1
|
Thio CLP, Shao JS, Luo CH, Chang YJ. Decoding innate lymphoid cells and innate-like lymphocytes in asthma: pathways to mechanisms and therapies. J Biomed Sci 2025; 32:48. [PMID: 40355861 PMCID: PMC12067961 DOI: 10.1186/s12929-025-01142-w] [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: 01/05/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Asthma is a chronic inflammatory lung disease driven by a complex interplay between innate and adaptive immune components. Among these, innate lymphoid cells (ILCs) and innate-like lymphocytes have emerged as crucial players in shaping the disease phenotype. Within the ILC family, group 2 ILCs (ILC2s), in particular, contribute significantly to type 2 inflammation through their rapid production of cytokines such as IL-5 and IL-13, promoting airway eosinophilia and airway hyperreactivity. On the other hand, innate-like lymphocytes such as invariant natural killer T (iNKT) cells can play either pathogenic or protective roles in asthma, depending on the stimuli and lung microenvironment. Regulatory mechanisms, including cytokine signaling, metabolic and dietary cues, and interactions with other immune cells, play critical roles in modulating their functions. In this review, we highlight current findings on the role of ILCs and innate-like lymphocytes in asthma development and pathogenesis. We also examine the underlying mechanisms regulating their function and their interplay with other immune cells. Finally, we explore current therapies targeting these cells and their effector cytokines for asthma management.
Collapse
Affiliation(s)
- Christina Li-Ping Thio
- Institute of Biomedical Sciences, Academia Sinica, No. 128 Academia Road, Section 2, Nankang, Taipei City, 115, Taiwan
| | - Jheng-Syuan Shao
- Institute of Biomedical Sciences, Academia Sinica, No. 128 Academia Road, Section 2, Nankang, Taipei City, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei City, 115, Taiwan
| | - Chia-Hui Luo
- Institute of Biomedical Sciences, Academia Sinica, No. 128 Academia Road, Section 2, Nankang, Taipei City, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei City, 115, Taiwan
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, No. 128 Academia Road, Section 2, Nankang, Taipei City, 115, Taiwan.
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung City, 404, Taiwan.
| |
Collapse
|
2
|
Bryant N, Muehling LM, Wavell K, Teague WG, Woodfolk JA. Rhinovirus as a driver of airway T cell dynamics in children with treatment-refractory recurrent wheeze. JCI Insight 2025; 10:e189480. [PMID: 40337866 DOI: 10.1172/jci.insight.189480] [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: 11/21/2024] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Severe asthma in children is notoriously difficult to treat, and its immunopathogenesis is complex. In particular, the contribution of T cells and relationships to antiviral immunity remain enigmatic. Here, we coupled deep phenotyping with machine learning methods to elucidate the dynamics of T cells in the lower airways of children with treatment-refractory recurrent wheeze, and examine rhinovirus (RV) as a driver. Our strategy revealed a T cell landscape dominated by type 1 and type 17 CD8+ signatures. Interrogation of phenotypic relationships coupled with trajectory mapping identified T cell migratory and differentiation pathways spanning the blood and airways that culminated in tissue residency, and involved transitions between type 1 and type 17 tissue-resident types. These dynamics were reflected in cytokine polyfunctionality. Use of machine learning tools to cross-compare T cell populations that were enriched in the airways of RV-positive children with those induced in the blood following experimental RV challenge precisely pinpointed RV-responsive signatures that contributed to T cell migratory and differentiation pathways. Despite their rarity, these signatures were also detected in the airways of RV-negative children. Together, our results underscore the aberrant nature of type 1 immunity in the airways of children with recurrent wheeze, and implicate an important viral trigger as a driver.
Collapse
Affiliation(s)
- Naomi Bryant
- Department of Medicine
- Department of Microbiology, Immunology, and Cancer Biology, and
| | | | - Kristin Wavell
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - W Gerald Teague
- Child Health Research Center, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Judith A Woodfolk
- Department of Medicine
- Department of Microbiology, Immunology, and Cancer Biology, and
| |
Collapse
|
3
|
Ye Q, Opoku G, Orlov M, Jaramillo AM, Holguin F, Vladar EK, Janssen WJ, Evans CM. Mucins and Their Roles in Asthma. Immunol Rev 2025; 331:e70034. [PMID: 40305069 DOI: 10.1111/imr.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Mucus is a crucial component of airway host defense. For optimal protection, its chief components-the mucins MUC5AC and MUC5B-need to be tightly regulated. Their expression localizes to specific secretory epithelial cell types capable of producing and secreting massive glycopolymers. In asthma, abnormal mucus is an important clinical problem that is effectively treated with therapies that directly target mucins. This review summarizes what is known about how mucin gene regulation, protein synthesis, and secretion are regulated in healthy and asthmatic lungs. Ultimately, a better understanding of these processes could help identify novel ways of preventing or reversing airway mucus dysfunction.
Collapse
Affiliation(s)
- Qihua Ye
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Gilda Opoku
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Marika Orlov
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Ana M Jaramillo
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Fernando Holguin
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Eszter K Vladar
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
| | - William J Janssen
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, USA
| | - Christopher M Evans
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, Colorado, USA
- Immunology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Integrated Physiology PhD Program, University of Colorado School of Medicine, Denver, Colorado, USA
- Research Service, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| |
Collapse
|
4
|
Li W, Marx N, Yang Q, Fang D, Zhang Y. Obesity: Next game changer of allergic airway diseases? Clin Transl Med 2025; 15:e70316. [PMID: 40329860 PMCID: PMC12056501 DOI: 10.1002/ctm2.70316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/05/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
Obesity and allergic diseases are global health concerns, both of which are seeing an increase in prevalence in recent years. Obesity has been recognised as an important comorbidity in subpopulations with allergic airway diseases, which represents a unique phenotype and endotype. Obesity-related allergic airway diseases are associated with exacerbated clinical symptom burden, altered immune response, increased disease severity and compromised predictive capability of conventional biomarkers for evaluating endotype and prognosis. Moreover, treatment of obesity-related allergic airway diseases is challenging because this unique endotype and phenotype is associated with poor response to standard therapeutic strategies. Therapeutic regimen that involves weight loss by non-surgical and surgical interventions, gut microbiome-targeted treatment, glucagon-like peptide-1 receptor agonist and other agents should be considered in this population. In this review, we outline the current knowledge of the impact of obesity on prevalence, endotypes, clinical symptom and management of allergic airway diseases. Increased understanding of the implications of obesity may contribute to better treatment options for the obesity-related refractory airway inflammation, particularly in precision medicine. KEY POINTS: Obesity can increase the prevalence of allergic airway diseases such as asthma, AR, and CRSwNP. Obesity alters the immune endotype and exacerbates clinical symptoms of respiratory allergic diseases. Obesity-related allergic airway diseases exhibit therapeutic resistance to standard treatment. Obesity-related allergic airway diseases constitute a distinct category of endotypes and phenotypes, requiring further in-depth research and novel therapeutic approaches.
Collapse
Affiliation(s)
- Wenlong Li
- Department of Otolaryngology‐Head and Neck SurgeryThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of AllergyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Otolaryngology‐Head and Neck SurgeryKey Laboratory of Airway Inflammatory Disease Research and Innovative Technology TranslationGuangzhouChina
- Naso‐Orbital‐Maxilla and Skull Base CenterThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Noah Marx
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Qintai Yang
- Department of Otolaryngology‐Head and Neck SurgeryThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of AllergyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Otolaryngology‐Head and Neck SurgeryKey Laboratory of Airway Inflammatory Disease Research and Innovative Technology TranslationGuangzhouChina
- Naso‐Orbital‐Maxilla and Skull Base CenterThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Deyu Fang
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Yana Zhang
- Department of Otolaryngology‐Head and Neck SurgeryThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of AllergyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
- Department of Otolaryngology‐Head and Neck SurgeryKey Laboratory of Airway Inflammatory Disease Research and Innovative Technology TranslationGuangzhouChina
- Naso‐Orbital‐Maxilla and Skull Base CenterThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
5
|
Simmalee K, Kawamatawong T, Vitte J, Demoly P, Lumjiaktase P. Exploring the pathogenesis and clinical implications of asthma, chronic obstructive pulmonary disease (COPD), and asthma-COPD overlap (ACO): a narrative review. Front Med (Lausanne) 2025; 12:1514846. [PMID: 40313547 PMCID: PMC12044671 DOI: 10.3389/fmed.2025.1514846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/26/2025] [Indexed: 05/03/2025] Open
Abstract
The complexity and diversity of the immune response in patients with asthma, chronic obstructive pulmonary disease (COPD), and asthma-COPD overlap present significant challenges for disease management. Relying on a limited number of biomarkers and clinical data is insufficient to fully reveal the immunopathogenesis of these diseases. However, in vitro technologies such as cell analysis, cytokine investigation, and nucleic acid sequencing have provided new insights into the underlying mechanisms of these diseases, leading to the discovery of several biomarkers-including cell degranulation, cell function, secreted cytokines, and single nucleotide polymorphisms-that have potential clinical implications. This paper reviews the immunopathogenesis in asthma, chronic obstructive pulmonary disease, and asthma-COPD overlap and examines the applications of recent in vitro models to detect candidate biomarkers that could enhance diagnostic precision, predict severity, monitor treatments, and develop new treatment strategies. A deeper understanding of the immune response in these diseases, along with the integration of in vitro models into clinical practice, could greatly improve the management of these respiratory diseases, making approaches more personalized and efficient.
Collapse
Affiliation(s)
- Kantapat Simmalee
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Theerasuk Kawamatawong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Joana Vitte
- Immunology Laboratory, University Hospital of Reims and INSERM UMR-S 1250 P3CELL, University of Reims Champagne-Ardenne, Reims, France
| | - Pascal Demoly
- Division of Allergy, University Hospital of Montpellier and IDESP, University of Montpellier - Inserm, Inria, Montpellier, France
| | - Putthapoom Lumjiaktase
- Department of Pathology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
6
|
Barkat MQ, Manzoor M, Xu C, Hussain N, Salawi A, Yang H, Hussain M. Severe asthma beyond bronchodilators: Emerging therapeutic approaches. Int Immunopharmacol 2025; 152:114360. [PMID: 40049087 DOI: 10.1016/j.intimp.2025.114360] [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: 01/21/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/24/2025]
Abstract
Asthma is characterized by reversible airway inflammation, obstruction, and structural remodeling, which lead to the eosinophils and lymphocytes accumulation at inflammation sites and the release of inflammatory cells, like mast cells and dendritic cells, from lungs' epithelial and smooth muscle cells that trigger the activation and release of cytokines and chemokines, attracting more cells and contributing to asthma development. Available pharmacological interventions, like bronchodilators and anti-inflammatory agents, are considered generally safe and effective to treat asthma, but many affected individuals with severe asthma still struggle with symptom control. This review highlights recent innovative therapies, such as chemoattractant receptor-homologous molecule expressed on Th2 cell (CRTH2) antagonists, S-nitrosoglutathione reductase (GSNOR) and phosphodiesterase (PDE) inhibitors, and other novel biological agents, which offer potential new strategies for managing severe asthma and may alter the disease's course. Kew words. Inflammation; CRTH2; GSNOR; PDE; Interleukins; Biological agents.
Collapse
Affiliation(s)
| | - Majid Manzoor
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310015, China
| | - Nadia Hussain
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, United Arab Emirates; AAU Health and Biomedical Research center, Al Ain University, Abu Dhabi, United Arab Emirates
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hao Yang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Musaddique Hussain
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
7
|
Kato A, Kita H. The immunology of asthma and chronic rhinosinusitis. Nat Rev Immunol 2025:10.1038/s41577-025-01159-0. [PMID: 40240657 DOI: 10.1038/s41577-025-01159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Asthma and chronic rhinosinusitis (CRS) are common chronic inflammatory diseases of the respiratory tract that have increased in prevalence over the past five decades. The clinical relationship between asthma and CRS has been well recognized, suggesting a common pathogenesis between these diseases. Both diseases are driven by complex airway epithelial cell and immune cell interactions that occur in response to environmental triggers such as allergens, microorganisms and irritants. Advances, including a growing understanding of the biology of the cells involved in the disease, the application of multiomics technologies and the performance of large-scale clinical studies, have led to a better understanding of the pathophysiology and heterogeneity of asthma and CRS. This research has promoted the concept that these diseases consist of several endotypes, in which airway epithelial cells, innate lymphoid cells, T cells, B cells, granulocytes and their mediators are distinctly involved in the immunopathology. Identification of the disease heterogeneity and immunological markers has also greatly improved the protocols for biologic therapies and the clinical outcomes in certain subsets of patients. However, many clinical and research questions remain. In this Review, we discuss recent advances in characterizing the immunological mechanisms of asthma and CRS, with a focus on the main cell types and molecules involved in these diseases.
Collapse
Affiliation(s)
- Atsushi Kato
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hirohito Kita
- Division of Allergy, Asthma, and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ, USA.
- Department of Immunology, Mayo Clinic Arizona, Scottsdale, AZ, USA.
| |
Collapse
|
8
|
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:S1933-0219(25)00029-7. [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] [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
|
9
|
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
|
10
|
Pastore D, Lupia C, D'Amato M, Bruni A, Garofalo E, Longhini F, Gallelli L, Vatrella A, Pelaia G, Pelaia C. Emerging biological treatments for asthma. Expert Opin Emerg Drugs 2025:1-11. [PMID: 39873193 DOI: 10.1080/14728214.2025.2460529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/24/2024] [Accepted: 01/27/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION Severe asthma is a chronic airway disease characterized by many pathomechanisms known as endotypes. Biological therapies targeting severe asthma endotypes have significantly improved the treatment of this disease, thus remarkably bettering patient quality of life. AREAS COVERED This review aims to describe current biological therapies for severe asthma, highlighting emerging ones. Several studies have confirmed the beneficial effects of currently available monoclonal antibodies targeting immunoglobulin E (IgE), interleukin-5 (IL-5) or its receptor, and interleukin-4 (IL-4)/interleukin-13 (IL-13) receptors (IL-4R/IL-13R). However, patients with T2-low asthma are not eligible for the above biological therapies. EXPERT OPINION New treatments are now moving toward targeting the upstream pathways of asthma pathogenesis, coordinated by innate cytokines such as alarmins. These key proinflammatory mediators orchestrate the activation of complex cellular networks including both innate and adaptive immune responses. Alarmins include thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and interleukin-33 (IL-33), which are released from injured airway epithelial cells. TSLP and the other alarmins are suitable targets of biological therapies which are effective for add-on treatment of type 2 asthma. Moreover, anti-alarmin monoclonal antibodies can be also beneficial for patients with T2-low, poorly controlled severe asthma.
Collapse
Affiliation(s)
- Daniela Pastore
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Chiara Lupia
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Maria D'Amato
- Department of Respiratory Medicine, "V. Monaldi University Hospital, Naples, Italy
| | - Andrea Bruni
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Eugenio Garofalo
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Federico Longhini
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Luca Gallelli
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Alessandro Vatrella
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Girolamo Pelaia
- Department of Health Sciences,University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
11
|
Yue M, Gaietto K, Han YY, Rosser FJ, Xu Z, Qoyawayma C, Acosta-Perez E, Canino G, Forno E, Chen W, Celedón JC. Transcriptomic Profiles in Nasal Epithelium and Asthma Endotypes in Youth. JAMA 2025; 333:307-318. [PMID: 39745770 PMCID: PMC11775736 DOI: 10.1001/jama.2024.22684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/07/2024] [Indexed: 01/29/2025]
Abstract
Importance T helper 2 (T2) cells and T helper 17 (T17) cells are CD4+ T cell subtypes involved in asthma. Characterizing asthma endotypes based on these cell types in diverse groups is important for developing effective therapies for youths with asthma. Objective To identify asthma endotypes in school-aged youths aged 6 to 20 years by examining the distribution and characteristics of transcriptomic profiles in nasal epithelium. Design, Setting, and Participants Cross-sectional analysis of nasal epithelial samples from 3 studies of youths with asthma aged 6 to 20 years: Stress and Treatment Response in Puerto Rican and African American Children with Asthma (STAR; N = 156), Epigenetic Variation and Childhood Asthma in Puerto Ricans (EVA-PR; N = 237), and Vitamin D Kids Asthma (VDKA; N = 66). Main Outcomes and Measures The primary outcome was nasal epithelial transcription profiles of 3 T2 and 5 T17 pathway genes. Clinical characteristics, total and allergen-specific immunoglobulin E (IgE), blood eosinophils, and lung function were compared across profiles in all studies. Results Mean ages for STAR, EVA-PR, and VDKA participants were 14.2, 15.4, and 10.3 years, respectively. The percentage of female participants ranged from 41% to 53.2% across studies. The predominant race or ethnicity was Puerto Rican in EVA-PR (100%) and Black or African American in STAR (71.8%) and VDKA (57.6%). Three transcriptomic profiles were identified: high T2 expression (T2HIGH), high T17 expression (T17HIGH), and low expression of both pathways (T2LOW/T17LOW). Across studies, T2HIGH was present in 23% to 29% of participants, T17HIGH in 35% to 47%, and T2LOW/T17LOW in 30% to 38%. In each study, median total IgE and blood eosinophils for the T2HIGH profile was higher than for the T2LOW profiles (IgE, 584-869 vs 105-382 IU/mL; eosinophils, 343-560 vs 164-413 cells/mL). Of the participants in all profiles, at least 50% had 1 or more positive allergen-specific IgEs. A differential expression meta-analysis identified 3516 and 2494 differentially expressed genes for the T2HIGH and T17HIGH profiles, respectively. The T17HIGH profile was associated with interleukin 17 and neutrophil signaling pathways and the T2HIGH profile was associated with interleukin 13 signaling pathways. Conclusions and Relevance Nasal transcriptomic profiles consistent with T2-high, T17-high, and T2-low/T17-low endotypes occurred in similar proportions across 3 studies of predominantly racially and ethnically minoritized youths with asthma. Most participants had T2-low asthma endotypes and sensitization to 1 or more allergens was common among these endotypes.
Collapse
Affiliation(s)
- Molin Yue
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kristina Gaietto
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yueh Ying Han
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Franziska J. Rosser
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhongli Xu
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher Qoyawayma
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Glorisa Canino
- Behavioral Sciences Research Institute, San Juan, Puerto Rico
- Department of Pediatrics, Medical Science Campus, University of Puerto Rico, San Juan
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wei Chen
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
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
|
13
|
Lindsley AW, Lugogo N, Reeh KAG, Spahn J, Parnes JR. Asthma Biologics Across the T2 Spectrum of Inflammation in Severe Asthma: Biomarkers and Mechanism of Action. J Asthma Allergy 2025; 18:33-57. [PMID: 39830595 PMCID: PMC11742565 DOI: 10.2147/jaa.s496630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Airway inflammation, a hallmark feature of asthma, drives many canonical features of the disease, including airflow limitation, mucus plugging, airway remodeling, and hyperresponsiveness. The T2 inflammatory paradigm is firmly established as the dominant mechanism of asthma pathogenesis, largely due to the success of inhaled corticosteroids and biologic therapies targeting components of the T2 pathway, including IL-4, IL-5, IL-13, and thymic stromal lymphopoietin (TSLP). However, up to 30% of patients may lack signatures of meaningful T2 inflammation (ie, T2 low). In T2-low asthma patients, T2 inflammation may be masked due to anti-inflammatory treatments or may be highly variable depending on exposure to common asthma triggers such as allergens, respiratory infections, and smoke or pollution. The epithelium and epithelial cytokines (TSLP, IL-33) are increasingly recognized as upstream drivers of canonical T2 pathways and as modulators of various effector cells, including mast cells, eosinophils, and neutrophils, which impact the pathological manifestations of airway smooth muscle hypertrophy, hypercontractility, and airway hyperresponsiveness. Approved biologics for severe asthma target several distinct mechanisms of action, leading to differential effects on the spectrum of T2 inflammation, inflammatory biomarkers, and treatment efficacy (reducing asthma exacerbations, improving lung function, and diminishing symptoms). The approved anti-asthma biologics primarily target T2 immune pathways, with little evidence suggesting a benefit of targeting non-T2 asthma-associated mediators. Indeed, many negative results challenge current assumptions about the etiology of non-T2 asthma and raise doubts about the viability of targeting popular alternative inflammatory pathways, such as T17. Novel data have emerged from the use of biologics to treat various inflammatory mediators and have furthered our understanding of pathogenic mechanisms that drive asthma. This review discusses inflammatory pathways that contribute to asthma, quantitatively outlines effects of available biologics on biomarkers, and summarizes data and challenges from clinical trials that address non-T2 mechanisms of asthma.
Collapse
Affiliation(s)
| | - Njira Lugogo
- Michigan Medicine Asthma Program, University of Michigan, Ann Arbor, MI, USA
| | | | | | | |
Collapse
|
14
|
Seluk L, Davis AE, Rhoads S, Wechsler ME. Novel asthma treatments: Advancing beyond approved novel step-up therapies for asthma. Ann Allergy Asthma Immunol 2025; 134:9-18. [PMID: 39393433 DOI: 10.1016/j.anai.2024.09.016] [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: 09/18/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/13/2024]
Abstract
Over the past 2 decades, the management of severe asthma has shifted from relying on inhaled corticosteroids and bronchodilators to more precise, targeted approaches. Monoclonal antibodies designed to address specific molecular pathways in asthma have transformed care for patients with severe asthma. Because therapy targeting IgE became the first biologic developed for allergic asthma in 2003, monoclonal antibodies targeting interleukin (IL)-5, IL-5 receptor, IL-4/-13 receptor, and thymic stromal lymphopoietin have been approved for treating difficult-to-treat asthma, improving symptoms, reducing exacerbations, and reducing oral corticosteroid dosing. Despite these advances, many patients continue to experience asthma exacerbations and symptoms and fail to achieve remission. To address this, pharmaceutical companies and researchers are exploring novel therapies targeting different aspects of asthma pathophysiology, including cytokines, enzymes, and cellular pathways. Innovative treatments such as inhaled biologics, ultra-long-acting biologics, and combination biologics are in development. New molecular targets, such as Bruton tyrosine kinase, OX-40 ligand, and Janus kinase, offer promise for addressing unmet needs in asthma care. Although many therapies have failed to get approval for use because of a lack of efficacy, trial design, or toxicity, these experiments still provide insights into asthma's underlying mechanisms. The future of asthma management looks promising, with emerging therapies aiming to improve patient outcomes. The challenge will lie in identifying the right therapy for each patient and developing personalized treatment strategies.
Collapse
Affiliation(s)
- Lior Seluk
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado
| | - Andrea E Davis
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado; Division of Pulmonary Sciences and Critical Care Medicine, The Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sarah Rhoads
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado; Division of Pulmonary Sciences and Critical Care Medicine, The Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michael E Wechsler
- Division of Pulmonary, Critical Care, and Sleep Medicine, National Jewish Health, Denver, Colorado.
| |
Collapse
|
15
|
Zidan E, Wilson G, Shin JJ, Chupp G. The next frontier: Defining and optimizing treatments for patients with type 2 low asthma. Ann Allergy Asthma Immunol 2025; 134:3-4. [PMID: 39414022 DOI: 10.1016/j.anai.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Affiliation(s)
- Elena Zidan
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gabriella Wilson
- Section of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Junghee Jenny Shin
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut; Section of Pulmonary, Allergy, Immunology & Sleep Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
16
|
Tashiro H, Kuwahara Y, Kurihara Y, Takahashi K. Molecular mechanisms and clinical impact of biologic therapies in severe asthma. Respir Investig 2025; 63:50-60. [PMID: 39642687 DOI: 10.1016/j.resinv.2024.11.015] [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: 09/30/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024]
Abstract
Severe asthma is a critical condition for patients with asthma, characterized by frequent exacerbations, decreased pulmonary function, and unstable symptoms related to asthma. Consequently, the administration of systemic corticosteroids, which cause secondary damage because of their adverse effects, is considered. Recently, several types of molecular-targeted biological therapies have become available for patients with severe asthma, and they have a capacity to improve the pathophysiology of severe asthma. However, several clinical reports indicate that the effects differ depending on the biological targets of asthma in individual patients. In this review, the molecular mechanisms and clinical impact of biologic therapies in severe asthma are described. In addition, molecules targeted by possible future biologics are also addressed. Better understanding of the mechanistic basis for the role of biologics in severe asthma could lead to new therapeutic options for these patients.
Collapse
Affiliation(s)
- Hiroki Tashiro
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kuwahara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Yuki Kurihara
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan
| | - Koichiro Takahashi
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Saga Prefecture, 849-8501, Japan.
| |
Collapse
|
17
|
Berkinbayeva M, Gu W, Chen Z, Gao P. Group 3 Innate Lymphoid Cells: A Potential Therapeutic Target for Steroid Resistant Asthma. Clin Rev Allergy Immunol 2024; 68:1. [PMID: 39751959 PMCID: PMC11698894 DOI: 10.1007/s12016-024-09012-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Asthma is a chronic airway inflammatory disease that affects millions globally. Although glucocorticoids are a mainstay of asthma treatment, a subset of patients show resistance to these therapies, resulting in poor disease control and increased morbidity. The complex mechanisms underlying steroid-resistant asthma (SRA) involve Th1 and Th17 lymphocyte activity, neutrophil recruitment, and NLRP3 inflammasome activation. Recent studies provided evidence that innate lymphoid cells type 3 (ILC3s) might be potential therapeutic targets for non-eosinophilic asthma (NEA) and SRA. Like Th17 cells, ILC3s play crucial roles in immune responses, inflammation, and tissue homeostasis, contributing to disease severity and corticosteroid resistance in NEA. Biologics targeting ILC3-related pathways have shown promise in managing Th2-low asthma, suggesting new avenues for SRA treatment. This review aims to explore the risk factors for SRA, discuss the challenges and mechanisms underlying SRA, consolidate current findings on innate lymphoid cells, and elucidate their role in respiratory conditions. We present the latest findings on the involvement of ILC3s in human diseases and explore their potential mechanisms in SRA development. Furthermore, we review emerging therapeutic biologics targeting ILC3-related pathways in managing NEA and SRA. This review highlights current challenges, and emerging therapeutic strategies, and addresses a significant gap in asthma research, with implications for improving the management of steroid-resistant asthma.
Collapse
Affiliation(s)
- Marzhan Berkinbayeva
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
| | - Wenjing Gu
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhifeng Chen
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, The Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Room 3B.71, Baltimore, MD, 21224, USA.
| |
Collapse
|
18
|
Emala CW, Saroya TK, Miao Y, Wang S, Sang S, DiMango EA. Low-Dose Oral Ginger Improves Daily Symptom Scores in Asthma. Pharmaceuticals (Basel) 2024; 17:1651. [PMID: 39770492 PMCID: PMC11728807 DOI: 10.3390/ph17121651] [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] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objective: A significant number of individuals with asthma have poorly controlled daily symptoms and utilize dietary supplements such as ginger in a quest for improved symptom control; however, its effectiveness at improving the control of symptoms is unproven. We questioned whether low-dose oral ginger would improve subjective and objective measurements of asthma control in mild-to-moderate asthmatics. Methods: We performed a randomized, placebo-controlled, double-blinded study of a low dose (1 g twice daily) of a dietary supplement of ginger in 32 mild-to-moderate uncontrolled asthmatics over a 2-month trial period while maintaining daily conventional asthma therapies. The planned primary outcomes included an increased tolerance to inhaled methacholine and decreased concentrations of fractional excretion of exhaled nitric oxide (FeNO). Secondary planned outcomes included measurements of asthma control by the Asthma Control Test (ACT), a 2-week symptom recall test, and the Juniper mini Asthma Quality of Life Questionnaire (AQLQ), and blood eosinophils and asthma-associated cytokines. Results: Exhaled nitric oxide or blood eosinophils were not changed by oral ginger. However, three different measures of asthma symptom control were improved by the 28-day time point of oral ginger. Asthma-associated serum cytokines (IL-13 and IL-17A) were modulated by oral ginger. Conclusions: This is the first demonstration that a small daily dose of a dietary supplement of ginger may improve asthma symptoms and reduce inflammation in human asthmatics. These findings support the need for additional studies using larger doses of ginger in specific endotypes of asthmatics that may identify a novel therapeutic for asthma.
Collapse
Affiliation(s)
- Charles W. Emala
- Department of Anesthesiology, Columbia University Vagelos College of Physicians and Surgeons, 628 W. 168th St. PH 505 Center, New York, NY 10032, USA
| | - Tarnjot K. Saroya
- Department of Medicine (Pulmonology, Allergy and Critical Care), Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yuqi Miao
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10027, USA
| | - Shuang Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10027, USA
| | - Shengmin Sang
- Laboratory for Functional Foods and Human Health, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Emily A. DiMango
- Department of Medicine (Pulmonology, Allergy and Critical Care), Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| |
Collapse
|
19
|
Rao X, Luo H, Luo K, Hu C. Silencing SMAD4 inhibits inflammation and ferroptosis in asthma by blocking the IL-17A signaling pathway. Respir Res 2024; 25:429. [PMID: 39643876 PMCID: PMC11622552 DOI: 10.1186/s12931-024-03052-1] [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: 07/29/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND The TGF-β/SMAD signaling pathway is crucial in the pathogenesis of asthma. However, SMAD family member 4 (SMAD4), a key mediator of TGF-β, its roles and underlying mechanisms in asthma remain unclear. METHODS The in vivo and in vitro roles of SMAD4 in asthma were investigated through an ovalbumin (OVA)-induced mouse model and an interleukin-13 (IL-13)-induced cell model. The molecular mechanism of SMAD4 influenced asthma was examined using transcriptome sequencing, followed by feedback experiments involving recombinant human interleukin 17 A (rhIL-17 A), an IL-17 A signaling pathway activator. RESULTS SMAD4 was highly expressed in the asthma models. SMAD4 silencing alleviated damage to lung tissue and decreased inflammatory infiltration. Expression levels of Caspase-3, IgG, and inflammatory factors were reduced after silencing SMAD4. Silencing SMAD4 suppressed ferroptosis. Silencing SMAD4 also enhanced IL-13-induced BEAS-2B cell proliferation and suppressed apoptosis. Furthermore. IL-17 A signaling pathway was promoted in the asthma models, as evidenced by elevated IL-17RA, IL-17 A, and Act1 protein levels. SMAD4 silencing inhibited the expression levels of these IL-17 A pathway-associated proteins. Moreover, rhIL-17 A treatment notably reversed the impacts of SMAD4 silencing on asthma in the IL-13-induced cell model and OVA-induced mouse model, indicating that silencing SMAD4 inhibited inflammation and ferroptosis in asthma by blocking the IL-17 A signaling pathway. CONCLUSION Silencing SMAD4 prevents inflammation and ferroptosis in asthma by inhibiting the IL-17 pathway, which provides a novel potential approach for asthma therapy.
Collapse
Affiliation(s)
- Xingyu Rao
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Hong Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Kaiyuan Luo
- Department of Pediatrics, First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Zhanggong District, Ganzhou, 341000, China
| | - Chaohua Hu
- Department of Surgery I, The Third Affiliated Hospital of Gannan Medical University/Affiliated Stomatological Hospital, No. 46, Jingjiu Road, Zhanggong District, Ganzhou, Jiangxi Province, 341000, China.
| |
Collapse
|
20
|
Mleczko M, Gerkowicz A, Krasowska D. Co-Occurrence of Psoriasis and Asthma in the Pediatric Population: A Systematic Review and Meta-Analysis. J Clin Med 2024; 13:6991. [PMID: 39598135 PMCID: PMC11594869 DOI: 10.3390/jcm13226991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction: The risk of asthma in patients with psoriasis and that of psoriasis in patients with asthma have increased, but relevant data for the pediatric population are lacking. Therefore, we performed a meta-analysis to assess the pooled association between psoriasis and asthma in children and adolescents. Methods: We conducted an extensive search of the medical literature databases through to July 2024. The estimated risk ratios (RRs) and corresponding 95% confidence intervals (CIs) were computed. Results: Three studies, involving 5310 children with psoriasis and 1,539,029 control participants, were included to evaluate the incidence of asthma in children with psoriasis. The meta-analysis indicated a significantly increased risk of asthma in children with psoriasis [RR 1.38 (95% CI, 1.28-1.49)]. Additionally, two studies involving 104,369 asthmatic children and 1,539,029 controls were included to evaluate the incidence of psoriasis in children with asthma. The meta-analysis indicated a significant increase in the risk of psoriasis in children with asthma [RR 1.17 (95% CI, 0.70-1.95)]. Conclusions: This meta-analysis offers evidence supporting the association between psoriasis and asthma in pediatric populations. Therefore, physicians should make patients aware of the connection between these two chronic diseases.
Collapse
Affiliation(s)
- Mateusz Mleczko
- Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 11 Staszica St., 20-081 Lublin, Poland
| | | | | |
Collapse
|
21
|
Bryant N, Muehling LM, Wavell K, Teague WG, Woodfolk JA. Rhinovirus as a Driver of Airway T-Cell Dynamics in Children with Severe Asthma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623877. [PMID: 39605344 PMCID: PMC11601360 DOI: 10.1101/2024.11.15.623877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Severe asthma in children is notoriously difficult to treat, and its immunopathogenesis is complex. In particular, the contribution of T cells and relationships to anti-viral immunity, remain enigmatic. Here, we coupled deep phenotyping with machine learning methods to resolve the dynamics of T cells in the diseased lower airways, and examined rhinovirus (RV) as a driver. Our strategy revealed a T-cell landscape dominated by type 1 and type 17 CD8+ signatures. Interrogation of phenotypic relationships coupled with trajectory mapping identified T-cell migratory and differentiation pathways spanning the blood and airways that culminated in tissue residency, and included transitions between type 1 and type 17 tissue-resident types. These T-cell dynamics were reflected in cytokine polyfunctionality in situ . Use of machine learning to cross-compare T-cell populations that were enriched in the airways of RV-positive children with those induced in the blood after RV challenge in an experimental infection model, precisely pinpointed RV-responsive signatures that mapped to T-cell differentiation pathways. Despite their rarity, these signatures were detected in the airways of uninfected children. Together, our results underscore the aberrant nature of type 1 immunity in the airways of children with severe asthma, and implicate an important viral trigger as a driver.
Collapse
|
22
|
Pasha MA, Hopp RJ, Habib N, Tang DD. Biomarkers in asthma, potential for therapeutic intervention. J Asthma 2024; 61:1376-1391. [PMID: 38805392 DOI: 10.1080/02770903.2024.2361783] [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: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/26/2024] [Indexed: 05/30/2024]
Abstract
Asthma is a heterogeneous disease characterized by multiple phenotypes with varying risk factors and therapeutic responses. This Commentary describes research on biomarkers for T2-"high" and T2-"low" inflammation, a hallmark of the disease. Patients with asthma who exhibit an increase in airway T2 inflammation are classified as having T2-high asthma. In this endotype, Type 2 cytokines interleukins (IL)-4, IL-5, and IL-13, plus other inflammatory mediators, lead to increased eosinophilic inflammation and elevated fractional exhaled nitric oxide (FeNO). In contrast, T2-low asthma has no clear definition. Biomarkers are considered valuable tools as they can help identify various phenotypes and endotypes, as well as treatment response to standard treatment or potential therapeutic targets, particularly for biologics. As our knowledge of phenotypes and endotypes expands, biologics are increasingly integrated into treatment strategies for severe asthma. These treatments block specific inflammatory pathways or single mediators. While single or composite biomarkers may help to identify subsets of patients who might benefit from these treatments, only a few inflammatory biomarkers have been validated for clinical application. One example is sputum eosinophilia, a particularly useful biomarker, as it may suggest corticosteroid responsiveness or reflect non-compliance to inhaled corticosteroids. As knowledge develops, a meaningful goal would be to provide individualized care to patients with asthma.
Collapse
Affiliation(s)
- M Asghar Pasha
- Department of Medicine, Division of Allergy and Immunology, Albany Medical College, Albany, NY, USA
| | - Russell J Hopp
- Department of Pediatrics, University of NE Medical Center and Children's Hospital and Medical Center, Omaha, NE, USA
| | - Nazia Habib
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
23
|
Brüggemann TR, Krishnamoorthy N, Hagner M, Matschiner G, Jaquin T, Tavares LP, Peh HY, Levy BD. A new Anticalin protein for IL-23 inhibits non-type 2 allergen-driven mouse lung inflammation and airway hyperresponsiveness. Am J Physiol Lung Cell Mol Physiol 2024; 327:L624-L633. [PMID: 39104317 PMCID: PMC11563638 DOI: 10.1152/ajplung.00295.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024] Open
Abstract
Severe asthma is a syndromic label assigned to patients based on clinical parameters, yet there are diverse underlying molecular endotypes in severe asthma pathobiology. Immunophenotyping of asthma biospecimens commonly includes a mixture of granulocytes and lymphocytes. Recently, a subset of patients with severe asthma was defined as non-type 2 with neutrophil-enriched inflammation associated with increased Th17 CD4+ T cells and IL-17 levels. Here, we used an allergen-driven mouse model of increased IL-17 and mixed granulocyte lung inflammation to determine the impact of upstream regulation by an Anticalin protein that specifically binds IL-23. Airway administration of the IL-23-binding Anticalin protein (AcIL-23) decreased lung neutrophils, eosinophils, macrophages, lymphocytes, IL-17+ CD4 T cells, mucous cell metaplasia, and methacholine-induced airway hyperresponsiveness. Selective targeting of IL-23 with a monoclonal antibody (IL-23p19; αIL-23) also decreased macrophages, IL-17+ CD4 T cells, and airway hyperresponsiveness. In contrast, a monoclonal antibody against IL-17A (αIL-17A) had no significant effect on airway hyperresponsiveness but did decrease lung neutrophils, macrophages, and IL-17+ CD4 T cells. Targeting the IL-23 pathway did not significantly change IL-5+ or IL-13+ CD4 T cells. Together, these data indicate that airway AcIL-23 mirrored the activity of systemic anti-IL-23 antibody to decrease airway hyperresponsiveness in addition to mixed granulocytic inflammation and that these protective actions were broader than blocking IL-17A or IL-5 alone, which selectively decreased airway neutrophils and eosinophils, respectively.NEW & NOTEWORTHY This is the first report of an Anticalin protein engineered to neutralize IL-23 (AcIL-23). Airway administration of AcIL-23 in mice regulated allergen-driven airway inflammation, mucous cell metaplasia, and methacholine-induced airway hyperresponsiveness. In mixed granulocytic allergic lung inflammation, immune regulation of IL-23 was broader than neutralization of either IL-17 or IL-5.
Collapse
Affiliation(s)
- Thayse R Brüggemann
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Nandini Krishnamoorthy
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | | | | | | | - Luciana P Tavares
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
24
|
Boyd JT, Khanwalkar AR. Biologics in Chronic Rhinosinusitis: Current and Emerging. Immunol Allergy Clin North Am 2024; 44:657-671. [PMID: 39389716 DOI: 10.1016/j.iac.2024.07.005] [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: 10/12/2024]
Abstract
Chronic rhinosinusitis (CRS) is categorized phenotypically into CRS with and without nasal polyps (CRSwNP, CRSsNP). Endotyping categorizes the disease based on immune cell activity and inflammatory mechanisms into Type 1, Type 2, and Type 3. The Type 2 endotype is the most researched and associated with asthma, atopic disease, and severe CRSwNP. For patients with poorly controlled CRSwNP, there are 3 approved biologic treatments: omalizumab, dupilumab, and mepolizumab. Many other biologics are being tested in Type 2, non-Type 2, and mixed endotypes in CRSwNP and CRSsNP. These studies will play a significant role in shaping the future of CRS management.
Collapse
Affiliation(s)
- Jacob T Boyd
- Department of Otolaryngology - Head and Neck Surgery, University of Colorado Anschutz School of Medicine, 12631 East 17th Avenue, MSB 205 Room 3001, Aurora, CO 80045, USA
| | - Ashoke R Khanwalkar
- Department of Otolaryngology - Head and Neck Surgery, University of Colorado Anschutz School of Medicine, 12631 East 17th Avenue, MSB 205 Room 3001, Aurora, CO 80045, USA.
| |
Collapse
|
25
|
Desai B, Adrish M, Mohan A, Lugogo NL. Biologics in Asthma: Emerging Biologics. Immunol Allergy Clin North Am 2024; 44:751-763. [PMID: 39389722 DOI: 10.1016/j.iac.2024.07.008] [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] [Indexed: 10/12/2024]
Abstract
Advances in our understanding of asthma pathophysiology have led to the advent of multiple targeted asthma therapies such as biologics. However, partial response to biologics occurs, indicating residual disease activity in some patients. Hence, there exists a need for new therapies that focus on novel pathways, alongside perhaps evaluation of combination biologic therapies and modulators of downstream cytokine activation. Therefore, although our current focus is on biologics; it is critical to take a more holistic approach including consideration for nonbiologic therapies that have the potential to significantly advance asthma care.
Collapse
Affiliation(s)
- Brinda Desai
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Muhammad Adrish
- Department of Pulmonary & Critical Care, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Arjun Mohan
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2d21, Ann Arbor, MI 48109, USA
| | - Njira L Lugogo
- Department of Medicine, University of Michigan, 300 North Ingalls Street, Suite 2c40, Ann Arbor, MI 48109, USA.
| |
Collapse
|
26
|
Feng Y, Xu L, Zhang J, Bin J, Pang X, He S, Fang L. Allergenic protein-induced type I hypersensitivity models: a review. FRONTIERS IN ALLERGY 2024; 5:1481011. [PMID: 39483683 PMCID: PMC11525013 DOI: 10.3389/falgy.2024.1481011] [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: 08/15/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
Context Type I hypersensitivity affects approximately one-third of the global population. As the pathophysiology underlying the development of type I hypersensitivity (asthma, food allergy, and anaphylactic shock, etc.) is complex and heterogeneous, animal model studies continue to be the key to identifying novel molecular pathways and providing therapeutic strategies. Objective Selection of the animal model should be done with careful consideration of the protocol variables, animal species, and strains to accurately reflect the clinical symptoms typical of humans. Methods The following databases were searched: PubMed and Web of Science. Results and conclusion Foreign allergens include allergenic proteins and chemical haptens. This review summarizes the various methods used for designing animal models of common allergenic protein-induced type I hypersensitivity, namely, passive anaphylaxis model, active systemic anaphylaxis/anaphylaxis shock model, food allergy model, asthma model, and IgE-mediated cell models. Additionally, we summarize shrimp tropomyosin-induced type I hypersensitivity models from our previous studies and discuss their advantages and limitations compared with that of ovalbumin-induced models.
Collapse
Affiliation(s)
- Yanhua Feng
- Paediatric Department, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China
| | - Liangyu Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jinming Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jinlian Bin
- Paediatric Department, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xialing Pang
- Paediatric Department, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China
| | - Sheng He
- Paediatric Department, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Guangxi Clinical Research Center for Pediatric Diseases, Nanning, China
- Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Lei Fang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research, Yangzhou University Medical College, Yangzhou, China
| |
Collapse
|
27
|
Hansi RK, Ranjbar M, Whetstone CE, Gauvreau GM. Regulation of Airway Epithelial-Derived Alarmins in Asthma: Perspectives for Therapeutic Targets. Biomedicines 2024; 12:2312. [PMID: 39457624 PMCID: PMC11505104 DOI: 10.3390/biomedicines12102312] [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: 09/23/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Asthma is a chronic respiratory condition predominantly driven by a type 2 immune response. Epithelial-derived alarmins such as thymic stromal lymphopoietin (TSLP), interleukin (IL)-33, and IL-25 orchestrate the activation of downstream Th2 cells and group 2 innate lymphoid cells (ILC2s), along with other immune effector cells. While these alarmins are produced in response to inhaled triggers, such as allergens, respiratory pathogens or particulate matter, disproportionate alarmin production by airway epithelial cells can lead to asthma exacerbations. With alarmins produced upstream of the type 2 inflammatory cascade, understanding the pathways by which these alarmins are regulated and expressed is critical to further explore new therapeutics for the treatment of asthmatic patients. This review emphasizes the critical role of airway epithelium and epithelial-derived alarmins in asthma pathogenesis and highlights the potential of targeting alarmins as a promising therapeutic to improve outcomes for asthma patients.
Collapse
Affiliation(s)
| | | | | | - Gail M. Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada; (R.K.H.); (M.R.); (C.E.W.)
| |
Collapse
|
28
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
29
|
Sim S, Choi Y, Park HS. Update on Inflammatory Biomarkers for Defining Asthma Phenotype. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:462-472. [PMID: 39363766 PMCID: PMC11450439 DOI: 10.4168/aair.2024.16.5.462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Asthma is a chronic heterogeneous disease characterized by various symptoms and persistent airway inflammation, resulting in progressive lung function decline. Classifying asthma phenotypes/endotypes is crucial because the underlying mechanisms and long-term outcomes vary from patient to patient. Recent trials have identified several biomarkers for classifying asthma phenotypes/endotypes, and current treatments have been developed on the basis of these biomarkers. Conventional biomarkers, including immunoglobulin E, blood/sputum eosinophil counts, airway obstruction or reversibility, and fractional exhaled nitric oxide, are widely used to diagnose asthma. However, these markers have some limitations, necessitating the discovery of additional biomarkers. Therefore, this review summarizes recently suggested biomarkers for representing type 2-high (eosinophilic) vs. type 2-low (neutrophilic) asthma, non-steroidal anti-inflammatory drug-exacerbated respiratory disease, and severe asthma. Additionally, we discuss the potential benefits of these biomarkers in classifying specific phenotypes/endotypes and managing asthmatic patients.
Collapse
Affiliation(s)
- Soyoon Sim
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
30
|
Li Y, Yang T, Jiang B. Neutrophil and neutrophil extracellular trap involvement in neutrophilic asthma: A review. Medicine (Baltimore) 2024; 103:e39342. [PMID: 39183388 PMCID: PMC11346896 DOI: 10.1097/md.0000000000039342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/21/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
Asthma is a highly prevalent chronic inflammatory disease characterized by variable airflow obstruction and airway hyperresponsiveness. Neutrophilic asthma (NA) is classified as "type 2 low" asthma, defined as 65% or more neutrophils in the total cell count. There is no clear consensus on the pathogenesis of NA, and the accumulation of neutrophils and release of neutrophil extracellular traps (NETs) may be responsible for its development. A NET is a large extracellular meshwork comprising cell membrane and granule proteins. It is a powerful antimicrobial defence system that traps, neutralizes, and kills bacteria, fungi, viruses, and parasites and prevents the spread of microorganisms. However, dysregulation of NETs may lead to chronic airway inflammation, is associated with worsening of asthma, and has been the subject of major research advances in chronic lung diseases in recent years. NA is insensitive to steroids, and there is a need to find effective biomarkers as targets for the treatment of NA to replace steroids. This review analyses the mechanisms of action between asthmatic neutrophil recruitment and NET formation and their impact on NA development. It also discusses their possible therapeutic significance in NA, summarizing the advances made in NA agents and providing strategies for the treatment of NA, provide a theoretical basis for the development of new therapeutic drugs, thereby improving the level of diagnosis and treatment, and promoting the research progress in the field of asthma.
Collapse
Affiliation(s)
- Yuemu Li
- Institutes of Integrative Medicine, Heilongjiang Provincial Hospital of Traditional Chinese Medicine, Heilongjiang, China
| | - Tianyi Yang
- Institutes of Integrative Medicine, Heilongjiang Provincial Hospital of Traditional Chinese Medicine, Heilongjiang, China
| | - Baihua Jiang
- Institutes of Integrative Medicine, Heilongjiang Provincial Hospital of Traditional Chinese Medicine, Heilongjiang, China
| |
Collapse
|
31
|
Sharma S, Gerber AN, Kraft M, Wenzel SE. Asthma Pathogenesis: Phenotypes, Therapies, and Gaps: Summary of the Aspen Lung Conference 2023. Am J Respir Cell Mol Biol 2024; 71:154-168. [PMID: 38635858 PMCID: PMC11299090 DOI: 10.1165/rcmb.2024-0082ws] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/17/2024] [Indexed: 04/20/2024] Open
Abstract
Although substantial progress has been made in our understanding of asthma pathogenesis and phenotypes over the nearly 60-year history of the Aspen Lung Conferences on asthma, many ongoing challenges exist in our understanding of the clinical and molecular heterogeneity of the disease and an individual patient's response to therapy. This report summarizes the proceedings of the 2023 Aspen Lung Conference, which was organized to review the clinical and molecular heterogeneity of asthma and to better understand the impact of genetic, environmental, cellular, and molecular influences on disease susceptibility, heterogeneity, and severity. The goals of the conference were to review new information about asthma phenotypes, cellular processes, and cellular signatures underlying disease heterogeneity and treatment response. The report concludes with ongoing gaps in our understanding of asthma pathobiology and provides some recommendations for future research to better understand the clinical and basic mechanisms underlying disease heterogeneity in asthma and to advance the development of new treatments for this growing public health problem.
Collapse
Affiliation(s)
- Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anthony N. Gerber
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, National Jewish Health, Denver, Colorado
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Health System, New York, New York; and
| | - Sally E. Wenzel
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Remcho TP, Kolls JK. Unfolding the Role of Th17 Cells in Neutrophilic Lung Inflammation. Am J Respir Cell Mol Biol 2024; 71:137-138. [PMID: 38747688 PMCID: PMC11299083 DOI: 10.1165/rcmb.2024-0180ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Affiliation(s)
- T Parks Remcho
- Center for Translational Research in Infection and Inflammation Tulane School of Medicine New Orleans, Louisiana
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation Tulane School of Medicine New Orleans, Louisiana
| |
Collapse
|
33
|
Ishmael L, Casale T, Cardet JC. Molecular Pathways and Potential Therapeutic Targets of Refractory Asthma. BIOLOGY 2024; 13:583. [PMID: 39194521 DOI: 10.3390/biology13080583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024]
Abstract
Asthma is a chronic inflammatory lung disease. Refractory asthma poses a significant challenge in management due to its resistance to standard therapies. Key molecular pathways of refractory asthma include T2 inflammation mediated by Th2 and ILC2 cells, eosinophils, and cytokines including IL-4, IL-5, and IL-13. Additionally, non-T2 mechanisms involving neutrophils, macrophages, IL-1, IL-6, and IL-17 mediate a corticosteroid resistant phenotype. Mediators including alarmins (IL-25, IL-33, TSLP) and OX40L have overlap between T2 and non-T2 inflammation and may signify unique pathways of asthma inflammation. Therapies that target these pathways and mediators have proven to be effective in reducing exacerbations and improving lung function in subsets of severe asthma patients. However, there are patients with severe asthma who do not respond to approved therapies. Small molecule inhibitors, such as JAK-inhibitors, and monoclonal antibodies targeting mast cells, IL-1, IL-6, IL-33, TNFα, and OX40L are under investigation for their potential to modulate inflammation involved in refractory asthma. Understanding refractory asthma heterogeneity and identifying mediators involved are essential in developing therapeutic interventions for patients unresponsive to currently approved biologics. Further investigation is needed to develop personalized treatments based on these molecular insights to potentially offer more effective treatments for this complex disease.
Collapse
Affiliation(s)
- Leah Ishmael
- Division of Pulmonary, Allergy, and Sleep Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Thomas Casale
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Juan Carlos Cardet
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
34
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [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: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Kuramoto K, Morishima Y, Yoshida K, Ano S, Kawashima K, Yabuuchi Y, Sakai C, Matsumura S, Nishino K, Yazaki K, Matsuyama M, Kiwamoto T, Ishii Y, Hizawa N. Nrf2 Deficiency Accelerates IL-17-Dependent Neutrophilic Airway Inflammation in Asthmatic Mice. Antioxidants (Basel) 2024; 13:818. [PMID: 39061887 PMCID: PMC11274244 DOI: 10.3390/antiox13070818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Asthma is a heterogeneous disease that can be broadly classified into type 2, which is primarily steroid-sensitive and eosinophilic, and non-type 2, which is primarily steroid-resistant and neutrophilic. While the mechanisms leading to the development of molecular-targeted therapies for type 2 asthma are being elucidated, much remains to be learned about non-type 2 asthma. To investigate the role of oxidative stress in refractory allergic airway inflammation, we compared asthma models generated by immunizing wild-type and nuclear factor erythroid-2-related factor 2 (Nrf2)-deficient mice with the house dust mite antigen. Both asthma models had similar levels of airway inflammation and hyperresponsiveness, but the Nrf2-deficient mice had increased oxidative stress and exacerbated neutrophilic airway inflammation compared with the wild-type mice. Type 2 cytokines and the expression of GATA3, a transcription factor that is important for Th2 cell differentiation, had decreased in Nrf2-deficient mice compared with the wild-type mice, whereas helper T (Th) 17 cytokines and the expression of RORγt, which is important for Th17 cell differentiation, had increased. Furthermore, the neutrophilic airway inflammation caused by Nrf2 deficiency was ameliorated by interleukin (IL)-17 neutralization. We have concluded that the disruption of the Nrf2-mediated antioxidant defense system contributed to the induction of Th17 differentiation and exacerbated allergic neutrophilic airway inflammation.
Collapse
Affiliation(s)
| | - Yuko Morishima
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan (C.S.); (K.N.); (Y.I.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pilkington AW, Buragamadagu B, Johnston RA. Weighted Breaths: Exploring Biologic and Non-Biologic Therapies for Co-Existing Asthma and Obesity. Curr Allergy Asthma Rep 2024; 24:381-393. [PMID: 38878250 PMCID: PMC11233394 DOI: 10.1007/s11882-024-01153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE OF REVIEW To discuss the effectiveness of biologics, some of which comprise the newest class of asthma controller medications, and non-biologics in the treatment of asthma co-existing with obesity. RECENT FINDINGS Our review of recent preliminary and published data from clinical trials revealed that obese asthmatics respond favorably to dupilumab, mepolizumab, omalizumab, and tezepelumab, which are biologics currently indicated as add-on maintenance therapy for severe asthma. Furthermore, clinical trials are ongoing to assess the efficacy of non-biologics in the treatment of obese asthma, including a glucagon-like peptide-1 receptor agonist, a Janus kinase inhibitor, and probiotics. Although many biologics presently indicated as add-on maintenance therapy for severe asthma exhibit efficacy in obese asthmatics, other phenotypes of asthma co-existing with obesity may be refractory to these medications. Thus, to improve quality of life and asthma control, it is imperative to identify therapeutic options for all existing phenotypes of obese asthma.
Collapse
Affiliation(s)
- Albert W Pilkington
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States Department of Health and Human Services, 1000 Frederick Lane, Morgantown, WV, 26508-5402, USA
| | - Bhanusowmya Buragamadagu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Richard A Johnston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, United States Department of Health and Human Services, 1000 Frederick Lane, Morgantown, WV, 26508-5402, USA.
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA.
- Department of Physiology, Pharmacology, and Toxicology, School of Medicine, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
37
|
Olejnik AE, Kuźnar-Kamińska B. Association of Obesity and Severe Asthma in Adults. J Clin Med 2024; 13:3474. [PMID: 38930006 PMCID: PMC11204497 DOI: 10.3390/jcm13123474] [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: 04/25/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The incidence of obesity and asthma continues to enhance, significantly impacting global public health. Adipose tissue is an organ that secretes hormones and cytokines, causes meta-inflammation, and contributes to the intensification of bronchial hyperreactivity, oxidative stress, and consequently affects the different phenotypes of asthma in obese people. As body weight increases, the risk of severe asthma increases, as well as more frequent exacerbations requiring the use of glucocorticoids and hospitalization, which consequently leads to a deterioration of the quality of life. This review discusses the relationship between obesity and severe asthma, the underlying molecular mechanisms, changes in respiratory function tests in obese people, its impact on the occurrence of comorbidities, and consequently, a different response to conventional asthma treatment. The article also reviews research on possible future therapies for severe asthma. The manuscript is a narrative review of clinical trials in severe asthma and comorbid obesity. The articles were found in the PubMed database using the keywords asthma and obesity. Studies on severe asthma were then selected for inclusion in the article. The sections: 'The classification connected with asthma and obesity', 'Obesity-related changes in pulmonary functional tests', and 'Obesity and inflammation', include studies on subjects without asthma or non-severe asthma, which, according to the authors, familiarize the reader with the pathophysiology of obesity-related asthma.
Collapse
Affiliation(s)
- Aneta Elżbieta Olejnik
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Szamarzewskiego 84 Street, 60-569 Poznan, Poland;
| | | |
Collapse
|
38
|
Leduc L, Leclère M, Lavoie JP. Towards personalized medicine for the treatment of equine asthma. Vet J 2024; 305:106125. [PMID: 38704018 DOI: 10.1016/j.tvjl.2024.106125] [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: 02/20/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Although horses with asthma share similar clinical signs, the heterogeneity of the disease in terms of severity, triggering factors, inflammatory profile, and pathological features has hindered our ability to define biologically distinct subgroups. The recognition of phenotypes and endotypes could enable the development of precision medicine, including personalized, targeted therapy, to benefit affected horses. While in its infancy in horses, this review outlines the phenotypes of equine asthma and discusses how knowledge gained from targeted therapy in human medicine can be applied to evaluate the potential opportunities for personalized medicine in equine asthma and to suggest avenues for research to advance this emerging field.
Collapse
Affiliation(s)
- Laurence Leduc
- Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Mathilde Leclère
- Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada
| | - Jean-Pierre Lavoie
- Department of Clinical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
39
|
He BX, Fang SB, Xie YC, Lou DX, Wu ZC, Li CG, Liu XQ, Zhou ZR, Huang LX, Tian T, Chen DH, Fu QL. Small extracellular vesicles derived from human mesenchymal stem cells prevent Th17-dominant neutrophilic airway inflammation via immunoregulation on Th17 cells. Int Immunopharmacol 2024; 133:112126. [PMID: 38669946 DOI: 10.1016/j.intimp.2024.112126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Type 17 helper T cells (Th17)-dominant neutrophilic airway inflammation is critical in the pathogenesis of steroid-resistant airway inflammation such as severe asthma. Small extracellular vesicles (sEV) derived from human mesenchymal stem cells (MSCs) display extensive therapeutic effects and advantages in many diseases. However, the role of MSC-sEV in Th17-dominant neutrophilic airway inflammation and the related mechanisms are still poorly studied. Here we found that MSC-sEV significantly alleviated the infiltration of inflammatory cells in peribronchial interstitial tissues and reduced levels of inflammatory cells, especially neutrophils, in bronchoalveolar lavage fluids (BALF) of mice with neutrophilic airway inflammation. Consistently, MSC-sEV significantly decreased levels of IL-17A in BALF and Th17 in lung tissues. Furthermore, we found that labelled MSC-sEV were taken up by human CD4+ T cells most obviously at 12 h after incubation, and distributed mostly in mouse lungs. More importantly, potential signaling pathways involved in the MSC-sEV mediated inhibition of Th17 polarization were found using RNA sequencing. Using Western blot, JAK2-STAT3 pathway was identified as an important role in the inhibition of Th17 polarization by MSC-sEV. We found that proteins in MSC-sEV were mostly involved in the therapeutic effects of MSC-sEV. In total, our study suggested that MSC-sEV could be a potential therapeutic strategy for the treatment of neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Bi-Xin He
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shu-Bing Fang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying-Chun Xie
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong-Xiao Lou
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zi-Cong Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Chan-Gu Li
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Qing Liu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Rou Zhou
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Long-Xin Huang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tian Tian
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Hua Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Division of Allergy, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
40
|
Hosoki K, Govindhan A, Knight JM, Sur S. Allosteric inhibition of CXCR1 and CXCR2 abrogates Th2/Th17-associated Allergic Lung Inflammation in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593638. [PMID: 38798651 PMCID: PMC11118468 DOI: 10.1101/2024.05.13.593638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Background IL4, IL5, IL13, and IL17-producing CD4 T helper 2 (Th2)-cells and IL17-producing CD4 T helper 17 (Th17)-cells contribute to chronic eosinophilic and neutrophilic airway inflammation in asthma and allergic airway inflammation. Chemokines and their receptors are upregulated in Th2/Th17-mediated inflammation. However, the ability of CXCR1 and CXCR2 modulate Th2 and Th17-cell-mediated allergic lung inflammation has not been reported. Methods Mice sensitized and challenged with cat dander extract (CDE) mount a vigorous Th2-Th17-mediated allergic lung inflammation. Allosteric inhibitor of CXCR1 and CXCR2, ladarixin was orally administered in this model. The ability of ladarixin to modulate allergen-challenge induced recruitment of CXCR1 and CXCR2-expressing Th2 and Th17-cells and allergic lung inflammation were examined. Results Allergen challenge in sensitized mice increased mRNA expression levels of Il4, Il5, Il13, Il6, Il1β, Tgfβ1, Il17, Il23, Gata3, and Rorc , and induced allergic lung inflammation characterized by recruitment of CXCR1- and CXCR2-expressing Th2-cells, Th17-cells, neutrophils, and eosinophils. Allosteric inhibition of CXCR1 and CXCR2 vigorously blocked each of these pro-inflammatory effects of allergen challenge. CXCL chemokines induced a CXCR1 and CXCR2-dependent proliferation of IL4, IL5, IL13, and IL17 expressing T-cells. Conclusion Allosteric inhibition of CXCR1 and CXCR2 abrogates blocks recruitment of CXCR1- and CXCR2-expressing Th2-cells, Th17-cells, neutrophils, and eosinophils in this mouse model of allergic lung inflammation. We suggest that the ability of allosteric inhibition of CXCR1 and CXCR2 to abrogate Th2 and Th17-mediated allergic inflammation should be investigated in humans.
Collapse
|
41
|
Kovach MA, Käck U, Che KF, Brundin B, Konradsen JR, Lindén A. Systemic IL-26 correlates with improved asthma control in children sensitized to dog allergen. Respir Res 2024; 25:163. [PMID: 38622712 PMCID: PMC11020994 DOI: 10.1186/s12931-024-02773-7] [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: 09/29/2023] [Accepted: 03/13/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Interleukin (IL)-26 is produced by T helper type 17 (Type 17) cells and exerts immunomodulatory plus antimicrobial effects. Previous studies show that local IL-26 concentrations in the airways are higher in patients with uncontrolled than in those with controlled asthma, and that this intriguing cytokine bears biomarker potential. Here, we determined how systemic IL-26 relates to allergen sensitization, asthma severity, and to IL-17 A in children. METHODS Serum samples were obtained from children with (n = 60) and without (n = 17) sensitization to dog allergen, and IL-26 and IL-17 A protein concentrations were measured using ELISA. Self-reported history, including medication use and validated symptom-based questionnaire scores, was recorded. RESULTS The serum concentrations of IL-26 were enhanced in allergen-sensitized subjects and correlated with those of IL-17 A in a positive manner. However, the IL-26 concentrations did not markedly differ between allergen-sensitized subjects with and without asthma, eczema, allergic rhinitis, or a history of food allergy. Notably, IL-26 concentrations correlated with increasing Asthma Control Test (ACT) scores in a positive manner and with inhaled corticosteroid in a negative manner, amongst sensitized subjects with asthma. Moreover, subjects with asthma requiring ≥ 1 course of oral corticosteroids in the preceding 12 months had decreased IL-26 concentrations. CONCLUSION This study forwards evidence that systemic IL-26, just like IL-17 A, is involved in allergen sensitization among children. The association of systemic IL-26 with improved asthma control is compatible with the cellular sources being recruited into the airways in severe asthma, which supports that this kinocidin bears potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Melissa A Kovach
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - Ulrika Käck
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Sachs´ Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Karlhans F Che
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Bettina Brundin
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Jon R Konradsen
- Department of Medicine Solna Immunology and Allergy Unit, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 171 77, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Prueitt RL, Goodman JE. Evidence evaluated by European Food Safety Authority does not support lowering the temporary tolerable daily intake for bisphenol A. Toxicol Sci 2024; 198:185-190. [PMID: 38265237 DOI: 10.1093/toxsci/kfad136] [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] [Indexed: 01/25/2024] Open
Abstract
The European Food Safety Authority (EFSA) recently derived a tolerable daily intake (TDI) for bisphenol A (BPA) of 0.2 ng/kg bw/day. There are several issues with EFSA's hazard assessment review process, including that it was based on a limited subset of relevant studies. Multiple public commenters on EFSA's draft evaluation of BPA, including several European regulatory agencies, noted these issues, yet they were not adequately addressed by EFSA in the final evaluation. The TDI for BPA was based on an intermediate immunotoxicity endpoint in mice that has not been observed in other species; there is no evidence that it is a precursor event to any downstream pathological outcome. The TDI is several orders of magnitude lower than estimates of safe doses of BPA established by agencies worldwide, including EFSA's temporary TDI (t-TDI) for BPA established in 2015. Overall, the EFSA hazard assessment review process has led to a conclusion that there are low-dose effects of BPA based on very few, lower quality experimental animal studies. This conclusion is not supported by the totality of the available evidence, which includes multiple high-quality studies not considered by EFSA and indicates that the t-TDI established in 2015 is protective of human health.
Collapse
|
43
|
Quoc QL, Choi Y, Hur GY, Park HS. New targets for type 2-low asthma. Korean J Intern Med 2024; 39:215-227. [PMID: 38317271 PMCID: PMC10918384 DOI: 10.3904/kjim.2023.299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 02/07/2024] Open
Abstract
Asthma is characterized by airway obstruction and inflammation, and presents significant diagnostic and treatment challenges. The concept of endotypes has improved understanding of the mechanisms of asthma and has stimulated the development of effective treatment strategies. Sputum profiles may be used to classify asthma into two major inflammatory types: type 2-high (T2H) and type 2-low (T2L) asthma. T2H, characterized by elevated type 2 inflammation, has been extensively studied and several effective biologic treatments have been developed. However, managing T2L is more difficult due to the lack of reliable biomarkers for accurate diagnosis and classification. Additionally, conventional anti-inflammatory therapy does not completely control the symptoms of T2L; therefore, further research is needed to identify effective biologic treatments. This review provides new insights into the clinical characteristics and underlying mechanisms of severe T2L and investigates potential therapeutic approaches to control the disease.
Collapse
Affiliation(s)
- Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon,
Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon,
Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang,
Korea
| | - Gyu-Young Hur
- Department of Internal Medicine, Korea University College of Medicine, Seoul,
Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon,
Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon,
Korea
| |
Collapse
|
44
|
Wang H, Yip KH, Keam SP, Vlahos R, Nichol K, Wark P, Toubia J, Kral AC, Cildir G, Pant H, Hercus TR, Wilson N, Owczarek C, Lopez AF, Bozinovski S, Tumes DJ. Dual inhibition of airway inflammation and fibrosis by common β cytokine receptor blockade. J Allergy Clin Immunol 2024; 153:672-683.e6. [PMID: 37931708 DOI: 10.1016/j.jaci.2023.10.021] [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: 03/25/2023] [Revised: 09/11/2023] [Accepted: 10/12/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Patients with severe asthma can present with eosinophilic type 2 (T2), neutrophilic, or mixed inflammation that drives airway remodeling and exacerbations and represents a major treatment challenge. The common β (βc) receptor signals for 3 cytokines, GM-CSF, IL-5, and IL-3, which collectively mediate T2 and neutrophilic inflammation. OBJECTIVE To determine the pathogenesis of βc receptor-mediated inflammation and remodeling in severe asthma and to investigate βc antagonism as a therapeutic strategy for mixed granulocytic airway disease. METHODS βc gene expression was analyzed in bronchial biopsy specimens from patients with mild-to-moderate and severe asthma. House dust mite extract and Aspergillus fumigatus extract (ASP) models were used to establish asthma-like pathology and airway remodeling in human βc transgenic mice. Lung tissue gene expression was analyzed by RNA sequencing. The mAb CSL311 targeting the shared cytokine binding site of βc was used to block βc signaling. RESULTS βc gene expression was increased in patients with severe asthma. CSL311 potently reduced lung neutrophils, eosinophils, and interstitial macrophages and improved airway pathology and lung function in the acute steroid-resistant house dust mite extract model. Chronic intranasal ASP exposure induced airway inflammation and fibrosis and impaired lung function that was inhibited by CSL311. CSL311 normalized the ASP-induced fibrosis-associated extracellular matrix gene expression network and strongly reduced signatures of cellular inflammation in the lung. CONCLUSIONS βc cytokines drive steroid-resistant mixed myeloid cell airway inflammation and fibrosis. The anti-βc antibody CSL311 effectively inhibits mixed T2/neutrophilic inflammation and severe asthma-like pathology and reverses fibrosis gene signatures induced by exposure to commonly encountered environmental allergens.
Collapse
Affiliation(s)
- Hao Wang
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Kwok Ho Yip
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia
| | - Simon P Keam
- Research and Development, CSL Limited, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Ross Vlahos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Kristy Nichol
- Immune Health Research Program, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter Wark
- Immune Health Research Program, Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - John Toubia
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia
| | - Anita C Kral
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia
| | - Gökhan Cildir
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia
| | - Harshita Pant
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia; Faculty of Medicine, University of Adelaide, Adelaide, Australia
| | - Timothy R Hercus
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia
| | - Nick Wilson
- Research and Development, CSL Limited, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Catherine Owczarek
- Research and Development, CSL Limited, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Angel F Lopez
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia; Faculty of Medicine, University of Adelaide, Adelaide, Australia
| | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia.
| | - Damon J Tumes
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide, Australia.
| |
Collapse
|
45
|
Quoc QL, Cao TBT, Jang JH, Shin YS, Choi Y, Park HS. ST2-Mediated Neutrophilic Airway Inflammation: A Therapeutic Target for Patients With Uncontrolled Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:22-41. [PMID: 38262389 PMCID: PMC10823144 DOI: 10.4168/aair.2024.16.1.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/05/2023] [Accepted: 08/05/2023] [Indexed: 01/25/2024]
Abstract
PURPOSE Suppression of tumorigenicity 2 (ST2) has been proposed as the receptor contributing to neutrophilic inflammation in patients with type 2-low asthma. However, the exact role of ST2 in neutrophil activation remains poorly understood. METHODS A total of 105 asthmatic patients (classified into 3 groups according to control status: the controlled asthma [CA], partly-controlled asthma [PA], and uncontrolled asthma [UA] groups), and 104 healthy controls were enrolled to compare serum levels of soluble ST2 (sST2) and interleukin (IL)-33. Moreover, the functions of ST2 in neutrophils and macrophages (Mϕ) were evaluated ex vivo and in vivo. RESULTS Serum sST2 levels were significantly higher in the UA group than in the CA or PA groups (P < 0.05 for all) with a negative correlation between serum sST2 and forced expiratory volume in 1 second % (r = -0.203, P = 0.038). Significantly higher expression of ST2 receptors on peripheral neutrophils was noted in the UA group than in the PA or CA groups. IL-33 exerted its effects on the production of reactive oxygen species, the formation of extracellular traps from neutrophils, and Mϕ polarization/activation. In neutrophilic asthmatic mice, treatment with anti-ST2 antibody significantly suppressed proinflammatory cytokines (tumor necrosis factor-alpha and IL-17A) as well as the numbers of immune cells (neutrophils, Mϕ, and group 3 innate lymphoid cells) in the lungs. CONCLUSIONS These results suggest that IL-33 induces the activation of neutrophils and Mϕ via ST2 receptors, leading to neutrophilic airway inflammation and poor control status of asthma. ST2 could be a therapeutic target for neutrophilic airway inflammation in patients with UA.
Collapse
Affiliation(s)
- Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Jae-Hyuk Jang
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomaterials Science, College of Natural Resources and Life Science, Pusan National University, Miryang, Korea.
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
46
|
Quan J, Wen X, Su G, Zhong Y, Huang T, Xiong Z, Huang J, Lv Y, Li S, Luo S, Luo C, Cai X, Lai X, Xiang Y, Zheng SG, Shao Y, Lin H, Gao X, Tang J, Lai T. Epithelial SIRT6 governs IL-17A pathogenicity and drives allergic airway inflammation and remodeling. Nat Commun 2023; 14:8525. [PMID: 38135684 PMCID: PMC10746710 DOI: 10.1038/s41467-023-44179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Dysregulation of IL-17A is closely associated with airway inflammation and remodeling in severe asthma. However, the molecular mechanisms by which IL-17A is regulated remain unclear. Here we identify epithelial sirtuin 6 (SIRT6) as an epigenetic regulator that governs IL-17A pathogenicity in severe asthma. Mice with airway epithelial cell-specific deletion of Sirt6 are protected against allergen-induced airway inflammation and remodeling via inhibiting IL-17A-mediated inflammatory chemokines and mesenchymal reprogramming. Mechanistically, SIRT6 directly interacts with RORγt and mediates RORγt deacetylation at lysine 192 via its PPXY motifs. SIRT6 promotes RORγt recruitment to the IL-17A gene promoter and enhances its transcription. In severe asthma patients, high expression of SIRT6 positively correlates with airway remodeling and disease severity. SIRT6 inhibitor (OSS_128167) treatment significantly attenuates airway inflammation and remodeling in mice. Collectively, these results uncover a function for SIRT6 in regulating IL-17A pathogenicity in severe asthma, implicating SIRT6 as a potential therapeutic target for severe asthma.
Collapse
Affiliation(s)
- Jingyun Quan
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiaoxia Wen
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Guomei Su
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yu Zhong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Tong Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhilin Xiong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jiewen Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yingying Lv
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Shihai Li
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Shuhua Luo
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chaole Luo
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Xin Cai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xianwen Lai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuanyuan Xiang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Song Guo Zheng
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Yiming Shao
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Haitao Lin
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiao Gao
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China.
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
47
|
Mao R, Jiang Z, Min Z, Wang G, Xie M, Gao P, Zhu L, Li H, Chen Z. Peripheral neutrophils and oxidative stress-associated molecules for predicting the severity of asthma: a cross-sectional study based on multidimensional assessment. Front Med (Lausanne) 2023; 10:1240253. [PMID: 38131042 PMCID: PMC10733438 DOI: 10.3389/fmed.2023.1240253] [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: 06/14/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Objectives This study aims to explore the relationship between the severity of asthma and neutrophils and related oxidative stress-associated molecules in peripheral blood and induced sputum. Methods A total of 67 subjects were included in this study, namely, 25 patients with severe asthma and 42 patients with non-severe asthma. Clinical data, induced sputum and peripheral blood were collected. Lung function and molecules related to oxidative stress in induced sputum and peripheral blood of asthma patients were detected. The relationship between neutrophils and asthma severity was analyzed. HDAC2 mRNA and protein expression levels and HDAC2 activity were also analyzed. Multivariate logistic regression was performed to select statistically significant variables. Results The absolute value of neutrophils and percentage of neutrophils were higher in the severe asthma patients. These two values were used to predict the severity of asthma by ROC analysis, with the best cutoff values being 4.55 × 109/L (sensitivity 83.3%, specificity 64.0%) and 55.15% (sensitivity 54.8%, specificity 88.0%). The ROS concentration of neutrophils in the induced sputum samples and the 8-iso-PGF2α concentration in the peripheral blood samples were higher in the severe asthma group (P = 0.012; P = 0.044), whereas there was reduced HDAC2 protein activity in PBMCs (P < 0.001). A logistic equation and a nomogram were created to give a precise prediction of disease severity. Conclusion Oxidative stress is increased in severe asthma patients. Peripheral blood neutrophils and 8-iso-PGF2α can be used as biomarkers to predict the severity of asthma. A prediction model was created for evaluating asthma severity.
Collapse
Affiliation(s)
- Ruolin Mao
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihui Min
- Research Center of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gang Wang
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Min Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Lei Zhu
- Department of Respiratory and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Huayin Li
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Institute of Respiratory Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
48
|
Bilal H, Khan MN, Khan S, Fang W, Chang W, Yin B, Song NJ, Liu Z, Zhang D, Yao F, Wang X, Wang Q, Cai L, Hou B, Wang J, Mao C, Liu L, Zeng Y. Risk of candidiasis associated with interleukin-17 inhibitors: Implications and management. Mycology 2023; 15:30-44. [PMID: 38558839 PMCID: PMC10977001 DOI: 10.1080/21501203.2023.2265664] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/27/2023] [Indexed: 04/04/2024] Open
Abstract
The application of interleukin-17 (IL-17) inhibitors, including secukinumab, ixekizumab, brodalumab, and bimekizumab, are associated with elevated risk of candidiasis. These medications interfere with the IL-17 pathway, which is essential for maintaining mucosal barriers and coordinating the immune response against Candida species. The observational data and clinical trials demonstrate the increased incidence of candidiasis in individuals treated with IL-17 inhibitors. Brodalumab and bimekizumab pose a greater risk than secukinumab in eliciting candidiasis, whereas the data regarding ixekizumab are equivocal. Higher doses and prolonged treatment duration of IL-17 inhibitors increase the risk of candidiasis by compromising the immune response against Candida species. Prior to prescribing IL-17 inhibitors, healthcare professionals should comprehensively evaluate patients' medical histories and assess their risk factors. Patients should be educated on the signs and symptoms of candidiasis to facilitate early detection and intervention. Future research should focus on identifying the risk factors associated with candidiasis in patients receiving IL-17 inhibitors. Prospective studies and long-term surveillance are required to explore the impact of specific inhibitors on the incidence and severity of candidiasis and to evaluate the effectiveness of combination therapies, such as concurrent use of IL-17 inhibitors and prophylactic antifungal agents.
Collapse
Affiliation(s)
- Hazrat Bilal
- Department of Dermatology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Muhammad Nadeem Khan
- Faculty of Biological Sciences, Department of Microbiology, Quaid-I-Azam University, Islamabad, Pakistan
| | - Sabir Khan
- Department of Dermatology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wenjie Fang
- Department of Dermatology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Wenqiang Chang
- School of Pharmacy, Shandong University, Qingdao, Shandong, China
| | - Bin Yin
- Department of Dermatovenereology, Chengdu Second People's Hospital, Chengdu, China
| | - Ning-Jing Song
- Department of Dermatology, Tongren Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhongrong Liu
- Department of Dermatology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dongxing Zhang
- Department of Dermatology, Meizhou Dongshan Hospital, Meizhou, Guangdong, China
- Department of Dermatology, Meizhou People's Hospital, Meizhou, Guangdong, China
| | - Fen Yao
- Department of Pharmacy, Shantou University School Medical College, Shantou, China
| | - Xun Wang
- Department of Dermatology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Qian Wang
- Department of Dermatology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Lin Cai
- Department of Dermatology, Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Bing Hou
- Department of Clinical Laboratory, Skin and Venereal Diseases Prevention and Control Hospital of Shantou City, Shantou, Guangdong, China
| | - Jiayue Wang
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyan Mao
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingxi Liu
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuebin Zeng
- Department of Dermatology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
49
|
Zeng X, Qing J, Li CM, Lu J, Yamawaki T, Hsu YH, Vander Lugt B, Hsu H, Busby J, McDowell PJ, Jackson DJ, Djukanovic R, Matthews JG, Arron JR, Bradding P, Brightling CE, Chaudhuri R, Choy DF, Cowan D, Fowler SJ, Hardman TC, Harrison T, Howarth P, Lordan J, Mansur AH, Menzies-Gow A, Pavord ID, Walker S, Woodcock A, Heaney LG. Blood transcriptomic signature in type-2 biomarker-low severe asthma and asthma control. J Allergy Clin Immunol 2023; 152:876-886. [PMID: 37315813 DOI: 10.1016/j.jaci.2023.05.023] [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: 10/31/2022] [Revised: 05/02/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Patients with type-2 (T2) cytokine-low severe asthma often have persistent symptoms despite suppression of T2 inflammation with corticosteroids. OBJECTIVES We sought to analyze whole blood transcriptome from 738 samples in T2-biomarker-high/-low patients with severe asthma to relate transcriptomic signatures to T2 biomarkers and asthma symptom scores. METHODS Bulk RNA-seq data were generated for blood samples (baseline, week 24, week 48) from 301 participants recruited to a randomized clinical trial of corticosteroid optimization in severe asthma. Unsupervised clustering, differential gene expression analysis, and pathway analysis were performed. Patients were grouped by T2-biomarker status and symptoms. Associations between clinical characteristics and differentially expressed genes (DEGs) associated with biomarker and symptom levels were investigated. RESULTS Unsupervised clustering identified 2 clusters; cluster 2 patients were blood eosinophil-low/symptom-high and more likely to be receiving oral corticosteroids (OCSs). Differential gene expression analysis of these clusters, with and without stratification for OCSs, identified 2960 and 4162 DEGs, respectively. Six hundred twenty-seven of 2960 genes remained after adjusting for OCSs by subtracting OCS signature genes. Pathway analysis identified dolichyl-diphosphooligosaccharide biosynthesis and assembly of RNA polymerase I complex as significantly enriched pathways. No stable DEGs were associated with high symptoms in T2-biomarker-low patients, but numerous associated with elevated T2 biomarkers, including 15 that were upregulated at all time points irrespective of symptom level. CONCLUSIONS OCSs have a considerable effect on whole blood transcriptome. Differential gene expression analysis demonstrates a clear T2-biomarker transcriptomic signature, but no signature was found in association with T2-biomarker-low patients, including those with a high symptom burden.
Collapse
Affiliation(s)
- Xue Zeng
- Amgen Research, Amgen, Inc, South San Francisco, Calif
| | - Jing Qing
- Amgen Research, Amgen, Inc, South San Francisco, Calif
| | - Chi-Ming Li
- Amgen Research, Amgen, Inc, South San Francisco, Calif
| | - Jiamiao Lu
- Amgen Research, Amgen, Inc, South San Francisco, Calif
| | | | | | | | - Hailing Hsu
- Amgen Research, Amgen, Inc, Thousand Oaks, Calif
| | - John Busby
- Wellcome-Wolfson Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, United Kingdom
| | - P J McDowell
- Wellcome-Wolfson Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, United Kingdom
| | - David J Jackson
- Guy's & St Thomas' NHS Trust and Department of Asthma, Allergy & Lung Biology, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Ratko Djukanovic
- School of Clinical and Experimental Sciences, University of Southampton, NIHR Southampton Biomedical Research Centre, Southampton, United Kingdom
| | | | | | - Peter Bradding
- Department of Respiratory Sciences, Institute for Lung Health and Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Christopher E Brightling
- Department of Respiratory Sciences, Institute for Lung Health and Leicester NIHR Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Rekha Chaudhuri
- Gartnavel General Hospital, Glasgow, and University of Glasgow, Glasgow, United Kingdom
| | | | - D Cowan
- NHS Greater Glasgow and Clyde, Stobhill Hospital, Glasgow, United Kingdom
| | - S J Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom; Manchester Academic Health Science Centre and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | | | - Tim Harrison
- Nottingham Respiratory NIHR Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Peter Howarth
- School of Clinical and Experimental Sciences, University of Southampton, NIHR Southampton Biomedical Research Centre, Southampton, United Kingdom
| | - James Lordan
- The Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - A H Mansur
- University of Birmingham and Heartlands Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | | | - Ian D Pavord
- Oxford Respiratory NIHR BRC, Nuffield Department of Medicine, The University of Oxford, Oxford, United Kingdom
| | - Samantha Walker
- Asthma UK & British Lung Foundation Partnership, London, United Kingdom
| | - Ashley Woodcock
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom; Manchester Academic Health Science Centre and NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Liam G Heaney
- Wellcome-Wolfson Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University, Belfast, United Kingdom.
| |
Collapse
|
50
|
Cai J, Tao H, Liu H, Hu Y, Han S, Pu W, Li L, Li G, Li C, Zhang J. Intrinsically bioactive and biomimetic nanoparticle-derived therapies alleviate asthma by regulating multiple pathological cells. Bioact Mater 2023; 28:12-26. [PMID: 37214258 PMCID: PMC10193170 DOI: 10.1016/j.bioactmat.2023.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Asthma is a serious global public health concern. Airway neutrophilic inflammation is closely related to severe asthma, for which effective and safe therapies remain to be developed. Here we report nanotherapies capable of simultaneously regulating multiple target cells relevant to the pathogenesis of neutrophilic asthma. A nanotherapy LaCD NP based on a cyclic oligosaccharide-derived bioactive material was engineered. LaCD NP effectively accumulated in the injured lungs of asthmatic mice and mainly distributed in neutrophils, macrophages, and airway epithelial cells after intravenous or inhalation delivery, thereby ameliorating asthmatic symptoms and attenuating pulmonary neutrophilic inflammation as well as reducing airway hyperresponsiveness, remodeling, and mucus production. Surface engineering via neutrophil cell membrane further enhanced targeting and therapeutic effects of LaCD NP. Mechanistically, LaCD NP can inhibit the recruitment and activation of neutrophils, especially reducing the neutrophil extracellular traps formation and NLRP3 inflammasome activation in neutrophils. Also, LaCD NP can suppress macrophage-mediated pro-inflammatory responses and prevent airway epithelial cell death and smooth muscle cell proliferation, by mitigating neutrophilic inflammation and its direct effects on relevant cells. Importantly, LaCD NP showed good safety performance. Consequently, LaCD-derived multi-bioactive nanotherapies are promising for effective treatment of neutrophilic asthma and other neutrophil-associated diseases.
Collapse
Affiliation(s)
- Jiajun Cai
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Hui Tao
- Department of Pharmacology, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Huan Liu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Yi Hu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Songling Han
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Wendan Pu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Lanlan Li
- Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Gang Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
- State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| |
Collapse
|