1
|
Park KS, Lässer C, Lötvall J. Extracellular vesicles and the lung: from disease pathogenesis to biomarkers and treatments. Physiol Rev 2025; 105:1733-1821. [PMID: 40125970 DOI: 10.1152/physrev.00032.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/14/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Nanosized extracellular vesicles (EVs) are released by all cells to convey cell-to-cell communication. EVs, including exosomes and microvesicles, carry an array of bioactive molecules, such as proteins and RNAs, encapsulated by a membrane lipid bilayer. Epithelial cells, endothelial cells, and various immune cells in the lung contribute to the pool of EVs in the lung microenvironment and carry molecules reflecting their cellular origin. EVs can maintain lung health by regulating immune responses, inducing tissue repair, and maintaining lung homeostasis. They can be detected in lung tissues and biofluids such as bronchoalveolar lavage fluid and blood, offering information about disease processes, and can function as disease biomarkers. Here, we discuss the role of EVs in lung homeostasis and pulmonary diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, pulmonary fibrosis, and lung injury. The mechanistic involvement of EVs in pathogenesis and their potential as disease biomarkers are discussed. Finally, the pulmonary field benefits from EVs as clinical therapeutics in severe pulmonary inflammatory disease, as EVs from mesenchymal stem cells attenuate severe respiratory inflammation in multiple clinical trials. Further, EVs can be engineered to carry therapeutic molecules for enhanced and broadened therapeutic opportunities, such as the anti-inflammatory molecule CD24. Finally, we discuss the emerging opportunity of using different types of EVs for treating severe respiratory conditions.
Collapse
Affiliation(s)
- Kyong-Su Park
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at the Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
2
|
Matloubi M, Sedaghat F, Shan L, Basu S, Halayko AJ, Gounni AS. The dichotomous impacts of Semaphorin3E deficiency on exacerbating airway hyperresponsiveness, remodelling, and inflammation in type-2 low and type-2 high asthma models. PLoS One 2025; 20:e0322353. [PMID: 40512736 PMCID: PMC12165363 DOI: 10.1371/journal.pone.0322353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 05/23/2025] [Indexed: 06/16/2025] Open
Abstract
Semaphorin3E has shown promise in alleviating the severity of asthma in preclinical studies; however, its role in the chronic features of type 2-low asthma remains unclear. Therefore, we aimed to investigate the role of Sema3E in a mouse model of severe asthma that exhibits a mix of granulocytic inflammation with neutrophils dominance and compared the results with those from the type-2 high eosinophilic asthma model. Sema3E knockout (KO) and wild-type (WT) mice were subjected to type-2 low and type-2 high regimens using house dust mite (HDM) combined with cyclic-di-GMP or HDM alone, respectively. Airway hyperresponsiveness parameters were measured using the FlexiVent ventilator. Bronchoalveolar lavage fluid cell phenotyping was performed by flowcytometry. Additionally, cytokines and antibodies were quantified using Mesoscale and ELISA. Mucus overproduction and goblet cell hyperplasia were visualized by Periodic-acid-Schiff staining. In comparison to WT mice, Sema3E KO mice exhibited an enhanced tissue resistance and tissue elastance in the type 2-low asthma model. Concurrently, Sema3E KO mice that were subjected to the type-2 low asthma model demonstrated an elevated presence of pulmonary neutrophils, dendritic cells, CD4 T cells, as well as increased levels of IL-17, TNF, IL-1β, CXCL-8, and MCP-1/CCL2 in comparison to their WT counterparts. However, in the type-2 high model, Sema3E KO mice exhibited a significant increase in goblet cell numbers and mucus overproduction, as well as enhancements in the number of eosinophils, IgE-producing B cells, and IL-4 levels compared to WT mice, highlighting the homeostatic role of Sema3E in the distinct immune niche of type-2 low and type-2 high asthma. Overall, our data showed that Sema3E is critical in modulating AHR, airway inflammation, and tissue remodelling in type 2 low and type 2 high phenotypes of asthma. The Sema3E regulatory network varies depending on the immunization regimen, affecting distinct parameters in type-2 low and type-2 high asthma models.
Collapse
Affiliation(s)
- Mojdeh Matloubi
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Fatemeh Sedaghat
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lianyu Shan
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Abdelilah S. Gounni
- Department of Immunology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
3
|
Dorscheid D, Gauvreau GM, Georas SN, Hiemstra PS, Varricchi G, Lambrecht BN, Marone G. Airway epithelial cells as drivers of severe asthma pathogenesis. Mucosal Immunol 2025; 18:524-536. [PMID: 40154790 DOI: 10.1016/j.mucimm.2025.03.003] [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: 05/20/2024] [Revised: 01/31/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Our understanding of the airway epithelium's role in driving asthma pathogenesis has evolved over time. From being regarded primarily as a physical barrier that could be damaged via inflammation, the epithelium is now known to actively contribute to asthma development through interactions with the immune system. The airway epithelium contains multiple cell types with specialized functions spanning barrier action, mucociliary clearance, immune cell recruitment, and maintenance of tissue homeostasis. Environmental insults may cause direct or indirect injury to the epithelium leading to impaired barrier function, epithelial remodelling, and increased release of inflammatory mediators. In severe asthma, the epithelial barrier repair process is inhibited and the response to insults is exaggerated, driving downstream inflammation. Genetic and epigenetic mechanisms also maintain dysregulation of the epithelial barrier, adding to disease chronicity. Here, we review the role of the airway epithelium in severe asthma and how targeting the epithelium can contribute to asthma treatment.
Collapse
Affiliation(s)
- Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steve N Georas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gilda Varricchi
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
| | - Bart N Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.
| | - Gianni Marone
- Department of Translational Medical Sciences (DiSMeT) and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy.
| |
Collapse
|
4
|
Althoff MD, Holguin F. Care of the Patient With Asthma. Ann Intern Med 2025; 178:ITC81-ITC96. [PMID: 40489781 DOI: 10.7326/annals-25-01034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/11/2025] Open
Abstract
Nearly 8% of the U.S. population is diagnosed with asthma, leading to more than 5 million office visits and 1 million emergency department visits annually. Both outpatient and inpatient internal medicine clinicians treat asthma frequently, but nuances in diagnosis and management have emerged. This article highlights many of these developments.
Collapse
Affiliation(s)
- Meghan D Althoff
- University of Colorado Anschutz Medical Campus, Aurora, Colorado (M.D.A., F.H.)
| | - Fernando Holguin
- University of Colorado Anschutz Medical Campus, Aurora, Colorado (M.D.A., F.H.)
| |
Collapse
|
5
|
Christopher-Hayes NJ, Ghetti S. Neurocognitive risks of asthma during childhood. Dev Cogn Neurosci 2025; 73:101564. [PMID: 40349572 PMCID: PMC12139513 DOI: 10.1016/j.dcn.2025.101564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 03/23/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
The impact of chronic medical conditions on the developing brain has gained recent attention, but the neurocognitive risks associated with asthma, which has high prevalence in childhood, are still largely unknown. Recent findings have underscored that children with asthma may be at higher risk for developing cognitive difficulties. In this review, we examine the pathophysiology of asthma and its associations with brain and cognitive development based on rodent models and relatively scant research in humans. We also examine risk factors that may exacerbate asthma symptoms and neurocognitive outcomes, and we discuss why children may be particularly vulnerable to asthma-related neurocognitive consequences. We conclude by providing a framework for future research.
Collapse
Affiliation(s)
- Nicholas J Christopher-Hayes
- Center for Mind and Brain, University of California, Davis, CA 95618, USA; Department of Psychology, University of California, Davis, CA 95616, USA.
| | - Simona Ghetti
- Center for Mind and Brain, University of California, Davis, CA 95618, USA; Department of Psychology, University of California, Davis, CA 95616, USA.
| |
Collapse
|
6
|
Rabin RL, Altman MC, Arshad SH, Beger RD, Frischmeyer-Guerrerio PA, Goleva E, Hamilton RG, Khurana Hershey GK, Shamji MH, Sampson HA, Santos AF, Shreffler WG, Togias A, Vieths S, Wambre E, Wenzel SE, Hise K, Lee J, Tripathi A, Slater JE. Biomarker-driven drug development for allergic diseases and asthma: An FDA public workshop. J Allergy Clin Immunol 2025; 155:1753-1766. [PMID: 40154576 PMCID: PMC12145241 DOI: 10.1016/j.jaci.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
The US Food and Drug Administration (FDA) hosted a workshop on February 22, 2024, to discuss the status of biomarkers in drug development for allergic asthma and food allergy. The workshop provided a forum for open discussion among regulators, academicians, National Institutes of Health staff and industry to inform stakeholders of the requirements for the FDA to adopt a biomarker as a surrogate end point for a clinical trial, and to inform FDA of the status of various biomarkers in development. The workshop was divided into 3 sessions: (1) FDA and European Union regulators discussing regulatory perspectives on use of biomarkers in drug development programs, (2) investigators discussing biomarkers for pediatric and adult asthma, and (3) investigators discussing biomarkers for food allergy. In this report, we review the information presented at the workshop and summarize the current status of potential biomarkers for these allergic diseases.
Collapse
Affiliation(s)
- Ronald L Rabin
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md.
| | | | - S Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Richard D Beger
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Ark
| | | | - Elena Goleva
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | | | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hugh A Sampson
- Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course and Population Sciences & Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, and the Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Wayne G Shreffler
- Food Allergy Center, Division of Pediatric Allergy and Immunology, and the Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Stefan Vieths
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Erik Wambre
- Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, School of Public Health, University of Pittsburgh, Pittsburgh, Pa
| | - Kathleen Hise
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Joohee Lee
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Anubha Tripathi
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| | - Jay E Slater
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Md
| |
Collapse
|
7
|
Zhang X, Tao Y, Song Z, Sun L, Sun Y, Jin R, Chang C. Antibody response to SARS-CoV-2 vaccine in patients with asthma. J Asthma 2025; 62:1082-1091. [PMID: 39932243 DOI: 10.1080/02770903.2025.2458523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/28/2024] [Accepted: 01/21/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Viral infections constitute a primary trigger for asthma exacerbations. While vaccines protect against viral infections by eliciting a Th1 immune response, the impact of the asthmatic immune milieu which is characterized by Th2 cytokine dominance and elevated IgE levels on post-vaccination antibody production remains elusive. Therefore, vaccination protocols tailored to asthma patients need to be formulated. METHODS The levels of IgG specific for SARS-CoV-2 S protein were measured in the sera of vaccinated and unvaccinated individuals by ELISA. The differences in antibody titers between asthma patients and healthy controls, as well as among distinct asthma subgroups were analyzed. RESULTS The vaccinated individuals had significantly elevated serum antibody levels compared to their unvaccinated counterparts. There were no significant differences in the antibody titers of asthma patients and healthy controls after completion of the three-dose vaccination regimen. Furthermore, no discernible variations in antibody titers were detected among the asthma subgroups. CONCLUSION Asthma patients can safely adhere to the same vaccination strategies as the general healthy population, negating the need for any specialized vaccination protocols based solely on the asthmatic immune landscape.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Yifei Tao
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhu Song
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Lina Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| | - Rong Jin
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Chun Chang
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing, China
| |
Collapse
|
8
|
Reza MI, Ambhore NS. Inflammation in Asthma: Mechanistic Insights and the Role of Biologics in Therapeutic Frontiers. Biomedicines 2025; 13:1342. [PMID: 40564060 DOI: 10.3390/biomedicines13061342] [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: 04/19/2025] [Revised: 05/25/2025] [Accepted: 05/28/2025] [Indexed: 06/28/2025] Open
Abstract
Asthma is a chronic and multifaceted respiratory condition that affects over 300 million individuals across the globe. It is characterized by persistent inflammation of the airways, which leads to episodes of wheezing, breathlessness, chest tightness, and coughing. The most prevalent form of asthma is classified as Type 2 or T2-high asthma. In this variant, the immune response is heavily driven by eosinophils, mast cells, and T-helper 2 (Th2) cells. These components release a cascade of cytokines, including interleukin-4 (IL-4), interleukin-5 (IL-5), and interleukin-13 (IL-13). This release promotes several processes: the production of immunoglobulin E (IgE), which is integral to allergic responses; the recruitment of eosinophils-white blood cells that contribute to inflammation and tissue damage. Conversely, non-Type 2 or T2-low asthma is typically associated with a different inflammatory profile characterized by neutrophilic inflammation. This type of asthma is driven by T-helper 1 (Th1) and T-helper 17 (Th17) immune responses, which are often present in older adults, smokers, and those suffering from more severe manifestations of the disease. Among asthmatic patients, approximately 80-85% of cases are classified as T2-high asthma, while only 15-20% are T2-low asthma. Treatment of asthma focuses heavily on controlling inflammation. Inhaled corticosteroids remain the cornerstone therapy for managing T2-high asthma. For more severe or treatment-resistant cases, biologic therapies targeting specific inflammatory pathways, such as anti-IgE (omalizumab), anti-IL-5 (mepolizumab, benralizumab), and anti-IL-4/IL-13 (dupilumab), have shown great promise. For T2-low asthma, macrolide antibiotics like azithromycin and other novel therapies are being explored. This article reviews the safety, efficacy, and indications of the currently approved biologics and discusses potential novel biologics for asthma.
Collapse
Affiliation(s)
- Mohammad Irshad Reza
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Nilesh S Ambhore
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
9
|
Donoghue LJ, Benner C, Chang D, Irudayanathan FJ, Pendergrass RK, Yaspan BL, Mahajan A, McCarthy MI. Integration of biobank-scale genetics and plasma proteomics reveals evidence for causal processes in asthma risk and heterogeneity. CELL GENOMICS 2025; 5:100840. [PMID: 40187354 DOI: 10.1016/j.xgen.2025.100840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Hundreds of genetic associations for asthma have been identified, yet translating these findings into mechanistic insights remains challenging. We leveraged plasma proteomics from the UK Biobank Pharma Proteomics Project (UKB-PPP) to identify biomarkers and effectors of asthma risk or heterogeneity using genetic causal inference approaches. We identified 609 proteins associated with asthma status (269 proteins after controlling for body mass index [BMI] and smoking). Analysis of genetically predicted protein levels identified 70 proteins with putative causal roles in asthma risk, including known drug targets and proteins without prior genetic evidence in asthma (e.g., GCHFR, TDRKH, and CLEC7A). The genetic architecture of causally associated proteins provided evidence for a Toll-like receptor (TLR)1-interleukin (IL)-27 asthma axis. Lastly, we identified evidence of causal relationships between proteins and heterogeneous aspects of asthma biology, including between TSPAN8 and neutrophil counts. These findings illustrate that integrating biobank-scale genetics and plasma proteomics can provide a framework to identify therapeutic targets and mechanisms underlying disease risk and heterogeneity.
Collapse
Affiliation(s)
| | | | - Diana Chang
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
10
|
Makrufardi F, Rusmawatiningtyas D, Murni IK, Arguni E, Lin YC, Ho KF, Chung KF, Lin SC, Chuang HC. Seasonal variation of pediatric asthma exacerbations and its association with asthma phenotypes. Pediatr Res 2025:10.1038/s41390-025-04073-2. [PMID: 40335642 DOI: 10.1038/s41390-025-04073-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND The objective of this study was to examine the associations of blood inflammatory phenotypes with acute pediatric asthma exacerbations during different seasons and the COVID-19 pandemic. METHODS A retrospective study was conducted involving 32,160 pediatric asthma patients from January 2008 to December 2021. Asthma blood inflammatory phenotypes were categorized based on low (L) and high (H) eosinophils (E) and neutrophils (N) (LBE/HBE: ≥ 0.25 × 109/L and LBN/HBN: ≥ 5 × 109/L, respectively) and logistic regression was used to examine the odds ratio (OR) of outcome variables. RESULTS A 109/L increase of neutrophils and eosinophils was associated with a 1.015-fold (95% CI: 1.009-1.021) and a 1.057-fold increase in the OR (95% CI: 1.026-1.088) for asthma exacerbations of hospitalized pediatric asthma patients. An increase in HBE/LBN phenotype was associated with a respective 1.232-fold (95% CI: 1.081-1.404) and 1.248-fold (95% CI: 1.101-1.414) increase in the OR for asthma exacerbations of hospitalized pediatric asthma patients before the COVID-19 pandemic in the winter and autumn seasons. However, an increase of LBE/LBN phenotype was associated with a respective 0.873-fold (95% CI: 0.769-0.991), 0.872-fold (95% CI: 0.771-0.986), and 0.813-fold (95% CI: 0.709-0.932) decrease in the OR for asthma exacerbations in the winter, spring and summer seasons. CONCLUSIONS HBE/LBN phenotype had a higher risk of asthma exacerbations among hospitalized pediatric asthma patients in the winter and autumn, while LBE/LBN phenotype had a lower risk in the winter, spring, and summer. IMPACT Blood eosinophils and neutrophils have been indicated to have a potential influence on pediatric asthma development and severity. HBE/LBN phenotype was associated with increased asthma exacerbations among hospitalized pediatric asthma patients during winter and autumn. Eosinophil and neutrophil predominance exhibited a higher influence on pediatric asthma exacerbations.
Collapse
Affiliation(s)
- Firdian Makrufardi
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Desy Rusmawatiningtyas
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Indah Kartika Murni
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Eggi Arguni
- Department of Child Health, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada - Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Yuan-Chien Lin
- Department of Civil Engineering, National Central University, Taoyuan City, Taiwan
| | - Kin-Fai Ho
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sheng-Chieh Lin
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Division of Allergy, Asthma, and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| | - Hsiao-Chi Chuang
- National Heart and Lung Institute, Imperial College London, London, UK.
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
11
|
Mahay G, Zysman M, Guibert N, Barnig C, Guilleminault L, Dupin C. Long-acting muscarinic antagonists (LAMA) in asthma: What is the best strategy? Respir Med Res 2025; 87:101157. [PMID: 39946978 DOI: 10.1016/j.resmer.2025.101157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/25/2024] [Accepted: 01/21/2025] [Indexed: 06/02/2025]
Abstract
The use of long-acting muscarinic antagonists (LAMA) in asthma is supported by their mechanism of action and evidence of drug synergy with inhaled corticosteroids ± long-acting β-agonists. This review discusses the scientific rationale, clinical data, and recommendations for the use of LAMA in the asthma therapeutic strategy. Adding a LAMA to a dual therapy with an inhaled corticosteroid and long-acting β-agonist has been shown to reduce exacerbations, increase asthma control, and improve quality of life, with a good safety profile. In addition, using a single inhaler device containing multiple drugs enhances patients' adherence to therapy. Some predictive factors of the efficacy of this triple therapy have been suggested in the literature: patients with a history of at least one exacerbation within the past 12 months, male patients, those younger than 65 years, and non-smokers have been reported to have a greater improvement from baseline forced expiratory volume in 1 second (FEV1) compared with patients without these characteristics, while patients with high bronchial hyperresponsiveness and persistent airway limitations (PAL) seem to show better gains in the exacerbation rate. However, eosinophil levels do not seem to predict the efficacy of LAMA. The role and long-term benefits of LAMA combined with biologic therapy in severe asthma remain uncertain, with more clinical data needed.
Collapse
Affiliation(s)
- Guillaume Mahay
- Service de Pneumologie, Oncologie thoracique et soins intensifs respiratoires, CHU Rouen, Rouen, France
| | - Maeva Zysman
- Service de Pneumologie, CHU Haut-Lévèque, Bordeaux, France; Univ. Bordeaux, Centre de Recherche cardio-thoracique, INSERM U1045, CIC 1401, Pessac, France; INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), France
| | - Nicolas Guibert
- Pôle des voies respiratoires, CHU de Toulouse, Toulouse, France
| | - Cindy Barnig
- INSERM, EFS BFC, LabEx LipSTIC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Univ. Bourgogne Franche-Comté, Besançon, France; Service de Pneumologie, Oncologie thoracique et allergologie respiratoire, CHRU Besançon, Besançon, France; INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), France
| | - Laurent Guilleminault
- Pôle des voies respiratoires, CHU de Toulouse, Toulouse, France; INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), France; Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, Toulouse, France
| | - Clairelyne Dupin
- INSERM, F-CRIN, Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science (CRISALIS), France; Service de Pneumologie A, Centre de Référence des Maladies Pulmonaires Rares, Hôpital Bichat, AP-HP, Paris, France; INSERM U1152, Paris, France.
| |
Collapse
|
12
|
Uehara S, Hirai K, Shirai T, Akamatsu T, Itoh K. PI3K pathway activation in severe asthma is linked to steroid insensitivity and adverse outcomes. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100439. [PMID: 40125453 PMCID: PMC11928809 DOI: 10.1016/j.jacig.2025.100439] [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: 09/27/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 03/25/2025]
Abstract
Background Patients with severe asthma may demonstrate reduced sensitivity to steroid treatment. However, the implications of this reduced responsiveness for clinical outcomes and the underlying mechanisms remain unclear. Objective The aim of this study was to investigate whether steroid sensitivity in patients with asthma is related to severity and clinical outcomes and to elucidate the role of inflammatory pathways in reducing steroid sensitivity. Methods This observational study of 169 asthma patients, with 161 followed for 1 year, involved isolation of peripheral blood mononuclear cells. These cells were treated with dexamethasone, and the mRNA expression of FKBP5, which is a marker of steroid sensitivity, was measured. To explore the mechanism underlying the reduced steroid sensitivity, cells were exposed to PI3K and MAPK inhibitors in combination with dexamethasone. Results A total of 53 patients diagnosed with severe asthma exhibited markedly diminished sensitivity to steroids compared with those with nonsevere asthma. Reduced steroid sensitivity has emerged as a critical risk factor for failure to experience clinical remission and exacerbation. This relationship between reduced steroid sensitivity and disease severity and adverse outcomes was confirmed at the 1-year follow-up. Mechanistic investigations revealed that the degree of recovery from steroid sensitivity after PI3Kδ/γ inhibitor treatment was significantly greater in patients with severe asthma than in those with nonsevere asthma, a finding confirmed at the 1-year follow-up. Conclusions Patients with severe asthma demonstrate reduced steroid sensitivity, which results in unfavorable clinical outcomes. Conversely, inhibition of the PI3K pathway significantly improves steroid sensitivity.
Collapse
Affiliation(s)
- Sekiko Uehara
- Department of Clinical Pharmacology and Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Keita Hirai
- Department of Clinical Pharmacology and Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
- Department of Clinical Pharmacology and Therapeutics, Shinshu University Graduate School of Medicine, Matsumoto, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan
| | - Toshihiro Shirai
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Taisuke Akamatsu
- Department of Respiratory Medicine, Shizuoka General Hospital, Shizuoka, Japan
| | - Kunihiko Itoh
- Department of Clinical Pharmacology and Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
- Laboratory of Clinical Pharmacogenomics, Shizuoka General Hospital, Shizuoka, Japan
| |
Collapse
|
13
|
Charles N, Blank U. IgE-Mediated Activation of Mast Cells and Basophils in Health and Disease. Immunol Rev 2025; 331:e70024. [PMID: 40165512 DOI: 10.1111/imr.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Type 2-mediated immune responses protect the body against environmental threats at barrier surfaces, such as large parasites and environmental toxins, and facilitate the repair of inflammatory tissue damage. However, maladaptive responses to typically nonpathogenic substances, commonly known as allergens, can lead to the development of allergic diseases. Type 2 immunity involves a series of prototype TH2 cytokines (IL-4, IL-5, IL-13) and alarmins (IL-33, TSLP) that promote the generation of adaptive CD4+ helper Type 2 cells and humoral products such as allergen-specific IgE. Mast cells and basophils are integral players in this network, serving as primary effectors of IgE-mediated responses. These cells bind IgE via high-affinity IgE receptors (FcεRI) expressed on their surface and, upon activation by allergens, release a variety of mediators that regulate tissue responses, attract and modulate other inflammatory cells, and contribute to tissue repair. Here, we review the biology and effector mechanisms of these cells, focusing primarily on their role in mediating IgE responses in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Nicolas Charles
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine Site Bichat, Paris, France
- Laboratoire d'Excellence Inflamex, Université Paris Cité, Paris, France
| | - Ulrich Blank
- Université Paris Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS EMR8252, Faculté de Médecine Site Bichat, Paris, France
- Laboratoire d'Excellence Inflamex, Université Paris Cité, Paris, France
| |
Collapse
|
14
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [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: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Ramonell RP, Oriss TB, McCreary-Partyka JC, Kale SL, Brandon NR, Ross MA, Gauthier MC, Yue M, Nee TJ, Das S, Chen W, Joglekar AV, Ray P, St Croix CM, Rajasundaram D, Wenzel SE, Ray A. CD8+ TEMRAs in severe asthma associate with asthma symptom duration and escape proliferation arrest. JCI Insight 2025; 10:e185061. [PMID: 40048261 PMCID: PMC12016929 DOI: 10.1172/jci.insight.185061] [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: 07/19/2024] [Accepted: 03/04/2025] [Indexed: 04/23/2025] Open
Abstract
Aberrant immune response is a hallmark of asthma, with 5%-10% of patients suffering from severe disease exhibiting poor response to standard treatment. A better understanding of the immune responses contributing to disease heterogeneity is critical for improving asthma management. T cells are major players in the orchestration of asthma, in both mild and severe disease, but it is unclear whether specific T cell subsets influence asthma symptom duration. Here we show a significant association of airway CD8+ effector memory T cells re-expressing CD45RA (TEMRAs), but not CD8+CD45RO+ or tissue-resident memory T cells, with asthma duration in patients with severe asthma (SA) but not mild to moderate asthma (MMA). Higher frequencies of IFN-γ+CD8+ TEMRAs compared with IFN-γ+CD45RO+ T cells were detected in SA airways, and the TEMRAs from patients with SA but not MMA proliferated ex vivo, although both expressed cellular senescence-associated biomarkers. Prompted by the transcriptomic profile of SA CD8+ TEMRAs and proliferative response to IL-15, airway IL15 expression was higher in patients with SA compared with MMA. Additionally, IL15 expression in asthmatic airways negatively correlated with lung function. Our findings add what we believe is a new dimension to understanding asthma heterogeneity, identifying IL-15 as a potential target for treatment.
Collapse
Affiliation(s)
- Richard P. Ramonell
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | - Timothy B. Oriss
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | - Sagar L. Kale
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | - Mark A. Ross
- Department of Cell Biology
- Center for Biological Imaging
| | - Marc C. Gauthier
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | | | - Taylor J. Nee
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | - Sudipta Das
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | | | - Prabir Ray
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
| | | | | | - Sally E. Wenzel
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anuradha Ray
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
| |
Collapse
|
16
|
Sahnoon L, Bajbouj K, Mahboub B, Hamoudi R, Hamid Q. Targeting IL-13 and IL-4 in Asthma: Therapeutic Implications on Airway Remodeling in Severe Asthma. Clin Rev Allergy Immunol 2025; 68:44. [PMID: 40257546 PMCID: PMC12011922 DOI: 10.1007/s12016-025-09045-2] [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: 03/19/2025] [Indexed: 04/22/2025]
Abstract
Asthma is a chronic respiratory disorder affecting individuals across all age groups. It is characterized by airway inflammation and remodeling and leads to progressive airflow restriction. While corticosteroids remain a mainstay therapy, their efficacy is limited in severe asthma due to genetic and epigenetic alterations, as well as elevated pro-inflammatory cytokines interleukin-4 (IL-4), interleukin-13 (IL-13), and interleukin-5 (IL-5), which drive structural airway changes including subepithelial fibrosis, smooth muscle hypertrophy, and goblet cell hyperplasia. This underscores the critical need for biologically targeted therapies. This review systematically examines the roles of IL-4 and IL-13, key drivers of type-2 inflammation, in airway remodeling and their potential as therapeutic targets. IL-4 orchestrates eosinophil recruitment, immunoglobulin class switching, and Th2 differentiation, whereas IL-13 directly modulates structural cells, including fibroblasts and epithelial cells, to promote mucus hypersecretion and extracellular matrix (ECM) deposition. Despite shared signaling pathways, IL-13 emerges as the dominant cytokine in remodeling processes including mucus hypersecretion, fibrosis and smooth muscle hypertrophy. While IL-4 primarily amplifies inflammatory cascades by driving IgE switching, promoting Th2 cell polarization that sustain cytokine release, and inducing chemokines to recruit eosinophils. In steroid-resistant severe asthma, biologics targeting IL-4/IL-13 show promise in reducing exacerbations and eosinophilic inflammation. However, their capacity to reverse established remodeling remains inconsistent, as clinical trials prioritize inflammatory biomarkers over long-term structural outcomes. This synthesis highlights critical gaps in understanding the durability of IL-4/IL-13 inhibition on airway structure and advocates for therapies combining biologics with remodeling-specific strategies. Through the integration of mechanistic insights and clinical evidence, this review emphasizes the need for long-term studies utilizing advanced imaging, histopathological techniques, and patient-reported outcomes to evaluate how IL-4/IL-13-targeted therapies alter airway remodeling and symptom burden, thereby informing more effective treatment approaches for severe, steroid-resistant asthma.
Collapse
Affiliation(s)
- Lina Sahnoon
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Bassam Mahboub
- Rashid Hospital, Dubai Health, 4545, Dubai, United Arab Emirates
| | - Rifat Hamoudi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Division of Surgery and Interventional Science, University College London, London, UK.
- Biomedically Informed Artificial Intelligence Laboratory, University of Sharjah, Sharjah, United Arab Emirates.
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Meakins-Christie Laboratories, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
17
|
Zhong X, Mitchell R, Billstrand C, Thompson EE, Sakabe NJ, Aneas I, Salamone IM, Gu J, Sperling AI, Schoettler N, Nóbrega MA, He X, Ober C. Integration of functional genomics and statistical fine-mapping systematically characterizes adult-onset and childhood-onset asthma genetic associations. Genome Med 2025; 17:35. [PMID: 40205616 PMCID: PMC11983851 DOI: 10.1186/s13073-025-01459-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified hundreds of loci underlying adult-onset asthma (AOA) and childhood-onset asthma (COA). However, the causal variants, regulatory elements, and effector genes at these loci are largely unknown. METHODS We performed heritability enrichment analysis to determine relevant cell types for AOA and COA, respectively. Next, we fine-mapped putative causal variants at AOA and COA loci. To improve the resolution of fine-mapping, we integrated ATAC-seq data in blood and lung cell types to annotate variants in candidate cis-regulatory elements (CREs). We then computationally prioritized candidate CREs underlying asthma risk, experimentally assessed their enhancer activity by massively parallel reporter assay (MPRA) in bronchial epithelial cells (BECs) and further validated a subset by luciferase assays. Combining chromatin interaction data and expression quantitative trait loci, we nominated genes targeted by candidate CREs and prioritized effector genes for AOA and COA. RESULTS Heritability enrichment analysis suggested a shared role of immune cells in the development of both AOA and COA while highlighting the distinct contribution of lung structural cells in COA. Functional fine-mapping uncovered 21 and 67 credible sets for AOA and COA, respectively, with only 16% shared between the two. Notably, one-third of the loci contained multiple credible sets. Our CRE prioritization strategy nominated 62 and 169 candidate CREs for AOA and COA, respectively. Over 60% of these candidate CREs showed open chromatin in multiple cell lineages, suggesting their potential pleiotropic effects in different cell types. Furthermore, COA candidate CREs were enriched for enhancers experimentally validated by MPRA in BECs. The prioritized effector genes included many genes involved in immune and inflammatory responses. Notably, multiple genes, including TNFSF4, a drug target undergoing clinical trials, were supported by two independent GWAS signals, indicating widespread allelic heterogeneity. Four out of six selected candidate CREs demonstrated allele-specific regulatory properties in luciferase assays in BECs. CONCLUSIONS We present a comprehensive characterization of causal variants, regulatory elements, and effector genes underlying AOA and COA genetics. Our results supported a distinct genetic basis between AOA and COA and highlighted regulatory complexity at many GWAS loci marked by both extensive pleiotropy and allelic heterogeneity.
Collapse
Affiliation(s)
- Xiaoyuan Zhong
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Robert Mitchell
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Emma E Thompson
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Isabella M Salamone
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Jing Gu
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nathan Schoettler
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Marcelo A Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
18
|
Jha RS. Achieving clinical remission in asthma with mepolizumab: a subanalysis on vitamin D as a predictor of response. J Asthma 2025; 62:549-553. [PMID: 39432687 DOI: 10.1080/02770903.2024.2419435] [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: 07/18/2024] [Revised: 09/12/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE Mepolizumab, an anti-IL-5 monoclonal antibody, has shown promise in reducing exacerbations and steroid dependency in severe eosinophilic asthma. This study aims to evaluate the effectiveness of mepolizumab in achieving clinical remission in asthma over 12 months and explore Vitamin D levels as a predictor of response. METHOD We assessed Asthma Control Questionnaire (ACQ) scores, spirometry, number of exacerbations, oral corticosteroid (OCS) use, and inhaled corticosteroid (ICS) use in 32 patients, observing significant clinical improvements. Data were collected 1 year prior to starting mepolizumab and one year after starting mepolizumab. Nasal polyps were not seen in all the patients, and computed tomography of para-nasal sinuses are not available for all the patients, so nasal polyps status are not evaluated to avoid any bias. RESULT Our results indicate that 12 patients achieved clinical remission after starting mepolizumab, with a strong correlation between higher Vitamin D levels and positive treatment outcomes. This suggests that optimizing Vitamin D levels could enhance the response to mepolizumab in asthma patients, and help in achieving better asthma control. CONCLUSION Mepolizumab is an effective treatment for severe eosinophilic asthma, significantly improving clinical outcomes and reducing corticosteroid use. This study highlights the importance of Vitamin D as a predictor of response to mepolizumab, suggesting that higher Vitamin D levels may enhance treatment efficacy.
Collapse
Affiliation(s)
- Ravi Shekhar Jha
- Department of Pulmonology, Fortis Escorts Hospital, Faridabad, India
| |
Collapse
|
19
|
Loponen J, Vähätalo I, Tuomisto LE, Niemelä O, Lehtimäki L, Hämäläinen M, Moilanen E, Kankaanranta H, Ilmarinen P. Physical exercise, systemic inflammation and adult-onset asthma: a 12-year follow-up study. J Asthma 2025; 62:714-724. [PMID: 39636329 DOI: 10.1080/02770903.2024.2438096] [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: 11/06/2023] [Revised: 09/30/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Objective: Physical exercise in treatment of asthma is scarcely studied with no clear exercise guidelines for asthmatics. We aimed to investigate the associations between physical exercise frequency, systemic inflammation and asthma control. This has not been previously studied in adult-onset asthma. Methods: This study is part of Seinäjoki Adult Asthma Study (SAAS), where 203 patients with adult-onset asthma were evaluated in 2012-2013. Exercise frequency was recorded with a structured lifestyle questionnaire. Study population was divided into two categories by exercise frequency: Low-frequency group exercised ≤2 times/week and high frequency group >2 times/week. Blood inflammatory markers were measured and IL-6 > 1.55 pg/ml and hs-CRP > 4.12 mg/l indicated systemic inflammation. Results: High-exercise frequency group had lower levels of hs-CRP (p = 0.007), IL-6 (p = 0.015), suPAR (p = 0.008) and adipsin (p = 0.031) and higher levels of adiponectin (p = 0.010) than low-exercise frequency group. In logistic multivariate regression models, higher-exercise frequency lowered odds for elevated hs-CRP (OR = 0.37, 95% CI 0.15-0.94) and IL-6 levels (OR = 0.43, 95% CI 0.20-0.91), after adjusting for possible confounding factors. There was no difference in lung function tests, asthma control test or airways questionnaire 20 scores between the exercise frequency groups. However, differences were found in single symptom questions; high-exercise frequency group had less symptoms during light housework and laughing but experienced more limitation of activity in self-reports. Conclusions: Higher-exercise frequency is associated with lower level of systemic inflammation in patients with adult-onset asthma but no clear association was found to asthma outcomes. Exercise frequency may be associated with lesser amount of some individual asthma symptoms.
Collapse
Affiliation(s)
- Juho Loponen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Health Centre Mehiläinen Tampere Keskusta, Tampere, Finland
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Iida Vähätalo
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Leena E Tuomisto
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Onni Niemelä
- Department of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital and University of Tampere, Seinäjoki, Finland
| | - Lauri Lehtimäki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Allergy Centre, Tampere University Hospital, Tampere, Finland
| | - Mari Hämäläinen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Eeva Moilanen
- The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Hannu Kankaanranta
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
- Krefting Research Centre, Institute of Medicine, Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Pinja Ilmarinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Respiratory Medicine, Seinäjoki Central Hospital, Seinäjoki, Finland
| |
Collapse
|
20
|
Ravi VR, Korkmaz FT, De Ana CL, Lu L, Shao FZ, Odom CV, Barker KA, Ramanujan A, Niszczak EN, Goltry WN, Martin IMC, Ha CT, Quinton LJ, Jones MR, Fine A, Welch JD, Chen F, Belkina AC, Mizgerd JP, Shenoy AT. Lung CD4 + resident memory T cells use airway secretory cells to stimulate and regulate onset of allergic airway neutrophilic disease. Cell Rep 2025; 44:115294. [PMID: 39965565 PMCID: PMC12011213 DOI: 10.1016/j.celrep.2025.115294] [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: 07/04/2024] [Revised: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Neutrophilic asthma is a vexing disease, but mechanistic and therapeutic advancements will require better models of allergy-induced airway neutrophilia. Here, we find that periodic ovalbumin (OVA) inhalation in sensitized mice elicits rapid allergic airway inflammation and pathophysiology mimicking neutrophilic asthma. OVA-experienced murine lungs harbor diverse clusters of CD4+ resident memory T (TRM) cells, including unconventional RORγtnegative/low T helper 17 (TH17) cells. Acute OVA challenge instigates interleukin (IL)-17A secretion from these TRM cells, driving CXCL5 production from Muc5achigh airway secretory cells, leading to destructive airway neutrophilia. The TRM and epithelial cell signals discovered herein are also observed in adult human asthmatic airways. Epithelial antigen presentation regulates this biology by skewing TRM cells toward TH2 and TH1 fates so that TH1-related interferon (IFN)-γ suppresses IL-17A-driven, CXCL5-mediated airway neutrophilia. Concordantly, in vivo IFN-γ supplementation improves disease outcomes. Thus, using our model of neutrophilic asthma, we identify lung epithelial-CD4+ TRM cell crosstalk as a key rheostat of allergic airway neutrophilia.
Collapse
Affiliation(s)
- Vijay Raaj Ravi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lu Lu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Feng-Zhi Shao
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Christine V Odom
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kimberly A Barker
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Aditya Ramanujan
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Emma N Niszczak
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wesley N Goltry
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ian M C Martin
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Catherine T Ha
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alan Fine
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Felicia Chen
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Anna C Belkina
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Anukul T Shenoy
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Hamada Y, Gibson PG, Clark VL, Lewthwaite H, Fricker M, Thomas D, McDonald VM. Dysfunctional Breathing and Depression Are Core Extrapulmonary and Behavior/Risk Factor Traits in Type 2-High Severe Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025:S2213-2198(25)00267-3. [PMID: 40120804 DOI: 10.1016/j.jaip.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Obesity and smoking are core treatable traits (TTs) in type 2 (T2)-low asthma, contributing to its pathophysiology. In contrast, core extrapulmonary and behavior/risk factor traits remain unclear in T2-high asthma. OBJECTIVE This study aimed to identify core extrapulmonary and behavior/risk factor traits for T2-high asthma. METHODS A cross-sectional study was conducted on 187 people (aged ≥18 years) with severe asthma who completed a multidimensional assessment. T2-high asthma was defined as blood eosinophils ≥150 cells/μL and/or fractional exhaled nitric oxide ≥20 ppb. Core TTs in T2-high asthma were identified among 9 extrapulmonary traits and 4 behavior/risk factor traits, using network analysis and dominance analysis for the Asthma Control Questionnaire scores, the Asthma Quality of Life Questionnaire scores, exacerbation frequency, and lung function. Associations between the identified core TTs and biomarkers were examined in participants with T2-high asthma. RESULTS Of 187 participants, 151 (80.7%) had T2-high severe asthma. Dysfunctional breathing and depression had higher values of node strength than other TTs, contributing most to worse asthma symptoms, poorer quality of life, and frequent exacerbations in T2-high asthma. These conditions in T2-high asthma were associated with elevated systemic inflammation, including blood neutrophils, neutrophil-lymphocyte ratio, and serum high-sensitivity C-reactive protein, independent of obesity, oral corticosteroid dose, and anxiety. CONCLUSIONS Core extrapulmonary and behavior/risk factor traits in T2-high severe asthma were dysfunctional breathing and depression, contributing to worse asthma outcomes, suggesting that core TTs may differ between asthma inflammatory phenotypes. Elevated systemic inflammation may help in recognizing the presence of dysfunctional breathing and depression in T2-high severe asthma.
Collapse
Affiliation(s)
- Yuto Hamada
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Japan.
| | - Peter G Gibson
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Vanessa L Clark
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; School of Nursing and Midwifery, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, NSW, Australia
| | - Hayley Lewthwaite
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Michael Fricker
- Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Dennis Thomas
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Vanessa M McDonald
- Center of Excellence in Treatable Traits, College of Health, Medicine and Wellbeing, the University of Newcastle, New Lambton Heights, NSW, Australia; Asthma and Breathing Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, NSW, Australia; School of Nursing and Midwifery, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
22
|
Biegański HM, Dąbrowski KM, Różańska-Walędziak A. Omentin-General Overview of Its Role in Obesity, Metabolic Syndrome and Other Diseases; Problem of Current Research State. Biomedicines 2025; 13:632. [PMID: 40149608 PMCID: PMC11940803 DOI: 10.3390/biomedicines13030632] [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: 01/31/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Omentin (omentin-1, intelectin-1, ITLN-1) is an adipokine considered to be a novel substance. Many chronic, inflammatory, or civilization diseases are linked to obesity, in which omentin plays a significant role. Methods: MEDLINE and SCOPUS databases were searched using the keywords "omentin" or "intelectin-1". Then the most recent articles providing new perspectives on the matter and the most important studies, which revealed crucial insight, were selected to summarize the current knowledge on the role of omentin in a literature review. Results and Conclusions: The valid role of this adipokine is evident in the course of metabolic syndrome. In most cases, elevated omentin expression is correlated with the better course of diseases, including: type 2 diabetes mellitus, polycystic ovary syndrome, rheumatoid arthritis, metabolic dysfunction-associated steatotic liver disease, Crohn's disease, ulcerative colitis, atherosclerosis, or ischemic stroke, for some of which it can be a better marker than the currently used ones. However, results of omentin studies are not completely one-sided. It was proven to participate in the development of asthma and atopic dermatitis and to have different concentration dynamics in various types of tumors. All of omentin's effects and properties make it an attractive subject of research, considering still unexplored inflammation mechanisms, in which it may play an important role. Omentin was proven to prevent osteoarthritis, hepatocirrhosis, and atherosclerosis in mouse models. All of the above places omentin among potential therapeutic products, and not only as a biomarker. However, the main problems with the omentin's research state are the lack of standardization, which causes many contradictions and disagreements in this field.
Collapse
Affiliation(s)
- Hubert Mateusz Biegański
- Medical Faculty, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland; (H.M.B.); (K.M.D.)
| | - Krzysztof Maksymilian Dąbrowski
- Medical Faculty, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland; (H.M.B.); (K.M.D.)
| | - Anna Różańska-Walędziak
- Departament of Human Physiology and Pathophysiology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland
| |
Collapse
|
23
|
Kuniyoshi Y, Tsujimoto Y, Banno M, Taito S, Ariie T, Kimoto T. Association of obesity or metabolic syndrome with various allergic diseases: An overview of reviews. Obes Rev 2025; 26:e13862. [PMID: 39663640 DOI: 10.1111/obr.13862] [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: 06/09/2023] [Revised: 09/17/2024] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
The relationship between obesity, metabolic syndrome, related disorders, and various allergic diseases remains unclear. An overview of reviews investigating potential associations between obesity or metabolic syndrome and various allergic diseases was conducted. MEDLINE, EMBASE, and the Cochrane Central Register of Controlled Trials were searched. Systematic reviews and meta-analyses with summary effect size and corresponding 95% confidence intervals for at least one outcome (asthma, atopic dermatitis, and various allergic diseases) were included. This study encompassed 17 systematic review articles and 29 eligible meta-analyses. All included meta-analyses indicated a positive association between obesity/overweight and asthma. Three meta-analyses from one review demonstrated a positive association between obesity/overweight and the risk of atopic dermatitis. However, no meta-analyses focused on the associations between obesity/overweight or metabolic syndrome and allergic rhinitis, allergic conjunctivitis, or other allergic conditions. All included reviews employed poor methodology according to the AMSTAR-2 assessment tools. Our findings suggest that obesity likely increases the risk of asthma. However, evidence for associations with other allergic diseases is limited. Furthermore, no meta-analyses were conducted to assess the relationship between metabolic syndrome and allergic diseases. Further studies are necessary to elucidate the associations between obesity and the full spectrum of allergic diseases.
Collapse
Affiliation(s)
- Yasutaka Kuniyoshi
- Department of Social Services and Healthcare Management, International University of Health and Welfare, Otawara, Tochigi, Japan
- Systematic Review Workshop Peer Support Group (SRWS-PSG), Osaka, Japan
| | - Yasushi Tsujimoto
- Systematic Review Workshop Peer Support Group (SRWS-PSG), Osaka, Japan
- Oku Medical Clinic, Osaka, Japan
| | - Masahiro Banno
- Systematic Review Workshop Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Psychiatry, Seichiryo Hospital, Nagoya, Japan
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shunsuke Taito
- Systematic Review Workshop Peer Support Group (SRWS-PSG), Osaka, Japan
- Division of Rehabilitation, Department of Clinical Practice and Support, Hiroshima University Hospital, Hiroshima, Japan
| | - Takashi Ariie
- Systematic Review Workshop Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Physical Therapy, School of Health Sciences at Fukuoka, International University of Health and Welfare, Fukuoka, Japan
| | - Takeru Kimoto
- Department of Pediatrics, Tsugaruhoken Medical COOP Kensei Hospital, Hirosaki, Japan
| |
Collapse
|
24
|
Kayikci H, Damadoglu E, Cihanbeylerden M, Tuccar C, Karakaya G, Kalyoncu AF. Clinical characteristics and biological treatment responses of patients with late-onset asthma phenotype. Allergy Asthma Proc 2025; 46:109-118. [PMID: 40011985 DOI: 10.2500/aap.2025.46.240105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Background: The data on subphenotypes and treatment responses to biologicals in late-onset asthma (LOA) is limited. This study aims to compare the clinical characteristics and treatment responses in severe asthma patients receiving biological treatments, categorized into early-onset asthma (EOA) and LOA groups. Methods: Patients treated with omalizumab or mepolizumab for at least six months at a tertiary care adult allergy clinic between December 2015 and December 2023 were included. Patients with persistent respiratory symptoms starting at age ≥40 years were categorized as LOA, while those with onset <40 years were categorized as EOA. Changes in Asthma Control Questionnaire (ACQ-6) scores, forced expiratory volume in one second (FEV1) percentages, and blood eosinophil counts were assessed at baseline and 6 months. The percentage change in FEV1 (liters) at 6 months relative to baseline was measured. Clinical remission rates were evaluated in those completing one year of treatment. Results: Among 87 patients, 38 (43.7%) had LOA and 49 (56.3%) had EOA. Of these, 22 (25.3%) received omalizumab and 65 (74.7%) received mepolizumab, with a mean treatment duration of 24.7 (±19.7) months. LOA patients had higher obesity rates and tobacco consumption compared to EOA patients (p = 0.041 and p = 0.024, respectively). There were no significant differences between LOA and EOA groups in ACQ scores, FEV1 percentage, the percentage change in FEV1 in liters and eosinophil counts (p = 0.531, p = 0.219, p = 0.632, p = 0.700, respectively). Within LOA patients, ACQ scores did not significantly differ between those treated with omalizumab and mepolizumab (p = 0.801). At 6 months, eosinophil counts significantly decreased with mepolizumab but not with omalizumab (p = 0.002). Conclusion: Biological treatment responses were similar between LOA and EOA groups. Omalizumab and mepolizumab showed comparable efficacy, with the exception of eosinophil count changes in LOA patients.
Collapse
|
25
|
Akca Sumengen A, Ozcevik Subasi D, Semerci R, Cakir GN. Effect of game-based asthma management interventions on pediatric asthma control, knowledge, attitudes, hospitalizations, and emergency visits: A systematic review and meta-analysis. J Pediatr Nurs 2025; 81:183-199. [PMID: 39743442 DOI: 10.1016/j.pedn.2024.12.009] [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: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Many studies have used game-based interventions to educate children about asthma. The study aims to determine the effectiveness of these games in improving asthma control and related outcomes in children. METHODS Seven databases were searched: PubMed, Cochrane Library, Scopus, CINAHL, Embase, Web of Science, and PsycINFO'. All research papers published until June 2023 were included. MeSH terms and keywords were used in the literature search. The Joanna Briggs Institute Meta-Analysis of Statistics Assessment and Review Instruments was used to assess the risk of bias. RESULTS This systematic review and meta-analysis included nine studies with a total sample size of 694 children. The effect size for hospitalization rates was not statistically significant (p > 0.05), although the association was significant (p = 0.004). Conversely, a statistically significant reduction in emergency visits was observed (p < 0.05), with an effect size estimate of 0.376. The analysis also revealed a significant improvement in asthma knowledge (p < 0.05), with an effect size estimate of 0.677 (95 % CI: 0.240 to 1.114, p = 0.002), and an increase in asthma control (p < 0.05), although the association was not statistically significant (p = 0.120) with an effect size estimate of 0.169 (95 % CI: -0.044 to 0.381). Conversely, no statistically significant effect was observed for asthma attitude (p > 0.05). CONCLUSION Game-based interventions have shown promise in improving asthma management in children by enhancing knowledge and control and reducing emergency visits. This approach is increasingly recommended in clinical settings, though there is notable heterogeneity in study design and participant demographics.
Collapse
Affiliation(s)
| | | | | | - Gokce Naz Cakir
- Faculty of Health Science Nursing Department, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
26
|
Laanesoo A, Mäe M, Remm A, Johnston SL, Altraja A, Bochenek G, Jakiela B, Rebane A. NLRP1 Is a Prominent Inflammasome Sensor Found in Bronchial Epithelial Cells in Asthma and Can Be Activated by Rhinovirus A16. Clin Exp Allergy 2025; 55:239-246. [PMID: 39934921 DOI: 10.1111/cea.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/14/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Asthma exacerbations are frequently triggered by human rhinoviruses (RVs). Among other pro-inflammatory responses, RV infection of airway epithelium promotes the activation of the inflammasome pathway, the role of which in asthma exacerbations and disease progression is still poorly understood. METHODS Bronchial brushing or biopsy specimens were collected from asthma patients and control subjects. Functional experiments were performed in cultured human bronchial epithelial cells (HBECs) using RV-A16, poly(I:C), and siRNA transfection. Gene expression was analysed by RNA-sequencing, RT-qPCR, immunofluorescence, western blot or ELISA. Caspase-1 activity was evaluated using FAM-FLICA assay. RESULTS The expression of NLRP1 was found to be the highest compared to other inflammasome sensors tested in brushed bronchial epithelium samples from asthma patients and control individuals, as well as in cultured primary HBECs. Additionally, we observed increased expression of CASP1 mRNA in bronchial epithelial cells from patients with neutrophilic asthma compared to those with paucigranulocytic and eosinophilic phenotypes. Changes in the expression of inflammasome pathway genes caused by RV-A16 infection were similar in HBEC cultures from asthma patients and controls, except for IL-1β, which showed increased response, and PYCARD, which exhibited decreased change in cells derived from asthma patients. Silencing of NLRP1 expression with siRNAs impeded RV-A16-induced activation of the inflammasome but had no effect on poly(I:C)-induced secretion of IL-1β and IL-18. CONCLUSION NLRP1 is highly expressed inflammasome sensor in both healthy and asthmatic bronchial epithelium and can be activated by RV-A16. RV-induced changes in the expression of inflammasome pathway genes suggest that there may be differences in HBECs derived from asthma patients, which may depend on the prevailing immunological phenotype of the disease.
Collapse
Affiliation(s)
- Anet Laanesoo
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Mariel Mäe
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Anu Remm
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - Alan Altraja
- Department of Pulmonology, University of Tartu, Tartu, Estonia
- Lung Clinic of the Tartu University Hospital, Tartu, Estonia
| | - Grazyna Bochenek
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Bogdan Jakiela
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
27
|
Fastiggi VA, Mank MM, Caporizzo MA, Poynter ME. Beta-Hydroxybutyrate Inhibits Bronchial Smooth Muscle Contraction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639075. [PMID: 40060651 PMCID: PMC11888348 DOI: 10.1101/2025.02.24.639075] [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: 03/17/2025]
Abstract
Asthma is a chronic respiratory condition characterized by airway inflammation, remodeling, and hyperresponsiveness to triggers causing airway constriction. Bronchial smooth muscle plays a critical role by narrowing airways, leading to obstruction and breathing difficulties, often exacerbated by mast cell infiltration and histamine release. Whereas current treatments, including bronchodilators, corticosteroids, and biologics provide effective management for most patients, alternative therapies are needed for difficult-to-treat asthma. Recent research highlights the potential of therapeutic ketosis, achieved through dietary interventions or supplementation with exogenous ketones, to reduce airway hyperresponsiveness and inflammation. Ketone bodies, known for providing energy during carbohydrate scarcity, also influence asthma by activating cell-surface receptors and transporters. In vivo, interventions like weight loss and caloric restriction increase ketone body levels, correlating with improved asthma symptoms, reduced oxidative stress, and inflammation. These effects suggest ketone bodies, particularly β-hydroxybutyrate, may play a therapeutic role in mitigating bronchoconstriction and smooth muscle contraction in asthma. We utilize human bronchial smooth muscle cells (in vitro) and mouse precision-cut lung slices (PCLS) (ex vivo) to assess the effects of BHB on histamine-induced bronchoconstriction. Brightfield microscopy showed that BHB reduces contraction in human bronchial smooth muscle cells, an effect involving free fatty acid receptor 3 (FFAR3) activation. Light microscopy of PCLS revealed that BHB inhibits airway narrowing and cellular extrusion, demonstrating its ability to mitigate bronchoconstriction by suppressing smooth muscle contraction. These results implicate bronchial smooth muscle as a cellular target of therapeutic ketosis, an important contributor to the beneficial effects of BHB in preclinical models of asthma.
Collapse
Affiliation(s)
- V Amanda Fastiggi
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
- Cellular, Molecular, and Biomedical Sciences Doctoral Program, University of Vermont, Burlington, VT, 05405, USA
| | - Madeleine M Mank
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| | - Matthew A Caporizzo
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05405, USA
| | - Matthew E Poynter
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| |
Collapse
|
28
|
Young N, Chen W, Chatterjee S, Gelzinnis S, Latheef AL, Simpson J, Wark PAB. Characterising airway inflammation in Aboriginal and Torres Strait Islander and non-Aboriginal and Torres Strait Islander adults with asthma and COPD. BMJ Open Respir Res 2025; 12:e002619. [PMID: 39986687 PMCID: PMC11848662 DOI: 10.1136/bmjresp-2024-002619] [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: 06/03/2024] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
OBJECTIVE To examine airway inflammatory cell profiles in Indigenous Australian adults with asthma and chronic obstructive pulmonary disease (COPD). DESIGN/SETTING A retrospective, cross-sectional study on data from a tertiary referral respiratory outpatient clinic. PARTICIPANTS Indigenous (n=23) and non-Indigenous (n=71) adults were matched according to diagnosis, gender and age to the ratio of 1:3. MAIN OUTCOME MEASURES Participants were defined by self-determined identification as Indigenous (Aboriginal) or non-Indigenous. A relevant history was taken, and lung function was measured by spirometry. In those with a diagnosis of asthma, symptom control was assessed by the Asthma Control Questionnaire, six items (ACQ6). In those with a diagnosis of COPD, symptoms were assessed by the COPD assessment test (CAT). Airway cell counts were obtained in all groups from bronchial lavage (BL) cell count. RESULTS Lung function and inhaled corticosteroid dose were similar between groups. Current smoking was three times more common in Indigenous people (35%) compared with non-Indigenous people (12%, p=0.009). In participants with asthma, ACQ6 scores were similar between Indigenous and non-Indigenous participants with asthma. In those with COPD, Indigenous participants had significantly higher total CAT scores as well as scores for cough and sputum with a score indicating a high impact on quality of life (CAT score ≥14, 85%-25%, p=0.017). There was no difference in BL cell differential counts. CONCLUSIONS Indigenous people with COPD had higher smoking rates, worsened CAT scores and more symptoms of cough and sputum production. There were no differences between the groups in airway inflammation, but neutrophilic inflammation was associated with poorly-controlled asthma.
Collapse
Affiliation(s)
- Nick Young
- Hunter New England Local Health District, New Lambton, Newcastle, Australia
| | - Winnie Chen
- Royal Australian College of General Practitioners, Sydney, New South Wales, Australia
| | - Shimul Chatterjee
- Central Coast Local Health District, Gosford, New South Wales, Australia
| | - Scott Gelzinnis
- Critical Care, Hunter New England Local Health District, New Lambton, New South Wales, Australia
| | | | - Jodie Simpson
- School of Medicine and Public Health, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle Hunter Medical Research Institute, New Lambton, New South Wales, Australia
| | - Peter A B Wark
- Immunology, Respiratory Medicine, Monash University Faculty of Medicine Nursing and Health Sciences, Prahran, Victoria, Australia
- Allergy, Immunology and Respiratory Medicine, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Fastiggi VA, Mank MM, Poynter ME. Beta-Hydroxybutyrate Attenuates Bronchial Smooth Muscle Pro-Inflammatory Cytokine Production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639048. [PMID: 40027689 PMCID: PMC11870512 DOI: 10.1101/2025.02.19.639048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Asthma is a common airway condition causing breathing difficulties due to reversible airflow obstruction. It often affects obese individuals, with symptoms triggered by environmental factors that induce immune responses, leading to inflammation and bronchoconstriction. Bronchial smooth muscle (BSM) plays a central role in airway narrowing, driven by type 2 immune responses involving cytokines like IL-4, IL-5, and IL-13, along with leukocytes including eosinophils and type 2 T-helper cells. These responses cause structural changes such as fibrosis and airway thickening, while BSM cells worsen asthma by releasing pro-inflammatory cytokines in response to allergens, microbial signals, or inflammatory cytokines from other cells. While current treatments manage asthma in most patients, alternative therapies are needed for difficult-to-treat cases, particularly prevalent in obese, allergic individuals. Emerging research suggests that therapeutic ketosis, induced by dietary changes or ketone supplementation, may reduce airway hyperresponsiveness and inflammation. The primary ketone body, β-hydroxybutyrate (BHB), produced during carbohydrate scarcity, acts via cell-surface receptors and transporters, potentially mitigating asthma symptoms. Weight loss and caloric restriction increase ketone levels, correlating with reduced inflammation and improved asthma outcomes. We hypothesized that β-hydroxybutyrate (BHB) reduces bronchoconstriction and inflammation in asthma by targeting bronchial smooth muscle. Using human bronchial smooth muscle cells (HBSMC) in vitro, we demonstrate herein that BHB suppresses IL-1β-induced pro-inflammatory cytokine production through Free Fatty Acid Receptor 3 (FFAR3) activation. These findings suggest that bronchial smooth muscle is a key target of therapeutic ketosis, supporting BHB's potential benefits in preclinical asthma models.
Collapse
Affiliation(s)
- V. Amanda Fastiggi
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
- Cellular, Molecular, and Biomedical Sciences Doctoral Program, University of Vermont, Burlington, VT, 05405, USA
| | - Madeleine M. Mank
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| | - Matthew E. Poynter
- Department of Medicine, Division of Pulmonary Disease and Critical Care, University of Vermont, and The Vermont Lung Center, Burlington, VT, 05405, USA
| |
Collapse
|
30
|
Zhong X, Mitchell R, Billstrand C, Thompson E, Sakabe NJ, Aneas I, Salamone IM, Gu J, Sperling AI, Schoettler N, Nóbrega MA, He X, Ober C. Integration of functional genomics and statistical fine-mapping systematically characterizes adult-onset and childhood-onset asthma genetic associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.11.25322088. [PMID: 40034789 PMCID: PMC11875274 DOI: 10.1101/2025.02.11.25322088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Genome-wide association studies (GWAS) have identified hundreds of loci underlying adult-onset asthma (AOA) and childhood-onset asthma (COA). However, the causal variants, regulatory elements, and effector genes at these loci are largely unknown. Methods We performed heritability enrichment analysis to determine relevant cell types for AOA and COA, respectively. Next, we fine-mapped putative causal variants at AOA and COA loci. To improve the resolution of fine-mapping, we integrated ATAC-seq data in blood and lung cell types to annotate variants in candidate cis-regulatory elements (CREs). We then computationally prioritized candidate CREs underlying asthma risk, experimentally assessed their enhancer activity by massively parallel reporter assay (MPRA) in bronchial epithelial cells (BECs) and further validated a subset by luciferase assays. Combining chromatin interaction data and expression quantitative trait loci, we nominated genes targeted by candidate CREs and prioritized effector genes for AOA and COA. Results Heritability enrichment analysis suggested a shared role of immune cells in the development of both AOA and COA while highlighting the distinct contribution of lung structural cells in COA. Functional fine-mapping uncovered 21 and 67 credible sets for AOA and COA, respectively, with only 16% shared between the two. Notably, one-third of the loci contained multiple credible sets. Our CRE prioritization strategy nominated 62 and 169 candidate CREs for AOA and COA, respectively. Over 60% of these candidate CREs showed open chromatin in multiple cell lineages, suggesting their potential pleiotropic effects in different cell types. Furthermore, COA candidate CREs were enriched for enhancers experimentally validated by MPRA in BECs. The prioritized effector genes included many genes involved in immune and inflammatory responses. Notably, multiple genes, including TNFSF4, a drug target undergoing clinical trials, were supported by two independent GWAS signals, indicating widespread allelic heterogeneity. Four out of six selected candidate CREs demonstrated allele-specific regulatory properties in luciferase assays in BECs. Conclusions We present a comprehensive characterization of causal variants, regulatory elements, and effector genes underlying AOA and COA genetics. Our results supported a distinct genetic basis between AOA and COA and highlighted regulatory complexity at many GWAS loci marked by both extensive pleiotropy and allelic heterogeneity.
Collapse
Affiliation(s)
- Xiaoyuan Zhong
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Robert Mitchell
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Emma Thompson
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J. Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Jing Gu
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I. Sperling
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nathan Schoettler
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Marcelo A. Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Xin He
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
31
|
Cuttone G, La Via L, Pappalardo F, Sorbello M, Paternò DS, Piattoli M, Gregoretti C, Misseri G. An Updated Review on the Use of Noninvasive Respiratory Supports in the Management of Severe Asthma Exacerbations. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:328. [PMID: 40005443 PMCID: PMC11857382 DOI: 10.3390/medicina61020328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Asthma is a reversible clinical condition characterized by airway obstruction due to bronchial smooth muscle contraction, inflammation and a hypersecretive state. Severe asthma exacerbations (SAE) may be a part of the natural history of this condition. Patients presenting with SAE are at higher risk of recurrent attacks, often nonresponsive to medical therapy and eventually requiring invasive mechanical ventilation (MV). The use of noninvasive respiratory supports (NRSs) may be beneficial in patients with SAE who are at risk of developing acute respiratory failure (ARF). However, their application is insufficiently supported by the evidence, as reports on their application in asthmatic patients are scarce and only a few retrospective studies with a limited number of participants have been published to date. This review discusses the potentialities of NRS in the treatment of SAE, with reference to the pathophysiological background and future perspectives on their use in asthma management.
Collapse
Affiliation(s)
- Giuseppe Cuttone
- Department of Anaesthesia and Trauma Center, Azienda Ospedaliera “Ospedali Riuniti Villa Sofia–Cervello”, 90146 Palermo, Italy;
| | - Luigi La Via
- Department of Anaesthesia and Intensive Care 1, University Hospital Policlinico “G. Rodolico–San Marco”, 95123 Catania, Italy;
| | - Federico Pappalardo
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (F.P.); (M.S.)
- Policlinico “G.B. Morgagni”, 95125 Catania, Italy
| | - Massimiliano Sorbello
- Faculty of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy; (F.P.); (M.S.)
- Department of Anaesthesia and Intensive Care, Giovanni Paolo II Hospital, 97100 Ragusa, Italy;
| | | | - Matteo Piattoli
- Faculty of Medicine and Dentistry, Università degli Studi di Roma “La Sapienza”, 00185 Rome, Italy;
- Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences “UniCamillus”, 00131 Rome, Italy;
| | - Cesare Gregoretti
- Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences “UniCamillus”, 00131 Rome, Italy;
- Department of Anaesthesia and Intensive Care, Fondazione Istituto “G. Giglio” Cefalù, 90015 Palermo, Italy
| | - Giovanni Misseri
- Department of Anaesthesia and Intensive Care, Fondazione Istituto “G. Giglio” Cefalù, 90015 Palermo, Italy
| |
Collapse
|
32
|
Grunwell JR, Fitzpatrick AM. Asthma Phenotypes and Biomarkers. Respir Care 2025. [PMID: 40013975 DOI: 10.1089/respcare.12352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Asthma experienced by both adults and children is a phenotypically heterogeneous condition. Severe asthma, characterized by ongoing symptoms and airway inflammation despite high doses of inhaled and/or systemic corticosteroids, is the focus of research efforts to understand this underlying heterogeneity. Clinical phenotypes in both adult and pediatric asthma have been determined using supervised definition-driven classification and unsupervised data-driven clustering methods. Efforts to understand the underlying inflammatory patterns of severe asthma have led to the seminal discovery of type 2-high versus type 2-low phenotypes and to the development of biologics targeted at type 2-high inflammation to reduce the rates of severe asthma exacerbations. Type 2-high asthma is characterized by upregulation of T helper 2 immune pathways including interleukin (IL)-4, IL-5, and IL-13 along with eosinophilic airway inflammation, sometimes allergic sensitization, and responsiveness to treatment with corticosteroids. Type 2-low asthma is poorly responsive to corticosteroids and is not as well characterized as type 2-high asthma. Type 2-low asthma is limited by being defined as the absence of type 2-high inflammatory markers. Choosing a biologic for the treatment of severe asthma involves the evaluation of a panel of biomarkers such as blood eosinophils, total and specific immunoglobulin E/allergic sensitization, and fractional exhaled nitric oxide. In this review, we focus on the underlying pathobiology of adult and pediatric asthma, discuss the different phenotype-based treatment options for adult and pediatric type 2-high with or without allergic asthma and type 2-low asthma, and describe a clinical phenotyping approach to patients to guide out-patient therapy. Finally, we end with a discussion of whether pediatric asthma exacerbations necessitating admission to an ICU constitute their own high-risk phenotype and/or whether it is a part of other previously defined high-risk subgroups such as difficult-to-control asthma, exacerbation-prone asthma, and severe treatment-resistant asthma.
Collapse
Affiliation(s)
- Jocelyn R Grunwell
- Dr. Grunwell is affiliated with Division of Critical Care Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| | - Anne M Fitzpatrick
- Dr. Fitzpatrick is affiliated with Division of Pulmonary, Allergy/Immunology, Cystic Fibrosis, and Sleep Medicine, Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
33
|
Wang X, Peng HM, Zhang MR, Li JJ, Zhao CP, Zhang YL, Wang SY, Zhu SY, Lu JK, Yin HL, Yin Q, Fang JB. Hyssopus cuspidatus volatile oil: a potential treatment for steroid-resistant asthma via inhibition of neutrophil extracellular traps. Chin Med 2025; 20:17. [PMID: 39901245 PMCID: PMC11792399 DOI: 10.1186/s13020-025-01069-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Steroid-resistant asthma (SRA) is a form of asthma resistant to corticosteroid therapy, which is characterized by the presence of neutrophil-predominant inflammatory response and neutrophil extracellular traps (NETs) formation. Hyssopus cuspidatus Boriss., a traditional Uyghur medicine, is known for its efficacy in treating inflammatory lung conditions such as asthma. However, the therapeutic impact and underlying mechanisms of Hyssopus cuspidatus Boriss.'s volatile oil (HVO) in SRA have not been fully elucidated. METHODS This study established an ovalbumin/lipopolysaccharide (OVA/LPS)-induced SRA mice model to evaluate the therapeutic effect of HVO on SRA. UPLC-QE-Orbitrap-MS was applied to analyze the serum compositions of HVO. Network pharmacology and molecular docking were employed to uncover the complex mechanisms of HVO in treating SRA and predict potential effective compounds in HVO. Furthermore, in vivo studies in SRA mice and in vitro studies using HL-60 cells and bone marrow neutrophils were conducted to validate the mechanism. RESULTS HVO could significantly ameliorate OVA/LPS-induced SRA symptoms, including airway hyperresponsiveness, airway inflammation, mucus overproduction and airway remodeling. 41 prototype compounds, 65 Phase I metabolites and 50 Phase II metabolites were identified in serum-containing HVO. The integration of network pharmacology with experimental validation revealed that HVO can inhibit the formation of NETs by targeting neutrophil elastase, thereby exerting a therapeutic influence on SRA. Meanwhile, molecular docking results showed that 3-methoxy-4-hydroxy mandelonitrile, 1,2,3,4-tetrahydro-1,5,7-trimethyl-naphthalene, cis-calamenene and aristol-1(10)-en-9-yl isovalerate may be the therapeutic compounds of HVO in treating SRA. CONCLUSION These findings suggest that HVO is a promising therapeutic candidate for neutrophil-dominant SRA by targeting NETs formation.
Collapse
Affiliation(s)
- Xu Wang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Ming Peng
- Department of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng-Ru Zhang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing-Jing Li
- Hubei Shizhen Laboratory, School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chuan-Peng Zhao
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ya-Li Zhang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Si-Yu Wang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Si-Ying Zhu
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Kang Lu
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hai-Long Yin
- Xinjiang Uygur Pharmaceutical Co., Ltd, No. 2, Shenyang Street, Urumqi Economic and Technological Development Zone (Toutunhe District), Xinjiang Uygur Autonomous Region, Urumqi, 830026, Xinjiang, China
| | - Qiang Yin
- Xinjiang Uygur Pharmaceutical Co., Ltd, No. 2, Shenyang Street, Urumqi Economic and Technological Development Zone (Toutunhe District), Xinjiang Uygur Autonomous Region, Urumqi, 830026, Xinjiang, China
| | - Jin-Bo Fang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
34
|
La Via L, Cuttone G, Misseri G, Sorbello M, Pappalardo F, Maniaci A, Duarte-Medrano G, Nuño-Lámbarri N, Zanza C, Gregoretti C. The use of noninvasive positive pressure ventilation for severe asthma: a systematic review and meta-analysis of randomized controlled trials with trial sequential analysis. Expert Rev Respir Med 2025; 19:165-173. [PMID: 39825601 DOI: 10.1080/17476348.2025.2454947] [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: 11/17/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
INTRODUCTION To evaluate the effectiveness of noninvasive positive pressure ventilation (NPPV) versus standard therapy in severe asthma exacerbations through meta-analysis. METHODS Nine randomized controlled trials (344 patients) were analyzed from inception to August 2024. Primary outcomes included respiratory rate, forced expiratory volume in first second (FEV1), and oxygen saturation (SpO2). Random effect models and trial sequential analyses were employed. RESULTS NPPV demonstrated significant reduction in respiratory rate versus standard therapy (mean difference [MD] -3.97, 95% CI -7.32 to -0.61, p = 0.02), though with high heterogeneity (I2 = 87%). FEV1 showed significant improvement with NPPV (MD 15.56%, 95% CI 6.86 to 24.26, p < 0.001) based on two studies. SpO2 showed no significant improvement (MD 0.62%, 95% CI -0.14 to 1.37, p = 0.11). No differences were found between pediatric and adult populations. Trial sequential analyses indicated insufficient evidence for definitive conclusions regarding respiratory rate and SpO2 improvements. CONCLUSIONS While NPPV may benefit severe asthma patients, particularly in reducing respiratory rate and improving FEV1, current evidence is insufficient for recommending routine clinical use. Larger randomized controlled trials are needed to establish NPPV's effectiveness in severe asthma exacerbation treatment. PROTOCOL REGISTRATION registration ID CRD42024580051.
Collapse
Affiliation(s)
- Luigi La Via
- Department of Anesthesia and Intensive Care 1, University Hospital Policlinico "G. Rodolico - San Marco", Catania, Italy
| | | | - Giovanni Misseri
- Department of Anesthesia and Intensive Care, Fondazione Istituto "G. Giglio" Cefalù, Cefalù, Italy
| | - Massimiliano Sorbello
- School of Medicine and Surgery, "Kore" University, Enna, Italy
- Department of Anesthesia and Intensive Care, Giovanni Paolo II Hospital, Ragusa, Italy
| | - Federico Pappalardo
- School of Medicine and Surgery, "Kore" University, Enna, Italy
- Policlinico Centro Cuore GB Morgagni, Catania, Italy
| | | | | | - Natalia Nuño-Lámbarri
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Surgery Department, Faculty of Medicine, The National Autonomous University of Mexico (UNAM), Mexico City, Mexico
| | - Christian Zanza
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cesare Gregoretti
- Department of Anesthesia and Intensive Care, Fondazione Istituto "G. Giglio" Cefalù, Cefalù, Italy
- Saint Camillus International Medical University (UniCamillus) Rome, Rome, Italy
| |
Collapse
|
35
|
Ameri S, Stang J, Walsted E, Price OJ. Mechanisms and Biomarkers of Exercise-induced Bronchoconstriction: Current Insights and Future Directions. Immunol Allergy Clin North Am 2025; 45:63-75. [PMID: 39608880 DOI: 10.1016/j.iac.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Exercise-induced bronchoconstriction (EIB) refers to temporary lower airway narrowing that occurs during or after vigorous physical exertion, with a high incidence in athletes and individuals with pre-existing asthma. The pathophysiology of EIB is not completely understood, but it is thought to involve a complex interplay among airway epithelial changes, immune responses, and environmental interactions. Phenotypic differences are apparent among those affected by EIB. This clinical review aims to summarize the complex mechanisms underlying EIB, explore the role of biomarkers in the diagnosis and management, and identify current gaps in knowledge to pave the way for future scientific discoveries.
Collapse
Affiliation(s)
- Sammy Ameri
- Department of Respiratory Medicine, Bispebjerg Hospital, Bispebjerg Bakke 23, Building 66, København NV 2400, Denmark.
| | - Julie Stang
- Department of Sports Medicine, Norwegian School of Sport Sciences, Sognsveien 220, Oslo 0863, Norway
| | - Emil Walsted
- Department of Respiratory Medicine, Bispebjerg Hospital, Bispebjerg Bakke 23, Building 66, København NV 2400, Denmark
| | - Oliver J Price
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, UK
| |
Collapse
|
36
|
Zhong X, Song J, Lei C, Wang X, Wang Y, Yu J, Dai W, Xu X, Fan J, Xia X, Zhang W. Machine learning-based screening of asthma biomarkers and related immune infiltration. FRONTIERS IN ALLERGY 2025; 6:1506608. [PMID: 39963184 PMCID: PMC11831286 DOI: 10.3389/falgy.2025.1506608] [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: 10/05/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Asthma has an annual increasing morbidity rate and imposes a heavy social burden on public healthcare systems. The aim of this study was to use machine learning to identify asthma-specific genes for the prediction and diagnosis of asthma. Methods Differentially expressed genes (DEGs) related to asthma were identified by examining public sequencing data from the Gene Expression Omnibus, coupled with the support vector machine recursive feature elimination and least absolute shrinkage and selection operator regression model. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene set enrichment analysis and correlation analyses between gene and immune cell levels were performed. An ovalbumin-induced asthma mouse model was established, and eukaryotic reference transcriptome high-throughput sequencing was performed to identify genes expressed in mouse lung tissues. Results Thirteen specific asthma genes were obtained from our dataset analysis (LOC100132287, CEACAM5, PRR4, CPA3, POSTN, LYPD2, TCN1, SCGB3A1, NOS2, CLCA1, TPSAB1, CST1, and C7orf26). The GO analysis demonstrated that DEGs linked to asthma were primarily related to positive regulation of guanylate cyclase activity, gpi anchor binding, peptidase activity and arginine binding. The renin-angiotensin system, arginine biosynthesis and arginine and proline metabolism were the key KEGG pathways of DEGs. Additionally, the genes CEACAM5, PRR4, CPA3, POSTN, CLCA1, and CST1 expression levels were positively associated with plasma cells and resting mast cells. The mouse model revealed elevated nos2 and clca1 expression in the asthmatic mouse group compared with that in normal mice, which was consistent with the findings in asthmatic patients. Discussion This study identified new marker genes for the prediction and diagnosis of asthma, which can be further validated and applied clinically.
Collapse
Affiliation(s)
- Xiaoying Zhong
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- The 2nd Ward of Pediatrics, Jinhua Maternal and Child Health Care Hospital, Jinhua, Zhejiang, China
| | - Jingjing Song
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Changyu Lei
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoming Wang
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yufei Wang
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiahui Yu
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Dai
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyi Xu
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junwen Fan
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaodong Xia
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weixi Zhang
- Allergy and Clinical Immunology Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
37
|
De Volder J, Bontinck A, Haelterman V, Boon L, Joos GF, Brusselle GG, Maes T. Anti-IL-5 treatment, but not neutrophil interference, attenuates inflammation in a mixed granulocytic asthma mouse model, elicited by air pollution. Respir Res 2025; 26:43. [PMID: 39875874 PMCID: PMC11773929 DOI: 10.1186/s12931-024-03082-9] [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: 07/05/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION Diesel exhaust particles (DEP) have been proven to aggravate asthma pathogenesis. We previously demonstrated that concurrent exposure to house dust mite (HDM) and DEP in mice increases both eosinophils and neutrophils in bronchoalveolar lavage fluid (BALF) and also results in higher levels of neutrophil-recruiting chemokines and neutrophil extracellular trap (NET) formation compared to sole HDM, sole DEP or saline exposure. We aimed to evaluate whether treatment with anti-IL-5 can alleviate the asthmatic features in this mixed granulocytic asthma model. Moreover, we aimed to unravel whether neutrophils modulate the DEP-aggravated eosinophilic airway inflammation. MATERIAL AND METHODS Female C57BL6/J mice were intranasally exposed to saline or HDM and DEP for 3 weeks (subacute model). Interference with eosinophils was performed by intraperitoneal administration of anti-IL-5 (TRFK5), which neutralizes IL-5. Interference with neutrophils and neutrophil elastase was performed by intraperitoneal anti-Ly6G and sivelestat administration, respectively. Outcome parameters included eosinophils subsets (homeostatic EOS and inflammatory EOS), proinflammatory cytokines, goblet cell hyperplasia and airway hyperresponsiveness. RESULTS The administration of anti-IL-5 significantly decreased eosinophilic responses, affecting both inflammatory and homeostatic eosinophil subsets, upon subacute HDM + DEP exposure while BAL neutrophils, NET formation and other asthma features remained present. Neutrophils were significantly reduced after anti-Ly6G administration in BALF, lung and blood without affecting the eosinophilic inflammation upon HDM + DEP exposure. Sivelestat treatment tended to decrease BALF inflammation, including eosinophils, upon HDM + DEP exposure, but did not affect lung inflammation. CONCLUSION Inhibition of IL-5 signalling, but not neutrophil interventions, significantly attenuates eosinophilic inflammation in a mouse model of mixed granulocytic asthma, elicited by air pollution exposure.
Collapse
Affiliation(s)
- Joyceline De Volder
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Annelies Bontinck
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Valerie Haelterman
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | | | - Guy F Joos
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Tania Maes
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Medical Research Building (MRB) II, Ghent University Hospital, 2 Floor, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
38
|
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
|
39
|
Lim J, Chang CJ, White AJ, Lo S, Wang H, Goodney G, Miao R, Barochia AV, Roger VL, Sandler DP, Wong JYY. Personal care product use and risk of adult-onset asthma: findings from the Sister Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.10.25320341. [PMID: 39830231 PMCID: PMC11741502 DOI: 10.1101/2025.01.10.25320341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND Personal care products (PCPs) contain endocrine-disrupting chemicals (EDCs) linked to hormonally-sensitive diseases. Population studies have found associations between prenatal EDC exposure and childhood asthma; however, few have investigated adult-onset asthma. OBJECTIVES We investigated the associations between commonly used PCPs and the risk of adult-onset asthma in a prospective cohort study of U.S. women. METHODS We analyzed 39,408 participants from the Sister Study (2003-2009). The participants self-reported their usage frequency of 41 PCPs in the 12-month period before baseline. Latent classes were used to identify groups with similar usage patterns ('infrequent', 'moderate', 'frequent') within types of products ('beauty', 'everyday hair', 'hygiene', and 'skincare'). Multivariable Cox regression models were used to assess the associations between PCP use and incident adult-onset asthma. RESULTS Over an average 12.5-year follow-up, 1,774 incident asthma cases were identified. Compared to infrequent users, moderate (hazard ratio [HR]=1.21 (95% confidence interval (CI):1.07,1.37)) and frequent (HR=1.22 (95%CI:1.08,1.38)) users of beauty products had significantly higher asthma risk. Similar associations were observed for hygiene (moderate: HR=1.14 (95%CI:1.01,1.29) and frequent: HR=1.20 (95%CI:1.06,1.36)) and skincare products (moderate: HR=1.21 (95%CI:1.06,1.38) and frequent: HR=1.20 (95%CI:1.06,1.35)). Several individual everyday hair products (hair spray, hair styling gel/mousse, and pomade or hair grease) were positively associated with asthma risk, but associations were not detected for everyday hair latent classes. DISCUSSION Our findings suggest that PCP use potentially contributes to future risk of adult-onset asthma among women. These novel findings reinforce the need for regulation of PCPs and their components to reduce the burden of asthma.
Collapse
|
40
|
Gu L, Zhu J, Nie Q, Xie B, Xue S, Zhang A, Li Q, Zhang Z, Li S, Li Y, Shi Q, Shi W, Zhao L, Liu S, Shi X. NLRP3 promotes inflammatory signaling and IL-1β cleavage in acute lung injury caused by cell wall extract of Lactobacillus casei. Commun Biol 2025; 8:20. [PMID: 39774843 PMCID: PMC11706994 DOI: 10.1038/s42003-025-07462-9] [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: 07/23/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025] Open
Abstract
Gram-positive bacterial pneumonia is a significant cause of hospitalization and death. Shortage of a good experimental model and therapeutic targets hinders the cure of acute lung injury (ALI). This study has established a mouse model of ALI using Gram-positive bacteria Lactobacillus casie cell wall extracts (LCWE) and identified the key regulator NLRP3. We show that LCWE induces TNF, NF-κB signaling, and so on pathways. Similar to lipopolysaccharide (LPS), LCWE induces the infiltration of CD11b-positive cells and inflammation in lungs. LCWE also triggers inflammatory signaling through TLR2, different from LPS through TLR4. It suggests that cytokines amplify inflammation signaling relying on NLRP3 in LCWE-induced ALI. NLRP3 deletion disrupts inflammation, IL-1β cleavage, and the infiltration of neutrophils and macrophages in the injured lung. Our study highlights an animal ALI model for Gram-positive bacterial pneumonia and that NLRP3 is a key therapeutic target to prevent inflammation and lung damage in LCWE-induced ALI.
Collapse
Affiliation(s)
- Lingui Gu
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Jinjin Zhu
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Qingbing Nie
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Binghua Xie
- The Fuyang Hospital, Anhui Medical University, Fuyang, Anhui, 236000, P. R. China
| | - Shuo Xue
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Ailing Zhang
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Qiangwei Li
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Zhengzhong Zhang
- The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Shupeng Li
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Yusen Li
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Qinquan Shi
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Weiwei Shi
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China
| | - Lei Zhao
- The Fuyang Hospital, Anhui Medical University, Fuyang, Anhui, 236000, P. R. China.
| | - Shuzhen Liu
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China.
| | - Xuanming Shi
- The School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, 230023, P. R. China.
| |
Collapse
|
41
|
Garcia FM, de Sousa VP, Silva-Dos-Santos PPE, Fernandes IS, Serpa FS, de Paula F, Mill JG, Bueno MRP, Errera FIV. Copy Number Variation in Asthma: An Integrative Review. Clin Rev Allergy Immunol 2025; 68:4. [PMID: 39755867 DOI: 10.1007/s12016-024-09015-0] [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] [Accepted: 11/13/2024] [Indexed: 01/06/2025]
Abstract
Asthma is a complex disease with varied clinical manifestations resulting from the interaction between environmental and genetic factors. While chronic airway inflammation and hyperresponsiveness are central features, the etiology of asthma is multifaceted, leading to a diversity of phenotypes and endotypes. Although most research into the genetics of asthma focused on the analysis of single nucleotide polymorphisms (SNPs), studies highlight the importance of structural variations, such as copy number variations (CNVs), in the inheritance of complex characteristics, but their role has not yet been fully elucidated in asthma. In this context, an integrative review was conducted to identify the genes and pathways involved, the location, size, and classes of CNVs, as well as their contribution to asthma risk, severity, control, and response to treatment. As a result of the review, 16 articles were analyzed, from different types of observational studies, such as case-control, cohort studies and genotyped-proband or trios design, that have been carried out in populations from different countries, ethnicities, and ages. Chromosomes 12 and 17 were the most studied in three publications each. CNVs located on 12 chromosomes were associated with asthma, the majority being found on chromosome 6p and 17q, of the deletion type, encompassing 30 different coding-protein genes and one pseudogene region. Six genes with CNVs were identified as significant expression quantitative locus (eQTLs) with mean expression in asthma-related tissues, such as the lung and whole blood. The phenotypic variability of asthma may hinder the clinical application of these findings, but the research shows the importance of investigating these genetic variations as possible biomarkers in asthma patients.
Collapse
Affiliation(s)
- Fernanda Mariano Garcia
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil.
| | - Valdemir Pereira de Sousa
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Priscila Pinto E Silva-Dos-Santos
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Izadora Silveira Fernandes
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Faradiba Sarquis Serpa
- Department of Medicine, School of Sciences of Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória, Espírito Santo, Brazil
- Hospital Santa Casa de Misericórdia de Vitória (HSCMV), Vitória, Espírito Santo, Brazil
| | - Flávia de Paula
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - José Geraldo Mill
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Physiological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Maria Rita Passos Bueno
- Department of Genetics and Evolutionary Biology, University of São Paulo (USP), São Paulo, São Paulo, Brazil
- Human Genome and Stem Cell Research Center, University of São Paulo (USP), São Paulo, São Paulo, Brazil
| | - Flávia Imbroisi Valle Errera
- Postgraduate Program in Biochemistry, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Postgraduate Program in Biotechnology, Northeast Network of Biotechnology (RENORBIO), Nucleator: Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
- Department of Biological Sciences, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| |
Collapse
|
42
|
Uehara S, Hirai K, Shirai T, Otaki H, Akamatsu T, Itoh K. Vitamin D Receptor rs2228570 Gene Polymorphism Is Associated with Asthma Severity and Exacerbations. Biol Pharm Bull 2025; 48:86-92. [PMID: 39894560 DOI: 10.1248/bpb.b24-00684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Vitamin D plays a crucial role in immune system function. Several studies have indicated that genetic variations in the vitamin D receptor (VDR) and vitamin D binding protein (VDBP, encoded by GC gene) increase the risk of developing asthma. However, the effect of these variations on the prognosis and clinical outcomes of asthma remains unclear. This study, involving 152 adult patients with asthma, aimed to assess the influence of VDR and GC polymorphisms on asthma severity and its exacerbation. Gene polymorphisms previously associated with asthma risk were analyzed, and VDR mRNA expression levels were evaluated in peripheral blood mononuclear cells. The AA genotype of the VDR rs2228570 polymorphism was associated with an elevated risk of severe asthma compared to the AG/GG genotype (odds ratio, 3.20; 95% confidence interval [CI], 1.24-8.28). Furthermore, patients with the rs2228570 AA genotype showed an elevated risk of exacerbation during the 1-year follow-up period (hazard ratio, 4.01; 95% CI, 1.75-9.15). VDR mRNA expression was significantly reduced in patients with the AA genotype. Furthermore, the mRNA expression levels of GLCCI1, HDAC2, NR3C1, and NFE2L2, which are associated with steroid response, were reduced in patients with the AA genotype. Our findings indicate that patients with the AA genotype of VDR rs2228570 are more likely to experience severe asthma and exacerbations. This polymorphism has the potential to reduce vitamin D efficacy by altering VDR function and expression, potentially resulting in increased inflammation and reduced steroid responsiveness in patients with asthma.
Collapse
Affiliation(s)
- Sekiko Uehara
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keita Hirai
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Department of Clinical Pharmacology and Therapeutics, Shinshu University Graduate School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Department of Pharmacy, Shinshu University Hospital, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Toshihiro Shirai
- Department of Respiratory Medicine, Shizuoka General Hospital, 4-27-1 Kita-Ando, Aoi-ku, Shizuoka 420-8527, Japan
| | - Hinako Otaki
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Taisuke Akamatsu
- Department of Respiratory Medicine, Shizuoka General Hospital, 4-27-1 Kita-Ando, Aoi-ku, Shizuoka 420-8527, Japan
| | - Kunihiko Itoh
- Department of Clinical Pharmacology & Genetics, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
- Laboratory of Clinical Pharmacogenomics, Shizuoka General Hospital, 4-27-1 Kita-Ando, Aoi-ku, Shizuoka 420-8527, Japan
| |
Collapse
|
43
|
Carriera L, Barone R, Ielo S, Coppola A. Simultaneous treatment with benralizumab and ustekinumab in a patient with severe asthma and ulcerative colitis. Lung India 2025; 42:49-52. [PMID: 39718916 PMCID: PMC11789949 DOI: 10.4103/lungindia.lungindia_337_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 12/26/2024] Open
Abstract
The burden of autoimmune diseases is rising worldwide. The expansion of the population of patients eligible for severe asthma biological therapy we are seeing in clinical practice could lead to the simultaneous use of different monoclonal antibodies. We present the case of biological combination therapy with ustekinumab and benralizumab in a patient with ulcerative colitis and severe eosinophilic asthma. The patient, already undergoing biological treatment for colitis, began to suffer from uncontrolled severe asthma. Since benralizumab was administered, the patient has not experienced any exacerbations requiring oral corticosteroids, emergency department visits, or hospital admissions, and the control of asthma symptoms and respiratory function considerably improved. Twelve months after the initiation of the combination, both diseases are well controlled, without any side effects or blood test abnormalities. To our knowledge, this is one of the first reported cases of patients simultaneously receiving a combination of biological therapy for ulcerative colitis and asthma.
Collapse
Affiliation(s)
- Lorenzo Carriera
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Barone
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Simone Ielo
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angelo Coppola
- UOC Pneumologia, Ospedale San Filippo Neri-ASL Roma 1, Rome, Italy
- UniCamillus, Saint Camillus International University of Health Sciences, Rome, Italy
| |
Collapse
|
44
|
Ramonell RP, Gauthier MC, Ray A, Wenzel SE. Biologic Medications for Severe Asthma: Implications for Understanding Pathogenic Heterogeneity and Endotypes. Annu Rev Med 2025; 76:339-355. [PMID: 39586024 DOI: 10.1146/annurev-med-070323-103158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Asthma is a chronic inflammatory disease of the airways long known for phenotypic heterogeneity. Phenotyping studies in asthma have led to a better characterization of disease pathogenesis, yet further work is needed to pair available treatments with disease endotypes. In this review, the biology of targeted pathways is discussed along with the efficacy of biologic therapies targeting those pathways. Results of asthma clinical trials are included, as well as results of trials in related diseases. This review then analyzes how biologics help to inform the complex immunobiology of asthma and further guide their use while identifying areas for future research.
Collapse
Affiliation(s)
- Richard P Ramonell
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Marc C Gauthier
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Asthma and Environmental Lung Health Institute at UPMC, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
45
|
Kim HB. Navigating the Asthma Maze in Children Through Trajectories With Allergic Comorbidities. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2025; 17:1-4. [PMID: 39895597 PMCID: PMC11791363 DOI: 10.4168/aair.2025.17.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025]
Affiliation(s)
- Hyo-Bin Kim
- Department of Pediatrics, Inha University Hospital, Inha University College of Medicine, Incheon, Korea.
| |
Collapse
|
46
|
Mdkhana B, Saheb Sharif-Askari N, Cagliani R, Al-Sheakly BKS, Ramakrishnan RK, Saheb Sharif-Askari F, Hachim IY, Hamid Q, Rawas-Qalaji M, Halwani R. Inhibiting DNA Sensing Pathway Controls Steroid Hyporesponsive Lung Inflammation. Adv Biol (Weinh) 2025; 9:e2400230. [PMID: 39601482 DOI: 10.1002/adbi.202400230] [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: 05/02/2024] [Revised: 10/24/2024] [Indexed: 11/29/2024]
Abstract
DNA damage underlies the progression of asthma toward a severe, steroid hyporesponsive phenotype. The accumulation of double-stranded DNA within the cytosol triggers the activation of cytosolic DNA-sensing pathways, notably the Stimulator of Interferon Genes (STING) pathway. However, the precise role of STING in driving steroid hyporesponsiveness remains elusive and warrants further investigation. This study evaluates STING levels in human bronchial fibroblasts from severe asthmatic patients and in lung homogenates from a steroid hyporesponsive lung inflammation mouse model. STING level is assessed at baseline, post house dust mites (HDM) stimulation, and following treatment with dexamethasone and STING inhibitor. The effect of STING inhibitors on regulating steroid hyporesponsiveness particularly glucocorticoid receptor (GR)-α/GR-β ratio is also examined. Severe asthmatic fibroblasts exhibit elevated STING/IFN-I pathway activation, further heightened by HDM and a similar pattern is seen in lung homogenates from steroid hyporesponsive mice. Dexamethasone combined with an STING inhibitor reduces STING activity, while dexamethasone alone is ineffective. Interestingly, the STING inhibitor restores steroid sensitivity by increasing the GRα/GRβ ratio. Furthermore, nanoparticle-encapsulated STING inhibitor more effectively reduces airway hyperresponsiveness and restores steroid sensitivity than the free inhibitor. These findings emphasize STING's role in severe asthma pathogenesis, proposing nanoparticle delivery of STING inhibitors as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Roberta Cagliani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biological Sciences, Khalifa University of Science & Technology, Abu Dhabi, UAE
| | | | - Rakhee K Ramakrishnan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ibrahim Yaseen Hachim
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mutasem Rawas-Qalaji
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Saudi Arabia
| |
Collapse
|
47
|
Wu T, Ji M, Li T, Luo L. The molecular and metabolic landscape of ferroptosis in respiratory diseases: Pharmacological aspects. J Pharm Anal 2025; 15:101050. [PMID: 40034685 PMCID: PMC11873008 DOI: 10.1016/j.jpha.2024.101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 03/05/2025] Open
Abstract
Ferroptosis is a form of cell death that occurs when there is an excess of reactive oxygen species (ROS), lipid peroxidation, and iron accumulation. The precise regulation of metabolic pathways, including iron, lipid, and amino acid metabolism, is crucial for cell survival. This type of cell death, which is associated with oxidative stress, is controlled by a complex network of signaling molecules and pathways. It is also implicated in various respiratory diseases such as asthma, chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), lung cancer, pulmonary fibrosis (PF), and the coronavirus disease 2019 (COVID-19). To combat drug resistance, it is important to identify appropriate biological markers and treatment targets, as well as intervene in respiratory disorders to either induce or prevent ferroptosis. The focus is on the role of ferroptosis in the development of respiratory diseases and the potential of targeting ferroptosis for prevention and treatment. The review also explores the interaction between immune cell ferroptosis and inflammatory mediators in respiratory diseases, aiming to provide more effective strategies for managing cellular ferroptosis and respiratory disorders.
Collapse
Affiliation(s)
- Tong Wu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Miaorong Ji
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| |
Collapse
|
48
|
Nishi K, Matsumoto H, Sunadome H, Nagasaki T, Oguma T, Tashima N, Hayashi Y, Terada S, Morita K, Yoshimura C, Nishizaka Y, Sano A, Iwanaga T, Sano H, Haraguchi R, Tohda Y, Kawaguchi T, Matsuda F, Hirai T. IL1RL1 variant may affect the response to type 2 biologics in patients with severe asthma. ERJ Open Res 2025; 11:00448-2024. [PMID: 39811553 PMCID: PMC11726575 DOI: 10.1183/23120541.00448-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/24/2024] [Indexed: 01/16/2025] Open
Abstract
Background Asthma is a heterogeneous disease with variable response to treatment. Genetic backgrounds are involved in the severity of type 2 asthma, but their effects on responses to biologics remain unknown. This study aimed to clarify the role of genetic factors in response to biologics in patients with severe asthma. Methods Adults with severe asthma receiving biologics were enrolled in this multicentre, observational, real-world study. The responses to biologics were evaluated using Physicians' Global Evaluation of Treatment Effectiveness (GETE). Optimal biologic for each patient was also determined based on the best GETE score for the biologic used or currently used biologic. Three single nucleotide polymorphisms (IL1RL1, rs1420101; IL4RA, rs8832; and TSLP rs1837253) were examined. Results Among the 113 patients analysed, 53 (46.9%) had an excellent GETE score for at least one biologic. These patients with an excellent GETE score for at least one biologic, particularly for benralizumab, had the risk genotype of rs1420101 more frequently than the remaining patients, independent of the clinical demographics. Regarding the optimal biologic for each patient, anti-IL-5 drugs were optimal for patients with the rs1420101 TT or rs8832 GG genotype. Furthermore, dupilumab was similarly effective, regardless of the risk genotypes examined in this study. Conclusion IL1RL1 rs1420101 TT genotype and/or IL4RA rs8832 GG genotype may predict an excellent or optimal response to biologic therapy in each patient, particularly to anti-interleukin-5 targeted therapy. The elucidation of genetic predisposition may improve the management of severe asthma in the era of biologics.
Collapse
Affiliation(s)
- Kenta Nishi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hironobu Sunadome
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tadao Nagasaki
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine and Allergology, Kindai University Nara Hospital, Nara, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine, Kyoto City Hospital, Kyoto, Japan
| | - Noriyuki Tashima
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Respiratory Medicine, Medical Research Institute Kitano Hospital, PIIF Tazuke-kofukai, Osaka, Japan
| | - Yusuke Hayashi
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoru Terada
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyohei Morita
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Chie Yoshimura
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Yasuo Nishizaka
- Department of Respiratory Medicine, Osaka Red Cross Hospital, Osaka, Japan
| | - Akiko Sano
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takashi Iwanaga
- Center for General Medical Education and Clinical Training, Kindai University Hospital, Osaka, Japan
| | - Hiroyuki Sano
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Ryuta Haraguchi
- Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yuji Tohda
- Kindai University Hospital, Osaka, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
49
|
Kuks PJ, Kole TM, Kraft M, Siddiqui S, Fabbri LM, Rabe KF, Papi A, Brightling C, Singh D, van der Molen T, Kocks JWW, Chung KF, Adcock IM, Bhavsar PK, Kermani NZ, Heijink IH, Pouwels SD, Kerstjens HA, Slebos DJ, van den Berge M. Neutrophilic inflammation in sputum or blood does not define a clinically distinct asthma phenotype in ATLANTIS. ERJ Open Res 2025; 11:00616-2024. [PMID: 39963164 PMCID: PMC11831683 DOI: 10.1183/23120541.00616-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/01/2024] [Indexed: 02/20/2025] Open
Abstract
Introduction Neutrophilic asthma has been suggested to be a clinically distinct phenotype characterised by more severe airflow obstruction and higher exacerbation risk. However, this has only been assessed in few and smaller studies, using different cut-offs to define neutrophilia, and with conflicting results. We used data from ATLANTIS, an observational longitudinal study including a large number of patients with asthma and healthy controls. The aim of the present study was to examine whether neutrophilic inflammation, either in sputum or blood, is more prevalent in asthma and whether it correlates with disease severity. Methods ATLANTIS included 773 asthma patients, with blood collected from 767 (99%) and sputum from 228 patients (30%). Data were available from 244 healthy controls, all providing blood and 126 (52%) providing sputum. Asthma patients were characterised, including parameters of large and small airways disease at baseline and after 6 and 12 months of follow-up. Sputum and blood neutrophilia were defined as values exceeding the upper quartile in asthma patients. Results The prevalence of sputum neutrophilia did not differ between asthma patients and healthy controls. Asthma patients with sputum neutrophilia did not display more severe symptoms, large or small airways disease or more frequent exacerbations. Blood neutrophilia was more common in asthma and was associated with higher body mass index, female sex, current smoking and systemic corticosteroid use. Patients with blood neutrophilia had a statistically significant, but small, increase in residual volume/total lung capacity. Blood neutrophilia was not associated with large or small airways disease or exacerbation risk. Conclusion Sputum and blood neutrophilia do not define a distinct clinical phenotype in asthma.
Collapse
Affiliation(s)
- Pauline J.M. Kuks
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tessa M. Kole
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Monica Kraft
- Samuel Bronfman Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Leonardo M. Fabbri
- Section of Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F. Rabe
- Dept of Medicine, Christian Albrechts University Kiel (member of the German Center for Lung Research (DZL)), Kiel, Germany
- Lungen Clinic Grosshansdorf (member of the DZL), Grosshansdorf, Germany
| | - Alberto Papi
- Research Centre on Asthma and COPD, University of Ferrara, Ferrara, Italy
| | - Chris Brightling
- Institute for Lung Health, National Institute for Health Research Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, University Hospital of South Manchester, University of Manchester, Manchester, UK
| | - Thys van der Molen
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Willem W.H. Kocks
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- General Practitioners Research Institute, Groningen, The Netherlands
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ian M. Adcock
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Pankaj K. Bhavsar
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Irene H. Heijink
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Simon D. Pouwels
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Huib A.M. Kerstjens
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dirk-Jan Slebos
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten van den Berge
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
50
|
Li Z, Liang Z, Wang P, Li W, Li Y, Liu N, Ma Q. SnS 2 QDs@MXene Ohmic Junction-Based Surface Plasmon Coupling ECL Sensor to Detect Saliva Exosome for the Diagnosis of Childhood Asthma. NANO LETTERS 2024; 24:15878-15885. [PMID: 39576655 DOI: 10.1021/acs.nanolett.4c04939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
This study represents a novel surface plasmon coupling electrochemiluminescence (SPC-ECL) method for detecting salivary exosomes and the diagnosis of childhood asthma. First, SnS2 QDs@MXene Ohmic junctions was developed as efficient ECL emitters. The Ohmic junction provided a low resistance to reduce the contact resistance and improve charge injection efficiency, which enhanced the ECL signal by 2.76 times. Furthermore, the self-assembled surface plasmonic Bi@SiO2 array was prepared. When the ECL of SnS2 QDs@MXene resonated with the electronic oscillations in the Bi@SiO2 NPs array, the luminescence intensity was enhanced and regulated into the directionally polarized signal by the SPC-ECL effect. Remarkably, the detection of CD9-exosomes in saliva was achieved successfully based on the above sensing system, which can be used to analysis the acute exacerbation and chronic persistence of childhood asthma.
Collapse
Affiliation(s)
- Zhenrun Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zihui Liang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Peilin Wang
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenyan Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Yameng Li
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Ning Liu
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| | - Qiang Ma
- Department of Analytical Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
| |
Collapse
|