1
|
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
|
2
|
Ramonell RP, Oriss TB, McCreary-Partyka JC, Kale SL, Brandon NR, Ross MA, Gauthier MC, Yue M, Nee TJ, Das S, Chen W, Joglekar AV, Ray P, St Croix CM, Rajasundaram D, Wenzel SE, Ray A. CD8+ TEMRAs in severe asthma associate with asthma symptom duration and escape proliferation arrest. JCI Insight 2025; 10:e185061. [PMID: 40048261 PMCID: PMC12016929 DOI: 10.1172/jci.insight.185061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 03/04/2025] [Indexed: 04/23/2025] Open
Abstract
Aberrant immune response is a hallmark of asthma, with 5%-10% of patients suffering from severe disease exhibiting poor response to standard treatment. A better understanding of the immune responses contributing to disease heterogeneity is critical for improving asthma management. T cells are major players in the orchestration of asthma, in both mild and severe disease, but it is unclear whether specific T cell subsets influence asthma symptom duration. Here we show a significant association of airway CD8+ effector memory T cells re-expressing CD45RA (TEMRAs), but not CD8+CD45RO+ or tissue-resident memory T cells, with asthma duration in patients with severe asthma (SA) but not mild to moderate asthma (MMA). Higher frequencies of IFN-γ+CD8+ TEMRAs compared with IFN-γ+CD45RO+ T cells were detected in SA airways, and the TEMRAs from patients with SA but not MMA proliferated ex vivo, although both expressed cellular senescence-associated biomarkers. Prompted by the transcriptomic profile of SA CD8+ TEMRAs and proliferative response to IL-15, airway IL15 expression was higher in patients with SA compared with MMA. Additionally, IL15 expression in asthmatic airways negatively correlated with lung function. Our findings add what we believe is a new dimension to understanding asthma heterogeneity, identifying IL-15 as a potential target for treatment.
Collapse
Affiliation(s)
- Richard P. Ramonell
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | - Timothy B. Oriss
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | - Sagar L. Kale
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | - Mark A. Ross
- Department of Cell Biology
- Center for Biological Imaging
| | - Marc C. Gauthier
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | | | - Taylor J. Nee
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
| | - Sudipta Das
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
| | | | | | - Prabir Ray
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
| | | | | | - Sally E. Wenzel
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anuradha Ray
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine
- Asthma and Environmental Lung Health Institute at UPMC
- Department of Immunology
| |
Collapse
|
3
|
Margelidon-Cozzolino V, Balsamelli J, Carrard J, Ait Yahia S, Gevaert MH, Demoulin-Alexikova S, Pichavant M, Tsicopoulos A, Chenivesse C, Lejeune S, de Nadai P. Dog allergen-induced asthma in mice: a relevant model of T2 low severe asthma with airway remodelling. Inflamm Res 2025; 74:52. [PMID: 40082266 PMCID: PMC11906515 DOI: 10.1007/s00011-025-02004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 03/16/2025] Open
Abstract
OBJECTIVE AND DESIGN Airway remodelling (AR) is a disabling phenomenon in patients with severe asthma, yet suitable models are lacking. We previously developed a dog allergen-induced murine asthma model characterized by T2low Th17-driven neutrophilic airway inflammation and AR. To assess its relevance to human AR associated with T2low severe asthma, a condition characterised by poor response to inhaled steroids, we tested the steroid sensitivity of the key features of this model. MATERIAL Asthma was induced in C57BL/6 J mice by intranasal sensitization, followed by a three-week challenge with dog allergen. TREATMENT Daily intraperitoneal 1 mg kg-1 dexamethasone was administrated during the last week of challenge. METHODS We measured airway resistances in response to methacholine, cellular inflammation in bronchoalveolar lavage, lung cytokines, and quantified AR features, in response to dexamethasone. RESULTS Dexamethasone-treated mice showed persistent airway hyperresponsiveness, neutrophilic inflammation, and Il17a overexpression, whereas Il22 expression was abrogated. Pathological AR features, including mucus hyperproduction, subepithelial fibrosis and smooth muscle hypertrophy were not eliminated by dexamethasone. CONCLUSIONS Our dog allergen-induced murine model of asthma mirrors the steroid-insensitive traits of human severe T2low asthma with AR, making it a relevant tool for identifying novel therapeutic targets in this orphan asthma subset.
Collapse
Affiliation(s)
- Victor Margelidon-Cozzolino
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France.
- Groupement Des Hôpitaux de L'Institut Catholique de Lille (GHICL), Lille, France.
- Service de Pneumologie, Hôpital Saint-Philibert, Rue du Grand But, 59160, Lomme, France.
| | - Joanne Balsamelli
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Julie Carrard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Saliha Ait Yahia
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Marie-Hélène Gevaert
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014-PLBS, Lille, France
| | - Silvia Demoulin-Alexikova
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Muriel Pichavant
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Anne Tsicopoulos
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| | - Cécile Chenivesse
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
- CRISALIS (Clinical Research Initiative In Severe Asthma: a Lever for Innovation & Science), F-CRIN Network, INSERM US015, Toulouse, France
| | - Stéphanie Lejeune
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
- Univ. Lille, Department of Pediatric Pulmonology and Allergy, Hôpital Jeanne de Flandre, CHU Lille, 59000, Lille, France
| | - Patricia de Nadai
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR9017 - CIIL-Center for Infection and Immunity of Lille, 59000, Lille, France
| |
Collapse
|
4
|
Ford ML, Reza MI, Ruwanpathirana A, Sathish V, Britt RD. Integrative Roles of Pro-Inflammatory Cytokines on Airway Smooth Muscle Structure and Function in Asthma. Immunol Rev 2025; 330:e70007. [PMID: 39991781 PMCID: PMC11848829 DOI: 10.1111/imr.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Asthma has become more appreciated for its heterogeneity with studies identifying type 2 and non-type 2 phenotypes/endotypes that ultimately lead to airflow obstruction, airway hyperresponsiveness, and remodeling. The pro-inflammatory environment in asthma influences airway smooth muscle (ASM) structure and function. ASM has a vast repertoire of inflammatory receptors that, upon activation, contribute to prominent features in asthma, notably immune cell recruitment and activation, hypercontractility, proliferation, migration, and extracellular matrix protein deposition. These pro-inflammatory responses in ASM can be mediated by both type 2 (e.g., IL-4, IL-13, and TSLP) and non-type 2 (e.g., TNFα, IFNγ, IL-17A, and TGFβ) cytokines, highlighting roles for ASM in type 2 and non-type 2 asthma phenotypes/endotypes. In recent years, there has been considerable advances in understanding how pro-inflammatory cytokines promote ASM dysfunction and impair responsiveness to asthma therapy, corticosteroids and long-acting β2-adrenergic receptor agonists (LABAs). Transcriptomic analyses on human ASM cells and tissues have expanded our knowledge in this area but have also raised new questions regarding ASM and its role in asthma. In this review, we discuss how pro-inflammatory cytokines, corticosteroids, and LABAs affect ASM structure and function, with particular focus on changes in gene expression and transcriptional programs in type 2 and non-type 2 asthma.
Collapse
Affiliation(s)
- Maria L. Ford
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Biomedical Sciences Graduate Program, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Mohammad Irshad Reza
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth DakotaUSA
| | - Anushka Ruwanpathirana
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Biomedical Sciences Graduate Program, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Venkatachalem Sathish
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth DakotaUSA
| | - Rodney D. Britt
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
5
|
Latayan J, Akkenapally SV, Madala SK. Emerging Concepts in Cytokine Regulation of Airway Remodeling in Asthma. Immunol Rev 2025; 330:e70020. [PMID: 40116139 PMCID: PMC11926778 DOI: 10.1111/imr.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/18/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Asthma, a chronic respiratory condition that has seen a dramatic rise in prevalence over the past few decades, now affects more than 300 million people globally and imposes a significant burden on healthcare systems. The key pathological features of asthma include inflammation, airway hyperresponsiveness, mucus cell metaplasia, smooth muscle hypertrophy, and subepithelial fibrosis. Cytokines released by lung epithelial cells, stromal cells, and immune cells during asthma are critical to pathological tissue remodeling in asthma. Over the past few decades, researchers have made great strides in understanding key cells involved in asthma and the cytokines that they produce. Epithelial cells as well as many adaptive and innate immune cells are activated by environmental signals to produce cytokines, namely, type 2 cytokines (IL-4, IL-5, IL-13), IFN-γ, IL-17, TGF-β, and multiple IL-6 family members. However, the precise mechanisms through which these cytokines contribute to airway remodeling remain elusive. Additionally, multiple cell types can produce the same cytokines, making it challenging to decipher how specific cell types and cytokines uniquely contribute to asthma pathogenesis. This review highlights recent advances and provides a comprehensive overview of the key cells involved in the production of cytokines and how these cytokines modulate airway remodeling in asthma.
Collapse
Affiliation(s)
- Jana Latayan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
- Immunology Graduate ProgramUniversity of CincinnatiCincinnatiOhioUSA
| | - Santhoshi V. Akkenapally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| | - Satish K. Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal MedicineThe University of CincinnatiCincinnatiOhioUSA
| |
Collapse
|
6
|
Gauthier M, Kale SL, Ray A. T1-T2 Interplay in the Complex Immune Landscape of Severe Asthma. Immunol Rev 2025; 330:e70011. [PMID: 39991821 PMCID: PMC11849004 DOI: 10.1111/imr.70011] [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: 11/21/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Asthma is orchestrated by an aberrant immune response involving a complex interplay between multiple inflammatory cell types. An increase in Th2 cells in the asthmatic airway is a hallmark of asthma, and biologics blocking their effector functions have been life-changing for many severe asthma patients who poorly respond to immunosuppression by corticosteroids. However, studies in the past decade have highlighted not only other cell types that also produce Th2 cytokines boosting the Type 2/T2 phenotype but also a heightened IFN-γ response, primarily from T cells, referred to as a Type 1/T1 immune response. Data derived from studies of immune cells in the airways and mouse models of severe asthma suggest a role of IFN-γ in corticosteroid resistance, airway hyperreactivity, and also airway remodeling via effects on other cell types including mast cells, eosinophils, airway epithelial cells, and airway smooth muscle cells. The simultaneous presence of T1 and T2 immune responses is detectable in the sickest of asthma patients in whom corticosteroids suppress the T2 but not the T1 response. This article has reviewed our current understanding of the complex T1-T2 interplay in severe asthma highlighting mediators that impact both arms which may be targeted alone or in combination for disease alleviation.
Collapse
Affiliation(s)
- Marc Gauthier
- Pulmonary Allergy Critical Care and Sleep Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Sagar L. Kale
- Pulmonary Allergy Critical Care and Sleep Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Anuradha Ray
- Pulmonary Allergy Critical Care and Sleep Medicine, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
7
|
Ramirez GA, Cardamone C, Lettieri S, Fredi M, Mormile I. Clinical and Pathophysiological Tangles Between Allergy and Autoimmunity: Deconstructing an Old Dichotomic Paradigm. Clin Rev Allergy Immunol 2025; 68:13. [PMID: 39932658 PMCID: PMC11814061 DOI: 10.1007/s12016-024-09020-3] [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] [Accepted: 12/26/2024] [Indexed: 02/14/2025]
Abstract
Allergic and autoimmune disorders are characterised by dysregulation of the immune responses to otherwise inert environmental substances and autoantigens, leading to inflammation and tissue damage. Their incidence has constantly increased in the last decades, and their co-occurrence defies current standards in patient care. For years, allergy and autoimmunity have been considered opposite conditions, with IgE and Th2 lymphocytes cascade driving canonical allergic manifestations and Th1/Th17-related pathways accounting for autoimmunity. Conversely, growing evidence suggests that these conditions not only share some common inciting triggers but also are subtended by overlapping pathogenic pathways. Permissive genetic backgrounds, along with epithelial barrier damage and changes in the microbiome, are now appreciated as common risk factors for both allergy and autoimmunity. Eosinophils and mast cells, along with autoreactive IgE, are emerging players in triggering and sustaining autoimmunity, while pharmacological modulation of B cells and Th17 responses has provided novel clues to the pathophysiology of allergy. By combining clinical and therapeutic evidence with data from mechanistic studies, this review provides a state-of-the-art update on the complex interplay between allergy and autoimmunity, deconstructing old dichotomic paradigms and offering potential clues for future research.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Cardamone
- Immunorheumatology Unit, University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Largo Città d'Ippocrate, Via San Leonardo 1, 84131, Salerno, Italy.
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Sara Lettieri
- Pulmonology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Mormile
- Division of Internal Medicine and Clinical Immunology, Department of Internal Medicine and Clinical Complexity, AOU Federico II, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
8
|
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
|
9
|
Fahy JV, Jackson ND, Sajuthi SP, Pruesse E, Moore CM, Everman JL, Rios C, Tang M, Gauthier M, Wenzel SE, Bleecker ER, Castro M, Comhair SA, Erzurum SC, Hastie AT, Moore W, Israel E, Levy BD, Denlinger L, Jarjour NN, Johansson MW, Mauger DT, Phillips BR, Sumino K, Woodruff PG, Peters MC, Seibold MA. Type 1 Immune Responses Related to Viral Infection Influence Corticosteroid Response in Asthma. Am J Respir Crit Care Med 2025; 211:194-204. [PMID: 39601762 PMCID: PMC11812531 DOI: 10.1164/rccm.202402-0403oc] [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: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
Rationale: Corticosteroid-responsive type 2 (T2) inflammation underlies the T2-high asthma endotype. However, we hypothesized that type 1 (T1) inflammation, possibly related to viral infection, may also influence corticosteroid response. Objectives: To determine the frequency and within-patient variability of T1-high, T2-high, and T1/T2-high asthma endotypes and whether virally influenced T1-high disease influences corticosteroid response in asthma. Methods: Patients in SARP-3 (Severe Asthma Research Program-3) had sputum collected at baseline, after intramuscular (triamcinolone acetonide) corticosteroid treatment, and at 1- and 3-year follow-ups. Sputum cell RNA was used for whole-transcriptome gene network and viral metagenomic analyses. We then profiled patients as highly expressing T1 and/or T2 gene networks and established the influence of these endotypes on corticosteroid responsiveness and the likelihood of viral transcript detection in the airways. Measurements and Main Results: We found that 22% and 35% of patients with asthma highly expressed T1 and T2 network genes, respectively, and that 8.5% highly expressed both networks. Asthma severity outcomes were worse in T2-high compared with T1-high asthma and most severe in the T1-high/T2-high subgroup. Corticosteroid treatment strongly suppressed T2 but poorly suppressed T1 gene expression, and corticosteroid-associated improvements in FEV1 occurred only in patients with T1-low/T2-high disease and not in patients with T1-high/T2-high disease. Viral metagenomic analyses uncovered that 24% of asthma sputum samples tested positive for a respiratory virus, and high viral carriage was associated with 14-fold increased risk of T1-high disease. Conclusions: Airway T1 immune responses are relatively common in asthma, are largely corticosteroid resistant, and are associated with subclinical viral infection.
Collapse
Affiliation(s)
- John V. Fahy
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California
| | | | | | | | | | | | - Cydney Rios
- Center for Genes, Environment, and Health and
| | - Monica Tang
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Marc Gauthier
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sally E. Wenzel
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eugene R. Bleecker
- Division of Genetics, Genomics, and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Arizona
| | - Mario Castro
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kansas School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas
| | - Suzy A. Comhair
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | | | - Annette T. Hastie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Wendy Moore
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Bruce D. Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Loren Denlinger
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nizar N. Jarjour
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Mats W. Johansson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - David T. Mauger
- Division of Statistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, Hershey, Pennsylvania
| | - Brenda R. Phillips
- Division of Statistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, Hershey, Pennsylvania
| | - Kaharu Sumino
- Division of Pulmonary Critical Care Medicine, Washington University, St. Louis, Missouri; and
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Michael C. Peters
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Max A. Seibold
- Center for Genes, Environment, and Health and
- Department of Pediatrics, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
10
|
Mdkhana B, Saheb Sharif-Askari N, Cagliani R, Al-Sheakly BKS, Ramakrishnan RK, Saheb Sharif-Askari F, Hachim IY, Hamid Q, Rawas-Qalaji M, Halwani R. Inhibiting DNA Sensing Pathway Controls Steroid Hyporesponsive Lung Inflammation. Adv Biol (Weinh) 2025; 9:e2400230. [PMID: 39601482 DOI: 10.1002/adbi.202400230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/24/2024] [Indexed: 11/29/2024]
Abstract
DNA damage underlies the progression of asthma toward a severe, steroid hyporesponsive phenotype. The accumulation of double-stranded DNA within the cytosol triggers the activation of cytosolic DNA-sensing pathways, notably the Stimulator of Interferon Genes (STING) pathway. However, the precise role of STING in driving steroid hyporesponsiveness remains elusive and warrants further investigation. This study evaluates STING levels in human bronchial fibroblasts from severe asthmatic patients and in lung homogenates from a steroid hyporesponsive lung inflammation mouse model. STING level is assessed at baseline, post house dust mites (HDM) stimulation, and following treatment with dexamethasone and STING inhibitor. The effect of STING inhibitors on regulating steroid hyporesponsiveness particularly glucocorticoid receptor (GR)-α/GR-β ratio is also examined. Severe asthmatic fibroblasts exhibit elevated STING/IFN-I pathway activation, further heightened by HDM and a similar pattern is seen in lung homogenates from steroid hyporesponsive mice. Dexamethasone combined with an STING inhibitor reduces STING activity, while dexamethasone alone is ineffective. Interestingly, the STING inhibitor restores steroid sensitivity by increasing the GRα/GRβ ratio. Furthermore, nanoparticle-encapsulated STING inhibitor more effectively reduces airway hyperresponsiveness and restores steroid sensitivity than the free inhibitor. These findings emphasize STING's role in severe asthma pathogenesis, proposing nanoparticle delivery of STING inhibitors as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Bushra Mdkhana
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Roberta Cagliani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Biological Sciences, Khalifa University of Science & Technology, Abu Dhabi, UAE
| | | | - Rakhee K Ramakrishnan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ibrahim Yaseen Hachim
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Qutayba Hamid
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mutasem Rawas-Qalaji
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Prince Abdullah Ben Khaled Celiac Disease Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Saudi Arabia
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Saheb Sharif-Askari N, Mdkhana B, Hafezi S, Khalil BA, Al-Sheakly BK, Halwani H, Saheb Sharif-Askari F, Halwani R. Calprotectin is regulated by IL-17A and induces steroid hyporesponsiveness in asthma. Inflamm Res 2024; 73:1875-1888. [PMID: 39212675 DOI: 10.1007/s00011-024-01937-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/25/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Calprotectin, a calcium-binding protein, plays a crucial role in inflammation and has been associated with various inflammatory diseases, including asthma. However, its regulation and impact on steroid hyporesponsiveness, especially in severe asthma, remain poorly understood. METHODS This study investigated the regulation of calprotectin proteins (S100A8 and S100A9) by IL-17 and its role in steroid hyporesponsiveness using in vitro and in vivo models. Calprotectin expression was assessed in primary bronchial fibroblasts from healthy controls and severe asthmatic patients, as well as in mouse models of steroid hyporesponsive lung inflammation induced by house dust mite (HDM) allergen and cyclic-di-GMP (cdiGMP) adjuvant. The effects of IL-17A stimulation on calprotectin expression and steroid response markers in bronchial epithelial and fibroblast cells were examined. Additionally, the therapeutic potential of paquinimod, a calprotectin inhibitor, in mitigating airway inflammation and restoring steroid response signatures in the mouse model was evaluated. RESULTS The results demonstrated upregulation of calprotectin expression in asthmatic bronchial fibroblasts compared to healthy controls, as well as in refractory asthma samples compared to non-refractory asthma. IL-17 stimulation induced calprotectin expression and dysregulated glucocorticoid response signatures in lung epithelial and fibroblast cells. Treatment with paquinimod reversed IL-17-induced dysregulation of steroid signatures, indicating the involvement of calprotectin in this process. In the HDM/cdiGMP mouse model, paquinimod significantly attenuated airway inflammation and hyperresponsiveness, and restored steroid response signatures, whereas dexamethasone showed limited efficacy. Mechanistically, paquinimod inhibited MAPK/ERK and NF-κB pathways downstream of calprotectin, leading to reduced lung inflammation. CONCLUSION These findings highlight calprotectin as a potential therapeutic target regulated by IL-17 in steroid hyporesponsive asthma. Targeting calprotectin may offer a promising approach to alleviate airway inflammation and restore steroid responsiveness in severe asthma. Further investigations are warranted to explore its therapeutic potential in clinical settings and elucidate its broader implications in steroid mechanisms of action.
Collapse
Affiliation(s)
- Narjes Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bushra Mdkhana
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Shirin Hafezi
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Bariaa A Khalil
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Baraa Khalid Al-Sheakly
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Hala Halwani
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Fatemeh Saheb Sharif-Askari
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Rabih Halwani
- Research Institute for Medical and Health Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
- Department of Pediatrics, Faculty of Medicine, Prince Abdullah Ben Khaled Celiac Disease Research Chair, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
14
|
Galliano I, Montanari P, Monti G, Dini M, Calvi C, Clemente A, Pau A, Gambarino S, Bergallo M. IL10 and CXCL10 mRNA expression in food protein-induced enterocolitis syndrome. Cytokine 2024; 182:156720. [PMID: 39133968 DOI: 10.1016/j.cyto.2024.156720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND Food protein-induced enterocolitis syndrome (FPIES) is a rare non-IgE-mediated food allergy that mainly impacts babies and 7toddlers. The exact mechanism of FPIES is not completely understood. By studying the expression of IL-10 and CXCL10 in pediatric FPIES patients, researchers can gain insights into the immune mechanisms underlying this disorder. METHODS Peripheral venous blood was collected and subsequently stabilized with RNA pro. Total RNA was extracted and mRNA levels of CXCL10 and IL-10 was determined with real time PCR. RESULTS Children with FPIES had significantly higher values than the healthy control group (HC) for CXCL10 while FPIES had a significant lower values than the control group for IL-10. CONCLUSIONS Our results show a high production of CXCL10 and a concomitant reduced production of IL-10 in FPIES subjects who have not yet reached tolerance. These data may represent a molecular diagnostic marker for FPIES.
Collapse
Affiliation(s)
- Ilaria Galliano
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Paola Montanari
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Giovanna Monti
- Pediatric Allergy Unit, Regina Margherita Children's Hospital, Piazza Polonia 94, Turin, Italy.
| | - Maddalena Dini
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Cristina Calvi
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Anna Clemente
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Anna Pau
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Stefano Gambarino
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| | - Massimiliano Bergallo
- Department of Pediatric Sciences and Public Health, University of Turin, Piazza Polonia 94, Turin, Italy.
| |
Collapse
|
15
|
Liao G, Yan Q, Zhang M, Zhang X, Yang J, Huang H, Liu X, Jiang Y, Gong J, Zhan S, Li D, Huang X. Integrative analysis of network pharmacology and proteomics reveal the protective effect of Xiaoqinglong Decotion on neutrophilic asthma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118102. [PMID: 38561057 DOI: 10.1016/j.jep.2024.118102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/10/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoqinglong Decotion (XQLD) is a commonly used Chinese herbal formula in clinical practice, especially for allergic diseases such as asthma. However, its intrinsic mechanism for the treatment of neutrophilic asthma (NA) remains unclear. AIM OF THE STUDY The aim of this study was to evaluate the efficacy and potential mechanisms of XQLD on NA using network pharmacology and in vivo experiments. MATERIALS AND METHODS First, the active compounds, potential targets and mechanisms of XQLD against NA were initially elucidated by network pharmacology. Then, OVA/CFA-induced NA mice were treated with XQLD to assess its efficacy. Proteins were then analyzed and quantified using a Tandem Mass Tags approach for differentially expressed proteins (DEPs) to further reveal the mechanisms of NA treatment by XQLD. Finally, the hub genes, critical DEPs and potential pathways were validated. RESULTS 176 active compounds and 180 targets against NA were identified in XQLD. Protein-protein interaction (PPI) network revealed CXCL10, CX3CR1, TLR7, NCF1 and FABP4 as hub genes. In vivo experiments showed that XQLD attenuated inflammatory infiltrates, airway mucus secretion and remodeling in the lungs of NA mice. Moreover, XQLD significantly alleviated airway neutrophil inflammation in NA mice by decreasing the expression of IL-8, MPO and NE. XQLD also reduced the levels of CXCL10, CX3CR1, TLR7, NCF1 and FABP4, which are closely associated with neutrophil inflammation. Proteomics analysis identified 28 overlapping DEPs in the control, NA and XQLD groups, and we found that XQLD inhibited ferroptosis signal pathway (elevated GPX4 and decreased ASCL3) as well as the expression of ARG1, MMP12 and SPP1, while activating the Rap1 signaling pathway. CONCLUSION This study revealed that inhibition of ARG1, MMP12 and SPP1 expression as well as ferroptosis pathways, and activation of the Rap1 signaling pathway contribute to the therapeutic effect of XQLD on NA.
Collapse
Affiliation(s)
- Gang Liao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Qian Yan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Miaofen Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Xinxin Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Jing Yang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Huiting Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Jing Gong
- Guangdong Provincial Second Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Shaofeng Zhan
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Detang Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Pharmacy, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Xiufang Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Clinical Research Academy of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
16
|
Radhouani M, Starkl P. Adjuvant-independent airway sensitization and infection mouse models leading to allergic asthma. FRONTIERS IN ALLERGY 2024; 5:1423938. [PMID: 39157265 PMCID: PMC11327155 DOI: 10.3389/falgy.2024.1423938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/05/2024] [Indexed: 08/20/2024] Open
Abstract
Asthma is a chronic respiratory disease of global importance. Mouse models of allergic asthma have been instrumental in advancing research and novel therapeutic strategies for patients. The application of relevant allergens and physiological routes of exposure in such models has led to valuable insights into the complexities of asthma onset and development as well as key disease mechanisms. Furthermore, environmental microbial exposures and infections have been shown to play a fundamental part in asthma pathogenesis and alter disease outcome. In this review, we delve into physiological mouse models of allergic asthma and explore literature reports on most significant interplays between microbial infections and asthma development with relevance to human disease.
Collapse
Affiliation(s)
- Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
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
|
18
|
Akenroye A, Nopsopon T, Hacker JJ, Laidlaw TM. Ratio of plasma IL-13/TNF- ∝ and CXCL10/CCL17 predicts mepolizumab and omalizumab response in asthma better than eosinophil count or immunoglobulin E level. Sci Rep 2024; 14:10404. [PMID: 38710930 PMCID: PMC11074109 DOI: 10.1038/s41598-024-60864-3] [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] [Received: 11/04/2023] [Accepted: 04/29/2024] [Indexed: 05/08/2024] Open
Abstract
To date, most studies to identify biomarkers associated with response to the anti-interleukin 5 agent, mepolizumab, and to the anti-immunoglobulin E agent, omalizumab have focused on clinically available biomarkers, such as the peripheral blood eosinophil counts (BEC) and total immunoglobulin E (IgE). However, these biomarkers often have low predictive accuracy, with many patients with eosinophilic or allergic asthma failing to demonstrate clinical response to mepolizumab or omalizumab respectively. In this study, we evaluated the association of baseline pre-biologic plasma levels of 26 cytokines and chemokines, including T-helper 1 (Th1)-, Th2-, Th17-related cytokines, and their ratios with subsequent clinical response to mepolizumab or omalizumab. We defined clinical response as a reduction in the baseline annual exacerbation rate by half or more over the one-year period following initiation of the biologic. Baseline levels of plasma IL-13 were differentially elevated in responders versus non-responders to mepolizumab and plasma CXCL10 levels were differentially elevated in responders to omalizumab. The ratio of IL-13/TNF-α had the best sensitivity and specificity in predicting response to mepolizumab and CXCL10/CCL17 to omalizumab, and these performed better as predictive biomarkers of response than BEC and IgE. Cytokines and chemokines associated with airway eosinophilia, allergic inflammation, or Th2 inflammation, such as IL-13 and CXCL10, may be better predictors of clinical response to mepolizumab and omalizumab, than IL-5 or IgE, the targets of mepolizumab and omalizumab.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Tanawin Nopsopon
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Jonathan J Hacker
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Tanya M Laidlaw
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 60 Fenwood Road, Boston, MA, 02115, USA
| |
Collapse
|
19
|
Cao TBT, Quoc QL, Jang JH, Park HS. Immune Cell-Mediated Autoimmune Responses in Severe Asthma. Yonsei Med J 2024; 65:194-201. [PMID: 38515356 PMCID: PMC10973555 DOI: 10.3349/ymj.2023.0432] [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: 10/26/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 03/23/2024] Open
Abstract
Severe asthma (SA) has heterogeneous inflammatory phenotypes characterized by persistent airway inflammation (eosinophilic and/or neutrophilic inflammation) and remodeling. Various immune cells (eosinophils, neutrophils, and macrophages) become more activated and release inflammatory mediators and extracellular traps, damaging the protective barrier of airway epithelial cells and further activating other immune and structural cells. These cells play a role in autoimmune responses in asthmatic airways, where the adaptive immune system generates autoantibodies, inducing immunoglobulin G-dependent airway inflammation. Recent studies have suggested that adult asthmatics had high titers of autoantibodies associated with asthma severity, although pathogenic factors or diagnostic criteria are not well-defined. This challenge is further compounded by asthmatics with the autoimmune responses showing therapy insensitivity or failure to current pharmacological and biological treatment. This review updates emerging mechanisms of autoimmune responses in asthmatic airways and provides insights into their roles, proposing potential biomarkers and therapeutic targets for SA.
Collapse
Affiliation(s)
- Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Quang Luu Quoc
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Jae-Hyuk Jang
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
20
|
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
|
21
|
Yin M, Wadhwa R, Marshall JE, Gillis CM, Kim RY, Dua K, Palsson-McDermott EM, Fallon PG, Hansbro PM, O’Neill LAJ. 4-Octyl Itaconate Alleviates Airway Eosinophilic Inflammation by Suppressing Chemokines and Eosinophil Development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:13-23. [PMID: 37991425 PMCID: PMC7617081 DOI: 10.4049/jimmunol.2300155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/20/2023] [Indexed: 11/23/2023]
Abstract
4-Octyl itaconate (4-OI) is a derivative of the Krebs cycle-derived metabolite itaconate and displays an array of antimicrobial and anti-inflammatory properties through modifying cysteine residues within protein targets. We have found that 4-OI significantly reduces the production of eosinophil-targeted chemokines in a variety of cell types, including M1 and M2 macrophages, Th2 cells, and A549 respiratory epithelial cells. Notably, the suppression of these chemokines in M1 macrophages was found to be NRF2-dependent. In addition, 4-OI can interfere with IL-5 signaling and directly affect eosinophil differentiation. In a model of eosinophilic airway inflammation in BALB/c mice, 4-OI alleviated airway resistance and reduced eosinophil recruitment to the lungs. Our findings suggest that itaconate derivatives could be promising therapeutic agents for the treatment of eosinophilic asthma.
Collapse
Affiliation(s)
- Maureen Yin
- School of Biochemistry and Immunology, Trinity Biomedical
Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute and University
of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South
Wales, Australia
| | - Jacqueline E Marshall
- Centre for Inflammation, Centenary Institute and University
of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South
Wales, Australia
| | - Caitlin M Gillis
- Centre for Inflammation, Centenary Institute and University
of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South
Wales, Australia
| | - Richard Y Kim
- School of Life Sciences, Faculty of Science, University of
Technology Sydney, Sydney, New South Wales, Australia
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute and University
of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South
Wales, Australia
| | - Eva M Palsson-McDermott
- School of Biochemistry and Immunology, Trinity Biomedical
Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Padraic G Fallon
- School of Biochemistry and Immunology, Trinity Biomedical
Sciences Institute, Trinity College Dublin, Dublin, Ireland
- School of Medicine, Trinity Biomedical Sciences Institute,
Trinity College Dublin, Dublin, Ireland
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University
of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney, New South
Wales, Australia
- Priority Research Centre for Immune Health, Hunter Medical
Research Institute and University of Newcastle, Newcastle, New South Wales,
Australia
| | - Luke AJ O’Neill
- School of Biochemistry and Immunology, Trinity Biomedical
Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Chow YH, Murphy RC, An D, Lai Y, Altemeier WA, Manicone AM, Hallstrand TS. Intravascular Leukocyte Labeling Refines the Distribution of Myeloid Cells in the Lung in Models of Allergen-induced Airway Inflammation. Immunohorizons 2023; 7:853-860. [PMID: 38099934 PMCID: PMC10759158 DOI: 10.4049/immunohorizons.2300059] [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: 08/03/2023] [Accepted: 11/15/2023] [Indexed: 12/18/2023] Open
Abstract
Innate immune cell populations are critical in asthma with different functional characteristics based on tissue location, which has amplified the importance of characterizing the precise number and location of innate immune populations in murine models of asthma. In this study, we performed premortem intravascular (IV) labeling of leukocytes in mice in two models of asthma to differentiate innate immune cell populations within the IV compartment versus those residing in the lung tissue or airway lumen. We performed spectral flow cytometry analysis of the blood, suspensions of digested lung tissue, and bronchoalveolar lavage fluid. We discovered that IV labeled leukocytes do not contaminate analysis of bronchoalveolar lavage fluid but represent a significant proportion of cells in digested lung tissue. Exclusion of IV leukocytes significantly improved the accuracy of the assessments of myeloid cells in the lung tissue and provided important insights into ongoing trafficking in both eosinophilic and neutrophilic asthma models.
Collapse
Affiliation(s)
- Yu-Hua Chow
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Ryan C. Murphy
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Dowon An
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Ying Lai
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - William A. Altemeier
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Anne M. Manicone
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| | - Teal S. Hallstrand
- Division of Pulmonary, Critical Care, and Sleep Medicine and Center for Lung Biology, Department of Medicine, University of Washington, Seattle, WA 98109
| |
Collapse
|
23
|
Holmdahl I, Chakraborty S, Hoyer A, Filiou A, Asarnoj A, Sjölander A, Borres MP, van Hage M, Hedlin G, Konradsen JR, Söderhäll C. Inflammatory related plasma proteins involved in acute preschool wheeze. Clin Transl Allergy 2023; 13:e12308. [PMID: 38006384 PMCID: PMC10618892 DOI: 10.1002/clt2.12308] [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: 05/19/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Preschool wheeze is a risk factor for asthma development. However, the molecular mechanism behind a wheezing episode is not well understood. OBJECTIVE Our aims were to assess the association of plasma proteins with acute preschool wheeze and to study the proteins with differential expression at the acute phase at revisit after 3 months. Additionally, to investigate the relationship between protein expression and clinical parameters. METHOD We measured 92 inflammatory proteins in plasma and clinical parameters from 145 children during an episode of preschool wheeze (PW) and at the revisit after 3 months (PW-R, n = 113/145) and 101 healthy controls (HC) aged 6-48 months in the GEWAC cohort using the antibody-mediated proximity extension-based assay (Olink Proteomics, Uppsala). RESULTS Of the 74 analysed proteins, 52 were differentially expressed between PW and HC. The expression profiles of the top 10 proteins, Oncostatin M (OSM), IL-10, IL-6, Fibroblast growth factor 21 (FGF21), AXIN1, CXCL10, SIRT2, TNFSF11, Tumour necrosis factor β (TNF-β) and CASP8, could almost entirely separate PW from HC. Five out of 10 proteins were associated with intake of oral corticosteroids (OCS) 24 h preceding blood sampling (OSM, CASP8, IL-10, TNF-β and CXCL10). No differences in protein expression were seen between PWs with or without OCS in comparison to HC. At the revisit after 3 months, differential protein expressions were still seen between PW-R and HC for three (IL-10, SIRT2 and FGF21) of the 10 proteins. CONCLUSION Our results contribute to unravelling potential immunopathological pathways shared between preschool wheeze and asthma.
Collapse
Affiliation(s)
- Idun Holmdahl
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Sandip Chakraborty
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Angela Hoyer
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Anastasia Filiou
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Anna Asarnoj
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | | | - Magnus P. Borres
- Thermo Fisher ScientificUppsalaSweden
- Department of Women's and Children's HealthUppsala UniversityUppsalaSweden
| | - Marianne van Hage
- Division of Immunology and AllergyDepartment of Medicine SolnaKarolinska Institutet and Karolinska University HospitalStockholmSweden
| | - Gunilla Hedlin
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Jon R. Konradsen
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| | - Cilla Söderhäll
- Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
- Astrid Lindgren's Children's HospitalKarolinska University HospitalStockholmSweden
| |
Collapse
|
24
|
Czimmerer Z, Nagy L. Epigenomic regulation of macrophage polarization: Where do the nuclear receptors belong? Immunol Rev 2023; 317:152-165. [PMID: 37074820 PMCID: PMC10524119 DOI: 10.1111/imr.13209] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023]
Abstract
Our laboratory has a long-standing research interest in understanding how lipid-activated transcription factors, nuclear hormone receptors, contribute to dendritic cell and macrophage gene expression regulation, subtype specification, and responses to a changing extra and intracellular milieu. This journey in the last more than two decades took us from identifying target genes for various RXR heterodimers to systematically mapping nuclear receptor-mediated pathways in dendritic cells to identifying hierarchies of transcription factors in alternative polarization in macrophages to broaden the role of nuclear receptors beyond strictly ligand-regulated gene expression. We detail here the milestones of the road traveled and draw conclusions regarding the unexpectedly broad role of nuclear hormone receptors as epigenomic components of dendritic cell and macrophage gene regulation as we are getting ready for the next challenges.
Collapse
Affiliation(s)
- Zsolt Czimmerer
- Institute of Genetics, Biological Research Centre, Eotvos Lorand Research Network, Szeged, Hungary
| | - Laszlo Nagy
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St Petersburg, FL, United States
| |
Collapse
|
25
|
Gauthier M, Kale SL, Oriss TB, Gorry M, Ramonell RP, Dalton K, Ray P, Fahy JV, Seibold MA, Castro M, Jarjour N, Gaston B, Bleecker ER, Meyers DA, Moore W, Hastie AT, Israel E, Levy BD, Mauger D, Erzurum S, Comhair SA, Wenzel SE, Ray A. CCL5 is a potential bridge between type 1 and type 2 inflammation in asthma. J Allergy Clin Immunol 2023; 152:94-106.e12. [PMID: 36893862 PMCID: PMC10330021 DOI: 10.1016/j.jaci.2023.02.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/06/2023] [Accepted: 02/13/2023] [Indexed: 03/09/2023]
Abstract
BACKGROUND Type 1 (T1) inflammation (marked by IFN-γ expression) is now consistently identified in subsets of asthma cohorts, but how it contributes to disease remains unclear. OBJECTIVE We sought to understand the role of CCL5 in asthmatic T1 inflammation and how it interacts with both T1 and type 2 (T2) inflammation. METHODS CCL5, CXCL9, and CXCL10 messenger RNA expression from sputum bulk RNA sequencing, as well as clinical and inflammatory data were obtained from the Severe Asthma Research Program III (SARP III). CCL5 and IFNG expression from bronchoalveolar lavage cell bulk RNA sequencing was obtained from the Immune Mechanisms in Severe Asthma (IMSA) cohort and expression related to previously identified immune cell profiles. The role of CCL5 in tissue-resident memory T-cell (TRM) reactivation was evaluated in a T1high murine severe asthma model. RESULTS Sputum CCL5 expression strongly correlated with T1 chemokines (P < .001 for CXCL9 and CXCL10), consistent with a role in T1 inflammation. CCL5high participants had greater fractional exhaled nitric oxide (P = .009), blood eosinophils (P < .001), and sputum eosinophils (P = .001) in addition to sputum neutrophils (P = .001). Increased CCL5 bronchoalveolar lavage expression was unique to a previously described T1high/T2variable/lymphocytic patient group in the IMSA cohort, with IFNG trending with worsening lung obstruction only in this group (P = .083). In a murine model, high expression of the CCL5 receptor CCR5 was observed in TRMs and was consistent with a T1 signature. A role for CCL5 in TRM activation was supported by the ability of the CCR5 inhibitor maraviroc to blunt reactivation. CONCLUSION CCL5 appears to contribute to TRM-related T1 neutrophilic inflammation in asthma while paradoxically also correlating with T2 inflammation and with sputum eosinophilia.
Collapse
Affiliation(s)
- Marc Gauthier
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| | - Sagar Laxman Kale
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Timothy B Oriss
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Michael Gorry
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Richard P Ramonell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Kathryn Dalton
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Prabir Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - John V Fahy
- Division of Pulmonary Allergy and Critical Care, University of California, San Francisco, Calif
| | - Max A Seibold
- Center for Genes, Environment, and Health and Department of Pediatrics, National Jewish Health, Denver, Colo; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colo
| | - Mario Castro
- Pulmonary, Critical Care and Sleep Medicine, University of Kansas School of Medicine, Kansas City, Kan
| | - Nizar Jarjour
- Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin School of Medicine, Madison, Wis
| | - Benjamin Gaston
- Riley Hospital for Children and Indiana University School of Medicine Department of Pediatrics, Indianapolis, Ind
| | - Eugene R Bleecker
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Ariz
| | - Deborah A Meyers
- Division of Genetics, Genomics and Precision Medicine, Department of Medicine, University of Arizona, Tucson, Ariz
| | - Wendy Moore
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC
| | - Annette T Hastie
- Section on Pulmonary, Critical Care, Allergy & Immunologic Diseases, Wake Forest School of Medicine, Winston-Salem, NC
| | - Elliot Israel
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - Bruce D Levy
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Mass
| | - David Mauger
- Division of Statistics and Bioinformatics, Department of Public Health Sciences, Pennsylvania State University, Hershey, Pa
| | - Serpil Erzurum
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Suzy A Comhair
- Lerner Research Institute, Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Environmental and Occupation Health, University of Pittsburgh School of Public Health, Pittsburgh, Pa
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| |
Collapse
|
26
|
Ginebaugh SP, Hagner M, Ray A, Erzurum SC, Comhair SAA, Denlinger LC, Jarjour NN, Castro M, Woodruff PG, Christenson SA, Bleecker ER, Meyers DA, Hastie AT, Moore WC, Mauger DT, Israel E, Levy BD, Wenzel SE, Camiolo MJ. Bronchial epithelial cell transcriptional responses to inhaled corticosteroids dictate severe asthmatic outcomes. J Allergy Clin Immunol 2023; 151:1513-1524. [PMID: 36796454 PMCID: PMC10257752 DOI: 10.1016/j.jaci.2023.01.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND Inhaled corticosteroids (CSs) are the backbone of asthma treatment, improving quality of life, exacerbation rates, and mortality. Although effective for most, a subset of patients with asthma experience CS-resistant disease despite receiving high-dose medication. OBJECTIVE We sought to investigate the transcriptomic response of bronchial epithelial cells (BECs) to inhaled CSs. METHODS Independent component analysis was performed on datasets, detailing the transcriptional response of BECs to CS treatment. The expression of these CS-response components was examined in 2 patient cohorts and investigated in relation to clinical parameters. Supervised learning was used to predict BEC CS responses using peripheral blood gene expression. RESULTS We identified a signature of CS response that was closely correlated with CS use in patients with asthma. Participants could be separated on the basis of CS-response genes into groups with high and low signature expression. Patients with low expression of CS-response genes, particularly those with a severe asthma diagnosis, showed worse lung function and quality of life. These individuals demonstrated enrichment for T-lymphocyte infiltration in endobronchial brushings. Supervised machine learning identified a 7-gene signature from peripheral blood that reliably identified patients with poor CS-response expression in BECs. CONCLUSIONS Loss of CS transcriptional responses within bronchial epithelium was related to impaired lung function and poor quality of life, particularly in patients with severe asthma. These individuals were identified using minimally invasive blood sampling, suggesting these findings may enable earlier triage to alternative treatments.
Collapse
Affiliation(s)
- Scott P Ginebaugh
- Integrative Systems Biology, University of Pittsburgh, Pittsburgh, Pa
| | | | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | | | | | - Loren C Denlinger
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Mario Castro
- University of Kansas School of Medicine, Kansas City, Mo
| | - Prescott G Woodruff
- University of California, San Francisco School of Medicine, San Francisco, Calif
| | | | - Eugene R Bleecker
- Division for Genetics, Genomics and Personalized Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | - Deborah A Meyers
- Division for Genetics, Genomics and Personalized Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | | | - Wendy C Moore
- Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Elliot Israel
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Bruce D Levy
- Department of Medicine, Divisions of Pulmonary & Critical Care Medicine & Allergy & Immunology, Brigham & Women's Hospital, Harvard Medical School, Boston, Mass
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pa; Department of Environmental Medicine and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | | |
Collapse
|
27
|
Wei C, Wang Y, Hu C. Bioinformatic analysis and experimental validation of the potential gene in the airway inflammation of steroid-resistant asthma. Sci Rep 2023; 13:8098. [PMID: 37208441 DOI: 10.1038/s41598-023-35214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
Steroid-resistant asthma is a troublesome clinical problem in public health. The pathogenesis of steroid-resistant asthma is complex and remains to be explored. In our work, the online Gene Expression Omnibus microarray dataset GSE7368 was used to explore differentially expressed genes (DEGs) between steroid-resistant asthma patients and steroid-sensitive asthma patients. Tissue-specific gene expression of DEGs was analyzed using BioGPS. The enrichment analyses were performed using GO, KEGG, and GSEA analysis. The protein-protein interaction network and key gene cluster were constructed using STRING, Cytoscape, MCODE, and Cytohubba. A steroid-resistant neutrophilic asthma mouse model was established using lipopolysaccharide (LPS) and ovalbumin (OVA). An LPS-stimulated J744A.1 macrophage model was prepared to validate the underlying mechanism of the interesting DEG gene using the quantitative reverse transcription-polymerase chain reaction (qRT-PCR). A total of 66 DEGs were identified, most of which were present in the hematologic/immune system. Enrichment analysis displayed that the enriched pathways were the IL-17 signaling pathway, MAPK signal pathway, Toll-like receptor signaling pathway, and so on. DUSP2, as one of the top upregulated DEGs, has not been clearly demonstrated in steroid-resistant asthma. In our study, we observed that the salubrinal administration (DUSP2 inhibitor) reversed neutrophilic airway inflammation and cytokine responses (IL-17A, TNF-α) in a steroid-resistant asthma mouse model. We also found that salubrinal treatment reduced inflammatory cytokines (CXCL10 and IL-1β) in LPS-stimulated J744A.1 macrophages. DUSP2 may be a candidate target for the therapy of steroid-resistant asthma.
Collapse
Affiliation(s)
- Chaochao Wei
- Department of Pulmonary and Critical Care Medicine, Hainan General Hospital, Haikou, People's Republic of China
- Department of Pulmonary and Critical Care Medicine, Affiliated Hainan Hospital of Hainan Medical University, Haikou, People's Republic of China
- Department of Oncology, Xiangya Hospital Central South University, Changsha, People's Republic of China
- Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, 571199, People's Republic of China
| | - Yang Wang
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Chengping Hu
- Department of Respiratory Medicine (Department of Respiratory and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
28
|
Li J, Zheng Z, Liu Y, Zhang H, Zhang Y, Gao J. IRAK-M has effects in regulation of lung epithelial inflammation. Respir Res 2023; 24:103. [PMID: 37029363 PMCID: PMC10082527 DOI: 10.1186/s12931-023-02406-5] [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: 11/19/2022] [Accepted: 03/22/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Epithelial barrier is important for asthma development by shaping immune responses. Airway expressing-IL-1 receptor-associated kinase (IRAK)-M of Toll-like receptor pathway was involved in immunoregulation of airway inflammation through influencing activities of macrophages and dendritic cells or T cell differentiation. Whether IRAK-M has effect on cellular immunity in airway epithelial cells upon stimulation remains unclear. METHODS We modeled cellular inflammation induced by IL-1β, TNF-α, IL-33, and house dust mite (HDM) in BEAS-2B and A549 cells. Cytokine production and pathway activation were used to reflect the effects of IRAK-M siRNA knockdown on epithelial immunity. Genotyping an asthma-susceptible IRAK-M SNP rs1624395 and measurement of serum CXCL10 levels were performed in asthma patients. RESULTS IRAK-M expression was significantly induced in BEAS-2B and A549 cells after inflammatory stimulation. IRAK-M knockdown increased the lung epithelial production of cytokines and chemokines, including IL-6, IL-8, CXCL10, and CXCL11, at both mRNA and protein levels. Upon stimulation, IRAK-M silencing led to overactivation of JNK and p38 MAPK in lung epithelial cells. While antagonizing JNK or p38 MAPK inhibited increased secretion of CXCL10 in IRAK-M silenced-lung epithelium. Asthma patients carrying G/G genotypes had significantly higher levels of serum CXCL10 than those carrying homozygote A/A. CONCLUSION Our findings suggested that IRAK-M has effect on lung epithelial inflammation with an influence on epithelial secretion of CXCL10 partly mediated through JNK and p38 MAPK pathways. IRAK-M modulation might indicate a new insight into asthma pathogenesis from disease origin.
Collapse
Affiliation(s)
- Jia Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Zhoude Zheng
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yi Liu
- Department of Respiratory Medicine, Civil Aviation General Hospital, Beijing, 100123, China
| | - Hongbing Zhang
- Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Youming Zhang
- Section of Genomic and Environmental Medicine National Heart and Lung Institute, Imperial College London, London, SW3 6LY, UK
| | - Jinming Gao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
29
|
Carroll OR, Pillar AL, Brown AC, Feng M, Chen H, Donovan C. Advances in respiratory physiology in mouse models of experimental asthma. Front Physiol 2023; 14:1099719. [PMID: 37008013 PMCID: PMC10060990 DOI: 10.3389/fphys.2023.1099719] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023] Open
Abstract
Recent advances in mouse models of experimental asthma coupled with vast improvements in systems that assess respiratory physiology have considerably increased the accuracy and human relevance of the outputs from these studies. In fact, these models have become important pre-clinical testing platforms with proven value and their capacity to be rapidly adapted to interrogate emerging clinical concepts, including the recent discovery of different asthma phenotypes and endotypes, has accelerated the discovery of disease-causing mechanisms and increased our understanding of asthma pathogenesis and the associated effects on lung physiology. In this review, we discuss key distinctions in respiratory physiology between asthma and severe asthma, including the magnitude of airway hyperresponsiveness and recently discovered disease drivers that underpin this phenomenon such as structural changes, airway remodeling, airway smooth muscle hypertrophy, altered airway smooth muscle calcium signaling, and inflammation. We also explore state-of-the-art mouse lung function measurement techniques that accurately recapitulate the human scenario as well as recent advances in precision cut lung slices and cell culture systems. Furthermore, we consider how these techniques have been applied to recently developed mouse models of asthma, severe asthma, and asthma-chronic obstructive pulmonary disease overlap, to examine the effects of clinically relevant exposures (including ovalbumin, house dust mite antigen in the absence or presence of cigarette smoke, cockroach allergen, pollen, and respiratory microbes) and to increase our understanding of lung physiology in these diseases and identify new therapeutic targets. Lastly, we focus on recent studies that examine the effects of diet on asthma outcomes, including high fat diet and asthma, low iron diet during pregnancy and predisposition to asthma development in offspring, and environmental exposures on asthma outcomes. We conclude our review with a discussion of new clinical concepts in asthma and severe asthma that warrant investigation and how we could utilize mouse models and advanced lung physiology measurement systems to identify factors and mechanisms with potential for therapeutic targeting.
Collapse
Affiliation(s)
- Olivia R. Carroll
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Amber L. Pillar
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Alexandra C. Brown
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Min Feng
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Chantal Donovan
- Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
- *Correspondence: Chantal Donovan,
| |
Collapse
|
30
|
Helou DG, Quach C, Fung M, Painter JD, Hurrell BP, Eddie Loh YH, Howard E, Shafiei-Jahani P, Soroosh P, Sharpe AH, Akbari O. Human PD-1 agonist treatment alleviates neutrophilic asthma by reprogramming T cells. J Allergy Clin Immunol 2023; 151:526-538.e8. [PMID: 35963455 PMCID: PMC9905221 DOI: 10.1016/j.jaci.2022.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neutrophilic asthma is associated with disease severity and corticosteroid insensitivity. Novel therapies are required to manage this life-threatening asthma phenotype. Programmed cell death protein-1 (PD-1) is a key homeostatic modulator of the immune response for T-cell effector functions. OBJECTIVE We sought to investigate the role of PD-1 in the regulation of acute neutrophilic inflammation in a murine model of airway hyperreactivity (AHR). METHODS House dust mite was used to induce and compare neutrophilic AHR in wild-type and PD-1 knockout mice. Then, the therapeutic potential of a human PD-1 agonist was tested in a humanized mouse model in which the PD-1 extracellular domain is entirely humanized. Single-cell RNA sequencing and flow cytometry were mainly used to investigate molecular and cellular mechanisms. RESULTS PD-1 was highly induced on pulmonary T cells in our inflammatory model. PD-1 deficiency was associated with an increased neutrophilic AHR and high recruitment of inflammatory cells to the lungs. Consistently, PD-1 agonist treatment dampened AHR, decreased neutrophil recruitment, and modulated cytokine production in a humanized PD-1 mouse model. Mechanistically, we demonstrated at the transcriptional and protein levels that the inhibitory effect of PD-1 agonist is associated with the reprogramming of pulmonary effector T cells that showed decreased number and activation. CONCLUSIONS PD-1 agonist treatment is efficient in dampening neutrophilic AHR and lung inflammation in a preclinical humanized mouse model.
Collapse
Affiliation(s)
- Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Christine Quach
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Marshall Fung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Yong-Hwee Eddie Loh
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, Calif
| | - Emily Howard
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | | | - Arlene H Sharpe
- Department of Immunology, Harvard Medical School, Boston, Mass
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Calif.
| |
Collapse
|
31
|
Britt RD, Porter N, Grayson MH, Gowdy KM, Ballinger M, Wada K, Kim HY, Guerau-de-Arellano M. Sterols and immune mechanisms in asthma. J Allergy Clin Immunol 2023; 151:47-59. [PMID: 37138729 PMCID: PMC10151016 DOI: 10.1016/j.jaci.2022.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The field of sterol and oxysterol biology in lung disease has recently gained attention, revealing a unique need for sterol uptake and metabolism in the lung. The presence of cholesterol transport, biosynthesis, and sterol/oxysterol-mediated signaling in immune cells suggests a role in immune regulation. In support of this idea, statin drugs that inhibit the cholesterol biosynthesis rate-limiting step enzyme, hydroxymethyl glutaryl coenzyme A reductase, show immunomodulatory activity in several models of inflammation. Studies in human asthma reveal contradicting results, whereas promising retrospective studies suggest benefits of statins in severe asthma. Here, we provide a timely review by discussing the role of sterols in immune responses in asthma, analytical tools to evaluate the role of sterols in disease, and potential mechanistic pathways and targets relevant to asthma. Our review reveals the importance of sterols in immune processes and highlights the need for further research to solve critical gaps in the field.
Collapse
Affiliation(s)
- Rodney D. Britt
- Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus
- Department of Pediatrics, The Ohio State University, Columbus
| | - Ned Porter
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville
| | - Mitchell H. Grayson
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus
| | - Kymberly M. Gowdy
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, Wexner Medical Center, Columbus
| | - Megan Ballinger
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine, Wexner Medical Center, Columbus
| | - Kara Wada
- Department of Otolaryngology, Wexner Medical Center, Columbus
| | - Hye-Young Kim
- Department of Chemistry and Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville
| | - Mireia Guerau-de-Arellano
- School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, Columbus
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus
- Department of Neuroscience, The Ohio State University, Columbus
| |
Collapse
|
32
|
McIntyre AP, Viswanathan RK. Phenotypes and Endotypes in Asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:119-142. [PMID: 37464119 DOI: 10.1007/978-3-031-32259-4_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Asthma is a broadly encompassing diagnosis of airway inflammation with significant variability in presentation and response. Advances in molecular techniques and imaging have unraveled the delicate mechanistic tapestry responsible for the underlying inflammatory pathways in asthma. The elucidation of biomarkers and cellular components specific to these inflammatory pathways allowed for the categorization of asthma from generic phenotypes to more specific mechanistic endotypes, with two prominent subgroups emerging based on the level of Type 2 inflammation present - T2 high and T2 low (or non-T2). Sophisticated modeling and cluster analyses using a combination of clinical, physiologic, and biomarker parameters have permitted the identification of subendotypes within the broader T2 umbrella. This mechanistic-driven classification schema for asthma has dramatically altered the landscape of asthma management with the discovery and approval of targeted biologic therapies and has ushered in a new era of personalized precision medicine in asthma.
Collapse
Affiliation(s)
- Amanda P McIntyre
- Division of Allergy, Pulmonary & Critical Care, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA
| | - Ravi K Viswanathan
- Division of Allergy, Pulmonary & Critical Care, Department of Medicine, University of Wisconsin School of Medicine & Public Health, Madison, WI, USA.
| |
Collapse
|
33
|
Ray A, Das J, Wenzel SE. Determining asthma endotypes and outcomes: Complementing existing clinical practice with modern machine learning. Cell Rep Med 2022; 3:100857. [PMID: 36543110 PMCID: PMC9798025 DOI: 10.1016/j.xcrm.2022.100857] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/24/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022]
Abstract
There is unprecedented opportunity to use machine learning to integrate high-dimensional molecular data with clinical characteristics to accurately diagnose and manage disease. Asthma is a complex and heterogeneous disease and cannot be solely explained by an aberrant type 2 (T2) immune response. Available and emerging multi-omics datasets of asthma show dysregulation of different biological pathways including those linked to T2 mechanisms. While T2-directed biologics have been life changing for many patients, they have not proven effective for many others despite similar biomarker profiles. Thus, there is a great need to close this gap to understand asthma heterogeneity, which can be achieved by harnessing and integrating the rich multi-omics asthma datasets and the corresponding clinical data. This article presents a compendium of machine learning approaches that can be utilized to bridge the gap between predictive biomarkers and actual causal signatures that are validated in clinical trials to ultimately establish true asthma endotypes.
Collapse
Affiliation(s)
- Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, 3459 Fifth Avenue, MUH 628 NW, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Jishnu Das
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Systems Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sally E Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, 3459 Fifth Avenue, MUH 628 NW, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Environmental Medicine and Occupational Health, School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
34
|
Bryant N, Muehling LM. T-cell responses in asthma exacerbations. Ann Allergy Asthma Immunol 2022; 129:709-718. [PMID: 35918022 PMCID: PMC9987567 DOI: 10.1016/j.anai.2022.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Asthma is a chronic lung disease comprising multiple endotypes and characterized by periodic exacerbations. A diverse array of T cells has been found to contribute to all endotypes of asthma in pathogenic and regulatory roles. Here, we review the contributions of CD4+, CD8+, and unconventional T cells in allergic and nonallergic asthma. DATA SOURCES Review of published literature pertaining to conventional and unconventional T-cell types in asthma. STUDY SELECTIONS Recent peer-reviewed articles pertaining to T cells in asthma, with additional peer-reviewed studies for context. RESULTS Much research in asthma has focused on the roles of CD4+ TH cells. Roles for TH2 cells in promoting allergic asthma pathogenesis have been well-described, and the recent description of pathogenic TH2A cells provides additional insight into these responses. Other TH types, notably TH1 and TH17, have been linked to neutrophilic and steroid-resistant asthma phenotypes. Beyond CD4+ T cells, CD8+ Tc2 cells are also strongly associated with allergic asthma. An emerging area for study is unconventional T-cell types, including γδT, invariant natural killer T, and mucosal-associated invariant T cells. Although data in asthma remain limited for these cells, their ability to bridge innate and adaptive responses likely makes them key players in asthma. A number of asthma therapies target T-cell responses, and, although data are limited, they seem to modulate T-cell populations. CONCLUSION Given the diversity and heterogeneity of asthma and T-cell responses, there remain many rich avenues for research to better understand the pathogenesis of asthma. Despite the breadth of T cells in asthma, approved therapeutics remain limited to TH2 networks.
Collapse
Affiliation(s)
- Naomi Bryant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
35
|
Current Limitations and Recent Advances in the Management of Asthma. Dis Mon 2022:101483. [DOI: 10.1016/j.disamonth.2022.101483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
36
|
Mantov N, Zrounba M, Brollo M, Grassin-Delyle S, Glorion M, David M, Naline E, Devillier P, Salvator H. Ruxolitinib inhibits cytokine production by human lung macrophages without impairing phagocytic ability. Front Pharmacol 2022; 13:896167. [PMID: 36059986 PMCID: PMC9437255 DOI: 10.3389/fphar.2022.896167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The Janus kinase (JAK) 1/2 inhibitor ruxolitinib has been approved in an indication of myelofibrosis and is a candidate for the treatment of a number of inflammatory or autoimmune diseases. We assessed the effects of ruxolitinib on lipopolysaccharide (LPS)- and poly (I:C)-induced cytokine production by human lung macrophages (LMs) and on the LMs’ phagocytic activity.Methods: Human LMs were isolated from patients operated on for lung carcinoma. The LMs were cultured with ruxolitinib (0.5 × 10−7 M to 10–5 M) or budesonide (10–11 to 10–8 M) and then stimulated with LPS (10 ng·ml−1) or poly (I:C) (10 μg·ml−1) for 24 h. Cytokines released by the LMs into the supernatants were measured using ELISAs. The phagocytosis of labelled bioparticles was assessed using flow cytometry.Results: Ruxolitinib inhibited both the LPS- and poly (I:C)-stimulated production of tumor necrosis factor alpha, interleukin (IL)-6, IL-10, chemokines CCL2, and CXCL10 in a concentration-dependent manner. Ruxolitinib also inhibited the poly (I:C)- induced (but not the LPS-induced) production of IL-1ß. Budesonide inhibited cytokine production more strongly than ruxolitinib but failed to mitigate the production of CXCL10. The LMs’ phagocytic activity was not impaired by the highest tested concentration (10–5 M) of ruxolitinib.Conclusion: Clinically relevant concentrations of ruxolitinib inhibited the LPS- and poly (I:C)-stimulated production of cytokines by human LMs but did not impair their phagocytic activity. Overall, ruxolitinib’s anti-inflammatory activities are less intense than (but somewhat different from) those of budesonide—particularly with regard to the production of the corticosteroid-resistant chemokine CXCL-10. Our results indicate that treatment with a JAK inhibitor might be a valuable anti-inflammatory strategy in chronic obstructive pulmonary disease, Th1-high asthma, and both viral and non-viral acute respiratory distress syndromes (including coronavirus disease 2019).
Collapse
Affiliation(s)
- Nikola Mantov
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
| | - Mathilde Zrounba
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
- Respiratory Diseases Department, Foch Hospital, Suresnes, France
| | - Marion Brollo
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
| | - S Grassin-Delyle
- Respiratory Diseases Department, Foch Hospital, Suresnes, France
- Infection and Inflammation, Health Biotechnology Department, Paris-Saclay University, UVSQ, INSERM, Montigny le Bretonneux, France
| | - Matthieu Glorion
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
- Thoracic Surgery Department, Foch Hospital, Suresnes, France
| | - Mélanie David
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
| | - Emmanuel Naline
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
| | - Philippe Devillier
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
- Respiratory Diseases Department, Foch Hospital, Suresnes, France
- Faculté des Sciences de la Santé Simone Veil, UVSQ Paris-Saclay University, Montigny-le-Bretonneux, France
| | - Hélène Salvator
- Laboratory of Research in Respiratory Pharmacology—Virologie et Immunologie Moleculaire (VIM) Suresnes, V2I—UMR-0892 Paris Saclay University, Suresnes, France
- Respiratory Diseases Department, Foch Hospital, Suresnes, France
- Faculté des Sciences de la Santé Simone Veil, UVSQ Paris-Saclay University, Montigny-le-Bretonneux, France
- *Correspondence: Hélène Salvator,
| |
Collapse
|
37
|
Rahimi RA, Cho JL, Jakubzick CV, Khader SA, Lambrecht BN, Lloyd CM, Molofsky AB, Talbot S, Bonham CA, Drake WP, Sperling AI, Singer BD. Advancing Lung Immunology Research: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2022; 67:e1-18. [PMID: 35776495 PMCID: PMC9273224 DOI: 10.1165/rcmb.2022-0167st] [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] [Indexed: 11/24/2022] Open
Abstract
The mammalian airways and lungs are exposed to a myriad of inhaled particulate matter, allergens, and pathogens. The immune system plays an essential role in protecting the host from respiratory pathogens, but a dysregulated immune response during respiratory infection can impair pathogen clearance and lead to immunopathology. Furthermore, inappropriate immunity to inhaled antigens can lead to pulmonary diseases. A complex network of epithelial, neural, stromal, and immune cells has evolved to sense and respond to inhaled antigens, including the decision to promote tolerance versus a rapid, robust, and targeted immune response. Although there has been great progress in understanding the mechanisms governing immunity to respiratory pathogens and aeroantigens, we are only beginning to develop an integrated understanding of the cellular networks governing tissue immunity within the lungs and how it changes after inflammation and over the human life course. An integrated model of airway and lung immunity will be necessary to improve mucosal vaccine design as well as prevent and treat acute and chronic inflammatory pulmonary diseases. Given the importance of immunology in pulmonary research, the American Thoracic Society convened a working group to highlight central areas of investigation to advance the science of lung immunology and improve human health.
Collapse
|
38
|
Ragipoglu D, Bülow J, Hauff K, Voss M, Haffner-Luntzer M, Dudeck A, Ignatius A, Fischer V. Mast Cells Drive Systemic Inflammation and Compromised Bone Repair After Trauma. Front Immunol 2022; 13:883707. [PMID: 35558068 PMCID: PMC9086903 DOI: 10.3389/fimmu.2022.883707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
There is evidence that mast cells contribute to inflammation induced by hemorrhagic shock, severe tissue injury or sepsis. Mast cells are highly responsive to alarm signals generated after trauma, and release many inflammatory mediators including interleukin-6, a key mediator of posttraumatic inflammation. An overwhelming posttraumatic inflammation causes compromised bone healing; however, the underlying cellular and molecular mechanisms are poorly understood. Recently, we found that mast cells trigger local and systemic inflammation after isolated fracture leading to uneventful bone repair. Here, we investigated whether mast cells critically contribute to trauma-induced compromised bone healing. Male Mcpt5-Cre+ R-DTA mice, which lack connective tissue type mast cells, and their mast cell-competent Cre- littermates underwent a femur fracture with/without thoracic trauma. Posttraumatic systemic and local inflammation and bone repair were assessed 3 h and 21 d post injury. Both, the systemic and pulmonary inflammation was significantly increased in mast cell-competent mice upon combined trauma compared to isolated fracture. In mast cell-deficient mice, the increase of inflammatory mediators in the circulation induced by the severe trauma was abolished. In the bronchoalveolar lavage fluid, the trauma-induced increase of inflammatory cytokines was not reduced, but the neutrophil invasion into the lungs was significantly diminished in the absence of mast cells. Locally in the fracture hematoma, mast cell-competent mice displayed reduced inflammatory mediator concentrations after combined trauma compared to isolated fracture, which was abolished in mast cell-deficient mice. Notably, while combined trauma resulted in compromised bone repair in mast cell-competent mice, indicated by significantly reduced bone and increased cartilage fracture callus contents, this was abolished in Mcpt5-Cre+ R-DTA mice. Therefore, mast cells contribute to trauma-induced compromised bone repair and could be a potential target for new treatment options to improve fracture healing in multiply injured patients.
Collapse
Affiliation(s)
- Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Jasmin Bülow
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Kristin Hauff
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Martin Voss
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Anne Dudeck
- Medical Faculty, Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Trauma Research Center Ulm (ZTF), Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
39
|
Jackson D, Walum J, Banerjee P, Lewis BW, Prakash YS, Sathish V, Xu Z, Britt RD. Th1 cytokines synergize to change gene expression and promote corticosteroid insensitivity in pediatric airway smooth muscle. Respir Res 2022; 23:126. [PMID: 35578269 PMCID: PMC9109364 DOI: 10.1186/s12931-022-02046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/07/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Corticosteroids remain a key therapy for treating children with asthma. Patients with severe asthma are insensitive, resistant, or refractory to corticosteroids and have poorly controlled symptoms that involve airway inflammation, airflow obstruction, and frequent exacerbations. While the pathways that mediate corticosteroid insensitivity in asthma remain poorly defined, recent studies suggest that enhanced Th1 pathways, mediated by TNFα and IFNγ, may play a role. We previously reported that the combined effects of TNFα and IFNγ promote corticosteroid insensitivity in developing human airway smooth muscle (ASM).
Methods
To further understand the effects of TNFα and IFNγ on corticosteroid sensitivity in the context of neonatal and pediatric asthma, we performed RNA sequencing (RNA-seq) on human pediatric ASM treated with fluticasone propionate (FP), TNFα, and/or IFNγ.
Results
We found that TNFα had a greater effect on gene expression (~ 1000 differentially expressed genes) than IFNγ (~ 500 differentially expressed genes). Pathway and transcription factor analyses revealed enrichment of several pro-inflammatory responses and signaling pathways. Interestingly, treatment with TNFα and IFNγ augmented gene expression with more than 4000 differentially expressed genes. Effects of TNFα and IFNγ enhanced several pro-inflammatory genes and pathways related to ASM and its contributions to asthma pathogenesis, which persisted in the presence of corticosteroids. Co-expression analysis revealed several gene networks related to TNFα- and IFNγ-mediated signaling, pro-inflammatory mediator production, and smooth muscle contractility. Many of the co-expression network hubs were associated with genes that are insensitive to corticosteroids.
Conclusions
Together, these novel studies show the combined effects of TNFα and IFNγ on pediatric ASM and implicate Th1-associated cytokines in promoting ASM inflammation and hypercontractility in severe asthma.
Collapse
|
40
|
Adulthood asthma as a consequence of childhood adversity: a systematic review of epigenetically affected genes. J Dev Orig Health Dis 2022; 13:674-682. [PMID: 35256035 DOI: 10.1017/s2040174422000083] [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: 11/06/2022]
Abstract
There is an accumulating data that shows relation between childhood adversity and vulnerability to chronic diseases as well as epigenetic influences that in turn give rise to these diseases. Asthma is one of the chronic diseases that is influenced from genetic regulation of the inflammatory biomolecules and therefore the hypothesis in this research was childhood adversity might have caused epigenetic differentiation in the asthma-related genes in the population who had childhood trauma. To test this hypothesis, the literature was systematically reviewed to extract epigenetically modified gene data of the adults who had childhood adversity, and affected genes were further evaluated for their association with asthma. PRISMA guidelines were adopted and PubMed and Google Scholar were included in the searched databases, to evaluate epigenetic modifications in asthma-related genes of physically, emotionally or sexually abused children. After retrieving a total of 5245 articles, 36 of them were included in the study. Several genes and pathways that may contribute to pathogenesis of asthma development, increased inflammation, or response to asthma treatment were found epigenetically affected by childhood traumas. Childhood adversity, causing epigenetic changes in DNA, may lead to asthma development or influence the course of the disease and therefore should be taken into account for the prolonged health consequences.
Collapse
|
41
|
Celle A, Esteves P, Cardouat G, Beaufils F, Eyraud E, Dupin I, Maurat E, Lacomme S, Ousova O, Begueret H, Thumerel M, Marthan R, Girodet PO, Berger P, Trian T. Rhinovirus infection of bronchial epithelium induces specific bronchial smooth muscle cell migration of severe asthmatic patients. J Allergy Clin Immunol 2022; 150:104-113. [PMID: 35143808 DOI: 10.1016/j.jaci.2022.01.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Patients with severe asthma show an increase in both exacerbation frequency and bronchial smooth muscle (BSM) mass. Rhinovirus (RV) infection of the bronchial epithelium (BE) is the main trigger of asthma exacerbations. Histological analysis of biopsies shows that a close connection between BE and hypertrophic BSM is a criterion for severity of asthma. OBJECTIVE We hypothesized that RV infection of BE specifically increases asthmatic BSM cell migration. METHODS Serum samples, biopsies or BSM cells were obtained from 86 patients with severe asthma and 31 non-asthmatic subjects. BE cells from non-asthmatic subjects were cultured in an air-liquid interface and exposed to RV-16. Migration of BSM cells was assessed in response to BE supernatant using chemotaxis assays. Chemokine concentrations were analyzed by transcriptomics and ELISAs. Immunocytochemistry, western blotting and flow cytometry were used to quantify CXCR3 isoform distribution. CXCR3 downstream signaling pathways were assessed by calcium imaging and western blots. RESULTS BSM cells from severe asthmatic patients specifically migrated toward RV-infected BE, whereas those from non-asthmatic subjects did not. This specific migration is driven by BE CXCL10, which was increased in vitro in response to RV infection as well as in vivo in serum from exacerbating patients with severe asthma. The mechanism is related to both decreased expression and activation of the CXCR3-B-specific isoform in severe asthmatic BSM cells. CONCLUSION We have demonstrated a novel mechanism of BSM remodeling in severe asthmatic patients following RV exacerbation. This study highlights the CXCL10/CXCR3-A axis as a potential therapeutic target in severe asthma.
Collapse
Affiliation(s)
- Alexis Celle
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Pauline Esteves
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Guillaume Cardouat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Fabien Beaufils
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Edmée Eyraud
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Isabelle Dupin
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Elise Maurat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Sabrina Lacomme
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France
| | - Olga Ousova
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France
| | - Hugues Begueret
- CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Matthieu Thumerel
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Roger Marthan
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Pierre-Olivier Girodet
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Patrick Berger
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France; CHU de Bordeaux, Service d'exploration fonctionnelle respiratoire, Service de chirurgie, CIC 1401
| | - Thomas Trian
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, CIC 1401, F-33000 Bordeaux, France.
| |
Collapse
|
42
|
Badi YE, Pavel AB, Pavlidis S, Riley JH, Bates S, Kermani NZ, Knowles R, Kolmert J, Wheelock CE, Worsley S, Uddin M, Alving K, Bakke PS, Behndig A, Caruso M, Chanez P, Fleming LJ, Fowler SJ, Frey U, Howarth P, Horváth I, Krug N, Maitland-van der Zee AH, Montuschi P, Roberts G, Sanak M, Shaw DE, Singer F, Sterk PJ, Djukanovic R, Dahlen SE, Guo YK, Chung KF, Guttman-Yassky E, Adcock IM. Mapping atopic dermatitis and anti-IL-22 response signatures to type 2-low severe neutrophilic asthma. J Allergy Clin Immunol 2022; 149:89-101. [PMID: 33891981 DOI: 10.1016/j.jaci.2021.04.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/11/2021] [Accepted: 04/09/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Transcriptomic changes in patients who respond clinically to biological therapies may identify responses in other tissues or diseases. OBJECTIVE We sought to determine whether a disease signature identified in atopic dermatitis (AD) is seen in adults with severe asthma and whether a transcriptomic signature for patients with AD who respond clinically to anti-IL-22 (fezakinumab [FZ]) is enriched in severe asthma. METHODS An AD disease signature was obtained from analysis of differentially expressed genes between AD lesional and nonlesional skin biopsies. Differentially expressed genes from lesional skin from therapeutic superresponders before and after 12 weeks of FZ treatment defined the FZ-response signature. Gene set variation analysis was used to produce enrichment scores of AD and FZ-response signatures in the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes asthma cohort. RESULTS The AD disease signature (112 upregulated genes) encompassing inflammatory, T-cell, TH2, and TH17/TH22 pathways was enriched in the blood and sputum of patients with asthma with increasing severity. Patients with asthma with sputum neutrophilia and mixed granulocyte phenotypes were the most enriched (P < .05). The FZ-response signature (296 downregulated genes) was enriched in asthmatic blood (P < .05) and particularly in neutrophilic and mixed granulocytic sputum (P < .05). These data were confirmed in sputum of the Airway Disease Endotyping for Personalized Therapeutics cohort. IL-22 mRNA across tissues did not correlate with FZ-response enrichment scores, but this response signature correlated with TH22/IL-22 pathways. CONCLUSIONS The FZ-response signature in AD identifies severe neutrophilic asthmatic patients as potential responders to FZ therapy. This approach will help identify patients for future asthma clinical trials of drugs used successfully in other chronic diseases.
Collapse
Affiliation(s)
- Yusef Eamon Badi
- National Heart and Lung Institute, the Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom; Data Science Institute, Imperial College London, London, United Kingdom
| | - Ana B Pavel
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Biomedical Engineering, The University of Mississippi, Oxford, Miss
| | - Stelios Pavlidis
- Data Science Institute, Imperial College London, London, United Kingdom
| | - John H Riley
- GSK Respiratory Therapeutic Area Unit, Stevenage, United Kingdom
| | - Stewart Bates
- GSK Respiratory Therapeutic Area Unit, Stevenage, United Kingdom
| | | | | | - Johan Kolmert
- Centre for Allergy Research, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Craig E Wheelock
- Division of Physiological Chemistry 2, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Sally Worsley
- GSK Value Evidence and Outcomes, Brentford, United Kingdom
| | - Mohib Uddin
- Respiratory Global Medicines Development, AstraZeneca, Gothenburg, Sweden
| | - Kjell Alving
- Department of Women's and Children's Health: Paediatric Research, Uppsala University, Uppsala, Sweden
| | - Per S Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Annelie Behndig
- Department of Public Health and Clinical Medicine, Division of Medicine/Respiratory Medicine, Umeå University, Umeå, Sweden
| | - Massimo Caruso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Pascal Chanez
- Aix-Marseille Universite, Assistance Publique des Hopitaux de Marseille, Clinic des Bronches, Allergies et Sommeil, Marseille, France
| | - Louise J Fleming
- National Heart and Lung Institute, the Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Manchester Academic Health Science Centre and NIHR Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Urs Frey
- University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Peter Howarth
- Clinical and Experimental Sciences and Human Development in Health, University of Southampton Faculty of Medicine, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, United Kingdom
| | - Ildikó Horváth
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | | | | | - Paolo Montuschi
- Pharmacology, Catholic University of the Sacred Heart, Agostino Gemelli University Hospital Foundation, Rome, Italy
| | - Graham Roberts
- Clinical and Experimental Sciences and Human Development in Health, University of Southampton Faculty of Medicine, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, United Kingdom
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dominick E Shaw
- University of Nottingham, NIHR Biomedical Research Centre, Nottingham, United Kingdom
| | - Florian Singer
- Division of Respiratory Medicine, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
| | - Peter J Sterk
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ratko Djukanovic
- Clinical and Experimental Sciences and Human Development in Health, University of Southampton Faculty of Medicine, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, United Kingdom
| | - Sven-Eric Dahlen
- Centre for Allergy Research, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Yi-Ke Guo
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, the Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ian M Adcock
- National Heart and Lung Institute, the Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom.
| |
Collapse
|
43
|
Gauthier M, Kale SL, Oriss TB, Scholl K, Das S, Yuan H, Hu S, Chen J, Camiolo M, Ray P, Wenzel S, Ray A. Dual role for CXCR3 and CCR5 in asthmatic type 1 inflammation. J Allergy Clin Immunol 2022; 149:113-124.e7. [PMID: 34146578 PMCID: PMC8674372 DOI: 10.1016/j.jaci.2021.05.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/17/2021] [Accepted: 05/27/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Many patients with severe asthma (SA) fail to respond to type 2 inflammation-targeted therapies. We previously identified a cohort of subjects with SA expressing type 1 inflammation manifesting with IFN-γ expression and variable type 2 responses. OBJECTIVE We investigated the role of the chemotactic receptors C-X-C chemokine receptor 3 (CXCR3) and C-C chemokine receptor 5 (CCR5) in establishing type 1 inflammation in SA. METHODS Bronchoalveolar lavage microarray data from the Severe Asthma Research Program I/II were analyzed for pathway expression and paired with clinical parameters. Wild-type, Cxcr3-/-, and Ccr5-/- mice were exposed to a type 1-high SA model with analysis of whole lung gene expression and histology. Wild-type and Cxcr3-/- mice were treated with a US Food and Drug Administration-approved CCR5 inhibitor (maraviroc) with assessment of airway resistance, inflammatory cell recruitment by flow cytometry, whole lung gene expression, and histology. RESULTS A cohort of subjects with increased IFN-γ expression showed higher asthma severity. IFN-γ expression was correlated with CXCR3 and CCR5 expression, but in Cxcr3-/- and Ccr5-/- mice type 1 inflammation was preserved in a murine SA model, most likely owing to compensation by the other pathway. Incorporation of maraviroc into the experimental model blunted airway hyperreactivity despite only mild effects on lung inflammation. CONCLUSIONS IFNG expression in asthmatic airways was strongly correlated with expression of both the chemokine receptors CXCR3 and CCR5. Although these pathways provide redundancy for establishing type 1 lung inflammation, inhibition of the CCL5/CCR5 pathway with maraviroc provided unique benefits in reducing airway hyperreactivity. Targeting this pathway may be a novel approach for improving lung function in individuals with type 1-high asthma.
Collapse
Affiliation(s)
- Marc Gauthier
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa.
| | - Sagar Laxman Kale
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Timothy B Oriss
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa; Department of Immunology, The University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Kathryn Scholl
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Sudipta Das
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Huijuan Yuan
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Sanmei Hu
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Jie Chen
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Matthew Camiolo
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa
| | - Prabir Ray
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa; Department of Immunology, The University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Sally Wenzel
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa; The University of Pittsburgh School of Environmental and Occupational Health, Pittsburgh, Pa
| | - Anuradha Ray
- Department of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Pittsburgh, Pa; Department of Immunology, The University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| |
Collapse
|
44
|
Fischer V, Ragipoglu D, Diedrich J, Steppe L, Dudeck A, Schütze K, Kalbitz M, Gebhard F, Haffner-Luntzer M, Ignatius A. Mast Cells Trigger Disturbed Bone Healing in Osteoporotic Mice. J Bone Miner Res 2022; 37:137-151. [PMID: 34633111 DOI: 10.1002/jbmr.4455] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/21/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022]
Abstract
Mast cells are important tissue-resident sensor and effector immune cells but also play a major role in osteoporosis development. Mast cells are increased in numbers in the bone marrow of postmenopausal osteoporotic patients, and mast cell-deficient mice are protected from ovariectomy (OVX)-induced bone loss. In this study, we showed that mast cell-deficient Mcpt5-Cre R-DTA mice were protected from OVX-induced disturbed fracture healing, indicating a critical role for mast cells in the pathomechanisms of impaired bone repair under estrogen-deficient conditions. We revealed that mast cells trigger the fracture-induced inflammatory response by releasing inflammatory mediators, including interleukin-6, midkine (Mdk), and C-X-C motif chemokine ligand 10 (CXCL10), and promote neutrophil infiltration into the fracture site in OVX mice. Furthermore, mast cells were responsible for reduced osteoblast and increased osteoclast activities in OVX mice callus, as well as increased receptor activator of NF-κB ligand serum levels in OVX mice. Additional in vitro studies with human cells showed that mast cells stimulate osteoclastogenesis by releasing the osteoclastogenic mediators Mdk and CXCL10 in an estrogen-dependent manner, which was mediated via the estrogen receptor alpha on mast cells. In conclusion, mast cells negatively affect the healing of bone fractures under estrogen-deficient conditions. Hence, targeting mast cells might provide a therapeutic strategy to improve disturbed bone repair in postmenopausal osteoporosis. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Verena Fischer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Deniz Ragipoglu
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Johanna Diedrich
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Lena Steppe
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anne Dudeck
- Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Konrad Schütze
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Department of Trauma and Orthopedic Surgery, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen-Nürnberg, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
45
|
Jiang Y, Deng S, Hu X, Luo L, Zhang Y, Zhang D, Li X, Feng J. Identification of potential biomarkers and immune infiltration characteristics in severe asthma. Int J Immunopathol Pharmacol 2022; 36:3946320221114194. [PMID: 35817495 PMCID: PMC9280849 DOI: 10.1177/03946320221114194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES We hope to identify key molecules that can be used as markers of asthma severity and investigate their correlation with immune cell infiltration in severe asthma. METHODS An asthma dataset was downloaded from the Gene Expression Omnibus database and then processed by R software to obtain differentially expressed genes (DEGs). First, multiple enrichment platforms were applied to analyze crucial biological processes and pathways and protein-protein interaction networks related to the DEGs. We next combined least absolute shrinkage and selection operator logistic regression and the support vector machine-recursive feature elimination algorithms to screen diagnostic markers of severe asthma. Then, a local cohort consisting of 40 asthmatic subjects (24 with moderate asthma and 16 with severe asthma) was used for biomarker validation. Finally, infiltration of immune cells in asthma bronchoalveolar lavage fluid and their correlation with the screened markers was evaluated by CIBERSORT. RESULTS A total of 97 DEGs were identified in this study. Most of these genes are enriched in T cell activation and immune response in the asthma biological process. CC-chemokine receptor 7 (CCR7) and natural killer cell protein 7(NKG7) were identified as markers of severe asthma. The highest area under the ROC curve (AUC) was from a new indicator combining CCR7 and NKG7 (AUC = 0.851, adj. p < 0.05). Resting and activated memory CD4 T cells, activated NK cells, and CD8 T cells were found to be significantly higher in the severe asthma group (adj. p < 0.01). CCR7 and NKG7 were significantly correlated with these infiltrated cells that showed differences between the two groups. In addition, CCR7 was found to be significantly positively correlated with eosinophils (r = 0.38, adj. p < 0.05) infiltrated in bronchoalveolar lavage fluid. CONCLUSION CCR7 and NKG7 might be used as potential markers for asthma severity, and their expression may be associated with differences in immune cell infiltration in the moderate and severe asthma groups.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Shuanglinzi Deng
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Xinyue Hu
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Lisha Luo
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Yingyu Zhang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Daimo Zhang
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Xiaozhao Li
- Department of Nephrology, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| | - Juntao Feng
- Center of Respiratory Medicine, Xiangya Hospital, 12570Central South University, Changsha, Hunan, China
| |
Collapse
|
46
|
Lewis BW, Jackson D, Amici SA, Walum J, Guessas M, Guessas S, Coneglio E, Boda AV, Guerau-de-Arellano M, Grayson MH, Britt RD. Corticosteroid insensitivity persists in the absence of STAT1 signaling in severe allergic airway inflammation. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1194-L1205. [PMID: 34755542 PMCID: PMC8715027 DOI: 10.1152/ajplung.00244.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Corticosteroid insensitivity in asthma limits the ability to effectively manage severe asthma, which is characterized by persistent airway inflammation, airway hyperresponsiveness (AHR), and airflow obstruction despite corticosteroid treatment. Recent reports indicate that corticosteroid insensitivity is associated with increased interferon-γ (IFN-γ) levels and T-helper (Th) 1 lymphocyte infiltration in severe asthma. Signal transducer and activator of transcription 1 (STAT1) activation by IFN-γ is a key signaling pathway in Th1 inflammation; however, its role in the context of severe allergic airway inflammation and corticosteroid sensitivity remains unclear. In this study, we challenged wild-type (WT) and Stat1-/- mice with mixed allergens (MA) augmented with c-di-GMP [bis-(3'-5')-cyclic dimeric guanosine monophosphate], an inducer of Th1 cell infiltration with increased eosinophils, neutrophils, Th1, Th2, and Th17 cells. Compared with WT mice, Stat1-/- had reduced neutrophils, Th1, and Th17 cell infiltration. To evaluate corticosteroid sensitivity, mice were treated with either vehicle, 1 or 3 mg/kg fluticasone propionate (FP). Corticosteroids significantly reduced eosinophil infiltration and cytokine levels in both c-di-GMP + MA-challenged WT and Stat1-/- mice. However, histological and functional analyses show that corticosteroids did not reduce airway inflammation, epithelial mucous cell abundance, airway smooth muscle mass, and AHR in c-di-GMP + MA-challenged WT or Stat1-/- mice. Collectively, our data suggest that increased Th1 inflammation is associated with a decrease in corticosteroid sensitivity. However, increased airway pathology and AHR persist in the absence of STAT1 indicate corticosteroid insensitivity in structural airway cells is a STAT1 independent process.
Collapse
Affiliation(s)
- Brandon W. Lewis
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Devine Jackson
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Stephanie A. Amici
- 5Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio
| | - Joshua Walum
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Manel Guessas
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Sonia Guessas
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Elise Coneglio
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Akhila V. Boda
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | - Mireia Guerau-de-Arellano
- 5Division of Medical Laboratory Science, Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University, Columbus, Ohio,6Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio,7Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio,8Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Mitchell H. Grayson
- 2Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,3Division of Allergy and Immunology, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,4Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Rodney D. Britt
- 1Center for Perinatal Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio,4Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
47
|
Subramanian H, Hashem T, Bahal D, Kammala AK, Thaxton K, Das R. Ruxolitinib Ameliorates Airway Hyperresponsiveness and Lung Inflammation in a Corticosteroid-Resistant Murine Model of Severe Asthma. Front Immunol 2021; 12:786238. [PMID: 34777398 PMCID: PMC8586657 DOI: 10.3389/fimmu.2021.786238] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/18/2021] [Indexed: 11/27/2022] Open
Abstract
Asthma prevalence has increased considerably over the decades and it is now considered as one of the most common chronic disorders in the world. While the current anti-asthmatic therapies are effective for most asthma patients, there are 5-10% subjects whose disease is not controlled by such agents and they account for about 50% of the asthma-associated healthcare costs. Such patients develop severe asthma (SA), a condition characterized by a dominant Th1/Th17 cytokine response that is accompanied by Type 2 (T2)-low endotype. As JAK (Janus Kinase) signaling is very important for the activation of several cytokine pathways, we examined whether inhibition of JAKs might lessen the clinical and laboratory manifestations of SA. To that end, we employed a recently described murine model that recapitulates the complex immune response identified in the airways of human SA patients. To induce SA, mice were sensitized with house dust mite extract (HDME) and cyclic (c)-di-GMP and then subsequently challenged with HDME and a lower dose of c-di-GMP. In this model, treatment with the JAK inhibitor, Ruxolitinib, significantly ameliorated all the features of SA, including airway hyperresponsiveness and lung inflammation as well as total IgE antibody titers. Thus, these studies highlight JAKs as critical targets for mitigating the hyper-inflammation that occurs in SA and provide the framework for their incorporation into future clinical trials for patients that have severe or difficult-to manage asthma.
Collapse
Affiliation(s)
- Hariharan Subramanian
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Tanwir Hashem
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Devika Bahal
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Ananth K Kammala
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Kanedra Thaxton
- College of Natural Science, Michigan State University, East Lansing, MI, United States
| | - Rupali Das
- Department of Physiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
48
|
Osman HM, El Basha NR, Mansour AF, Hanna MOF. Serum IFN-γ-induced protein 10 (IP10/CXCL10): association with asthma exacerbations and severity in children. J Asthma 2021; 59:2135-2142. [PMID: 34752186 DOI: 10.1080/02770903.2021.1999465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Objective: Asthma is a chronic disorder of the airways, in which chemokines coordinate airway inflammation and determine its severity. We aimed to study the chemokine interferon-γ-inducible protein 10 kDa (IP10/CXCL10), a member of the CXC receptor 3 (CXCR3) ligand family, at the protein level in the serum of children, to evaluate the association between CXCL10 and exacerbations of childhood asthma.Methods: Patients experiencing an asthma exacerbation (42 patients) and stable patients (43 patients) were investigated for serum CXCL10 levels.Results: Patients with an asthma exacerbation expressed significantly higher CXCL10 levels in the serum than stable patients (P = <0.001). Additionally, CXCL10 values were elevated in severe asthma compared with moderate and mild disease (P = <0.001). In patients experiencing asthma exacerbations, higher values of CXCL10 were observed in atopic patients compared with non-atopic patients (P = 0.027) and in uncontrolled and partly controlled patients compared with controlled patients (P = 0.046).Conclusions: CXCL10 is proposed as an inflammatory serum marker for asthma exacerbations and worsening asthma symptoms. The levels of CXCL10 are representative of the clinical severity of asthma.
Collapse
Affiliation(s)
- Hanan M Osman
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noussa R El Basha
- Department of Pediatrics, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Mariam Onsy F Hanna
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
49
|
Camiolo MJ, Zhou X, Wei Q, Trejo Bittar HE, Kaminski N, Ray A, Wenzel SE. Machine learning implicates the IL-18 signaling axis in severe asthma. JCI Insight 2021; 6:e149945. [PMID: 34591794 PMCID: PMC8663569 DOI: 10.1172/jci.insight.149945] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/29/2021] [Indexed: 12/30/2022] Open
Abstract
Asthma is a common disease with profoundly variable natural history and patient morbidity. Heterogeneity has long been appreciated, and much work has focused on identifying subgroups of patients with similar pathobiological underpinnings. Previous studies of the Severe Asthma Research Program (SARP) cohort linked gene expression changes to specific clinical and physiologic characteristics. While invaluable for hypothesis generation, these data include extensive candidate gene lists that complicate target identification and validation. In this analysis, we performed unsupervised clustering of the SARP cohort using bronchial epithelial cell gene expression data, identifying a transcriptional signature for participants suffering exacerbation-prone asthma with impaired lung function. Clinically, participants in this asthma cluster exhibited a mixed inflammatory process and bore transcriptional hallmarks of NF-κB and activator protein 1 (AP-1) activation, despite high corticosteroid exposure. Using supervised machine learning, we found a set of 31 genes that classified patients with high accuracy and could reconstitute clinical and transcriptional hallmarks of our patient clustering in an external cohort. Of these genes, IL18R1 (IL-18 Receptor 1) negatively associated with lung function and was highly expressed in the most severe patient cluster. We validated IL18R1 protein expression in lung tissue and identified downstream NF-κB and AP-1 activity, supporting IL-18 signaling in severe asthma pathogenesis and highlighting this approach for gene and pathway discovery.
Collapse
Affiliation(s)
- Matthew J. Camiolo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xiuxia Zhou
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Qi Wei
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sally E. Wenzel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
50
|
Camiolo MJ, Kale SL, Oriss TB, Gauthier M, Ray A. Immune responses and exacerbations in severe asthma. Curr Opin Immunol 2021; 72:34-42. [PMID: 33773471 PMCID: PMC8460694 DOI: 10.1016/j.coi.2021.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Asthma as a clinical entity manifests with a broad spectrum of disease severity. Unlike milder asthma, severe disease is poorly controlled by inhaled corticosteroids, the current standard of care. Transcriptomic data, along with patient characteristics and response to biologics show that though Type 2 (T2) immune response remains an integral feature of asthma, additional molecular and immunologic factors may play important roles in pathogenesis. Mechanisms of T2 development, cellular sources of T2 cytokines and their relationship to additional immune pathways concurrently activated may distinguish several different subphenotypes, and perhaps endotypes of asthma, with differential response to non-specific and targeted anti-inflammatory therapies. Recent data have also associated non-T2 cytokines derived from T cells, particularly IFN-γ, and epithelial mediators with severe asthma. These topics and their relationships to acute asthma exacerbations are discussed in this review.
Collapse
Affiliation(s)
- Matthew J Camiolo
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sagar L Kale
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Timothy B Oriss
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marc Gauthier
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|