almost all cytokines in BAL in cwSA with AD. In conclusion, this study shows the major immune differences between cwSA with and without AD in BAL and blood suggesting that distinct endotypes may be implicated in the inflammatory responses observed in these pediatric patients.
Collapse
Affiliation(s)
- Guillaume Lezmi
- Université de Paris, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Inserm UMR1151, CNRS UMR8253, F-75015, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, F-75015, Paris, France.
| | - Clément Poirault
- Université de Paris, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Inserm UMR1151, CNRS UMR8253, F-75015, Paris, France; AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, F-75015, Paris, France
| | - Marta Grauso
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Immuno-Allergie Alimentaire, F-91191, Gif-sur-Yvette, France
| | - Céline Dietrich
- Université de Paris, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Inserm UMR1151, CNRS UMR8253, F-75015, Paris, France
| | - Karine Adel-Patient
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Laboratoire d'Immuno-Allergie Alimentaire, F-91191, Gif-sur-Yvette, France
| | - Maria Leite-de-Moraes
- Université de Paris, Institut Necker Enfants Malades, Equipe Immunorégulation et Immunopathologie, Inserm UMR1151, CNRS UMR8253, F-75015, Paris, France.
| |
Collapse
|
22
|
Yan Q, Zhang X, Xie Y, Yang J, Liu C, Zhang M, Zheng W, Lin X, Huang HT, Liu X, Jiang Y, Zhan SF, Huang X. Bronchial epithelial transcriptomics and experimental validation reveal asthma severity-related neutrophilc signatures and potential treatments. Commun Biol 2024; 7:181. [PMID: 38351296 PMCID: PMC10864370 DOI: 10.1038/s42003-024-05837-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024] Open
Abstract
Airway epithelial transcriptome analysis of asthma patients with different severity was used to disentangle the immune infiltration mechanisms affecting asthma exacerbation, which may be advantageous to asthma treatment. Here we introduce various bioinformatics methods and develop two models: an OVA/CFA-induced neutrophil asthma mouse model and an LPS-induced human bronchial epithelial cell damage model. Our objective is to investigate the molecular mechanisms, potential targets, and therapeutic strategies associated with asthma severity. Multiple bioinformatics methods identify meaningful differences in the degree of neutrophil infiltration in asthma patients with different severity. Then, PTPRC, TLR2, MMP9, FCGR3B, TYROBP, CXCR1, S100A12, FPR1, CCR1 and CXCR2 are identified as the hub genes. Furthermore, the mRNA expression of 10 hub genes is determined in vivo and in vitro models. Reperixin is identified as a pivotal drug targeting CXCR1, CXCR2 and MMP9. We further test the potential efficiency of Reperixin in 16HBE cells, and conclude that Reperixin can attenuate LPS-induced cellular damage and inhibit the expression of them. In this study, we successfully identify and validate several neutrophilic signatures and targets associated with asthma severity. Notably, Reperixin displays the ability to target CXCR1, CXCR2, and MMP9, suggesting its potential therapeutic value for managing deteriorating asthma.
Collapse
Affiliation(s)
- Qian Yan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xinxin Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Yi Xie
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Yang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Chengxin Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miaofen Zhang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Wenjiang Zheng
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xueying Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui-Ting Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China.
| | - Shao-Feng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xiufang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
23
|
Kim HJ, Dinh DTT, Yang J, Herath KHINM, Seo SH, Son YO, Kang I, Jee Y. High sucrose intake exacerbates airway inflammation through pathogenic Th2 and Th17 response in ovalbumin (OVA)-induced acute allergic asthma in C57BL/6 mice. J Nutr Biochem 2024; 124:109504. [PMID: 37944673 DOI: 10.1016/j.jnutbio.2023.109504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023]
Abstract
Asthma is an inflammatory disease characterized by chronic inflammation in lung tissues and excessive mucus production. High-fat diets have long been assumed to be a potential risk factor for asthma. However, to date, very few direct evidence indicating the involvement of high sucrose intake (HSI) in asthma progression exists. In this study, we investigate the effect of HSI on ovalbumin (OVA)-sensitized allergic asthma mice. We observed that HSI increased the expression of inflammatory genes (IL-1β, IL-6, TNF-α) in adipose tissues and led to reactive oxygen species generation in the liver and lung. In addition, HSI accelerated the TLR4/NF-κB signaling pathway leading to MMP9 activation, which promotes the chemokines and TGF-β secretion in the lungs of OVA-sensitized allergic asthma mice. More importantly, HSI significantly promoted the pathogenic Th2 and Th17 responses. The increase of IL-17A secretion by HSI increased the expression of chemokines (MCP-1, CXCL1, CXCL5, CXCL8). It resulted in eosinophil and mast cell infiltration in the lung and trachea. We also demonstrated that HSI increased mucus hypersecretion, which was validated by increased main mucin protein (MUC5AC) secreted in the lungs. Our findings suggest that HSI exacerbates the development of Th2/Th17-predominant asthma by upregulating the TLR4-mediated NF-κB pathway, leading to excessive MMP9 production.
Collapse
Affiliation(s)
- Hyo Jin Kim
- Department of Food Bioengineering, Jeju National University, Jeju, Republic of Korea
| | - Duong Thi Thuy Dinh
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea
| | - Jiwon Yang
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea; Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University
| | | | - Seok Hee Seo
- Department of Food Science and Nutrition, Jeju National University, Jeju, Republic of Korea
| | - Young-Ok Son
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea; Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University
| | - Inhae Kang
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea; Department of Food Science and Nutrition, Jeju National University, Jeju, Republic of Korea.
| | - Youngheun Jee
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, Republic of Korea; Department of Veterinary Medicine and Veterinary Medical Research Institute, Jeju National University, Jeju, Republic of Korea.
| |
Collapse
|
24
|
Li J, Bao T, Cao L, Ma M, Yu B, Zhang Y, Wu R, Zhu H, Tian Z. Establishment of a juvenile mouse asthma model induced by postnatal hyperoxia exposure combined with early OVA sensitization. Heliyon 2024; 10:e23291. [PMID: 38148813 PMCID: PMC10750071 DOI: 10.1016/j.heliyon.2023.e23291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Objective To establish a juvenile mouse asthma model by postnatal hyperoxia exposure combined with early ovalbumin (OVA) sensitization. Methods Female C57BL/6J newborn mice were exposed to hyperoxia (95 % O2) from postnatal day-1 (PND1) to PND7; intraperitoneally injected with OVA suspension on PND21, PND28; and stimulated by nebulized inhalation of 1 % OVA from PND36 to PND42. Within 48 h of the last challenge, we observed their activity performance and evaluated airway responsiveness (AHR). All mice were executed at PND44. Female (n = 32) were divided into four groups as follows: room air(RA)+phosphate-buffered saline (PBS) group; O2 (hyperoxia, 95 % O2) + PBS group; RA + OVA group; O2+OVA group. We obtained the serum, bronchoalveolar lavage fluid (BALF), and lung tissues. The Wright-Giemsa staining was performed for leukocyte classification in BALF and HE staining for pathological examination. The levels of IL-2, IL-5, IL-13, IL-17A and IL-10 in BALF and tIgE and sIgE in serum were detected by ELISA. Results Compared with OVA sensitization or hyperoxia exposure alone, the mice in the model group (O2+OVA) showed asthma-like symptoms and increased airway hyperreactivity,The levels of IL-5,IL-13 IL-17A were increased in BLAF,and total leukocyte and eosinophil counts were also significant increasesed. The levels of tIgE and sIgE in serum were increased. Conclusion Postnatal hyperoxia exposure combined with early OVA sensitization might establish a juvenile mouse asthma model.
Collapse
Affiliation(s)
- Jingyan Li
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Linxia Cao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Bingrui Yu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Yuan Zhang
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Rong Wu
- Neonatal Medical Center, Huaian Maternity and Child Healthcare Hospital, Anhui Medical University, Huai'an, Jiangsu, 223002, China
| | - Haiyan Zhu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai 'an, Jiangsu, 223300, China
| |
Collapse
|
25
|
Pathak MP, Patowary P, Chattopadhyay P, Barbhuiyan PA, Islam J, Gogoi J, Wankhar W. Obesity-associated Airway Hyperresponsiveness: Mechanisms Underlying Inflammatory Markers and Possible Pharmacological Interventions. Endocr Metab Immune Disord Drug Targets 2024; 24:1053-1068. [PMID: 37957906 DOI: 10.2174/0118715303256440231028072049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 08/14/2023] [Accepted: 09/15/2023] [Indexed: 11/15/2023]
Abstract
Obesity is rapidly becoming a global health problem affecting about 13% of the world's population affecting women and children the most. Recent studies have stated that obese asthmatic subjects suffer from an increased risk of asthma, encounter severe symptoms, respond poorly to anti-asthmatic drugs, and ultimately their quality-of-life decreases. Although, the association between airway hyperresponsiveness (AHR) and obesity is a growing concern among the public due to lifestyle and environmental etiologies, however, the precise mechanism underlying this association is yet to establish. Apart from aiming at the conventional antiasthmatic targets, treatment should be directed towards ameliorating obesity pathogenesis too. Understanding the pathogenesis underlying the association between obesity and AHR is limited, however, a plethora of obesity pathologies have been reported viz., increased pro-inflammatory and decreased anti-inflammatory adipokines, depletion of ROS controller Nrf2/HO-1 axis, NLRP3 associated macrophage polarization, hypertrophy of WAT, and down-regulation of UCP1 in BAT following down-regulated AMPKα and melanocortin pathway that may be correlated with AHR. Increased waist circumference (WC) or central obesity was thought to be related to severe AHR, however, some recent reports suggest body mass index (BMI), not WC tends to exaggerate airway closure in AHR due to some unknown mechanisms. This review aims to co-relate the above-mentioned mechanisms that may explain the copious relation underlying obesity and AHR with the help of published reports. A proper understanding of these mechanisms discussed in this review will ensure an appropriate treatment plan for patients through advanced pharmacological interventions.
Collapse
Affiliation(s)
| | - Pompy Patowary
- Division of Pharmaceutical Technology, Defence Research Laboratory, Tezpur, India
| | | | | | - Johirul Islam
- Department of Pharmaceutical Sciences, School of Health Sciences, Assam Kaziranga University, Jorhat, India
| | - Jyotchna Gogoi
- Department of Biochemistry, Faculty of Science, Assam Down Town University, Guwahati, India
| | - Wankupar Wankhar
- Department of Dialysis, Faculty of Paramedical Science, Assam Down Town University, Guwahati, India
| |
Collapse
|
26
|
Quan J, Wen X, Su G, Zhong Y, Huang T, Xiong Z, Huang J, Lv Y, Li S, Luo S, Luo C, Cai X, Lai X, Xiang Y, Zheng SG, Shao Y, Lin H, Gao X, Tang J, Lai T. Epithelial SIRT6 governs IL-17A pathogenicity and drives allergic airway inflammation and remodeling. Nat Commun 2023; 14:8525. [PMID: 38135684 PMCID: PMC10746710 DOI: 10.1038/s41467-023-44179-x] [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: 02/13/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Dysregulation of IL-17A is closely associated with airway inflammation and remodeling in severe asthma. However, the molecular mechanisms by which IL-17A is regulated remain unclear. Here we identify epithelial sirtuin 6 (SIRT6) as an epigenetic regulator that governs IL-17A pathogenicity in severe asthma. Mice with airway epithelial cell-specific deletion of Sirt6 are protected against allergen-induced airway inflammation and remodeling via inhibiting IL-17A-mediated inflammatory chemokines and mesenchymal reprogramming. Mechanistically, SIRT6 directly interacts with RORγt and mediates RORγt deacetylation at lysine 192 via its PPXY motifs. SIRT6 promotes RORγt recruitment to the IL-17A gene promoter and enhances its transcription. In severe asthma patients, high expression of SIRT6 positively correlates with airway remodeling and disease severity. SIRT6 inhibitor (OSS_128167) treatment significantly attenuates airway inflammation and remodeling in mice. Collectively, these results uncover a function for SIRT6 in regulating IL-17A pathogenicity in severe asthma, implicating SIRT6 as a potential therapeutic target for severe asthma.
Collapse
Affiliation(s)
- Jingyun Quan
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiaoxia Wen
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Guomei Su
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yu Zhong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Tong Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhilin Xiong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jiewen Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yingying Lv
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Shihai Li
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Shuhua Luo
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chaole Luo
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Xin Cai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xianwen Lai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuanyuan Xiang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Song Guo Zheng
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Yiming Shao
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Haitao Lin
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiao Gao
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China.
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
27
|
Galeana-Cadena D, Gómez-García IA, Lopez-Salinas KG, Irineo-Moreno V, Jiménez-Juárez F, Tapia-García AR, Boyzo-Cortes CA, Matías-Martínez MB, Jiménez-Alvarez L, Zúñiga J, Camarena A. Winds of change a tale of: asthma and microbiome. Front Microbiol 2023; 14:1295215. [PMID: 38146448 PMCID: PMC10749662 DOI: 10.3389/fmicb.2023.1295215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/15/2023] [Indexed: 12/27/2023] Open
Abstract
The role of the microbiome in asthma is highlighted, considering its influence on immune responses and its connection to alterations in asthmatic patients. In this context, we review the variables influencing asthma phenotypes from a microbiome perspective and provide insights into the microbiome's role in asthma pathogenesis. Previous cohort studies in patients with asthma have shown that the presence of genera such as Bifidobacterium, Lactobacillus, Faecalibacterium, and Bacteroides in the gut microbiome has been associated with protection against the disease. While, the presence of other genera such as Haemophilus, Streptococcus, Staphylococcus, and Moraxella in the respiratory microbiome has been implicated in asthma pathogenesis, indicating a potential link between microbial dysbiosis and the development of asthma. Furthermore, respiratory infections have been demonstrated to impact the composition of the upper respiratory tract microbiota, increasing susceptibility to bacterial diseases and potentially triggering asthma exacerbations. By understanding the interplay between the microbiome and asthma, valuable insights into disease mechanisms can be gained, potentially leading to the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- David Galeana-Cadena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Karen Gabriel Lopez-Salinas
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Valeria Irineo-Moreno
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Red de Medicina para la Educación, el Desarrollo y la Investigación Científica de Iztacala, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Alberto Boyzo-Cortes
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Luis Jiménez-Alvarez
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Inmunobiología y Genética, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas (INER), Mexico City, Mexico
| |
Collapse
|
28
|
Jie XL, Luo ZR, Yu J, Tong ZR, Li QQ, Wu JH, Tao Y, Feng PS, Lan JP, Wang P. Pi-Pa-Run-Fei-Tang alleviates lung injury by modulating IL-6/JAK2/STAT3/IL-17 and PI3K/AKT/NF-κB signaling pathway and balancing Th17 and Treg in murine model of OVA-induced asthma. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116719. [PMID: 37268260 DOI: 10.1016/j.jep.2023.116719] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pi-Pa-Run-Fei-Tang (PPRFT) is an empirical TCM prescription for treating asthma. However, the underlying mechanisms of PPRFT in asthma treatment have yet to be elucidated. Recent advances have revealed that some natural components could ameliorate asthma injury by affecting host metabolism. Untargeted metabolomics can be used to better understand the biological mechanisms underlying asthma development and identify early biomarkers that can help advance treatment. AIM OF THE STUDY The aim of this study was to verification the efficacy of PPRFT in the treatment of asthma and to preliminarily explore its mechanism. MATERIALS AND METHODS A mouse asthma model was built by OVA induction. Inflammatory cell in BALF was counted. The level of IL-6, IL-1β, and TNF-α in BALF were measured. The levels of IgE in the serum and EPO, NO, SOD, GSH-Px, and MDA in the lung tissue were measured. Furthermore, pathological damage to the lung tissues was detected to evaluate the protective effects of PPRFT. The serum metabolomic profiles of PPRFT in asthmatic mice were determined by GC-MS. The regulatory effects on mechanism pathways of PPRFT in asthmatic mice were explored via immunohistochemical staining and western blotting analysis. RESULTS PPRFT displayed lung-protective effects through decreasing oxidative stress, airway inflammation, and lung tissue damage in OVA-induced mice, which was demonstrated by decreasing inflammatory cell levels, IL-6, IL-1β, and TNF-α levels in BALF, and IgE levels in serum, decreasing EPO, NO, and MDA levels in lung tissue, elevating SOD and GSH-Px levels in lung tissue and lung histopathological changes. In addition, PPRFT could regulate the imbalance in Th17/Treg cell ratios, suppress RORγt, and increase the expression of IL-10 and Foxp3 in the lung. Moreover, PPRFT treatment led to decreased expression of IL-6, p-JAK2/Jak2, p-STAT3/STAT3, IL-17, NF-κB, p-AKT/AKT, and p-PI3K/PI3K. Serum metabolomics analysis revealed that 35 metabolites were significantly different among different groups. Pathway enrichment analysis indicated that 31 pathways were involved. Moreover, correlation analysis and metabolic pathway analysis identified three key metabolic pathways: galactose metabolism; tricarboxylic acid cycle; and glycine, serine, and threonine metabolism. CONCLUSION This research indicated that PPRFT treatment not only attenuates the clinical symptoms of asthma but is also involved in regulating serum metabolism. The anti-asthmatic activity of PPRFT may be associated with the regulatory effects of IL-6/JAK2/STAT3/IL-17 and PI3K/AKT/NF-κB mechanistic pathways.
Collapse
Affiliation(s)
- Xiao-Lu Jie
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zi-Rui Luo
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jin Yu
- Hangzhou Zhongmei Huadong Pharmaceutical Co., Ltd., Hangzhou, 310014, China
| | - Zhe-Ren Tong
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Qiao-Qiao Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Jia-Hui Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yi Tao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Pei-Shi Feng
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Ji-Ping Lan
- Experiment Center for Teaching & Learning Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
29
|
Song J, Zhang H, Tong Y, Wang Y, Xiang Q, Dai H, Weng C, Wang L, Fan J, Shuai Y, Lai C, Fang X, Chen M, Bao J, Zhang W. Molecular mechanism of interleukin-17A regulating airway epithelial cell ferroptosis based on allergic asthma airway inflammation. Redox Biol 2023; 68:102970. [PMID: 38035662 PMCID: PMC10711239 DOI: 10.1016/j.redox.2023.102970] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
Interleukin-17A (IL-17A) levels are elevated in patients with asthma. Ferroptosis has been identified as the non-apoptotic cell death type associated with asthma. Data regarding the relation of ferroptosis with asthma and the effect of IL-17A on modulating ferroptosis in asthma remain largely unclear. The present work focused on investigating the role of IL-17A in allergic asthma-related ferroptosis and its associated molecular mechanisms using public datasets, clinical samples, human bronchial epithelial cells, and an allergic asthma mouse model. We found that IL-17A was significantly upregulated within serum in asthma cases. Adding IL-17A significantly increased ferroptosis within human bronchial epithelial cells (BEAS-2B). In ovalbumin (OVA)-induced allergic asthmatic mice, IL-17A regulated and activated lipid peroxidation induced ferroptosis, whereas IL-17A knockdown effectively inhibited ferroptosis in vivo by protection of airway epithelial cells via the xCT-GSH-GPX4 antioxidant system and reduced airway inflammation. Mouse mRNA sequencing results indicated that the tumor necrosis factor (TNF) pathway was the differential KEGG pathway in the OVA group compared to healthy controls and the OVA group compared to the IL-17A knockout OVA group. We further used N-acetylcysteine (TNF inhibitor) to inhibit the TNF signaling pathway, which was found to protect BEAS-2B cells from IL-17A induced lipid peroxidation and ferroptosis damage. Our findings reveal a novel mechanism for the suppression of ferroptosis in airway epithelial cells, which may represent a new strategy for the use of IL-17A inhibitors against allergic asthma.
Collapse
Affiliation(s)
- Jingjing Song
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yu Tong
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yufei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Qiangwei Xiang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Cuiye Weng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Lei Wang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Junwen Fan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yilong Shuai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Chuqiao Lai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoxiao Fang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Mingxin Chen
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiali Bao
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
30
|
Ye X, Li Y, Fang B, Yuan Y, Feng D, Chen H, Li J, Meng Q, Xiong S, Ye D, Jiao L, Chen D, Chen R, Lei W, Gao Y, Li C. Type 17 mucosal-associated invariant T cells contribute to neutrophilic inflammation in patients with nasal polyps. J Allergy Clin Immunol 2023; 152:1153-1166.e12. [PMID: 37437744 DOI: 10.1016/j.jaci.2023.06.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/21/2023] [Accepted: 06/01/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND Immune regulation in chronic rhinosinusitis with nasal polyps (CRSwNP) with a neutrophilic endotype remains unclear. Mucosal-associated invariant T (MAIT) cells are tissue-resident innate T lymphocytes that respond quickly to pathogens and promote chronic mucosal inflammation. OBJECTIVE We aimed to investigate the roles of MAIT cells in neutrophilic CRSwNP. METHODS Nasal tissues were obtained from 113 patients with CRSwNP and 29 control subjects. Peripheral and tissue MAIT cells and their subsets were analyzed by flow cytometry. Polyp-derived MAIT cells were analyzed by RNA sequencing to study their effects on neutrophils. RESULTS Endotypes of CRSwNP were classified as paucigranulocytic (n = 21), eosinophilic (n = 29), neutrophilic (n = 39), and mixed granulocytic (n = 24). Frequencies of MAIT cells were significantly higher in neutrophilic (3.62%) and mixed granulocytic (3.60%) polyps than in control mucosa (1.78%). MAIT cell percentages positively correlated with local neutrophil counts. MAIT cells were more enriched in tissues than in matched PBMCs. The frequencies of MAIT1 subset or IFN-γ+ MAIT cells were comparable among control tissues and CRSwNP subtypes. The proportions of MAIT17 subset or IL-17A+ MAIT cells were significantly increased in neutrophilic or mixed granulocytic polyps compared with controls. RNA sequencing revealed type 17 and pro-neutrophil profiles in neutrophilic polyp-derived MAIT cells. In patients with neutrophilic CRSwNP, the proportions of MAIT and MAIT17 cells were positively correlated with local proinflammatory cytokines and symptom severity. In vitro experiments demonstrated that neutrophilic polyp-derived MAIT cells promoted neutrophil migration, survival, and activation. CONCLUSIONS MAIT cells from neutrophilic CRSwNP demonstrate type 17 functional properties and promote neutrophil infiltration in nasal mucosa.
Collapse
Affiliation(s)
- Xiaoyan Ye
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, China
| | - Yachun Li
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bixing Fang
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yizhang Yuan
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Danni Feng
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hexin Chen
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian Li
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning, China
| | - Qingxiang Meng
- Department of Otorhinolaryngology Head and Neck Surgery, Guangzhou First People's Hospital, Guangzhou, China
| | - Shaobing Xiong
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongmei Ye
- Organ Transplantation Centre, Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Linyi Jiao
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dehua Chen
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ruchong Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Department of Allergy and Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Wenbin Lei
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yifang Gao
- Organ Transplantation Centre, Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chunwei Li
- Department of Otolaryngology, Department of Allergy, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
31
|
Fan X, Shu P, Wang Y, Ji N, Zhang D. Interactions between neutrophils and T-helper 17 cells. Front Immunol 2023; 14:1279837. [PMID: 37920459 PMCID: PMC10619153 DOI: 10.3389/fimmu.2023.1279837] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/05/2023] [Indexed: 11/04/2023] Open
Abstract
Neutrophils comprise the majority of immune cells in human peripheral circulation, have potent antimicrobial activities, and are clinically significant in their abundance, heterogeneity, and subcellular localization. In the past few years, the role of neutrophils as components of the innate immune response has been studied in numerous ways, and these cells are crucial in fighting infections, autoimmune diseases, and cancer. T-helper 17 (Th17) cells that produce interleukin 17 (IL-17) are critical in fighting infections and maintaining mucosal immune homeostasis, whereas they mediate several autoimmune diseases. Neutrophils affect adaptive immune responses by interacting with adaptive immune cells. In this review, we describe the physiological roles of both Th17 cells and neutrophils and their interactions and briefly describe the pathological processes in which these two cell types participate. We provide a summary of relevant drugs targeting IL-17A and their clinical trials. Here, we highlight the interactions between Th17 cells and neutrophils in diverse pathophysiological situations.
Collapse
Affiliation(s)
- Xinzou Fan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Panyin Shu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ying Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Chatziparasidis G, Bush A, Chatziparasidi MR, Kantar A. Airway epithelial development and function: A key player in asthma pathogenesis? Paediatr Respir Rev 2023; 47:51-61. [PMID: 37330410 DOI: 10.1016/j.prrv.2023.04.005] [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: 02/05/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 06/19/2023]
Abstract
Though asthma is a common and relatively easy to diagnose disease, attempts at primary or secondary prevention, and cure, have been disappointing. The widespread use of inhaled steroids has dramatically improved asthma control but has offered nothing in terms of altering long-term outcomes or reversing airway remodeling and impairment in lung function. The inability to cure asthma is unsurprising given our limited understanding of the factors that contribute to disease initiation and persistence. New data have focused on the airway epithelium as a potentially key factor orchestrating the different stages of asthma. In this review we summarize for the clinician the current evidence on the central role of the airway epithelium in asthma pathogenesis and the factors that may alter epithelial integrity and functionality.
Collapse
Affiliation(s)
- Grigorios Chatziparasidis
- Paediatric Respiratory Unit, IASO Hospital, Larissa, Thessaly, Greece; Faculty of Nursing, Thessaly University, Greece.
| | - Andrew Bush
- National Heart and Lung Institute, Royal Brompton & Harefield NHS Foundation Trust, London, UK
| | | | - Ahmad Kantar
- Pediatric Asthma and Cough Centre, Instituti Ospedalieri Bergamaschi, University and Research Hospitals, Bergamo, Italy
| |
Collapse
|
33
|
Caffarelli C, Gracci S, Giannì G, Bernardini R. Are Babies Born Preterm High-Risk Asthma Candidates? J Clin Med 2023; 12:5400. [PMID: 37629440 PMCID: PMC10455600 DOI: 10.3390/jcm12165400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Among preterm infants, the risk of developing asthma is a matter of debate. This review discusses the state of the art of poorly understood prematurity-associated asthma. Impaired pulmonary function is common in children born prematurely. Preterm infants are prone to developing viral respiratory tract infections, bronchiolitis in the first year of life, and recurrent viral wheezing in preschool age. All of these conditions may precede asthma development. We also discuss the role of both atopic sensitization and intestinal microbiome and, consequently, immune maturation. Diet and pollution have been considered to better understand how prematurity could be associated with asthma. Understanding the effect of factors involved in asthma onset may pave the way to improve the prediction of this asthma phenotype.
Collapse
Affiliation(s)
- Carlo Caffarelli
- Clinica Pediatrica, Azienda Ospedaliero-Universitaria, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Serena Gracci
- Pediatric Unit, San Giuseppe Hospital, 50053 Empoli, Italy
| | - Giuliana Giannì
- Clinica Pediatrica, Azienda Ospedaliero-Universitaria, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | | |
Collapse
|
34
|
Ntinopoulou M, Cassimos D, Roupakia E, Kolettas E, Panopoulou M, Mantadakis E, Konstantinidis T, Chrysanthopoulou A. Ιnterleukin-17A-Enriched Neutrophil Extracellular Traps Promote Immunofibrotic Aspects of Childhood Asthma Exacerbation. Biomedicines 2023; 11:2104. [PMID: 37626601 PMCID: PMC10452671 DOI: 10.3390/biomedicines11082104] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
Childhood asthma is a chronic inflammatory airway disorder that can drive tissue remodeling. Neutrophils are amongst the most prominent inflammatory cells contributing to disease manifestations and may exert a potent role in the progression of inflammation to fibrosis. However, their role in asthma exacerbation is still understudied. Here, we investigate the association between neutrophil extracellular traps (NETs) and lung fibroblasts in childhood asthma pathophysiology using serum samples from pediatric patients during asthma exacerbation. Cell-based assays and NETs/human fetal lung fibroblast co-cultures were deployed. Increased levels of NETs and interleukin (IL)-17A were detected in the sera of children during asthma exacerbation. The in vitro stimulation of control neutrophils using the sera from pediatric patients during asthma exacerbation resulted in IL-17A-enriched NET formation. The subsequent co-incubation of lung fibroblasts with in vitro-generated IL-17A-enriched NETs led fibroblasts to acquire a pre-fibrotic phenotype, as assessed via enhanced CCN2 expression, migratory/healing capacity, and collagen release. These data uncover the important pathogenic role of the NET/IL-17A axis in asthma exacerbation, linking lung inflammation to fibroblast dysfunction and fibrosis.
Collapse
Affiliation(s)
- Maria Ntinopoulou
- Laboratory of Molecular Immunology, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (M.N.); (T.K.)
| | - Dimitrios Cassimos
- Department of Pediatrics, University General Hospital of Alexandroupolis, Democritus University of Thrace Medical School, 68100 Thrace, Greece; (D.C.); (E.M.)
| | - Eugenia Roupakia
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.R.); (E.K.)
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
| | - Evangelos Kolettas
- Laboratory of Biology, School of Medicine, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.R.); (E.K.)
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
| | - Maria Panopoulou
- Department of Microbiology, University General Hospital of Alexandroupolis, Democritus University of Thrace Medical School, 68100 Thrace, Greece;
| | - Elpis Mantadakis
- Department of Pediatrics, University General Hospital of Alexandroupolis, Democritus University of Thrace Medical School, 68100 Thrace, Greece; (D.C.); (E.M.)
| | - Theocharis Konstantinidis
- Laboratory of Molecular Immunology, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (M.N.); (T.K.)
| | - Akrivi Chrysanthopoulou
- Laboratory of Molecular Immunology, Department of Biological Applications and Technology, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (M.N.); (T.K.)
| |
Collapse
|
35
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances TH17 Responses and Promotes Neutrophilic Airway Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547286. [PMID: 37461622 PMCID: PMC10349957 DOI: 10.1101/2023.06.30.547286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Treatment-refractory severe asthma manifests a neutrophilic phenotype associated with TH17 responses. Heightened unfolded protein responses (UPRs) are associated with the risk of asthma, including severe asthma. However, how UPRs participate in the deregulation of TH17 cells leading to this type of asthma remains elusive. In this study, we investigated the role of the UPR sensor IRE1 in TH17 cell function and neutrophilic airway inflammation. We found that IRE1 is induced in fungal asthma and is highly expressed in TH17 cells relative to naïve CD4+ T cells. Cytokine (e.g. IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by TH17 cells. Ern1 (encoding IRE1)-deficiency decreases the expression of ER stress factors and impairs the differentiation and cytokine secretion of TH17 cells. Genetic ablation of Ern1 leads to alleviated TH17 responses and airway neutrophilia in a Candida albicans asthma model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances TH17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPRmediated secretory function of TH17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in TH17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kourtney M. Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
36
|
Peters K, Ernst S, Peters M. Interaction of Interleukin-17A with a Th2 Response in a Mouse Model of Allergic Airway Inflammation. Cells 2023; 12:1774. [PMID: 37443808 PMCID: PMC10340318 DOI: 10.3390/cells12131774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
BACKGROUND A total of 262 million people worldwide suffer from asthma and 461000 people died from it in 2019. Asthma is a disease with different endotypes defined by the granulocytes found in the asthmatic lung. In allergic asthma, the eosinophilic endotype is present, driven by a TH2 response. A TH17 immune response leads to the neutrophil endotype. This often causes uncontrolled asthma and is triggered by pollutants, microbes, and oxidative stress. It has been described that a significant number of patients with eosinophilic asthma develop mixed granulocytic asthma over time. The severity of asthma in the mixed endotype is related to the proportion of neutrophils in the lungs. PURPOSE In this report, we address the question of how a TH2 response interacts with IL-17A in allergic asthma. METHODS To this end, we used a mouse model to induce allergic asthma followed by an aerosol challenge with ovalbumin. To investigate the role of IL-17A, we administered IL-17A intranasally during the challenge phase. RESULTS IL-17A alone did not elicit an immune response, whereas in combination with allergic asthma, it resulted in a shift of the asthmatic endotype from eosinophilic to neutrophilic. TGFβ1 was increased in these lungs compared to asthmatic lungs without IL-17A, as was the expression of the IL-17A receptor subunits IL-17RA and IL-17RC. In cultures with human cells, we also found that IL-17A increased the expression of its receptors only in combination with IL-13. We also found this effect for IL-8, which attracts neutrophils in humans. CONCLUSIONS The TH2 response increased the sensitivity to IL-17A in a mouse asthma model as well as in human cell lines.
Collapse
Affiliation(s)
- Karin Peters
- Department of Molecular Immunology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Stefanie Ernst
- Department of Experimental Pneumology, Ruhr-University Bochum, D-44780 Bochum, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr-University Bochum, D-44780 Bochum, Germany
| |
Collapse
|
37
|
Lee EY, Choi W, Burkholder AB, Perera L, Mack JA, Miller FW, Fessler MB, Cook DN, Karmaus PWF, Nakano H, Garantziotis S, Madenspacher JH, House JS, Akhtari FS, Schmitt CS, Fargo DC, Hall JE, Motsinger-Reif AA. Race/ethnicity-stratified fine-mapping of the MHC locus reveals genetic variants associated with late-onset asthma. Front Genet 2023; 14:1173676. [PMID: 37415598 PMCID: PMC10321602 DOI: 10.3389/fgene.2023.1173676] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/09/2023] [Indexed: 07/08/2023] Open
Abstract
Introduction: Asthma is a chronic disease of the airways that impairs normal breathing. The etiology of asthma is complex and involves multiple factors, including the environment and genetics, especially the distinct genetic architecture associated with ancestry. Compared to early-onset asthma, little is known about genetic predisposition to late-onset asthma. We investigated the race/ethnicity-specific relationship among genetic variants within the major histocompatibility complex (MHC) region and late-onset asthma in a North Carolina-based multiracial cohort of adults. Methods: We stratified all analyses by self-reported race (i.e., White and Black) and adjusted all regression models for age, sex, and ancestry. We conducted association tests within the MHC region and performed fine-mapping analyses conditioned on the race/ethnicity-specific lead variant using whole-genome sequencing (WGS) data. We applied computational methods to infer human leukocyte antigen (HLA) alleles and residues at amino acid positions. We replicated findings in the UK Biobank. Results: The lead signals, rs9265901 on the 5' end of HLA-B, rs55888430 on HLA-DOB, and rs117953947 on HCG17, were significantly associated with late-onset asthma in all, White, and Black participants, respectively (OR = 1.73, 95%CI: 1.31 to 2.14, p = 3.62 × 10-5; OR = 3.05, 95%CI: 1.86 to 4.98, p = 8.85 × 10-6; OR = 19.5, 95%CI: 4.37 to 87.2, p = 9.97 × 10-5, respectively). For the HLA analysis, HLA-B*40:02 and HLA-DRB1*04:05, HLA-B*40:02, HLA-C*04:01, and HLA-DRB1*04:05, and HLA-DRB1*03:01 and HLA-DQB1 were significantly associated with late-onset asthma in all, White, and Black participants. Conclusion: Multiple genetic variants within the MHC region were significantly associated with late-onset asthma, and the associations were significantly different by race/ethnicity group.
Collapse
Affiliation(s)
- Eunice Y. Lee
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Wonson Choi
- Genomics and Bioinformatics Laboratory, Seoul National University, Seoul, Republic of Korea
| | - Adam B. Burkholder
- National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Lalith Perera
- Genomic Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Jasmine A. Mack
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
- Department of Obstetrics and Gynecology, University of Cambridge, Cambridge, United Kingdom
| | - Frederick W. Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Michael B. Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Donald N. Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
- Immunogenetics Group, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Peer W. F. Karmaus
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Stavros Garantziotis
- Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Jennifer H. Madenspacher
- Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - John S. House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Farida S. Akhtari
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
- Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Charles S. Schmitt
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - David C. Fargo
- National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Janet E. Hall
- Clinical Research Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| | - Alison A. Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, United States
| |
Collapse
|
38
|
Benazzouz S, Amri M, Ketfi A, Boutemine IM, Sellam SL, Benkhelifa S, Ameur F, Djebbara S, Achour K, Soufli I, Belguendouz H, Gharnaout M, Touil-Boukoffa C. Ex vivo Immuno-modulatory effect of Echinococcus granulosus laminated layer during allergic rhinitis and allergic asthma: A study in Algerian Patients. Exp Parasitol 2023; 250:108535. [PMID: 37116772 DOI: 10.1016/j.exppara.2023.108535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/08/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
The effect of helminthic infections on allergic diseases and asthma is still inconclusive. Moreover, there is considerable evidence suggesting that nitric oxide (NO), metalloproteinases and pro-inflammatory cytokines play a significant role in the physiopathology of these diseases. In this sense, the aim of our study is to investigate the ex vivo immunomodulatory effect of the laminated layer (LL, outside layer of parasitic cyst) of the helminth Echinococcus granulosus on NO, IL-17A and IL-10 production. In the first step of our study, we evaluated in vivo the NO, MMP-9, IL-17A, IL-10 levels in Algerian patients with allergic asthma and allergic rhinitis and their changes in relation with exacerbation status of the patients. In the principal part of our work, we assessed NO, IL-10 and IL-17A levels in supernatants of patients PBMCs cultures before and after stimulation with LL. Our results indicate a significant reduction in NO production by PBMCs of patients with allergic rhinitis and allergic asthma whether mild, moderate or severe after stimulation with LL. Interestingly, LL induces a significant decrease in the production of NO and IL17-A levels as well as an increase in the production of IL-10 in the cultures performed with PBMC of patients with severe allergic asthma. Importantly, our data indicate that LL exert a down-modulatory effect on inflammatory mediators (NO, IL-17A) and up immune-regulatory effect on IL-10 production. Collectively, our study supports the hygiene hypothesis suggesting that Echinococcus granulosus infection like other helminths could prevent and/or modulate inflammation responses during inflammatory diseases.
Collapse
Affiliation(s)
- Sara Benazzouz
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Manel Amri
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | | | - Insaf-Meriem Boutemine
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Sarah Leila Sellam
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Sarra Benkhelifa
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Fahima Ameur
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Sara Djebbara
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Karima Achour
- Service of Thoracic Surgery, University Hospital Center Lamine Debaghine, Bab El Oued, Algiers, Algeria
| | - Imene Soufli
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | - Houda Belguendouz
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria
| | | | - Chafia Touil-Boukoffa
- Team "Cytokines and NOsynthase, Immunity and Pathogenicity", Laboratory of Cellular and Molecular Biology, Faculty of Biological Sciences, University of Science and Technology Houari Boumediene, BP 32 El Alia, Bab Ezzouar, 16111, Algiers, Algeria.
| |
Collapse
|
39
|
Th17/Treg Imbalance: Implications in Lung Inflammatory Diseases. Int J Mol Sci 2023; 24:ijms24054865. [PMID: 36902294 PMCID: PMC10003150 DOI: 10.3390/ijms24054865] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/27/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Regulatory T cells (Tregs) and T helper 17 cells (Th17) are two CD4+ T cell subsets with antagonist effects. Th17 cells promote inflammation, whereas Tregs are crucial in maintaining immune homeostasis. Recent studies suggest that Th17 cells and Treg cells are the foremost players in several inflammatory diseases. In this review, we explore the present knowledge on the role of Th17 cells and Treg cells, focusing on lung inflammatory diseases, such as chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), sarcoidosis, asthma, and pulmonary infectious diseases.
Collapse
|
40
|
Yu H, Huang X, Zhu HH, Wang N, Xie C, Zhou YL, Shi HL, Chen MM, Wu YR, Ruan ZH, Lyu YB, Luo QL, Dong JC. Apigenin ameliorates non-eosinophilic inflammation, dysregulated immune homeostasis and mitochondria-mediated airway epithelial cell apoptosis in chronic obese asthma via the ROS-ASK1-MAPK pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154646. [PMID: 36645975 DOI: 10.1016/j.phymed.2023.154646] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/21/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Obese asthma is one of the important asthma phenotypes that have received wide attention in recent years. Excessive oxidative stress and different inflammatory endotypes may be important reasons for the complex symptoms, frequent aggravation, and resistance to traditional treatments of obese asthma. Apigenin (API), is a flavonoid natural small molecule compound with good anti-inflammatory and antioxidant activity in various diseases and proved to have the potential efficacy to combat obese asthma. METHODS In vivo, this study fed C57BL/6 J mice with high-fat diets(HFD)for 12 weeks and then stimulated them with OVA for 6 weeks to establish a model of chronic obese asthma, while different doses of oral API or dexamethasone were used for therapeutic interventions. In vitro, this study used HDM to stimulate human bronchial cells (HBEs) to establish the model and intervened with API or Selonsertib (SEL). RESULTS This study clarified that OVAinduced a type of mixed granulocytic asthma with elevated neutrophils and eosinophils in obese male mice fed with long-term HFD, which also exhibited mixed TH17/TH1/TH2 inflammation. Apigenin effectively suppressed this complex inflammation and acted as a regulator of immune homeostasis. Meanwhile, apigenin reduced AHR, inflammatory cell infiltration, airway epithelial cell apoptosis, airway collagen deposition, and lung oxidative stress via the ROS-ASK1-MAPK pathway in an obese asthma mouse model. In vitro, this study found that apigenin altered the binding status of TRAF6 to ASK1, inhibited ASK1 phosphorylation, and protected against ubiquitin-dependent degradation of ASK1, suggesting that ROS-activated ASK1 may be an important target for apigenin to exert anti-inflammatory and anti-apoptotic effects. To further verify the intervention mechanism, this study clarified that apigenin improved cell viability and mitochondrial function and inhibited apoptosis by interfering with the ROS-ASK1-MAPK pathway. CONCLUSIONS This study demonstrates for the first time the therapeutic effect of apigenin in chronic obese asthma and further clarifies its potential therapeutic targets. In addition, this study clarifies the specificity of chronic obese asthma and provides new options for its treatment.
Collapse
Affiliation(s)
- Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Hua-He Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Cong Xie
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yao-Long Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Han-Lin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Meng-Meng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yue-Ren Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Zhen-Hui Ruan
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Yu-Bao Lyu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Qing-Li Luo
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China; Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai, China.
| | - Jing-Cheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institute of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Fraga-Silva TFDC, Boko MMM, Martins NS, Cetlin AA, Russo M, Vianna EO, Bonato VLD. Asthma-associated bacterial infections: Are they protective or deleterious? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:14-22. [PMID: 37780109 PMCID: PMC10510013 DOI: 10.1016/j.jacig.2022.08.003] [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: 04/19/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 10/03/2023]
Abstract
Eosinophilic, noneosinophilic, or mixed granulocytic inflammations are the hallmarks of asthma heterogeneity. Depending on the priming of lung immune and structural cells, subjects with asthma might generate immune responses that are TH2-prone or TH17-prone immune response. Bacterial infections caused by Haemophilus, Moraxella, or Streptococcus spp. induce the secretion of IL-17, which in turn recruit neutrophils into the airways. Clinical studies and experimental models of asthma indicated that neutrophil infiltration induces a specific phenotype of asthma, characterized by an impaired response to corticosteroid treatment. The understanding of pathways that regulate the TH17-neutrophils axis is critical to delineate and develop host-directed therapies that might control asthma and its exacerbation episodes that course with infectious comorbidities. In this review, we outline clinical and experimental studies on the role of airway epithelial cells, S100A9, and high mobility group box 1, which act in concert with the IL-17-neutrophil axis activated by bacterial infections, and are related with asthma that is difficult to treat. Furthermore, we report critically our view in the light of these findings in an attempt to stimulate further investigations and development of immunotherapies for the control of severe asthma.
Collapse
Affiliation(s)
| | - Mèdéton Mahoussi Michaël Boko
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Núbia Sabrina Martins
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Andrea Antunes Cetlin
- Pulmonary Division, Department of Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Momtchilo Russo
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Elcio Oliveira Vianna
- Pulmonary Division, Department of Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Vania Luiza Deperon Bonato
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Basic and Applied Immunology Program, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
42
|
Chen D, Chen Q, Zhao K, Guo Y, Huang Y, Yuan Z, Cai Y, Li S, Xu J, Lin X. Exploring the mechanism of Xiaoqinglong decoction in the treatment of infantile asthma based on network pharmacology and molecular docking. Medicine (Baltimore) 2023; 102:e32623. [PMID: 36637916 PMCID: PMC9839235 DOI: 10.1097/md.0000000000032623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/20/2022] [Indexed: 01/14/2023] Open
Abstract
To explore the mechanism of Xiaoqinglong decoction (XQLD) in the treatment of infantile asthma (IA) based on network pharmacology and molecular docking. The active ingredients of fdrugs in XQLD were retrieved from Traditional Chinese Medicine Systems Pharmacology database and then the targets of drug ingredients were screened. The disease targets of IA were obtained from OMIM and Gencards databases, and the intersection targets of XQLD in the treatment of IA were obtained by Venny 2.1 mapping of ingredient targets and disease targets. Cytoscape software was used to construct active ingredient-intersection target network. The potential targets of XQLD in the treatment of IA were analyzed by protein-protein interaction network using STRING platform, and the Gene Ontology function and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were obtained by R Studio software. AutoDock was used to perform molecular docking for verification. In this study, 150 active ingredients of XQLD were obtained, including quercetin, kaempferol, β-sitosterol, luteolin, stigmasterol, and so on. And 92 intersection targets of drugs and diseases were obtained, including interleukin 6 (IL6), cystatin 3, estrogen receptor 1, hypoxia inducible factor 1A, HSP90AA1, epidermal growth factor receptor and so on. There were 127 items of Gene Ontology enrichment analysis and 125 Kyoto Encyclopedia of Genes and Genomes enrichment results, showing that apoptosis, IL-17 signaling pathway, tumor necrosis factor signaling pathway, P13K-Akt signaling pathway and other pathways may play a key role in the treatment of IA by XQLD. The results of molecular docking showed that the key active ingredients including quercetin, kaempferol, β-sitosterol, luteolin, stigmasterol, and the core targets including IL6, cystatin 3, estrogen receptor 1, hypoxia inducible factor 1A, HSP90AA1, and epidermal growth factor receptor had good binding activity. Through network pharmacology and molecular docking, the potential targets and modern biological mechanisms of XQLD in the treatment of IA were preliminarily revealed in the study, which will provide reference for subsequent animal experiments and clinical trials.
Collapse
Affiliation(s)
- Daman Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Qiqi Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kaibo Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongqi Guo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuxin Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehuan Yuan
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujia Cai
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sitong Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jiarong Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Lin
- Traditional Chinese Medicine Hospital of Guangdong Province, Guangzhou, China
| |
Collapse
|
43
|
Kaur S, Mishra J, Sehrawat A, Bhatti GK, Navik U, Reddy PH, Bhatti JS. Epigenetic Regulators of Inflammatory Gene Expression. TARGETING EPIGENETICS IN INFLAMMATORY LUNG DISEASES 2023:57-88. [DOI: https:/doi.org/10.1007/978-981-99-4780-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
|
44
|
Martinez-Paredes JF, Donaldson AM, Marino M, Choby G, Olomu O, Alfakir R, Stokken JK, O'Brien E, Lal D. Sinonasal Outcomes Using Oral Corticosteroids in Patients with Chronic Rhinosinusitis with Nasal Polyps and Positive Sinonasal Cultures. Int Arch Otorhinolaryngol 2022; 27:e286-e295. [PMID: 37125375 PMCID: PMC10147476 DOI: 10.1055/s-0042-1743275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Abstract
Introduction Patients with chronic rhinosinusitis with nasal polyps (CRSwNP) and positive sinonasal bacterial cultures may be recalcitrant to topical therapy alone due to the additional local inflammatory burden associated with bacterial infection/colonization.
Objective To evaluate sinonasal outcomes in CRSwNP patients with a positive perioperative bacterial culture, who were treated with postoperative intranasal corticosteroids (INCS) alone versus INCS in combination with a short-term course of oral corticosteroids (OCS).
Methods This is a retrospective chart review of CRSwNP patients. A total of 59 patients met inclusion criteria, including positive perioperative bacterial culture and treatment with INCS with or without concomitant use of OCS. Two cohorts were formed based on the chosen postoperative medical treatment; 32 patients underwent postoperative INCS alone, while 27 underwent INCS plus a ≤ 2-week course of OCS. The 22-item sinonasal outcome test (SNOT-22) scores and Lund-Kennedy scores (LKS) were assessed preoperatively, and at 2-week, 4-week, and 4 to 6 months after endoscopic sinus surgery (ESS).
Results There were no statistically significant differences in postoperative sinonasal symptoms or endoscopic scores between the cohorts treated with INCS plus OCS versus those prescribed INCS alone (p > 0.05). Our regression model failed to demonstrate a relationship between the use of OCS and better sinonasal outcomes at 2-week, 4-week, and 4 to 6 months after ESS (p > 0.05).
Conclusion Our study suggests that in a cohort of CRSwNP patients with recent bacterial infections, the postoperative use of combined OCS and INCS did not result in a statistical improvement of endoscopic and symptomatic outcomes over INCS irrigation alone. However, both treatment groups had a clinically significant improvement based on the Minimal Clinically Important Difference.
Collapse
Affiliation(s)
- Jhon F. Martinez-Paredes
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Jacksonville, Florida, United States
- Department of Surgery, University of Texas – Rio Grande Valley, Edinburg, Texas, United States
| | - Angela M. Donaldson
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Jacksonville, Florida, United States
| | - Michael Marino
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Phoenix, Arizona, United States
| | - Garret Choby
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - Osarenoma Olomu
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Jacksonville, Florida, United States
| | - Razan Alfakir
- Department of Speech, Language & Hearing Sciences, Auburn University, Alabama, United States
| | - Janalee K. Stokken
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - Erin O'Brien
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - Devyani Lal
- Department of Otolaryngology – Head and Neck Surgery, Mayo Clinic, Phoenix, Arizona, United States
| |
Collapse
|
45
|
Kaplan AG, Kim JW. Asthma Exacerbations and Glucagon-Like Peptide-1 Receptor Agonists: a Review of the Current Evidence. Pulm Ther 2022; 8:343-358. [DOI: 10.1007/s41030-022-00203-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/17/2022] [Indexed: 11/24/2022] Open
|
46
|
Recent Advances in Nanomaterials for Asthma Treatment. Int J Mol Sci 2022; 23:ijms232214427. [PMID: 36430906 PMCID: PMC9696023 DOI: 10.3390/ijms232214427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease with complex mechanisms, and these patients often encounter difficulties in their treatment course due to the heterogeneity of the disease. Currently, clinical treatments for asthma are mainly based on glucocorticoid-based combination drug therapy; however, glucocorticoid resistance and multiple side effects, as well as the occurrence of poor drug delivery, require the development of more promising treatments. Nanotechnology is an emerging technology that has been extensively researched in the medical field. Several studies have shown that drug delivery systems could significantly improve the targeting, reduce toxicity and improve the bioavailability of drugs. The use of multiple nanoparticle delivery strategies could improve the therapeutic efficacy of drugs compared to traditional delivery methods. Herein, the authors presented the mechanisms of asthma development and current therapeutic methods. Furthermore, the design and synthesis of different types of nanomaterials and micromaterials for asthma therapy are reviewed, including polymetric nanomaterials, solid lipid nanomaterials, cell membranes-based nanomaterials, and metal nanomaterials. Finally, the challenges and future perspectives of these nanomaterials are discussed to provide guidance for further research directions and hopefully promote the clinical application of nanotherapeutics in asthma treatment.
Collapse
|
47
|
Investigational Treatments in Phase I and II Clinical Trials: A Systematic Review in Asthma. Biomedicines 2022; 10:biomedicines10092330. [PMID: 36140430 PMCID: PMC9496184 DOI: 10.3390/biomedicines10092330] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Inhaled corticosteroids (ICS) remain the mainstay of asthma treatment, along with bronchodilators serving as control agents in combination with ICS or reliever therapy. Although current pharmacological treatments improve symptom control, health status, and the frequency and severity of exacerbations, they do not really change the natural course of asthma, including disease remission. Considering the highly heterogeneous nature of asthma, there is a strong need for innovative medications that selectively target components of the inflammatory cascade. The aim of this review was to systematically assess current investigational agents in Phase I and II randomised controlled trials (RCTs) over the last five years. Sixteen classes of novel therapeutic options were identified from 19 RCTs. Drugs belonging to different classes, such as the anti-interleukin (IL)-4Rα inhibitors, anti-IL-5 monoclonal antibodies (mAbs), anti-IL-17A mAbs, anti-thymic stromal lymphopoietin (TSLP) mAbs, epithelial sodium channel (ENaC) inhibitors, bifunctional M3 receptor muscarinic antagonists/β2-adrenoceptor agonists (MABAs), and anti-Fel d 1 mAbs, were found to be effective in the treatment of asthma, with lung function being the main assessed outcome across the RCTs. Several novel investigational molecules, particularly biologics, seem promising as future disease-modifying agents; nevertheless, further larger studies are required to confirm positive results from Phase I and II RCTs.
Collapse
|
48
|
Chen S, Yu L, Deng Y, Liu Y, Wang L, Li D, Yang K, Liu S, Tao A, Chen R. Early IL-17A Prevention Rather Than Late IL-17A Neutralization Attenuates Toluene Diisocyanate-Induced Mixed Granulocytic Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:528-548. [PMID: 36174994 PMCID: PMC9523423 DOI: 10.4168/aair.2022.14.5.528] [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: 03/19/2022] [Revised: 06/10/2022] [Accepted: 07/05/2022] [Indexed: 06/16/2023]
Abstract
PURPOSE Interleukin (IL)-17A plays a critical role in the pathogenesis of allergic airway inflammation. Yet, the exact roles of IL-17A in asthma are still controversial. Thus, the aim of this study was to dissect the roles of IL-17A in toluene diisocyanate (TDI)-induced mixed granulocytic asthma and to assess the effects of neutralizing antibody in different effector phases on TDI-induced asthma. METHODS IL-17A functions in allergic airway inflammation were evaluated using mice deficient in IL-17A (Il17a-/-) or IL-17A monoclonal antibody (IL-17A mab, intraperitoneally, 50 μg per mouse, 100 μg per mouse). Moreover, the effects of exogenous recombinant IL (rIL)-17A in vivo (murine rIL-17A, intranasally, 1 μg per mouse) and in vitro (human rIL-17A, 100 ng/mL) were investigated. RESULTS TDI-induced mixed granulocytic airway inflammation was IL-17A-dependent because airway hyperreactivity, neutrophil and eosinophil infiltration, airway smooth muscle thickness, epithelium injury, dysfunctional T helper (Th) 2 and Th17 responses, granulocytic chemokine production and mucus overproduction were more markedly reduced in the Il17a-/- mice or by IL-17A neutralization during the sensitization phase of wild-type (WT) mice. By contrast, IL-17A neutralization during the antigen-challenge phase aggravated TDI-induced eosinophils recruitment, with markedly elevated Th2 response. In line with this, instillation of rIL-17 during antigen sensitization exacerbated airway inflammation by promoting neutrophils aggregation, while rIL-17A during the antigen-challenge phase protected the mice from TDI-induced airway eosinophilia. Moreover, rIL-17A exerted distinct effects on eosinophil- or neutrophil-related signatures in vitro. CONCLUSIONS Our data demonstrated that IL-17A was required for the initiation of TDI-induced asthma, but functioned as a negative regulator of established allergic inflammation, suggesting that early abrogation of IL-17A signaling, but not late IL-17A neutralization, may prevent the progression of TDI-induced asthma and could be used as a therapeutic strategy for severe asthmatics in clinical settings.
Collapse
Affiliation(s)
- Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yao Deng
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yuanyuan Liu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lingwei Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Difei Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Kai Yang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ailin Tao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen Key Laboratory of Respiratory Diseases, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China.
| |
Collapse
|
49
|
Luo W, Hu J, Xu W, Dong J. Distinct spatial and temporal roles for Th1, Th2, and Th17 cells in asthma. Front Immunol 2022; 13:974066. [PMID: 36032162 PMCID: PMC9411752 DOI: 10.3389/fimmu.2022.974066] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022] Open
Abstract
Immune response in the asthmatic respiratory tract is mainly driven by CD4+ T helper (Th) cells, represented by Th1, Th2, and Th17 cells, especially Th2 cells. Asthma is a heterogeneous and progressive disease, reflected by distinct phenotypes orchestrated by τh2 or non-Th2 (Th1 and Th17) immune responses at different stages of the disease course. Heterogeneous cytokine expression within the same Th effector state in response to changing conditions in vivo and interlineage relationship among CD4+ T cells shape the complex immune networks of the inflammatory airway, making it difficult to find one panacea for all asthmatics. Here, we review the role of three T helper subsets in the pathogenesis of asthma from different stages, highlighting timing is everything in the immune system. We also discuss the dynamic topography of Th subsets and pathogenetic memory Th cells in asthma.
Collapse
Affiliation(s)
- Weihang Luo
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jindong Hu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Weifang Xu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
- *Correspondence: Jingcheng Dong, ; Weifang Xu,
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- *Correspondence: Jingcheng Dong, ; Weifang Xu,
| |
Collapse
|
50
|
Wang W, Li Y, Fan J, Qu X, Shang D, Qin Q, Xu T, Hamid Q, Dang X, Chang Y, Xu D. MiR-365-3p is a negative regulator in IL-17-mediated asthmatic inflammation. Front Immunol 2022; 13:953714. [PMID: 35958620 PMCID: PMC9361323 DOI: 10.3389/fimmu.2022.953714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/30/2022] [Indexed: 12/07/2022] Open
Abstract
Background Interleukin-17, the major proinflammatory cytokine secreted by Th17 cells, makes essential contribution to pathogenesis of severe asthma, while the detailed mechanisms, especially the involvement of microRNAs which are also important participants in asthma progression, remains largely unclear. Methods In this study, we established a house dust mite (HDM) extract-induced murine asthmatic models and the miRNA expression in the lung tissues of mice were profiled by miRNA microarray assay. The effect of miR-365-3p on IL-17-mediated inflammation was examined by qRT-PCR and immunoblotting analysis. The involvement of ARRB2 as target gene of miR-365-3p was verified by overexpression or RNA interference. Results HDM extract-induced asthmatic inflammation was proved to be IL17-mediated and miR-365-3p was screened out to be the only miRNA exclusively responsive to IL-17. miR-365-3p, whose expression was significantly downregulated upon IL-17 stimulation, was demonstrated to exert remarkable anti-inflammatory effect to decrease IL-17-provoked inflammatory cytokines (KC/IL-8 and IL-6) in both airway epithelial cells and macrophages of murine and human origins, verifying its universal antagonizing activity against IL-17-initiated inflammation across the two species. ARRB2 was characterized as the key target of miR-365-3p to negate IL-17-induced inflammatory cytokines. Conclusion Taken together, our data supported the notion that miR-365-3p, which was diminished by IL-17 in murine and human asthmatic pathogenesis, functioned as an essential negative mediator in IL-17-stimuated inflammatory response by targeting ARRB2, which would shed new light to the understanding and therapeutics thereof of asthmatic inflammation.
Collapse
Affiliation(s)
- Weijia Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Ying Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Jiaqi Fan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Xiaoyan Qu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Dong Shang
- Department of Respiration, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiaohong Qin
- Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Tun Xu
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Qutayba Hamid
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, QC, Canada
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Xiaomin Dang
- Department of Respiration, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ying Chang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Dan Xu, ; Ying Chang,
| | - Dan Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Dan Xu, ; Ying Chang,
| |
Collapse
|