101
|
Bhavsar PK. Role of Humoral Defense in Severe Asthma. Am J Respir Crit Care Med 2019; 197:1369-1371. [PMID: 29664685 DOI: 10.1164/rccm.201802-0288ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Pankaj K Bhavsar
- 1 National Heart and Lung Institute Imperial College London London, United Kingdom
| |
Collapse
|
102
|
Qin L, Gibson PG, Simpson JL, Baines KJ, McDonald VM, Wood LG, Powell H, Fricker M. Dysregulation of sputum columnar epithelial cells and products in distinct asthma phenotypes. Clin Exp Allergy 2019; 49:1418-1428. [PMID: 31264263 DOI: 10.1111/cea.13452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Dysfunction of the bronchial epithelium plays an important role in asthma; however, its measurement is challenging. Columnar epithelial cells are often quantified, yet rarely analysed, in induced sputum studies. OBJECTIVE We aimed to test whether sputum columnar epithelial cell proportion and count are altered in asthma, and whether they are associated with clinical and inflammatory variables. We aimed to test whether sputum-based measures could provide a relatively non-invasive means through which to monitor airway epithelial activation status. METHODS We examined the relationship of sputum columnar epithelial cells with clinical and inflammatory variables of asthma in a large retrospective cross-sectional cohort (901 participants with asthma and 138 healthy controls). In further studies, we used flow cytometry, microarray, qPCR and ELISA to characterize sputum columnar epithelial cells and their products. RESULTS Multivariate analysis and generation of 90th centile cut-offs (≥11% or ≥18.1 × 104 /mL) to identify columnar epithelial cell "high" asthma revealed a significant relationship between elevated sputum columnar cells and male gender, severe asthma and non-neutrophilic airway inflammation. Flow cytometry showed viable columnar epithelial cells were present in all sputum samples tested. An epithelial gene signature (SCGB3A1, LDLRAD1, FOXJ1, DNALI1, CFAP157, CFAP53) was detected in columnar epithelial cell-high sputum. CLCA1 mRNA and periostin protein, previously identified biomarkers of IL-13-mediated epithelial activation, were elevated in columnar epithelial cell-high sputum samples, but only when accompanied by eosinophilia. CONCLUSIONS & CLINICAL RELEVANCE Sputum columnar epithelial cells are related to important clinical and inflammatory variables in asthma. Measurement of epithelial biomarkers in sputum samples could allow non-invasive assessment of altered bronchial epithelium status in asthma.
Collapse
Affiliation(s)
- Ling Qin
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,Department of Respiratory Medicine (Department of Pulmonary and Critical Care Medicine), Xiangya Hospital, Central South University, Changsha, China
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Vanessa M McDonald
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Heather Powell
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, The University of Newcastle, New Lambton Heights, NSW, Australia.,National Health and Medical Research Council Centre of Excellence in Severe Asthma, Newcastle, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| |
Collapse
|
103
|
Gelfand EW, Hinks TSC. Is there a role for type 2 CD8 + T cells in patients with steroid-resistant asthma? J Allergy Clin Immunol 2019; 144:648-650. [PMID: 31376407 DOI: 10.1016/j.jaci.2019.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/01/2022]
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo.
| | - Timothy S C Hinks
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Department of Medicine Experimental Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
104
|
Kuruvilla ME, Lee FEH, Lee GB. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin Rev Allergy Immunol 2019; 56:219-233. [PMID: 30206782 DOI: 10.1007/s12016-018-8712-1] [Citation(s) in RCA: 747] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The model of asthma as a single entity has now been replaced by a much more complex biological network of distinct and interrelating inflammatory pathways. The term asthma is now considered an umbrella diagnosis for several diseases with distinct mechanistic pathways (endotypes) and variable clinical presentations (phenotypes). The precise definition of these endotypes is central to asthma management due to inherent therapeutic and prognostic implications. This review presents the molecular mechanisms behind the heterogeneity of airway inflammation in asthmatic patients. Asthma endotypes may be broadly regarded as type 2 (T2) high or T2-low. Several biologic agents have been approved for T2-high asthma, with numerous other therapeutics that are incipient and similarly targeted at specific molecular mechanisms. Collectively, these advances have shifted existing paradigms in the approach to asthma to tailor novel therapies.
Collapse
Affiliation(s)
- Merin E Kuruvilla
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, 615 Michael St, NE Suite 205, Atlanta, 30322, GA, USA.,Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University, 2015 Uppergate Dr. NE, Suite 326, Atlanta, GA, 30322, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, 615 Michael St, NE Suite 205, Atlanta, 30322, GA, USA.,Lowance Center for Human Immunology, Emory University, 615 Michael Street, Atlanta, 30322, GA, USA
| | - Gerald B Lee
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Medicine, Emory University, 615 Michael St, NE Suite 205, Atlanta, 30322, GA, USA. .,Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis, and Sleep, Department of Pediatrics, Emory University, 2015 Uppergate Dr. NE, Suite 326, Atlanta, GA, 30322, USA.
| |
Collapse
|
105
|
Miyake MM, Workman AD, Nocera AL, Wu D, Mueller SK, Finn K, Amiji MM, Bleier BS. Discriminant analysis followed by unsupervised cluster analysis including exosomal cystatins predict presence of chronic rhinosinusitis, phenotype, and disease severity. Int Forum Allergy Rhinol 2019; 9:1069-1076. [PMID: 31322841 DOI: 10.1002/alr.22380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Cystatins are epithelial protease inhibitors that participate in sinonasal immunity and inflammation. Nasal mucus-derived exosomes (NMDEs) are small vesicles secreted by epithelial cells that carry protein cargo reflective of their host cell. NMDEs have been used as a noninvasive biomarker source to study chronic rhinosinusitis with nasal polyps (CRSwNP) proteomics with superior sensitivity to whole mucus. The purpose of this study was to noninvasively quantify exosomal cystatins in a heterogenous population to determine their utility in predicting phenotype and disease severity. METHODS This was an Institutional Review Board-approved study in which NMDEs were purified from 105 patients undergoing sinonasal surgery by ultracentrifugation. Demographic and clinical variables were collected and phenotypes were assigned a priori. Linear discriminant analysis was executed based on normalized Cystatin values as phenotype predictor variables. Unsupervised cluster analysis was performed using Ward's linkage followed by Duda/Hart Je(2)/Je(1) index cluster stopping rules. Analysis of variance (ANOVA), Welch's test, and Fisher's exact tests were used for continuous and categorical variables. RESULTS NMDE Cystatin-2 expression segregated by phenotype (mean ± standard error [SEM]): control (23.4 ± 4.2 pg/µg, n = 32); CRS without NP (CRSsNP) (56.6 ± 8.3 pg/µg, n = 33); and CRSwNP (130.5 ± 16.7 pg/µg, n = 40) (p < 0.0001). Seven clusters were identified among patients where the highest NMDE Cystatin-2 levels clustered with asthma, tissue eosinophilia, and aspirin-exacerbated respiratory disease (AERD). CONCLUSION Cystatin levels in NMDEs predict CRS phenotype and disease severity. As a "liquid biopsy," noninvasive NMDE collection offers a promising opportunity to study disease pathophysiology, discriminate disease states, and potentially reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Michelle M Miyake
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA
| | - Alan D Workman
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA
| | - Angela L Nocera
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA.,Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA
| | - Dawei Wu
- Department of Otolaryngology, Beijing Anzhen Hospital, Capital Medical University, Beijing, P. R. China
| | - Sarina K Mueller
- Department of Otorhinolaryngology, Head and Neck Surgery, Friedrich-Alexander University Erlangen-Nurnberg, Erlangen, Germany
| | - Kristen Finn
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA
| |
Collapse
|
106
|
Liu G, Betts C, Cunoosamy DM, Åberg PM, Hornberg JJ, Sivars KB, Cohen TS. Use of precision cut lung slices as a translational model for the study of lung biology. Respir Res 2019; 20:162. [PMID: 31324219 PMCID: PMC6642541 DOI: 10.1186/s12931-019-1131-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 07/09/2019] [Indexed: 12/28/2022] Open
Abstract
Animal models remain invaluable for study of respiratory diseases, however, translation of data generated in genetically homogeneous animals housed in a clean and well-controlled environment does not necessarily provide insight to the human disease situation. In vitro human systems such as air liquid interface (ALI) cultures and organ-on-a-chip models have attempted to bridge the divide between animal models and human patients. However, although 3D in nature, these models struggle to recreate the architecture and complex cellularity of the airways and parenchyma, and therefore cannot mimic the complex cell-cell interactions in the lung. To address this issue, lung slices have emerged as a useful ex vivo tool for studying the respiratory responses to inflammatory stimuli, infection, and novel drug compounds. This review covers the practicality of precision cut lung slice (PCLS) generation and benefits of this ex vivo culture system in modeling human lung biology and disease pathogenesis.
Collapse
Affiliation(s)
- Guanghui Liu
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine Betts
- Pathology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Danen M Cunoosamy
- Bioscience, Respiratory Inflammation and Autoimmunity, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Present Address: Sanofi, Cambridge, MA, USA
| | - Per M Åberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jorrit J Hornberg
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Kinga Balogh Sivars
- RIA Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Taylor S Cohen
- Microbial Sciences, BioPharmaceuticals R&D, AstraZeneca, One Medimmune Way, Gaithersburg, MD, 20877, USA.
| |
Collapse
|
107
|
Lodge KM, Knolle MD, Jha A. Asthma: From Diagnosis to Endotype to Treatment. Am J Respir Crit Care Med 2019; 197:1065-1067. [PMID: 29494210 DOI: 10.1164/rccm.201710-2139rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Katharine M Lodge
- 1 Respiratory Medicine Division, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Martin D Knolle
- 2 Department of Respiratory Medicine, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| | - Akhilesh Jha
- 3 Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom; and.,4 Imperial College London, London, United Kingdom
| |
Collapse
|
108
|
Chung KF. Predicting Response to Triamcinolone in Severe Asthma by Machine Learning. Solving the Enigma. Am J Respir Crit Care Med 2019; 199:1299-1300. [PMID: 30789751 PMCID: PMC6543711 DOI: 10.1164/rccm.201902-0320ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Kian Fan Chung
- 1 National Heart & Lung Institute Imperial College London London, United Kingdom
| |
Collapse
|
109
|
Hassoun D, Moui A, Colas L, Blanc FX, Magnan A. [Update in severe asthma physiopathology and treatments]. Rev Med Interne 2019; 40:508-516. [PMID: 31128859 DOI: 10.1016/j.revmed.2019.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/26/2019] [Accepted: 04/12/2019] [Indexed: 11/17/2022]
Abstract
Asthma is an inflammatory airway disease which presentation is highly heterogeneous. Last two decades provided new clinical and basic data concerning asthma physiopathology that make global understanding much complex. Phenotypes based on clinical settings and paraclinical investigations from large cohorts confirm old paradigm (eosinophilic vs. non-eosinophilic asthma) but also introduce new concepts (obesity-related asthma, late onset asthma, etc.). Conversely, improvement of big data analytics allows to initiate new cohorts aiming at better understanding the pathophysiology underlying those phenotypes and unraveling new ones. However, clinical and therapeutic impacts of those big data need to be further detailed. In parallel, biotherapies and innovative techniques as bronchial thermoplasty become available for severe asthmatic patients who did not respond to specific treatment in the past. Development of a personalized medicine in severe asthma becomes an important challenge for tomorrow. This review will focus on new pathophysiological concepts arisen from large cohorts and new therapeutic strategies available and in progress for severe asthma.
Collapse
Affiliation(s)
- D Hassoun
- Service de pneumologie, l'institut du thorax, hôpital Guillaume et René LAENNEC, CHU de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex 1, France; Inserm UMR 1087, CNRS UMR 6291, l'institut du thorax, université de Nantes, 8, quai Moncousu, 44007 Nantes, France.
| | - A Moui
- Service de pneumologie, l'institut du thorax, hôpital Guillaume et René LAENNEC, CHU de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex 1, France; Inserm UMR 1087, CNRS UMR 6291, l'institut du thorax, université de Nantes, 8, quai Moncousu, 44007 Nantes, France
| | - L Colas
- Inserm UMR 1087, CNRS UMR 6291, l'institut du thorax, université de Nantes, 8, quai Moncousu, 44007 Nantes, France; Plate-forme transversale d'allergologie, l'institut du thorax, Hôpital Hôtel-Dieu, CHU de Nantes, 1, place Alexis-Ricordeau, 44093 Nantes cedex 1, France; Centre de recherche en transplantation et immunologie UMR1064, Inserm, université de Nantes, 30, boulevard Jean-Monnet, 44093 Nantes cedex 1, France
| | - F X Blanc
- Service de pneumologie, l'institut du thorax, hôpital Guillaume et René LAENNEC, CHU de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex 1, France; Inserm UMR 1087, CNRS UMR 6291, l'institut du thorax, université de Nantes, 8, quai Moncousu, 44007 Nantes, France
| | - A Magnan
- Service de pneumologie, l'institut du thorax, hôpital Guillaume et René LAENNEC, CHU de Nantes, boulevard Jacques-Monod, 44093 Nantes cedex 1, France; Inserm UMR 1087, CNRS UMR 6291, l'institut du thorax, université de Nantes, 8, quai Moncousu, 44007 Nantes, France
| |
Collapse
|
110
|
Altman MC, Gill MA, Whalen E, Babineau DC, Shao B, Liu AH, Jepson B, Gruchalla RS, O'Connor GT, Pongracic JA, Kercsmar CM, Khurana Hershey GK, Zoratti EM, Johnson CC, Teach SJ, Kattan M, Bacharier LB, Beigelman A, Sigelman SM, Presnell S, Gern JE, Gergen PJ, Wheatley LM, Togias A, Busse WW, Jackson DJ. Transcriptome networks identify mechanisms of viral and nonviral asthma exacerbations in children. Nat Immunol 2019; 20:637-651. [PMID: 30962590 PMCID: PMC6472965 DOI: 10.1038/s41590-019-0347-8] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 02/11/2019] [Indexed: 12/14/2022]
Abstract
Respiratory infections are common precursors to asthma exacerbations in children, but molecular immune responses that determine whether and how an infection causes an exacerbation are poorly understood. By using systems-scale network analysis, we identify repertoires of cellular transcriptional pathways that lead to and underlie distinct patterns of asthma exacerbation. Specifically, in both virus-associated and nonviral exacerbations, we demonstrate a set of core exacerbation modules, among which epithelial-associated SMAD3 signaling is upregulated and lymphocyte response pathways are downregulated early in exacerbation, followed by later upregulation of effector pathways including epidermal growth factor receptor signaling, extracellular matrix production, mucus hypersecretion, and eosinophil activation. We show an additional set of multiple inflammatory cell pathways involved in virus-associated exacerbations, in contrast to squamous cell pathways associated with nonviral exacerbations. Our work introduces an in vivo molecular platform to investigate, in a clinical setting, both the mechanisms of disease pathogenesis and therapeutic targets to modify exacerbations.
Collapse
Affiliation(s)
- Matthew C Altman
- Department of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA.
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA.
| | - Michelle A Gill
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth Whalen
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | | | - Baomei Shao
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew H Liu
- Department of Allergy and Immunology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Rebecca S Gruchalla
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - George T O'Connor
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | | | | | | - Meyer Kattan
- Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Leonard B Bacharier
- Division of Allergy, Immunology, and Pulmonary Medicine, Washington University, St. Louis, MO, USA
| | - Avraham Beigelman
- Division of Allergy, Immunology, and Pulmonary Medicine, Washington University, St. Louis, MO, USA
| | - Steve M Sigelman
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Scott Presnell
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - James E Gern
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter J Gergen
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Lisa M Wheatley
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Alkis Togias
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - William W Busse
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Daniel J Jackson
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
111
|
Östling J, van Geest M, Schofield JPR, Jevnikar Z, Wilson S, Ward J, Lutter R, Shaw DE, Bakke PS, Caruso M, Dahlen SE, Fowler SJ, Horváth I, Krug N, Montuschi P, Sanak M, Sandström T, Sun K, Pandis I, Auffray C, Sousa AR, Guo Y, Adcock IM, Howarth P, Chung KF, Bigler J, Sterk PJ, Skipp PJ, Djukanović R, Vaarala O. IL-17-high asthma with features of a psoriasis immunophenotype. J Allergy Clin Immunol 2019; 144:1198-1213. [PMID: 30998987 DOI: 10.1016/j.jaci.2019.03.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 03/06/2019] [Accepted: 03/18/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The role of IL-17 immunity is well established in patients with inflammatory diseases, such as psoriasis and inflammatory bowel disease, but not in asthmatic patients, in whom further study is required. OBJECTIVE We sought to undertake a deep phenotyping study of asthmatic patients with upregulated IL-17 immunity. METHODS Whole-genome transcriptomic analysis was performed by using epithelial brushings, bronchial biopsy specimens (91 asthmatic patients and 46 healthy control subjects), and whole blood samples (n = 498) from the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort. Gene signatures induced in vitro by IL-17 and IL-13 in bronchial epithelial cells were used to identify patients with IL-17-high and IL-13-high asthma phenotypes. RESULTS Twenty-two of 91 patients were identified with IL-17, and 9 patients were identified with IL-13 gene signatures. The patients with IL-17-high asthma were characterized by risk of frequent exacerbations, airway (sputum and mucosal) neutrophilia, decreased lung microbiota diversity, and urinary biomarker evidence of activation of the thromboxane B2 pathway. In pathway analysis the differentially expressed genes in patients with IL-17-high asthma were shared with those reported as altered in psoriasis lesions and included genes regulating epithelial barrier function and defense mechanisms, such as IL1B, IL6, IL8, and β-defensin. CONCLUSION The IL-17-high asthma phenotype, characterized by bronchial epithelial dysfunction and upregulated antimicrobial and inflammatory response, resembles the immunophenotype of psoriasis, including activation of the thromboxane B2 pathway, which should be considered a biomarker for this phenotype in further studies, including clinical trials targeting IL-17.
Collapse
Affiliation(s)
- Jörgen Östling
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marleen van Geest
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - James P R Schofield
- Centre for Proteomic Research, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Zala Jevnikar
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Susan Wilson
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jonathan Ward
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Rene Lutter
- AUMC, Department of Experimental Immunology, University of Amsterdam, Amsterdam, The Netherlands; AUMC, Department of Respiratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Dominick E Shaw
- Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom
| | - Per S Bakke
- Institute of Medicine, University of Bergen, Bergen, Norway
| | - Massimo Caruso
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sven-Erik Dahlen
- Centre for Allergy Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen J Fowler
- Respiratory and Allergy Research Group, University of Manchester, Manchester, United Kingdom
| | - Ildikó Horváth
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine Hannover, Hannover, Germany
| | - Paolo Montuschi
- Faculty of Medicine, Catholic University of the Sacred Heart, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy
| | - Marek Sanak
- Laboratory of Molecular Biology and Clinical Genetics, Medical College, Jagiellonian University, Krakow, Poland
| | - Thomas Sandström
- Department of Medicine, Department of Public Health and Clinical Medicine Respiratory Medicine Unit, Umeå University, Umeå, Sweden
| | - Kai Sun
- Data Science Institute, Imperial College, London, United Kingdom
| | - Ioannis Pandis
- Data Science Institute, Imperial College, London, United Kingdom
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Université de Lyon, Lyon, France
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GlaxoSmithKline, Stockley Park, United Kingdom
| | - Yike Guo
- Data Science Institute, Imperial College, London, United Kingdom
| | - Ian M Adcock
- Experimental Studies, Airways Disease Section, National Heart & Lung institute, Imperial College London, London, United Kingdom
| | - Peter Howarth
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Kian Fan Chung
- Experimental Studies, Airways Disease Section, National Heart & Lung institute, Imperial College London, London, United Kingdom
| | | | - Peter J Sterk
- AUMC, Department of Respiratory Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Skipp
- Centre for Proteomic Research, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom
| | - Ratko Djukanović
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Research, University of Southampton, Southampton, United Kingdom.
| | - Outi Vaarala
- Respiratory, Inflammation, Autoimmunity IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
112
|
Biomarkers and asthma management: analysis and potential applications. Curr Opin Allergy Clin Immunol 2019; 18:96-108. [PMID: 29389730 DOI: 10.1097/aci.0000000000000426] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Asthma features a high degree of heterogeneity in both pathophysiology and therapeutic response, resulting in many asthma patients being treated inadequately. Biomarkers indicative of underlying pathological processes could be used to identify disease subtypes, determine prognosis and to predict or monitor treatment response. However, the newly identified as well as more established biomarkers have different applications and limitations. RECENT FINDINGS Conventional markers for type 2-high asthma, such as blood eosinophils, fraction of exhaled nitric oxide, serum IgE and periostin, feature limited sensitivity and specificity despite their significant correlations. More distinctive models have been developed by combining biomarkers and/or using omics techniques. Recently, a model with a positive predictive value of 100% for identification of type 2-high asthma based on a combination of minimally invasive biomarkers was developed. SUMMARY Individualisation of asthma treatment regimens on the basis of biomarkers is necessary to improve asthma control. However, the suboptimal properties of currently available conventional biomarkers limit its clinical utility. Newly identified biomarkers and models based on combinations and/or omics analysis must be validated and standardised before they can be routinely applied in clinical practice. The development of robust biomarkers will allow development of more efficacious precision medicine-based treatment approaches for asthma.
Collapse
|
113
|
Reddel HK, Gerhardsson de Verdier M, Agustí A, Anderson G, Beasley R, Bel EH, Janson C, Make B, Martin RJ, Pavord I, Price D, Keen C, Gardev A, Rennard S, Sveréus A, Bansal AT, Brannman L, Karlsson N, Nuevo J, Nyberg F, Young SS, Vestbo J. Prospective observational study in patients with obstructive lung disease: NOVELTY design. ERJ Open Res 2019; 5:00036-2018. [PMID: 30723727 PMCID: PMC6355976 DOI: 10.1183/23120541.00036-2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) have overlapping clinical features and share pathobiological mechanisms but are often considered distinct disorders. Prospective, observational studies across asthma, COPD and asthma–COPD overlap are limited. NOVELTY is a global, prospective observational 3-year study enrolling ∼12 000 patients ≥12 years of age from primary and specialist clinical practices in 19 countries (ClinicalTrials.gov identifier: NCT02760329). NOVELTY's primary objectives are to describe patient characteristics, treatment patterns and disease burden over time, and to identify phenotypes and molecular endotypes associated with differential outcomes over time in patients with a diagnosis/suspected diagnosis of asthma and/or COPD. NOVELTY aims to recruit real-world patients, unlike clinical studies with restrictive inclusion/exclusion criteria. Data collected at yearly intervals include clinical assessments, spirometry, biospecimens, patient-reported outcomes (PROs) and healthcare utilisation (HCU). PROs and HCU will also be collected 3-monthly via internet/telephone. Data will be used to identify phenotypes and endotypes associated with different trajectories for symptom burden, clinical progression or remission and HCU. Results may allow patient classification across obstructive lung disease by clinical outcomes and biomarker profile, rather than by conventional diagnostic labels and severity categories. NOVELTY will provide a rich data source on obstructive lung disease, to help improve patient outcomes and aid novel drug development. NOVELTY is a global study to characterise patients with asthma and/or COPD and identify novel phenotypes and endotypeshttp://ow.ly/QFiH30n3IBF
Collapse
Affiliation(s)
- Helen K Reddel
- Woolcock Institute of Medical Research, University of Sydney, Sydney, Australia
| | | | - Alvar Agustí
- Respiratory Institute, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERES, Barcelona, Spain
| | - Gary Anderson
- Lung Health Research Centre, University of Melbourne, Melbourne, Australia
| | - Richard Beasley
- Medical Research Institute of New Zealand, Wellington, New Zealand
| | - Elisabeth H Bel
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Christer Janson
- Dept of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Barry Make
- National Jewish Health and University of Colorado Denver, Denver, CO, USA
| | - Richard J Martin
- National Jewish Health and University of Colorado Denver, Denver, CO, USA
| | - Ian Pavord
- Nuffield Dept of Medicine, University of Oxford, Oxford, UK
| | - David Price
- Observational and Pragmatic Research Institute, Singapore and Centre of Academic Primary Care, University of Aberdeen, Aberdeen, UK
| | - Christina Keen
- Early Clinical Development IMED Biotech Unit, AstraZeneca, Mölndal, Sweden
| | | | - Stephen Rennard
- Early Clinical Development, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Alecka Sveréus
- Respiratory TA, Global Medical Affairs, AstraZeneca, Mölndal, Sweden
| | | | | | - Niklas Karlsson
- Patient Reported Outcomes, Medical Evidence and Observational Research, Global Medical Affairs, AstraZeneca, Mölndal, Sweden
| | - Javier Nuevo
- Respiratory TA, Global Medical Affairs, AstraZeneca, Madrid, Spain
| | - Fredrik Nyberg
- Medical Evidence and Observational Research, Global Medical Affairs, AstraZeneca, Mölndal, Sweden
| | - Simon S Young
- Precision Medicine and Genomics, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Jørgen Vestbo
- School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
114
|
The impact of cigarette smoke exposure, COPD, or asthma status on ABC transporter gene expression in human airway epithelial cells. Sci Rep 2019; 9:153. [PMID: 30655622 PMCID: PMC6336805 DOI: 10.1038/s41598-018-36248-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
ABC transporters are conserved in prokaryotes and eukaryotes, with humans expressing 48 transporters divided into 7 classes (ABCA, ABCB, ABCC, ABCD, ABDE, ABCF, and ABCG). Throughout the human body, ABC transporters regulate cAMP levels, chloride secretion, lipid transport, and anti-oxidant responses. We used a bioinformatic approach complemented with in vitro experimental methods for validation of the 48 known human ABC transporters in airway epithelial cells using bronchial epithelial cell gene expression datasets available in NCBI GEO from well-characterized patient populations of healthy subjects and individuals that smoke cigarettes, or have been diagnosed with COPD or asthma, with validation performed in Calu-3 airway epithelial cells. Gene expression data demonstrate that ABC transporters are variably expressed in epithelial cells from different airway generations, regulated by cigarette smoke exposure (ABCA13, ABCB6, ABCC1, and ABCC3), and differentially expressed in individuals with COPD and asthma (ABCA13, ABCC1, ABCC2, ABCC9). An in vitro cell culture model of cigarette smoke exposure was able to recapitulate select observed in situ changes. Our work highlights select ABC transporter candidates of interest and a relevant in vitro model that will enable a deeper understanding of the contribution of ABC transporters in the respiratory mucosa in lung health and disease.
Collapse
|
115
|
Takeshima H, Horie M, Mikami Y, Makita K, Miyashita N, Matsuzaki H, Noguchi S, Urushiyama H, Hiraishi Y, Mitani A, Borok Z, Nagase T, Yamauchi Y. CISH is a negative regulator of IL-13-induced CCL26 production in lung fibroblasts. Allergol Int 2019; 68:101-109. [PMID: 30197185 DOI: 10.1016/j.alit.2018.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/02/2018] [Accepted: 08/05/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bronchial asthma is a chronic airway disease characterized by eosinophilic airway inflammation. Lung fibroblasts activated by IL-13 serve as important sources of chemokines, such as eotaxins, contributing to persistent eosinophilic inflammation. Src-homology 2-containing protein (CISH), belonging to the suppressor of cytokine signaling (SOCS) family, acts as a negative regulator of cytokine induction. The aim of this study was to elucidate the role of CISH in the production of eosinophil chemotactic chemokines in human lung fibroblasts. METHODS Normal human lung fibroblasts were stimulated by IL-13, and global gene expression profile was assessed by cDNA microarray. Expression changes and downstream of IL-13 signaling were evaluated by quantitative RT-PCR, ELISA or western blotting. Loss- and gain-of-function analyses of CISH were performed by small interfering RNA and vector overexpression, respectively. RESULTS Ingenuity pathway analysis revealed that IL-13 induced chemokine signaling, including the eotaxin family, while significantly suppressing IFN-α/β signaling. Among eight SOCS family members, CISH was most strongly induced by IL-13 via phosphorylation of signal transducer and activator of transcription 6 (STAT6). Loss- and gain-of-function studies demonstrated that CISH negatively regulated the expression of CCL26. CONCLUSIONS These findings suggest that CISH plays a key role in the eosinophilic inflammation associated with bronchial asthma by regulating IL-13-induced CCL26 production. Augmentation of CISH function could be a novel approach for treating eosinophilic inflammation in severe asthma.
Collapse
|
116
|
Abstract
The recent Lancet commission has highlighted that "asthma" should be used to describe a clinical syndrome of wheeze, breathlessness, chest tightness, and sometimes cough. The next step is to deconstruct the airway into components of fixed and variable airflow obstruction, inflammation, infection and altered cough reflex, setting the airway disease in the context of extra-pulmonary co-morbidities and social and environmental factors. The emphasis is always on delineating treatable traits, including variable airflow obstruction caused by airway smooth muscle constriction (treated with short- and long-acting β-2 agonists), eosinophilic airway inflammation (treated with inhaled corticosteroids) and chronic bacterial infection (treated with antibiotics with benefit if it is driving the disease). It is also important not to over-treat the untreatable, such as fixed airflow obstruction. These can all be determined using simple, non-invasive tests such as spirometry before and after acute administration of a bronchodilator (reversible airflow obstruction); peripheral blood eosinophil count, induced sputum, exhaled nitric oxide (airway eosinophilia); and sputum or cough swab culture (bacterial infection). Additionally, the pathophysiology of risk domains must be considered: these are risk of an asthma attack, risk of poor airway growth, and in pre-school children, risk of progression to eosinophilic school age asthma. Phenotyping the airway will allow more precise diagnosis and targeted treatment, but it is important to move to endotypes, especially in the era of increasing numbers of biologicals. Advances in -omics technology allow delineation of pathways, which will be particularly important in TH2 low eosinophilic asthma, and also pauci-inflammatory disease. It is very important to appreciate the difficulties of cluster analysis; a patient may have eosinophilic airway disease because of a steroid resistant endotype, because of non-adherence to basic treatment, and a surge in environmental allergen burden. Sophisticated -omics approaches will be reviewed in this manuscript, but currently they are not being used in clinical practice. However, even while they are being evaluated, management of the asthmas can and should be improved by considering the pathophysiologies of the different airway diseases lumped under that umbrella term, using simple, non-invasive tests which are readily available, and treating accordingly.
Collapse
Affiliation(s)
- Andrew Bush
- Departments of Paediatrics and Paediatric Respiratory Medicine, Royal Brompton Harefield NHS Foundation Trust and Imperial College, London, United Kingdom
| |
Collapse
|
117
|
Obeso D, Mera-Berriatua L, Rodríguez-Coira J, Rosace D, Fernández P, Martín-Antoniano IA, Santaolalla M, Marco Martín G, Chivato T, Fernández-Rivas M, Ramos T, Blanco C, Alvarado MI, Domínguez C, Angulo S, Barbas C, Barber D, Villaseñor A, Escribese MM. Multi-omics analysis points to altered platelet functions in severe food-associated respiratory allergy. Allergy 2018; 73:2137-2149. [PMID: 30028518 DOI: 10.1111/all.13563] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Prevalence and severity of allergic diseases have increased worldwide. To date, respiratory allergy phenotypes are not fully characterized and, along with inflammation progression, treatment is increasingly complex and expensive. Profilin sensitization constitutes a good model to study the progression of allergic inflammation. Our aim was to identify the underlying mechanisms and the associated biomarkers of this progression, focusing on severe phenotypes, using transcriptomics and metabolomics. METHODS Twenty-five subjects were included in the study. Plasma samples were analyzed using gas and liquid chromatography coupled to mass spectrometry (GC-MS and LC-MS, respectively). Individuals were classified in four groups-"nonallergic," "mild," "moderate," and "severe"-based on their clinical history, their response to an oral challenge test with profilin, and after a refinement using a mathematical metabolomic model. PBMCs were used for microarray analysis. RESULTS We found a set of transcripts and metabolites that were specific for the "severe" phenotype. By metabolomics, a decrease in carbohydrates and pyruvate and an increase in lactate were detected, suggesting aerobic glycolysis. Other metabolites were incremented in "severe" group: lysophospholipids, sphingosine-1-phosphate, sphinganine-1-phosphate, and lauric, myristic, palmitic, and oleic fatty acids. On the other hand, carnitines were decreased along severity. Significant transcripts in the "severe" group were found to be downregulated and were associated with platelet functions, protein synthesis, histone modification, and fatty acid metabolism. CONCLUSION We have found evidence that points to the association of severe allergic inflammation with platelet functions alteration, together with reduced protein synthesis, and switch of immune cells to aerobic glycolysis.
Collapse
Affiliation(s)
- David Obeso
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
- CEMBIO; Centro de Excelencia en Metabolómica y Bioanálisis; Facultad de Farmacia; Universidad San Pablo CEU; Madrid España
| | - Leticia Mera-Berriatua
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | - Juan Rodríguez-Coira
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
- CEMBIO; Centro de Excelencia en Metabolómica y Bioanálisis; Facultad de Farmacia; Universidad San Pablo CEU; Madrid España
| | - Domenico Rosace
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | - Paloma Fernández
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | - Isabel Adoración Martín-Antoniano
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
- Departamento de Ciencias Médicas Clínicas; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | | | | | - Tomás Chivato
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
- Departamento de Ciencias Médicas Clínicas; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | | | - Tania Ramos
- Hospital Universitario de La Princesa; Instituto de Investigación Sanitaria Princesa (IP); Madrid España
| | - Carlos Blanco
- Hospital Universitario de La Princesa; Instituto de Investigación Sanitaria Princesa (IP); Madrid España
| | | | | | - Santiago Angulo
- Departamento de Matemática Aplicada y Estadística; Universidad San Pablo CEU; Madrid España
| | - Coral Barbas
- CEMBIO; Centro de Excelencia en Metabolómica y Bioanálisis; Facultad de Farmacia; Universidad San Pablo CEU; Madrid España
| | - Domingo Barber
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | - Alma Villaseñor
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| | - María M. Escribese
- IMMA; Instituto de Medicina Molecular Aplicada; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
- Departamento de Ciencias Médicas Básicas; Facultad de Medicina; Universidad San Pablo CEU; Madrid España
| |
Collapse
|
118
|
Lu Y, Kared H, Tan SW, Becht E, Newell EW, Van Bever HPS, Ng TP, Larbi A. Dynamics of helper CD4 T cells during acute and stable allergic asthma. Mucosal Immunol 2018; 11:1640-1652. [PMID: 30087444 DOI: 10.1038/s41385-018-0057-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/14/2018] [Accepted: 06/12/2018] [Indexed: 02/04/2023]
Abstract
Asthma comprises heterogeneous clinical subtypes driven by diverse pathophysiological mechanisms. We characterized the modulation of the inflammatory environment with the phenotype, gene expression, and function of helper CD4 T cells among acutely exacerbated and stable asthma patients. Systemic Th2 immune deviation (IgE and Th2 cytokines) and inflammation (IL-6, CRP) were associated with increased Th17 cells during acute asthma. Th2/Th17 cell differentiation during acute asthma was regulated by the enhanced expression of transcription factors (c-MAF, IRF-4). The development of pathogenic Th2 cells during acute asthma was characterized by the secretion of inflammatory cytokines coupled with Th2 molecules and PPARγ expression. The acquisition of CD15S, CD39, CD101, and CCR4 contributed to the increased heterogeneity of Regulatory T cells during asthma. Two clusters were derived from above cytokines, CD4 T cell phenotypes, and clinical data. Cluster 1, characterized by high eosinophils, Th2 and ILC2 frequencies, and higher exacerbation rates, may represent Th2-high subtype. Cluster 2 represents a more complex subtype; it is constituted by higher neutrophils or Th17 frequencies, higher inhaled corticosteroids dose and poor asthma control. In conclusion, we characterized systematically and longitudinally Th2-high and non-Th2 asthma subtypes and the heterogeneity of CD4 T cells in stable and acute asthma.
Collapse
Affiliation(s)
- Yanxia Lu
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore. .,Department of Clinical Psychology and Psychiatry/School of Public Health, Zhejiang University College of Medicine, Hangzhou, China.
| | - Hassen Kared
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Etienne Becht
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hugo P S Van Bever
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tze Pin Ng
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Immunos Building at Biopolis, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Biology, Faculty of Science, University Tunis El Manar, Tunis, Tunisia.,Department of Medicine, Research Center on Aging, University of Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
119
|
Giovannini-Chami L, Paquet A, Sanfiorenzo C, Pons N, Cazareth J, Magnone V, Lebrigand K, Chevalier B, Vallauri A, Julia V, Marquette CH, Marcet B, Leroy S, Barbry P. The "one airway, one disease" concept in light of Th2 inflammation. Eur Respir J 2018; 52:13993003.00437-2018. [PMID: 30190271 DOI: 10.1183/13993003.00437-2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/13/2018] [Indexed: 12/19/2022]
Abstract
In line with the pathophysiological continuum described between nose and bronchus in allergic respiratory diseases, we assessed whether nasal epithelium could mirror the Type 2 T-helper cell (Th2) status of bronchial epithelium.Nasal and bronchial cells were collected by brushing from healthy controls (C, n=13), patients with allergic rhinitis and asthma (AR, n=12), and patients with isolated allergic rhinitis (R, n=14). Cellular composition was assessed by flow cytometry, gene expression was analysed by RNA sequencing and Th2, Type 17 T-helper cell (Th17) and interferon (IFN) signatures were derived from the literature.Infiltration by polymorphonuclear neutrophils (PMN) in the nose excluded 30% of the initial cohort. All bronchial samples from the AR group were Th2-high. The gene expression profile of nasal samples from the AR group correctly predicted the paired bronchial sample Th2 status in 71% of cases. Nevertheless, nasal cells did not appear to be a reliable surrogate for the Th2 response, in particular due to a more robust influence of the IFN response in 14 out of 26 nasal samples. The Th2 scores in the nose and bronchi correlated with mast cell count (both p<0.001) and number of sensitisations (p=0.006 and 0.002), while the Th17 scores correlated with PMN count (p=0.006 and 0.003).The large variability in nasal cell composition and type of inflammation restricts its use as a surrogate for assessing bronchial Th2 inflammation in AR patients.
Collapse
Affiliation(s)
- Lisa Giovannini-Chami
- Pediatric Pulmonology and Allergology Dept, Hôpitaux Pédiatriques de Nice CHU-Lenval, Université Côte d'Azur, Nice, France.,Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| | - Agnès Paquet
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| | - Céline Sanfiorenzo
- Pulmonology Dept, FHU Oncoage, CHU de Nice, Université Côte d'Azur, Nice, France
| | - Nicolas Pons
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Julie Cazareth
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Virginie Magnone
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Kévin Lebrigand
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Benoit Chevalier
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Ambre Vallauri
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Valérie Julia
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | | | - Brice Marcet
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France
| | - Sylvie Leroy
- Pulmonology Dept, FHU Oncoage, CHU de Nice, Université Côte d'Azur, Nice, France.,These authors contributed equally to this work
| | - Pascal Barbry
- Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Sophia Antipolis, France.,These authors contributed equally to this work
| |
Collapse
|
120
|
Gelfand EW, Schedel M. Molecular Endotypes Contribute to the Heterogeneity of Asthma. Immunol Allergy Clin North Am 2018; 38:655-665. [PMID: 30342586 DOI: 10.1016/j.iac.2018.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Diagnosis and management of asthma is commonly implemented based on clinical assessment. Although these nonmolecular biomarkers have been useful, limited resolution of the heterogeneity among asthmatic patients and little information regarding the underlying pathobiology of disease in individuals have been provided. Molecular endotying using global transcriptome expression profiling associated with clinical features of asthma has improved our understanding of disease mechanisms, risk assessment of asthma exacerbations, and treatment responses, especially in patients with type 2 inflammation. Further advances in establishing pathobiological subgroups, bioactive pathways, and true disease endotypes hold potential for a more personalized medical approach in asthmatic patients.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA.
| |
Collapse
|
121
|
Hurgobin B, de Jong E, Bosco A. Insights into respiratory disease through bioinformatics. Respirology 2018; 23:1117-1126. [PMID: 30218470 DOI: 10.1111/resp.13401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/18/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022]
Abstract
Respiratory diseases such as asthma, chronic obstructive pulmonary disease and lung cancer represent a critical area for medical research as millions of people are affected globally. The development of new strategies for treatment and/or prevention, and the identification of biomarkers for patient stratification and early detection of disease inception are essential to reducing the impact of lung diseases. The successful translation of research into clinical practice requires a detailed understanding of the underlying biology. In this regard, the advent of next-generation sequencing and mass spectrometry has led to the generation of an unprecedented amount of data spanning multiple layers of biological regulation (genome, epigenome, transcriptome, proteome, metabolome and microbiome). Dealing with this wealth of data requires sophisticated bioinformatics and statistical tools. Here, we review the basic concepts in bioinformatics and genomic data analysis and illustrate the application of these tools to further our understanding of lung diseases. We also highlight the potential for data integration of multi-omic profiles and computational drug repurposing to define disease subphenotypes and match them to targeted therapies, paving the way for personalized medicine.
Collapse
Affiliation(s)
- Bhavna Hurgobin
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Emma de Jong
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
122
|
Synergistic activation of pro-inflammatory type-2 CD8 + T lymphocytes by lipid mediators in severe eosinophilic asthma. Mucosal Immunol 2018; 11:1408-1419. [PMID: 29907870 PMCID: PMC6448764 DOI: 10.1038/s41385-018-0049-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 04/12/2018] [Accepted: 05/06/2018] [Indexed: 02/04/2023]
Abstract
Human type-2 CD8+ T cells are a cell population with potentially important roles in allergic disease. We investigated this in the context of severe asthma with persistent airway eosinophilia-a phenotype associated with high exacerbation risk and responsiveness to type-2 cytokine-targeted therapies. In two independent cohorts we show that, in contrast to Th2 cells, type-2 cytokine-secreting CD8+CRTH2+ (Tc2) cells are enriched in blood and airways in severe eosinophilic asthma. Concentrations of prostaglandin D2 (PGD2) and cysteinyl leukotriene E4 (LTE4) are also increased in the airways of the same group of patients. In vitro PGD2 and LTE4 function synergistically to trigger Tc2 cell recruitment and activation in a TCR-independent manner. These lipids regulate diverse genes in Tc2 cells inducing type-2 cytokines and many other pro-inflammatory cytokines and chemokines, which could contribute to eosinophilia. These findings are consistent with an important innate-like role for human Tc2 cells in severe eosinophilic asthma and suggest a potential target for therapeutic intervention in this and other diseases.
Collapse
|
123
|
Abstract
PURPOSE OF REVIEW Asthma is a heterogeneous disease consisting of different phenotypes that are driven by different mechanistic pathways. The purpose of this review is to emphasize the important role of precision medicine in asthma management. RECENT FINDINGS Despite asthma heterogeneity, the approach to management has been on the basis of disease severity, with the most severe patients reserved for the maximum treatments with corticosteroids and bronchodilators. At the severe end, the recent availability of biologic therapies in the form of anti-IgE (omalizumab) and anti-IL5 therapies (mepolizumab and reslizumab) has driven the adaptation of precision medicine. These therapies are reserved for severe asthma with defined either allergic or eosinophilic background, respectively. SUMMARY Unbiased definition of phenotypes or endotypes (which are phenotypes defined by mechanisms) is an important step towards the use of precision medicine in asthma. Although T2-high asthma has been defined with targets becoming available for treating allergic or eosinophilic asthma, the definition of non-T2 phenotypes remains a priority. Precision medicine is also dependent on the definition of biomarkers that can help differentiate between these phenotypes and pinpoint patients suitable for specific-targeted therapies. Thus, precision medicine links phenotypes (endotypes) to targeted treatments for better outcomes.
Collapse
|
124
|
Pité H, Morais-Almeida M, Rocha SM. Metabolomics in asthma: where do we stand? Curr Opin Pulm Med 2018; 24:94-103. [PMID: 29059088 DOI: 10.1097/mcp.0000000000000437] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Metabolomics has been used to uncover the metabolic signatures of asthma, both for biomarker identification and pathophysiologic mechanisms research. We aimed to review recent advances in this field, published since 2016, and discuss these findings implications to future research and application into clinical practice. RECENT FINDINGS Experimental asthma models and clinical studies in both children and adults supported independent metabolic signatures of asthma. Common reported pathways included purine, glycerophospholipid, glutathione, fatty acids, and arginine and proline metabolism. Metabolomics-based studies identified candidate biomarkers related to asthma severity and corticosteroid resistance, and supported the definition of the obesity-related phenotype at the molecular level. A systematic review with meta-analysis and recent prospective studies favored exhaled volatile organic compounds as one of the most promising biomarkers in asthma diagnosis and monitoring. SUMMARY Metabolomics has provided unique and novel insights into asthma profiling at the molecular level. Current challenges include procedures standardization and control of potentially confounding variables for external validation. Point-of-care technology developments bring metabolomics closer to clinical practice. In addition to biomarkers identification, relating metabolites to their biologic role will serve as critical foundations for understanding the biology underpinning asthma heterogeneity and for specific-targeted therapies. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Helena Pité
- Allergy Center, CUF Descobertas Hospital and CUF Infante Santo Hospital.,CEDOC, Chronic Diseases Research Center, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon
| | | | - Sílvia M Rocha
- Department of Chemistry & QOPNA, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
125
|
Translating Asthma: Dissecting the Role of Metabolomics, Genomics and Personalized Medicine. Indian J Pediatr 2018; 85:643-650. [PMID: 29185231 DOI: 10.1007/s12098-017-2520-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/27/2017] [Indexed: 01/26/2023]
Abstract
The management of asthma has largely stagnated over the last 25 years, but we are at the dawning of a new age wherein -omics technology can help us manage the disease objectively and rationally. Even in this new scientific age, getting the basics of asthma management right remains essential. The new technologies which can be applied to multiple biological samples include genomics (study of the genome), transcriptomics (gene transcription), lipidomics, proteomics and metabolomics (lipids, proteins and metabolites, respectively) and breathomics, using exhaled breath as a source of biomarkers, which is of particular interest in view of its non-invasive nature in pediatrics. Important applications will include the diagnosis of airways disease, including its components; the pathways driving airway pathology; monitoring the response to treatment; and measuring future risk (asthma attacks, poor lung growth trajectory). With the advent of a wide range of novel biologicals to treat asthma, -omics technology to personalize therapy will be especially important. The U-BIOPRED (Europe) and SARP (USA) groups have been most active in this field, especially using bronchoscopically obtained samples to perform cluster analyses to define new asthma endotypes. However, stability over time and consistency between investigators is imperfect. This is perhaps unsurprising; results of biomarker studies in asthma will be a composite of the underlying disease, the (variable) effects of adverse drivers such as allergen exposure and pollution, the effects of treatment, and the effects of adherence or otherwise to treatment. Ultimately, the aim should be an exhaled breath based tool with a rapid result that can be used as a routine in the clinic. However, at the moment, there are as yet no clinical applications in children of -omics technology.
Collapse
|
126
|
Abstract
The introduction of 16s ribosomal RNA sequencing as a nonculture technique has led to the discovery of the presence of microbiota in the lower airways of healthy individuals. These bacterial communities may originate from the mouth and nasopharynx or from the environment by inhalation. The microbial composition of the lower airways may be modulated by dietary factors, antibiotic therapy, and microbial infections, particularly in early life. In addition, circulatory products from gut microbiota may influence the lung microbiota to maintain mucosal immunity. Recent studies have revealed that, in asthma, the lower airway microbiota show reduced diversity and community composition that is linked to severity and inflammatory phenotype. There is also a greater prevalence of proteobacteria, including Haemophilus, in symptomatic asthma. Microbial dysbiosis may contribute to both the inception and progression of asthma in infants and children, and to corticosteroid resistance in asthma. A better understanding of the regulation of the lung and gut microbiota in asthma may pave the way for targeting microbiota to prevent and treat asthma.
Collapse
|
127
|
U-BIOPRED: evaluation of the value of a public-private partnership to industry. Drug Discov Today 2018; 23:1622-1634. [PMID: 29936248 DOI: 10.1016/j.drudis.2018.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/24/2018] [Accepted: 06/15/2018] [Indexed: 02/07/2023]
Abstract
Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) was initiated in the first year of the Innovative Medicines Initiative (IMI). It was an ambitious plan to tackle the understanding of asthma through an integration of clinical and multi-'omics approaches that necessitated the bringing together of industry, academic, and patient representatives because it was too large to be managed by any one of the partners in isolation. It was a novel experience for all concerned. In this review, we describe the main features of the U-BIOPRED experience from the industry perspective. We list some of the key advantages and learnings from the perspective of the authors, and also improvements that we feel could be made in future projects.
Collapse
|
128
|
Jevnikar Z, Östling J, Ax E, Calvén J, Thörn K, Israelsson E, Öberg L, Singhania A, Lau LCK, Wilson SJ, Ward JA, Chauhan A, Sousa AR, De Meulder B, Loza MJ, Baribaud F, Sterk PJ, Chung KF, Sun K, Guo Y, Adcock IM, Payne D, Dahlen B, Chanez P, Shaw DE, Krug N, Hohlfeld JM, Sandström T, Djukanovic R, James A, Hinks TSC, Howarth PH, Vaarala O, van Geest M, Olsson H. Epithelial IL-6 trans-signaling defines a new asthma phenotype with increased airway inflammation. J Allergy Clin Immunol 2018; 143:577-590. [PMID: 29902480 DOI: 10.1016/j.jaci.2018.05.026] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 04/15/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Although several studies link high levels of IL-6 and soluble IL-6 receptor (sIL-6R) to asthma severity and decreased lung function, the role of IL-6 trans-signaling (IL-6TS) in asthmatic patients is unclear. OBJECTIVE We sought to explore the association between epithelial IL-6TS pathway activation and molecular and clinical phenotypes in asthmatic patients. METHODS An IL-6TS gene signature obtained from air-liquid interface cultures of human bronchial epithelial cells stimulated with IL-6 and sIL-6R was used to stratify lung epithelial transcriptomic data (Unbiased Biomarkers in Prediction of Respiratory Disease Outcomes [U-BIOPRED] cohorts) by means of hierarchical clustering. IL-6TS-specific protein markers were used to stratify sputum biomarker data (Wessex cohort). Molecular phenotyping was based on transcriptional profiling of epithelial brushings, pathway analysis, and immunohistochemical analysis of bronchial biopsy specimens. RESULTS Activation of IL-6TS in air-liquid interface cultures reduced epithelial integrity and induced a specific gene signature enriched in genes associated with airway remodeling. The IL-6TS signature identified a subset of patients with IL-6TS-high asthma with increased epithelial expression of IL-6TS-inducible genes in the absence of systemic inflammation. The IL-6TS-high subset had an overrepresentation of frequent exacerbators, blood eosinophilia, and submucosal infiltration of T cells and macrophages. In bronchial brushings Toll-like receptor pathway genes were upregulated, whereas expression of cell junction genes was reduced. Sputum sIL-6R and IL-6 levels correlated with sputum markers of remodeling and innate immune activation, in particular YKL-40, matrix metalloproteinase 3, macrophage inflammatory protein 1β, IL-8, and IL-1β. CONCLUSIONS Local lung epithelial IL-6TS activation in the absence of type 2 airway inflammation defines a novel subset of asthmatic patients and might drive airway inflammation and epithelial dysfunction in these patients.
Collapse
Affiliation(s)
- Zala Jevnikar
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| | - Jörgen Östling
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Ax
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, Krefting Research Centre, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Calvén
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Kristofer Thörn
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Elisabeth Israelsson
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Lisa Öberg
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Akul Singhania
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom
| | - Laurie C K Lau
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom
| | - Susan J Wilson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Jonathan A Ward
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; Histochemistry Research Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anoop Chauhan
- Portsmouth Hospitals NHS Trust, Portsmouth, United Kingdom
| | - Ana R Sousa
- Discovery Medicine, GlaxoSmithKline, Brentford, United Kingdom
| | - Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CIRI UMR5308, CNRS-ENS-UCBL-INSERM, Université de Lyon, Lyon, France
| | | | | | - Peter J Sterk
- Department of Respiratory Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London UK & Royal Brompton Biomedical Research Unit at Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Kai Sun
- Department of Computing & Data Science Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Department of Computing & Data Science Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London UK & Royal Brompton Biomedical Research Unit at Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, London, United Kingdom
| | - Debbie Payne
- Centre for Integrated Genomic Medical Research, University of Manchester, Manchester, United Kingdom
| | - Barbro Dahlen
- Karolinska University Hospital & Centre for Allergy Research, Karolinska Institute, Stockholm, Sweden
| | | | - Dominick E Shaw
- Respiratory Biomedical Research Unit, University of Nottingham, Nottingham, United Kingdom
| | - Norbert Krug
- Fraunhofer Institute of Toxicology and Experimental Medicine, Member of the German Center for Lung Research, Hannover, Germany
| | - Jens M Hohlfeld
- Fraunhofer Institute of Toxicology and Experimental Medicine, Member of the German Center for Lung Research, Hannover, Germany; Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Thomas Sandström
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Ratko Djukanovic
- NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Anna James
- Experimental Asthma and Allergy Research, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Timothy S C Hinks
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom; Respiratory Medicine Unit, NDM Experimental Medicine, University of OxfordJohn Radcliffe Hospital, Oxford, United Kingdom
| | - Peter H Howarth
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton University Hospital, Southampton, United Kingdom; NIHR Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Outi Vaarala
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Marleen van Geest
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Henric Olsson
- Department of Bioscience, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | |
Collapse
|
129
|
Turner JH, Li P, Chandra RK. Mucus T helper 2 biomarkers predict chronic rhinosinusitis disease severity and prior surgical intervention. Int Forum Allergy Rhinol 2018; 8:1175-1183. [PMID: 29883053 DOI: 10.1002/alr.22160] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a diverse clinical syndrome with a heterogeneous pathophysiology. Early attempts to identify CRS endotypes and biomarkers have largely relied on analysis of surgically obtained tissue, thus limiting their practical utility. This study examined the ability of mucus T helper 2 (Th2) biomarkers to predict CRS disease severity and clinical characteristics. METHODS CRS (n = 90) and healthy control subjects (n = 17) were prospectively enrolled prior to surgical intervention and mucus levels of interleukin (IL)-4, IL-5, and IL-13 were determined using a multiplex cytometric bead assay. Data for relevant cytokines was then scaled, normalized, and later combined to develop standardized metrics indicative of Th2-associated inflammation. Th2-high and Th2-low subgroups were consequently identified and validated against factors associated with disease severity and clinical outcomes. RESULTS Mucus levels of IL-5 and IL-13 were elevated in CRS subjects compared to controls, while no significant difference was noted for IL-4. IL-5 and IL-13 high CRS were associated with worse objective measures of disease severity and greater rates of revision surgery. Similar relationships were noted for both cytokines when CRS with nasal polyps (CRSwNP) patients were analyzed separately. Th2-high CRS and Th2-low CRS were then categorized using a scaled IL-5/IL-13 metric. Th2-high CRS was characterized by an increased number of subjects with nasal polyps and comorbid asthma, and worse symptom and computed tomography (CT) scores. CONCLUSION The Th2-associated cytokines, IL-5 and IL-13, are detectable in sinonasal mucus and their levels can be used to define Th2-high and Th2-low CRS. Identification of Th2-high and Th2-low endotypes using mucus-based biomarkers could facilitate stratification of CRS subgroups and guide personalized therapies.
Collapse
Affiliation(s)
- Justin H Turner
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, TN
| | - Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, TN
| | - Rakesh K Chandra
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
130
|
Zeki AA, Elbadawi-Sidhu M. Innovations in asthma therapy: is there a role for inhaled statins? Expert Rev Respir Med 2018; 12:461-473. [PMID: 29575963 PMCID: PMC6018057 DOI: 10.1080/17476348.2018.1457437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/22/2018] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Asthma manifests as chronic airflow obstruction with persistent inflammation and airway hyperresponsiveness. The immunomodulatory and anti-inflammatory properties of the HMG-CoA reductase (HMGCR) inhibitors (a.k.a. statins), suggest a therapeutic role in chronic inflammatory lung diseases. However, despite positive laboratory investigations and promising epidemiological data, clinical trials using statins for the treatment of asthma have yielded conflicting results. Inadequate statin levels in the airway compartment could explain these findings. Areas covered: HMGCR is in the mevalonate (MA) pathway and MA signaling is fundamental to lung biology and asthma. This article will discuss clinical trials of oral statins in asthma, review lab investigations relevant to the systemic versus inhaled administration of statins, address the advantages and disadvantages of inhaled statins, and answer the question: is there a role for inhaled statins in the treatment of asthma? Expert commentary: If ongoing investigations show that oral administration of statins has no clear clinical benefits, then repurposing statins for delivery via inhalation is a logical next step. Inhalation of statins bypasses first-pass metabolism by the liver, and therefore, allows for delivery of significantly lower doses to the airways at greater potency. Statins could become the next major class of novel inhalers for the treatment of asthma.
Collapse
Affiliation(s)
- Amir A. Zeki
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, Davis, CA, USA
| | - Mona Elbadawi-Sidhu
- NIH West Coast Metabolomics Center, Genome and Biomedical Sciences Facility, University of California, Davis, CA, USA
| |
Collapse
|
131
|
Farzan N, Vijverberg SJ, Kabesch M, Sterk PJ, Maitland-van der Zee AH. The use of pharmacogenomics, epigenomics, and transcriptomics to improve childhood asthma management: Where do we stand? Pediatr Pulmonol 2018; 53:836-845. [PMID: 29493882 DOI: 10.1002/ppul.23976] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 02/01/2018] [Indexed: 01/11/2023]
Abstract
Asthma is a complex multifactorial disease and it is the most common chronic disease in children. There is a high variability in response to asthma treatment, even in patients with good adherence to maintenance treatment, and a correct inhalation technique. Distinct underlying disease mechanisms in childhood asthma might be the reason of this heterogeneity. A deeper knowledge of the underlying molecular mechanisms of asthma has led to the recent development of advanced and mechanism-based treatments such as biologicals. However, biologicals are recommended only for patients with specific asthma phenotypes who remain uncontrolled despite high dosages of conventional asthma treatment. One of the main unmet needs in their application is lack of clinically available biomarkers to individualize pediatric asthma management and guide treatment. Pharmacogenomics, epigenomics, and transcriptomics are three omics fields that are rapidly advancing and can provide tools to identify novel asthma mechanisms and biomarkers to guide treatment. Pharmacogenomics focuses on variants in the DNA, epigenomics studies heritable changes that do not involve changes in the DNA sequence but lead to alteration of gene expression, and transcriptomics investigates gene expression by studying the complete set of mRNA transcripts in a cell or a population of cells. Advances in high-throughput technologies and statistical tools together with well-phenotyped patient inclusion and collaborations between different centers will expand our knowledge of underlying molecular mechanisms involved in disease onset and progress. Furthermore, it could help to select and stratify appropriate therapeutic strategies for subgroups of patients and hopefully bring precision medicine to daily practice.
Collapse
Affiliation(s)
- Niloufar Farzan
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Susanne J Vijverberg
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Peter J Sterk
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Anke H Maitland-van der Zee
- Department of Respiratory Medicine, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
132
|
Takahashi K, Pavlidis S, Ng Kee Kwong F, Hoda U, Rossios C, Sun K, Loza M, Baribaud F, Chanez P, Fowler SJ, Horvath I, Montuschi P, Singer F, Musial J, Dahlen B, Dahlen SE, Krug N, Sandstrom T, Shaw DE, Lutter R, Bakke P, Fleming LJ, Howarth PH, Caruso M, Sousa AR, Corfield J, Auffray C, De Meulder B, Lefaudeux D, Djukanovic R, Sterk PJ, Guo Y, Adcock IM, Chung KF. Sputum proteomics and airway cell transcripts of current and ex-smokers with severe asthma in U-BIOPRED: an exploratory analysis. Eur Respir J 2018; 51:13993003.02173-2017. [PMID: 29650557 DOI: 10.1183/13993003.02173-2017] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 02/22/2018] [Indexed: 12/14/2022]
Abstract
Severe asthma patients with a significant smoking history have airflow obstruction with reported neutrophilia. We hypothesise that multi-omic analysis will enable the definition of smoking and ex-smoking severe asthma molecular phenotypes.The U-BIOPRED cohort of severe asthma patients, containing current-smokers (CSA), ex-smokers (ESA), nonsmokers and healthy nonsmokers was examined. Blood and sputum cell counts, fractional exhaled nitric oxide and spirometry were obtained. Exploratory proteomic analysis of sputum supernatants and transcriptomic analysis of bronchial brushings, biopsies and sputum cells was performed.Colony-stimulating factor (CSF)2 protein levels were increased in CSA sputum supernatants, with azurocidin 1, neutrophil elastase and CXCL8 upregulated in ESA. Phagocytosis and innate immune pathways were associated with neutrophilic inflammation in ESA. Gene set variation analysis of bronchial epithelial cell transcriptome from CSA showed enrichment of xenobiotic metabolism, oxidative stress and endoplasmic reticulum stress compared to other groups. CXCL5 and matrix metallopeptidase 12 genes were upregulated in ESA and the epithelial protective genes, mucin 2 and cystatin SN, were downregulated.Despite little difference in clinical characteristics, CSA were distinguishable from ESA subjects at the sputum proteomic level, with CSA patients having increased CSF2 expression and ESA patients showing sustained loss of epithelial barrier processes.
Collapse
Affiliation(s)
- Kentaro Takahashi
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK.,Research Centre for Allergy and Clinical Immunology, Asahi General Hospital, Matsudo, Japan
| | - Stelios Pavlidis
- Dept of Computing and Data Science Institute, Imperial College London, London, UK
| | - Francois Ng Kee Kwong
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Uruj Hoda
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Christos Rossios
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Kai Sun
- Dept of Computing and Data Science Institute, Imperial College London, London, UK
| | - Matthew Loza
- Janssen Research and Development, High Wycombe, UK
| | | | - Pascal Chanez
- Assistance Publique des Hôpitaux de Marseille, Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Marseille, France
| | - Steve J Fowler
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, University of Manchester and University Hospital of South Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | | | | | - Florian Singer
- Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jacek Musial
- Dept of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Barbro Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Eric Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Thomas Sandstrom
- Dept of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Dominic E Shaw
- Respiratory Research Unit, University of Nottingham, Nottingham, UK
| | - Rene Lutter
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Per Bakke
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Louise J Fleming
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Peter H Howarth
- NIHR Southampton Respiratory Biomedical Research Unit, Clinical and Experimental Sciences and Human Development and Health, Southampton, UK
| | - Massimo Caruso
- Dept Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GSK, Stockley Park, UK
| | - Julie Corfield
- AstraZeneca R&D, Molndal, Sweden.,Areteva R&D, Nottingham, UK
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Diane Lefaudeux
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Ratko Djukanovic
- NIHR Southampton Respiratory Biomedical Research Unit, Clinical and Experimental Sciences and Human Development and Health, Southampton, UK
| | - Peter J Sterk
- Dept of Clinical Science, University of Bergen, Bergen, Norway
| | - Yike Guo
- Dept of Computing and Data Science Institute, Imperial College London, London, UK
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK.,Dept of Computing and Data Science Institute, Imperial College London, London, UK
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK.,Dept of Computing and Data Science Institute, Imperial College London, London, UK
| | | |
Collapse
|
133
|
Hastie AT, Steele C, Dunaway CW, Moore WC, Rector BM, Ampleford E, Li H, Denlinger LC, Jarjour N, Meyers DA, Bleecker ER. Complex association patterns for inflammatory mediators in induced sputum from subjects with asthma. Clin Exp Allergy 2018. [PMID: 29520864 DOI: 10.1111/cea.13129] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND The release of various inflammatory mediators into the bronchial lumen is thought to reflect both the type and degree of airway inflammation, eosinophilic Th2, and Th9, or neutrophilic Th1, and Th17, in patients with asthma. AIMS We investigated whether cytokines and chemokines differed in sputum from subjects with more severe compared with milder asthma and whether unbiased factor analysis of cytokine and chemokine groupings indicates specific inflammatory pathways. METHODS Cell-free supernatants from induced sputum were obtained from subjects with a broad range of asthma severity (n = 158) and assessed using Milliplex® Cytokines/Chemokine kits I, II and III, measuring 75 individual proteins. Each cytokine, chemokine or growth factor concentration was examined for differences between asthma severity groups, for association with leucocyte counts, and by factor analysis. RESULTS Severe asthma subjects had 9 increased and 4 decreased proteins compared to mild asthma subjects and fewer differences compared to moderate asthma. Twenty-six mediators were significantly associated with an increasing single leucocyte type: 16 with neutrophils (3 interleukins [IL], 3 CC chemokines, 4 CXC chemokines, 4 growth factors, TNF-α and CX3CL1/Fractalkine); 5 with lymphocytes (IL-7, IL-16, IL-23, IFN-α2 and CCL4/MIP1β); IL-15 and CCL15/MIP1δ with macrophages; IL-5 with eosinophils; and IL-4 and TNFSF10/TRAIL with airway epithelial cells. Factor analysis grouped 43 cytokines, chemokines and growth factors which had no missing data onto the first 10 factors, containing mixes of Th1, Th2, Th9 and Th17 inflammatory and anti-inflammatory proteins. CONCLUSIONS Sputum cytokines, chemokines and growth factors were increased in severe asthma, primarily with increased neutrophils. Factor analysis identified complex inflammatory protein interactions, suggesting airway inflammation in asthma is characterized by overlapping immune pathways. Thus, focus on a single specific inflammatory mediator or pathway may limit understanding the complexity of inflammation underlying airway changes in asthma and selection of appropriate therapy.
Collapse
Affiliation(s)
- A T Hastie
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - C Steele
- Lung Immunology of Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C W Dunaway
- Lung Immunology of Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - W C Moore
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - B M Rector
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - E Ampleford
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - H Li
- University of Arizona College of Medicine, Tucson, AZ, USA
| | - L C Denlinger
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - N Jarjour
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - D A Meyers
- University of Arizona College of Medicine, Tucson, AZ, USA
| | - E R Bleecker
- University of Arizona College of Medicine, Tucson, AZ, USA
| | | |
Collapse
|
134
|
Singhania A, Wallington JC, Smith CG, Horowitz D, Staples KJ, Howarth PH, Gadola SD, Djukanović R, Woelk CH, Hinks TSC. Multitissue Transcriptomics Delineates the Diversity of Airway T Cell Functions in Asthma. Am J Respir Cell Mol Biol 2018; 58:261-270. [PMID: 28933920 DOI: 10.1165/rcmb.2017-0162oc] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Asthma arises from the complex interplay of inflammatory pathways in diverse cell types and tissues. We sought to undertake a comprehensive transcriptomic assessment of the epithelium and airway T cells that remain understudied in asthma and investigate interactions between multiple cells and tissues. Epithelial brushings and flow-sorted CD3+ T cells from sputum and BAL were obtained from healthy subjects (n = 19) and patients with asthma (mild, moderate, and severe asthma; n = 46). Gene expression was assessed using Affymetrix HT HG-U133+ PM GeneChips, and results were validated by real-time quantitative PCR. In the epithelium, IL-13 response genes (POSTN, SERPINB2, and CLCA1), mast cell mediators (CPA3 and TPSAB1), inducible nitric oxide synthase, and cystatins (CST1, CST2, and CST4) were upregulated in mild asthma, but, except for cystatins, were suppressed by corticosteroids in moderate asthma. In severe asthma-with predominantly neutrophilic phenotype-several distinct processes were upregulated, including neutrophilia (TCN1 and MMP9), mucins, and oxidative stress responses. The majority of the disease signature was evident in sputum T cells in severe asthma, where 267 genes were differentially regulated compared with health, highlighting compartmentalization of inflammation. This signature included IL-17-inducible chemokines (CXCL1, CXCL2, CXCL3, IL8, and CSF3) and chemoattractants for neutrophils (IL8, CCL3, and LGALS3), T cells, and monocytes. A protein interaction network in severe asthma highlighted signatures of responses to bacterial infections across tissues (CEACAM5, CD14, and TLR2), including Toll-like receptor signaling. In conclusion, the activation of innate immune pathways in the airways suggests that activated T cells may be driving neutrophilic inflammation and steroid-insensitive IL-17 response in severe asthma.
Collapse
Affiliation(s)
- Akul Singhania
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Joshua C Wallington
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Caroline G Smith
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Daniel Horowitz
- 3 Janssen Research and Development, High Wycombe, Buckinghamshire, United Kingdom
| | - Karl J Staples
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Peter H Howarth
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Stephan D Gadola
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,4 Roche, F. Hoffman-La Roche AG, Konzern-Hauptsitz, Basel, Switzerland; and
| | - Ratko Djukanović
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Christopher H Woelk
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Timothy S C Hinks
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom.,5 Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, United Kingdom
| |
Collapse
|
135
|
Abstract
Asthma-one of the most common chronic, non-communicable diseases in children and adults-is characterised by variable respiratory symptoms and variable airflow limitation. Asthma is a consequence of complex gene-environment interactions, with heterogeneity in clinical presentation and the type and intensity of airway inflammation and remodelling. The goal of asthma treatment is to achieve good asthma control-ie, to minimise symptom burden and risk of exacerbations. Anti-inflammatory and bronchodilator treatments are the mainstay of asthma therapy and are used in a stepwise approach. Pharmacological treatment is based on a cycle of assessment and re-evaluation of symptom control, risk factors, comorbidities, side-effects, and patient satisfaction by means of shared decisions. Asthma is classed as severe when requiring high-intensity treatment to keep it under control, or if it remains uncontrolled despite treatment. New biological therapies for treatment of severe asthma, together with developments in biomarkers, present opportunities for phenotype-specific interventions and realisation of more personalised treatment. In this Seminar, we provide a clinically focused overview of asthma, including epidemiology, pathophysiology, clinical diagnosis, asthma phenotypes, severe asthma, acute exacerbations, and clinical management of disease in adults and children older than 5 years. Emerging therapies, controversies, and uncertainties in asthma management are also discussed.
Collapse
Affiliation(s)
- Alberto Papi
- Research Centre on Asthma and COPD, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.
| | - Christopher Brightling
- Institute for Lung Health, Leicester National Institute for Health Research Biomedical Research Centre, Department of Infection, Immunity, and Inflammation, University of Leicester and University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Søren E Pedersen
- Department of Paediatrics, University of Southern Denmark, Kolding Hospital, Kolding, Denmark
| | - Helen K Reddel
- Clinical Management Group and NHMRC Centre of Research Excellence in Severe Asthma, Woolcock Institute of Medical Research, University of Sydney, NSW, Australia
| |
Collapse
|
136
|
Turner JH, Chandra RK, Li P, Bonnet K, Schlundt DG. Identification of clinically relevant chronic rhinosinusitis endotypes using cluster analysis of mucus cytokines. J Allergy Clin Immunol 2018; 141:1895-1897.e7. [PMID: 29452200 DOI: 10.1016/j.jaci.2018.02.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/25/2018] [Accepted: 02/05/2018] [Indexed: 02/03/2023]
Affiliation(s)
- Justin H Turner
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tenn.
| | - Rakesh K Chandra
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Kemberlee Bonnet
- Department of Psychological Sciences, Vanderbilt University School of Arts and Sciences, Nashville, Tenn; Vanderbilt Center for Qualitative Research, Nashville, Tenn
| | - David G Schlundt
- Department of Psychological Sciences, Vanderbilt University School of Arts and Sciences, Nashville, Tenn; Vanderbilt Center for Qualitative Research, Nashville, Tenn
| |
Collapse
|
137
|
Kuo CHS, Liu CY, Pavlidis S, Lo YL, Wang YW, Chen CH, Ko HW, Chung FT, Lin TY, Wang TY, Lee KY, Guo YK, Wang TH, Yang CT. Unique Immune Gene Expression Patterns in Bronchoalveolar Lavage and Tumor Adjacent Non-Neoplastic Lung Tissue in Non-Small Cell Lung Cancer. Front Immunol 2018; 9:232. [PMID: 29483918 PMCID: PMC5816075 DOI: 10.3389/fimmu.2018.00232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/26/2018] [Indexed: 12/13/2022] Open
Abstract
Background The immune cells in the local environments surrounding non-small cell lung cancer (NSCLC) implicate the balance of pro- and antitumor immunity; however, their transcriptomic profiles remain poorly understood. Methods A transcriptomic microarray study of bronchoalveolar lavage (BAL) cells harvested from tumor-bearing lung segments was performed in a discovery group. The findings were validated (1) in published microarray datasets, (2) in an independent group by RT-qPCR, and (3) in non-diseased and tumor adjacent non-neoplastic lung tissue by immunohistochemistry and in BAL cell lysates by immunoblotting. Result The differential expression of 129 genes was identified in the discovery group. These genes revealed functional enrichment in Fc gamma receptor-dependent phagocytosis and circulating immunoglobulin complex among others. Microarray datasets analysis (n = 607) showed that gene expression of BAL cells of tumor-bearing lung segment was also the unique transcriptomic profile of tumor adjacent non-neoplastic lung of early stage NSCLC and a significantly gradient increase of immunoglobulin genes’ expression for non-diseased lungs, tumor adjacent non-neoplastic lungs, and tumors was identified (ANOVA, p < 2 × 10−16). A 53-gene signature was determined with significant correlation with inhibitory checkpoint PDCD1 (r = 0.59, p = 0.0078) among others, where the nine top genes including IGJ and IGKC were RT-qPCR validated with high diagnostic performance (AUC: 0.920, 95% CI: 0.831–0.985, p = 2.98 × 10−7). Increased staining and expression of IGKC revealed by immunohistochemistry and immunoblotting in tumor adjacent non-neoplastic lung tissues (Wilcoxon signed-rank test, p < 0.001) and in BAL cell lysates (p < 0.01) of NSCLC, respectively, were noted. Conclusion The BAL cells of tumor-bearing lung segments and tumor adjacent non-neoplastic lung tissues present a unique gene expression characterized by IGKC in relation to inhibitory checkpoints. Further study of humoral immune responses to NSCLC is warranted.
Collapse
Affiliation(s)
- Chih-Hsi Scott Kuo
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Chien-Ying Liu
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Stelios Pavlidis
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Yu-Lun Lo
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yen-Wen Wang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Hung Chen
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - How-Wen Ko
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Fu-Tsai Chung
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tin-Yu Lin
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Tsai-Yu Wang
- Division of Airway Diseases, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Kang-Yun Lee
- Division of Thoracic Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Yi-Ke Guo
- Department of Computing, Imperial College London, Data Science Institute, London, United Kingdom
| | - Tzu-Hao Wang
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Cheng-Ta Yang
- Division of Lung Cancer and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
138
|
Beghé B, Fabbri LM, Contoli M, Papi A. Update in Asthma 2016. Am J Respir Crit Care Med 2017; 196:548-557. [PMID: 28530112 DOI: 10.1164/rccm.201702-0318up] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Bianca Beghé
- 1 Department of Medical and Surgical Sciences, University of Modena Reggio Emilia, Modena, Italy
| | - Leonardo M Fabbri
- 2 Research Centre on Asthma and Chronic Obstructive Pulmonary Disease, Department of Medical Sciences, University of Ferrara, Ferrara, Italy; and.,3 Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marco Contoli
- 2 Research Centre on Asthma and Chronic Obstructive Pulmonary Disease, Department of Medical Sciences, University of Ferrara, Ferrara, Italy; and
| | - Alberto Papi
- 2 Research Centre on Asthma and Chronic Obstructive Pulmonary Disease, Department of Medical Sciences, University of Ferrara, Ferrara, Italy; and
| |
Collapse
|
139
|
Modena BD, Wenzel SE. Consistency of T2 Gene Signatures in Severe Asthma. Key to Effective Treatments or Merely the Tip of the Iceberg? Am J Respir Crit Care Med 2017; 195:411-412. [PMID: 28199166 DOI: 10.1164/rccm.201609-1854ed] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Brian D Modena
- 1 Department of Molecular and Experimental Medicine The Scripps Research Institute La Jolla, California.,2 Division of Allergy, Asthma, and Immunology Scripps Health San Diego, California and
| | - Sally E Wenzel
- 3 Division of Pulmonary, Allergy, and Critical Care Medicine University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center Pittsburgh, Pennsylvania
| |
Collapse
|
140
|
Affiliation(s)
- Elliot Israel
- From the Pulmonary and Critical Care Medicine Division-Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston (E.I.); and the Clinical Management Group and Centre for Research Excellence in Severe Asthma, Woolcock Institute of Medical Research, University of Sydney, Sydney (H.K.R.)
| | - Helen K Reddel
- From the Pulmonary and Critical Care Medicine Division-Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston (E.I.); and the Clinical Management Group and Centre for Research Excellence in Severe Asthma, Woolcock Institute of Medical Research, University of Sydney, Sydney (H.K.R.)
| |
Collapse
|
141
|
Gelfand EW, Joetham A, Wang M, Takeda K, Schedel M. Spectrum of T-lymphocyte activities regulating allergic lung inflammation. Immunol Rev 2017; 278:63-86. [PMID: 28658551 PMCID: PMC5501488 DOI: 10.1111/imr.12561] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite advances in the treatment of asthma, optimization of symptom control remains an unmet need in many patients. These patients, labeled severe asthma, are responsible for a substantial fraction of the disease burden. In these patients, research is needed to define the cellular and molecular pathways contributing to disease which in large part are refractory to corticosteroid treatment. The causes of steroid-resistant asthma are multifactorial and result from complex interactions of genetics, environmental factors, and innate and adaptive immunity. Adaptive immunity, addressed here, integrates the activities of distinct T-cell subsets and by definition is dynamic and responsive to an ever-changing environment and the influences of epigenetic modifications. These T-cell subsets exhibit different susceptibilities to the actions of corticosteroids and, in some, corticosteroids enhance their functional activation. Moreover, these subsets are not fixed in lineage differentiation but can undergo transcriptional reprogramming in a bidirectional manner between protective and pathogenic effector states. Together, these factors contribute to asthma heterogeneity between patients but also in the same patient at different stages of their disease. Only by carefully defining mechanistic pathways, delineating their sensitivity to corticosteroids, and determining the balance between regulatory and effector pathways will precision medicine become a reality with selective and effective application of targeted therapies.
Collapse
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, USA
| |
Collapse
|
142
|
Abstract
INTRODUCTION Severe therapy-resistant asthma is an area where there has been recent advances in understanding that is percolating into improvements in management. Areas covered: This review covers the recent definition and approach to the diagnosis of severe asthma and its differentiation from difficult-to-treat asthma. The recent advances in phenotyping severe asthma and in ensuing changes in management approaches together with the introduction of new therapies are covered from a review of the recent literature. Expert commentary: After ascertaining the diagnosis of severe asthma, patients need to be treated adequately with existing therapies. The management approach to severe asthma now comprises of a phenotyping step for the definition of either an allergic or eosinophilic severe asthma for which targeted therapies are currently available. This will lead to a precision medicine approach to the management of severe asthma.
Collapse
Affiliation(s)
- Kian Fan Chung
- a Airways Disease , National Heart & Lung Institute, Imperial College London , London , UK.,b Biomedical Research Unit , Royal Brompton & Harefield NHS Trust , London , UK
| |
Collapse
|
143
|
Fricker M, Heaney LG, Upham JW. Can biomarkers help us hit targets in difficult-to-treat asthma? Respirology 2017; 22:430-442. [PMID: 28248008 DOI: 10.1111/resp.13014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 01/19/2017] [Accepted: 01/21/2017] [Indexed: 12/16/2022]
Abstract
Biomarkers may be a key foundation for the precision medicine of the future. In this article, we review current knowledge regarding biomarkers in difficult-to-treat asthma and their ability to guide the use of both conventional asthma therapies and novel (targeted) therapies. Biomarkers (as measured by tests including prednisolone and cortisol assays and the fractional exhaled nitric oxide (NO) suppression test) show promise in the assessment and management of non-adherence to inhaled and oral corticosteroids. Multiple markers of type 2 inflammation have been developed, including eosinophils in sputum and blood, exhaled NO, serum IgE and periostin. Although these show potential in guiding the selection of novel interventions for refractory type 2 inflammation in asthma, and in determining if the desired response is being achieved, it is becoming clear that different biomarkers reflect distinct components of the complex type 2 inflammatory pathways. Less progress has been made in identifying biomarkers for use in difficult-to-treat asthma that is not associated with type 2 inflammation. The future is likely to see further biomarker discovery, direct measurements of individual cytokines rather than surrogates of their activity and the increasing use of biomarkers in combination. If the promise of biomarkers is to be fulfilled, they will need to provide useful information that aids clinical decision-making, rather than being 'just another test' for clinicians to order.
Collapse
Affiliation(s)
- Michael Fricker
- Centre of Excellence in Severe Asthma, School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales, Australia
| | - Liam G Heaney
- Centre for Experimental Medicine, Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - John W Upham
- Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.,Department of Respiratory and Sleep Medicine, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
144
|
Agache I, Rogozea L. Asthma Biomarkers: Do They Bring Precision Medicine Closer to the Clinic? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2017; 9:466-476. [PMID: 28913985 PMCID: PMC5603474 DOI: 10.4168/aair.2017.9.6.466] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 03/03/2017] [Accepted: 03/13/2017] [Indexed: 12/11/2022]
Abstract
Measurement of biomarkers has been incorporated within clinical research of asthma to characterize the population and to associate the disease with environmental and therapeutic effects. Regrettably, at present, there are no specific biomarkers, none is validated or qualified, and endotype-driven choices overlap. Biomarkers have not yet reached clinical practice and are not included in current asthma guidelines. Last but not least, the choice of the outcome upholding the value of the biomarkers is extremely difficult, since it has to reflect the mechanistic intervention while being relevant to both the disease and the particular person. On the verge of a new age of asthma healthcare standard, we must embrace and adapt to the key drivers of change. Disease endotypes, biomarkers, and precision medicine represent an emerging model of patient care building on large-scale biologic databases, omics and diverse cellular assays, health information technology, and computational tools for analyzing sizable sets of data. A profound transformation of clinical and research pattern from population to individual risk and from investigator-imposed subjective disease clustering (hypothesis driven) to unbiased, data-driven models is facilitated by the endotype/biomarker-driven approach.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University of Brasov, Brasov, Romania.
| | - Liliana Rogozea
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University of Brasov, Brasov, Romania
| |
Collapse
|
145
|
Bush A. Persistent Bacterial Bronchitis: Time to Venture beyond the Umbrella. Front Pediatr 2017; 5:264. [PMID: 29322037 PMCID: PMC5732151 DOI: 10.3389/fped.2017.00264] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 11/27/2017] [Indexed: 01/23/2023] Open
Abstract
Chronic cough in children is common and frequently mismanaged. In the past, cough was diagnosed as asthma and inappropriate asthma therapies prescribed and escalated. It has been realized that persistent bacterial bronchitis (PBB) is a common cause of wet cough and responds to oral antibiotics. The initial definition comprised a history of chronic wet cough, positive bronchoalveolar (BAL) cultures for a respiratory pathogen and response to a 2-week course of oral amoxicillin-clavulanic acid. This is now termed PBB-micro; PBB-clinical eliminates the need for BAL. PBB-extended is PBB-micro or PBB-clinical but resolution necessitating 4 weeks of antibiotics; and recurrent PBB is >3 attacks of PBB-micro or-clinical/year. However, the airway has only a limited range of responses to chronic inflammation and infection, and neutrophilic airway disease is seen in many other conditions, such as cystic fibrosis and primary ciliary dyskinesia, both chronic suppurative lung disease endotypes, whose recognition has led to huge scientific and clinical advances. There is an urgent need to extend endotyping into PBB, especially PBB-recurrent. We need to move from associative studies and, in particular, deploy sophisticated modern -omics technologies and systems biology, rather as has been done in the context of asthma in U-BIOPRED. In summary, the use of the term PBB has done signal service in pointing us away from prescribing asthma therapies to children with infected airways, but we now need to move beyond a simple description to teasing out underlying endotypes.
Collapse
Affiliation(s)
- Andrew Bush
- Imperial College London, London, United Kingdom.,National Heart and Lung Institute, London, United Kingdom.,Royal Brompton Harefield NHS Foundation Trust, London, United Kingdom
| |
Collapse
|