1
|
Wilson GE, Knight J, Liu Q, Shelar A, Stewart E, Wang X, Yan X, Sanders J, Visness C, Gill M, Gruchalla R, Liu AH, Kattan M, Khurana Hershey GK, Togias A, Becker PM, Altman MC, Busse WW, Jackson DJ, Montgomery RR, Chupp GL. Activated sputum eosinophils associated with exacerbations in children on mepolizumab. J Allergy Clin Immunol 2024; 154:297-307.e13. [PMID: 38485057 PMCID: PMC11305967 DOI: 10.1016/j.jaci.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Accepted: 01/30/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND MUPPITS-2 was a randomized, placebo-controlled clinical trial that demonstrated mepolizumab (anti-IL-5) reduced exacerbations and blood and airway eosinophils in urban children with severe eosinophilic asthma. Despite this reduction in eosinophilia, exacerbation risk persisted in certain patients treated with mepolizumab. This raises the possibility that subpopulations of airway eosinophils exist that contribute to breakthrough exacerbations. OBJECTIVE We aimed to determine the effect of mepolizumab on airway eosinophils in childhood asthma. METHODS Sputum samples were obtained from 53 MUPPITS-2 participants. Airway eosinophils were characterized using mass cytometry and grouped into subpopulations using unsupervised clustering analyses of 38 surface and intracellular markers. Differences in frequency and immunophenotype of sputum eosinophil subpopulations were assessed based on treatment arm and frequency of exacerbations. RESULTS Median sputum eosinophils were significantly lower among participants treated with mepolizumab compared with placebo (58% lower, 0.35% difference [95% CI 0.01, 0.74], P = .04). Clustering analysis identified 3 subpopulations of sputum eosinophils with varied expression of CD62L. CD62Lint and CD62Lhi eosinophils exhibited significantly elevated activation marker and eosinophil peroxidase expression, respectively. In mepolizumab-treated participants, CD62Lint and CD62Lhi eosinophils were more abundant in participants who experienced exacerbations than in those who did not (100% higher for CD62Lint, 0.04% difference [95% CI 0.0, 0.13], P = .04; 93% higher for CD62Lhi, 0.21% difference [95% CI 0.0, 0.77], P = .04). CONCLUSIONS Children with eosinophilic asthma treated with mepolizumab had significantly lower sputum eosinophils. However, CD62Lint and CD62Lhi eosinophils were significantly elevated in children on mepolizumab who had exacerbations, suggesting that eosinophil subpopulations exist that contribute to exacerbations despite anti-IL-5 treatment.
Collapse
Affiliation(s)
- Gabriella E Wilson
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - James Knight
- Department of Genetics and Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Conn
| | - Qing Liu
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Ashish Shelar
- Department of Genetics and Yale Center for Genome Analysis, Yale School of Medicine, New Haven, Conn
| | - Emma Stewart
- Committee on Immunology, University of Chicago, Chicago, Ill
| | - Xiaomei Wang
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Xiting Yan
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | | | | | - Michelle Gill
- Department of Pediatric Infectious Diseases, Washington University in St Louis School of Medicine, St Louis, Mo
| | - Rebecca Gruchalla
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Andrew H Liu
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, Colo
| | - Meyer Kattan
- Department of Pediatric Pulmonology, Columbia University Irving Medical Center, New York, NY
| | | | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Patrice M Becker
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | | | - William W Busse
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn
| | - Geoffrey L Chupp
- Department of Internal Medicine, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
2
|
Castillo JA, Plaza V, Rodrigo G, Juliá B, Picado C, Fernández C, Mullol J. Chronic rhinosinusitis with nasal polyps and allergic rhinitis as different multimorbid treatable traits in asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100134. [PMID: 37781668 PMCID: PMC10510007 DOI: 10.1016/j.jacig.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 10/03/2023]
Abstract
Background Respiratory multimorbidities are linked to asthma, such as allergic rhinitis (AR) with early allergic asthma and chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) with late nonallergic asthma. Objective Our aim was to investigate the association of asthma severity and control with specific upper airway phenotypes. Method Patients with asthma were prospectively recruited from 23 pulmonology and ear, nose, and throat clinics. Asthma severity and control, as well as upper airway comorbidities (AR and non-AR [NAR], CRSwNP, and CRS without nasal polyps [CRSsNP]) were assessed according to international consensus guidelines definitions. Results A total of 492 asthmatic patients were included. Half of the asthmatic patients (49.6%) had associated rhinitis (37.0% had AR and 12.6% had NAR) and 36.2% had CRS (16.7% had CRSsNP and 19.5% had CRSwNP), whereas 14.2% had no sinonasal symptoms. Most cases of AR (78%) and NAR (84%) were present in patients with mild-to-moderate asthma, whereas CRSwNP was more frequent in patients with severe asthma (35% [P < .001]), mainly nonatopic asthma (44% [P < .001]). Patients with severe asthma with CRSwNP had worse asthma control, which was correlated (r = 0.249 [P = .034]) with sinus occupancy. Multiple logistic regression analysis showed that late-onset asthma, intolerance of aspirin and/or nonsteroidal anti-inflammatory drugs, and CRSwNP were independently associated with severe asthma. Conclusion Severe asthma is associated with CRSwNP, with sinus occupancy affecting asthma control. This study has identified 2 main different upper airway treatable traits, AR and CRSwNP, which need further evaluation to improve management and control of patients with asthma.
Collapse
Affiliation(s)
- José Antonio Castillo
- Pneumology Department, Hospital Universitari Dexeus, Barcelona, Spain
- CIBER of Respiratory Diseases, Spain
- Group of Rhinitis, Rhinosinusitis, and Nasal Polyps, Area of Asthma, SEPAR, Spain
| | - Vicente Plaza
- Pneumology Department, Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Gustavo Rodrigo
- Emergency Departament, Hospital Central de las Fuerzas Armadas, Montevideo, Uruguay
| | | | - César Picado
- CIBER of Respiratory Diseases, Spain
- Pneumology Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Cristina Fernández
- Preventive Medicine Department, Complejo Hospitalario Universitario de Santiago de Compostela, Instituto de Investigación Sanitaria Santiago de Compostela, Fundación IMAS, Santiago de Compostela, Spain
| | - Joaquim Mullol
- CIBER of Respiratory Diseases, Spain
- Group of Rhinitis, Rhinosinusitis, and Nasal Polyps, Area of Asthma, SEPAR, Spain
- Clinical and Experimental Respiratory Immunoallergy, IDIBAPS & Rhinology Unite and Smell Clinic, ENT Department, Hospital Clínic Barcelona, Universitat de Barcelona, Spain
| |
Collapse
|
3
|
Gonzalez-Uribe V, Romero-Tapia SJ, Castro-Rodriguez JA. Asthma Phenotypes in the Era of Personalized Medicine. J Clin Med 2023; 12:6207. [PMID: 37834850 PMCID: PMC10573947 DOI: 10.3390/jcm12196207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Asthma is a widespread disease affecting approximately 300-million people globally. This condition leads to significant morbidity, mortality, and economic strain worldwide. Recent clinical and laboratory research advancements have illuminated the immunological factors contributing to asthma. As of now, asthma is understood to be a heterogeneous disease. Personalized medicine involves categorizing asthma by its endotypes, linking observable characteristics to specific immunological mechanisms. Identifying these endotypic mechanisms is paramount in accurately profiling patients and tailoring therapeutic approaches using innovative biological agents targeting distinct immune pathways. This article presents a synopsis of the key immunological mechanisms implicated in the pathogenesis and manifestation of the disease's phenotypic traits and individualized treatments for severe asthma subtypes.
Collapse
Affiliation(s)
- Victor Gonzalez-Uribe
- Alergia e Inmunología Clínica, Hospital Infantil de México Federico Gómez, Ciudad de Mexico 06720, Mexico;
- Facultad Mexicana de Medicina, Universidad La Salle México, Ciudad de Mexico 14000, Mexico
| | - Sergio J. Romero-Tapia
- Health Sciences Academic Division (DACS), Universidad Juárez Autónoma de Tabasco, Villahermosa 86040, Mexico;
| | - Jose A. Castro-Rodriguez
- Department of Pediatric Pulmonology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| |
Collapse
|
4
|
Perez MF, Yurieva M, Poddutoori S, Mortensen EM, Crotty Alexander LE, Williams A. Transcriptomic responses in the blood and sputum of cigarette smokers compared to e-cigarette vapers. Respir Res 2023; 24:134. [PMID: 37208747 PMCID: PMC10196320 DOI: 10.1186/s12931-023-02438-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 04/27/2023] [Indexed: 05/21/2023] Open
Abstract
RATIONALE Electronic (e)-cigarettes are popular among youth and cigarette smokers attempting to quit. Studies to date have focused on the utility of e-cigarettes as a smoking cessation tool, but the biological effects are largely unknown. OBJECTIVES To identify transcriptomic differences in the blood and sputum of e-cigarette users compared to conventional cigarettes smokers and healthy controls and describe biological pathways affected by these tobacco products. METHODS Cross-sectional analysis of whole blood and sputum RNA-sequencing data from 8 smokers, 9 e-cigarette users (e-cigs) and 4 controls. Weighted gene co-network analysis (WGCNA) identified gene module associations. Ingenuity Pathway Analysis (IPA) identified canonical pathways associated with tobacco products. MAIN RESULTS In blood, a three-group comparison showed 16 differentially expressed genes (DEGs); pair-wise comparison showed 7 DEGs between e-cigs and controls, 35 DEGs between smokers and controls, and 13 DEGs between smokers and e-cigs. In sputum, 438 DEGs were in the three-group comparison. In pair-wise comparisons, there were 2 DEGs between e-cigs and controls, 270 DEGs between smokers and controls, and 468 DEGs between smokers and e-cigs. Only 2 genes in the smokers vs. control comparison overlapped between blood and sputum. Most gene modules identified through WGCNA associated with tobacco product exposures also were associated with cotinine and exhaled CO levels. IPA showed more canonical pathways altered by conventional cigarette smoking than by e-cigarette use. CONCLUSION Cigarette smoking and e-cigarette use led to transcriptomic changes in both blood and sputum. However, conventional cigarettes induced much stronger transcriptomic responses in both compartments.
Collapse
Affiliation(s)
- Mario F Perez
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | | | - Eric M Mortensen
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Laura E Crotty Alexander
- Division of Pulmonary Critical Care, Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
5
|
Suzukawa M, Ohta K, Fukutomi Y, Hashimoto H, Endo T, Abe M, Kamide Y, Yoshida M, Kikuchi Y, Kita T, Chibana K, Tanimoto Y, Hyodo K, Takata S, Inui T, Yasui M, Harada Y, Sato T, Sakakibara Y, Minakata Y, Inoue Y, Tamaki S, Shinohara T, Takami K, Tsubakihara M, Oki M, Wakamatsu K, Horiba M, Ideura G, Hidaka K, Saito AM, Kobayashi N, Taniguchi M. Classifications of moderate to severe asthma phenotypes in Japan and analysis of serum biomarkers: A Nationwide Cohort Study in Japan (NHOM Asthma Study). Allergol Int 2023; 72:63-74. [PMID: 35791991 DOI: 10.1016/j.alit.2022.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/17/2022] [Accepted: 05/25/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Asthma is a heterogeneous disease, and phenotyping can facilitate understanding of disease pathogenesis and direct appropriate asthma treatment. This nationwide cohort study aimed to phenotype asthma patients in Japan and identify potential biomarkers to classify the phenotypes. METHODS Adult asthma patients (n = 1925) from 27 national hospitals in Japan were enrolled and divided into Global Initiative for Asthma (GINA) steps 4 or 5 (GINA 4, 5) and GINA Steps 1, 2, or 3 (GINA 1-3) for therapy. Clinical data and questionnaires were collected. Biomarker levels among GINA 4, 5 patients were measured. Ward's minimum variance hierarchical clustering method and tree analysis were performed for phenotyping. Analysis of variance, the Kruskal-Wallis, and chi-square tests were used to compare cluster differences. RESULTS The following five clusters were identified: 1) late-onset, old, less-atopic; 2) late-onset, old, eosinophilic, low FEV1; 3) early-onset, long-duration, atopic, poorly controlled; 4) early-onset, young, female-dominant, atopic; and 5) female-dominant, T1/T2-mixed, most severe. Age of onset, disease duration, blood eosinophils and neutrophils, asthma control questionnaire Sum 6, number of controllers, FEV1, body mass index (BMI), and hypertension were the phenotype-classifying variables determined by tree analysis that assigned 79.5% to the appropriate cluster. Among the cytokines measured, IL-1RA, YKL40/CHI3L1, IP-10/CXCL10, RANTES/CCL5, and TIMP-1 were useful biomarkers for classifying GINA 4, 5 phenotypes. CONCLUSIONS Five distinct phenotypes were identified for moderate to severe asthma and may be classified using clinical and molecular variables (Registered in UMIN-CTR; UMIN000027776.).
Collapse
Affiliation(s)
- Maho Suzukawa
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, Tokyo, Japan.
| | - Ken Ohta
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, Tokyo, Japan; Japan Anti-Tuberculosis Association, JATA Fukujuji Hospital, Tokyo, Japan.
| | - Yuma Fukutomi
- Clinical Research Center, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Hiroya Hashimoto
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan; Core Laboratory, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takeo Endo
- National Hospital Organization Mito Medical Center, Ibaraki, Japan
| | - Masahiro Abe
- National Hospital Organization Ehime Medical Center, Ehime, Japan
| | - Yosuke Kamide
- Clinical Research Center, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Makoto Yoshida
- National Hospital Organization Fukuoka National Hospital, Fukuoka, Japan
| | | | - Toshiyuki Kita
- National Hospital Organization Kanazawa Medical Center, Ishikawa, Japan
| | - Kenji Chibana
- National Hospital Organization Okinawa National Hospital, Okinawa, Japan
| | - Yasushi Tanimoto
- National Hospital Organization Minami-Okayama Medical Center, Okayama, Japan
| | - Kentaro Hyodo
- National Hospital Organization Ibarakihigashi National Hospital, Ibaraki, Japan
| | - Shohei Takata
- National Hospital Organization Fukuokahigashi Medical Center, Fukuoka, Japan
| | - Toshiya Inui
- National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Masahide Yasui
- National Hospital Organization Nanao National Hospital, Ishikawa, Japan
| | - Yoshinori Harada
- Department of Rheumatology & Allergology, National Hospital Organization Osaka Minami Medical Center, Osaka, Japan
| | - Toshio Sato
- National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Yumi Sakakibara
- Federation of National Public Service Personnel Mutual Aid Associations Hiratsuka Kyosai Hospital, Kanagawa, Japan
| | | | - Yoshikazu Inoue
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Shinji Tamaki
- National Hospital Organization Nara Medical Center, Nara, Japan
| | - Tsutomu Shinohara
- National Hospital Organization Kochi National Hospital, Kochi, Japan
| | - Kazutaka Takami
- Kanto Central Hospital of the Mutual Aid Association of Public School Teachers, Tokyo, Japan
| | | | - Masahide Oki
- Department of Respiratory Medicine, National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | - Kentaro Wakamatsu
- National Hospital Organization Omuta National Hospital, Fukuoka, Japan
| | - Masahide Horiba
- Division of Respiratory Medicine, National Hospital Organization Higashisaitama National Hospital, Saitama, Japan
| | - Gen Ideura
- National Hospital Organization Shinshu Ueda Medical Center, Nagano, Japan
| | - Koko Hidaka
- National Hospital Organization Kokura Medical Center, Fukuoka, Japan
| | - Akiko M Saito
- Clinical Research Center, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Nobuyuki Kobayashi
- Clinical Research Center, National Hospital Organization Tokyo National Hospital, Tokyo, Japan; Fureai Machida Hospital, Tokyo, Japan
| | - Masami Taniguchi
- Clinical Research Center, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan; Shonan Kamakura General Hospital, Kanagawa, Japan
| |
Collapse
|
6
|
Functional immunophenotyping of children with critical status asthmaticus identifies differential gene expression responses in neutrophils exposed to a poly(I:C) stimulus. Sci Rep 2022; 12:19644. [PMID: 36385161 PMCID: PMC9666940 DOI: 10.1038/s41598-022-24261-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022] Open
Abstract
The host immune response to a viral immune stimulus has not been examined in children during a life-threatening asthma attack. We determined whether we could identify clusters of children with critical asthma by functional immunophenotyping using an intracellular viral analog stimulus. We performed a single-center, prospective, observational cohort study of 43 children ages 6-17 years admitted to a pediatric intensive care unit for an asthma attack between July 2019 to February 2021. Neutrophils were isolated from children, stimulated overnight with LyoVec poly(I:C), and mRNA was analyzed using a targeted Nanostring immunology array. Network analysis of the differentially expressed transcripts for the paired LyoVec poly(I:C) samples was performed. We identified two clusters by functional immunophenotyping that differed by the Asthma Control Test score. Cluster 1 (n = 23) had a higher proportion of children with uncontrolled asthma in the four weeks prior to PICU admission compared with cluster 2 (n = 20). Pathways up-regulated in cluster 1 versus cluster 2 included chemokine receptor/chemokines, interleukin-10 (IL-10), IL-4, and IL-13 signaling. Larger validation studies and clinical phenotyping of children with critical asthma are needed to determine the predictive utility of these clusters in a larger clinical setting.
Collapse
|
7
|
Abstract
The traditional one-size-fits all approach based on asthma severity is archaic. Asthma is a heterogenous syndrome rather than a single disease entity. Studies evaluating observable characteristics called phenotypes have elucidated this heterogeneity. Asthma clusters demonstrate overlapping features, are generally stable over time and are reproducible. What the identification of clusters may have failed to do, is move the needle of precision medicine meaningfully in asthma. This may be related to the lack of a straightforward and clinically meaningful way to apply what we have learned about asthma clusters. Clusters are based on both clinical factors and biomarkers. The use of biomarkers is slowly gaining popularity, but phenotyping based on biomarkers is generally greatly underutilized even in subspecialty care. Biomarkers are more often used to evaluate type 2 (T2) inflammatory signatures and eosinophils (sputum and blood), fractional exhaled nitric oxide (FeNO) and serum total and specific immunoglobulin (Ig) E reliably characterize the underlying inflammatory pathways. Biomarkers perform variably and clinicians must be familiar with their advantages and disadvantages to accurately apply them in clinical care. In addition, it is increasingly clear that clinical features are critical in understanding not only phenotypic characterization but in predicting response to therapy and future risk of poor outcomes. Strategies for asthma management will need to leverage our knowledge of biomarkers and clinical features to create composite scores and risk prediction tools that are clinically applicable. Despite significant progress, many questions remain, and more work is required to accurately identify non-T2 biomarkers. Adoption of phenotyping and more consistent use of biomarkers is needed, and we should continue to encourage this incorporation into practice.
Collapse
Affiliation(s)
- Arjun Mohan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Njira L Lugogo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
8
|
Wang T, He C, Hu M, Wu H, Ou S, Li Y, Fan C. Subtyping children with asthma by clustering analysis of mRNA expression data. Front Genet 2022; 13:974936. [PMID: 36159986 PMCID: PMC9500203 DOI: 10.3389/fgene.2022.974936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Asthma is a heterogeneous disease. There are several phenotypic classifications for childhood asthma. Methods: Unsupervised consensus cluster analysis was used to classify 36 children with persistent asthma from the GSE65204 dataset. The differentially expressed genes (DEGs) between different asthma subtypes were identified, and weighted gene co-expression network analysis (WGCNA) was carried out. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis was performed for DEGs and critical gene modules. Protein–protein interactions (PPI) were constructed to obtain the hub genes. Finally, differences in the immune microenvironment were analyzed between different subtypes. Results: Two subtypes (C1, C2) were identified using unsupervised consensus clustering. The DEGs between different asthma subtypes were mainly enriched in immune regulation and the release of inflammatory mediators. The important modular genes screened by WGCNA were mainly enriched in aspects of inflammatory mediator regulation. PPI analysis found 10 hub genes (DRC1, TTC25, DNALI1, DNAI1, DNAI2, PIH1D3, ARMC4, RSPH1, DNAAF3, and DNAH5), and ROC analysis demonstrated that 10 hub genes had a reliably ability to distinguish C1 from C2. And we observed differences between C1 and C2 in their immune microenvironment. Conclusion: Using the gene expression profiles of children’s nasal epithelium, we identified two asthma subtypes that have different gene expression patterns, biological characteristics, and immune microenvironments. This will provide a reference point for future childhood asthma typing and personalized therapy.
Collapse
Affiliation(s)
- Ting Wang
- The Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Changhui He
- The Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ming Hu
- Department of Pediatrics, Shenzhen Children’s Hospital, Shenzhen, Guangdong, China
| | - Honghua Wu
- Department of Pediatrics, Chenzhou No 1 People’s Hospital, Chenzhou, Hunan, China
| | - Shuteng Ou
- Department of Pediatrics, Chenzhou No 1 People’s Hospital, Chenzhou, Hunan, China
| | - Yuke Li
- The Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Chuping Fan
- The Affiliated Chenzhou Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Pediatrics, Chenzhou No 1 People’s Hospital, Chenzhou, Hunan, China
- *Correspondence: Chuping Fan,
| |
Collapse
|
9
|
Calhoun WJ, Chupp GL. The new era of add-on asthma treatments: where do we stand? ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2022; 18:42. [PMID: 35598022 PMCID: PMC9124422 DOI: 10.1186/s13223-022-00676-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/12/2022] [Indexed: 11/20/2022]
Abstract
Globally, a small proportion (5-12%) of asthma patients are estimated to have severe disease. However, severe asthma accounts for disproportionately high healthcare resource utilization. The Global Initiative for Asthma (GINA) management committee recommends treating patients with asthma with inhaled corticosteroids plus long-acting β2-agonists and, when needed, adding a long-acting muscarinic receptor antagonist or biologic agent. Five biologics, targeting different effectors in the type 2 inflammatory pathway, are approved for asthma treatment. However, biologics have not been compared against each other or add-on inhaled therapies in head-to-head clinical trials. As a result, their positioning versus that of current and anticipated small-molecule strategies is largely unknown. Furthermore, with the emergence of biomarkers for predicting response to biologics, a more personalized treatment approach-currently lacking with inhaled therapies-may be possible. To gain perspective, we reviewed recent advances in asthma pathophysiology, phenotypes, and biomarkers; the place of biologics in the management and personalized treatment of severe asthma; and the future of biologics and small-molecule drugs. We propose an algorithm for the stepwise treatment of severe asthma based on recommendations in the GINA strategy document that accounts for the broad range of phenotypes targeted by inhaled therapies and the specificity of biologics. In the future, both biologics and small molecules will continue to play key roles in the individualized treatment of severe asthma. However, as targeted therapies, their application will continue to be focused on patients with certain phenotypes who meet the specific criteria for use as identified in pivotal clinical trials.
Collapse
Affiliation(s)
- William J Calhoun
- Divisions of Pulmonary, Critical Care, and Sleep Medicine, and Allergy/Immunology; and Institute for Translational Sciences, University of Texas Medical Branch, 4.116 John Sealy Annex, 301 University Blvd, Galveston, TX, 77555-0568, USA.
| | - Geoffrey L Chupp
- Division of Pulmonary, Critical Care, and Sleep Medicine, Yale Center for Asthma and Airway Disease, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
10
|
Chen F, Liang Y, Zeng Z, Du L, Xu C, Guo Y, Xie C. Association of increased basic salivary proline-rich protein 1 levels in induced sputum with type 2-high asthma. Immun Inflamm Dis 2022; 10:e602. [PMID: 35344278 PMCID: PMC8959441 DOI: 10.1002/iid3.602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The aim of this study is to reveal whether basic salivary proline-rich protein BstNI subfamily 1 (PRB1) may be used as a diagnostic biomarker for type 2-high asthma. METHODS PRB1 protein levels in the induced sputum of 67 subjects with asthma and 27 controls were determined by an enzyme-linked immunosorbent assay. Correlation analyses between PRB1 in the induced sputum and airway inflammatory indicators were also performed. RESULTS PRB1 protein levels were significantly upregulated in the induced sputum of asthmatic patients (p =0.0098) and correlated with clinical eosinophil-related indicators and type 2 airway inflammation. These results indicate that PRB1 is a promising biomarker for type 2-high asthma. CONCLUSIONS The expression of PRB1 in induced sputum is a potential biomarker for type 2-high asthma. The results of this study present new insights into the diagnosis and treatment of asthma.
Collapse
Affiliation(s)
- Fengjia Chen
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yuxia Liang
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Zhimin Zeng
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Lijuan Du
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Changyi Xu
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Yubiao Guo
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Canmao Xie
- Division of Pulmonary and Critical Care MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
11
|
Pijnenburg MW, Frey U, De Jongste JC, Saglani S. Childhood asthma- pathogenesis and phenotypes. Eur Respir J 2021; 59:13993003.00731-2021. [PMID: 34711541 DOI: 10.1183/13993003.00731-2021] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/15/2021] [Indexed: 11/05/2022]
Abstract
In the pathogenesis of asthma in children there is a pivotal role for a type 2 inflammatory response to early life exposures or events. Interactions between infections, atopy, genetic susceptibility, and environmental exposures (such as farmyard environment, air pollution, tobacco smoke exposure) influence the development of wheezing illness and the risk for progression to asthma. The immune system, lung function and the microbiome in gut and airways develop in parallel and dysbiosis of the microbiome may be a critical factor in asthma development. Increased infant weight gain and preterm birth are other risk factors for development of asthma and reduced lung function. The complex interplay between these factors explains the heterogeneity of asthma in children. Subgroups of patients can be identified as phenotypes based on clinical parameters, or endotypes, based on a specific pathophysiological mechanism. Paediatric asthma phenotypes and endotypes may ultimately help to improve diagnosis of asthma, prediction of asthma development and treatment of individual children, based on clinical, temporal, developmental or inflammatory characteristics. Unbiased, data-driven clustering, using a multidimensional or systems biology approach may be needed to better define phenotypes. The present knowledge on inflammatory phenotypes of childhood asthma has now been successfully applied in the treatment with biologicals of children with severe therapy resistant asthma, and it is to be expected that more personalized treatment options may become available.
Collapse
Affiliation(s)
- Mariëlle W Pijnenburg
- Department of Paediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Urs Frey
- University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Johan C De Jongste
- Department of Paediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Sejal Saglani
- National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
12
|
Repetitive aeroallergen challenges elucidate maladaptive epithelial and inflammatory traits that underpin allergic airway diseases. J Allergy Clin Immunol 2021; 148:533-549. [PMID: 33493557 PMCID: PMC8298629 DOI: 10.1016/j.jaci.2021.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Signifying the 2-compartments/1-disease paradigm, allergic rhinoconjunctivitis (ARC) and asthma (AA) are prevalent, comorbid conditions triggered by environmental factors (eg, house dust mites [HDMs]). However, despite the ubiquity of triggers, progression to severe ARC/AA is infrequent, suggesting either resilience or adaptation. OBJECTIVE We sought to determine whether ARC/AA severity relates to maladaptive responses to disease triggers. METHODS Adults with HDM-associated ARC were challenged repetitively with HDMs in an aeroallergen challenge chamber. Mechanistic traits associated with disease severity were identified. RESULTS HDM challenges evoked maladaptive (persistently higher ARC symptoms), adaptive (progressive symptom reduction), and resilient (resistance to symptom induction) phenotypes. Symptom severity in the natural environment was an imprecise correlate of the phenotypes. Nasal airway traits, defined by low inflammation-effectual epithelial integrity, moderate inflammation-effectual epithelial integrity, and higher inflammation-ineffectual epithelial integrity, were hallmarks of the resilient, adaptive, and maladaptive evoked phenotypes, respectively. Highlighting a crosstalk mechanism, peripheral blood inflammatory tone calibrated these traits: ineffectual epithelial integrity associated with CD8+ T cells, whereas airway inflammation associated with both CD8+ T cells and eosinophils. Hallmark peripheral blood maladaptive traits were increased natural killer and CD8+ T cells, lower CD4+ mucosal-associated invariant T cells, and deficiencies along the TLR-IRF-IFN antiviral pathway. Maladaptive traits tracking HDM-associated ARC also contributed to AA risk and severity models. CONCLUSIONS Repetitive challenges with HDMs revealed that maladaptation to disease triggers may underpin ARC/AA disease severity. A combinatorial therapeutic approach may involve reversal of loss-of-beneficial-function traits (ineffectual epithelial integrity, TLR-IRF-IFN deficiencies), mitigation of gain-of-adverse-function traits (inflammation), and blocking of a detrimental crosstalk between the peripheral blood and airway compartments.
Collapse
|
13
|
Khanal S, Webster M, Niu N, Zielonka J, Nunez M, Chupp G, Slade MD, Cohn L, Sauler M, Gomez JL, Tarran R, Sharma L, Dela Cruz CS, Egan M, Laguna T, Britto CJ. SPLUNC1: a novel marker of cystic fibrosis exacerbations. Eur Respir J 2021; 58:13993003.00507-2020. [PMID: 33958427 DOI: 10.1183/13993003.00507-2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/29/2021] [Indexed: 11/05/2022]
Abstract
Acute pulmonary Exacerbations (AE) are episodes of clinical worsening in cystic fibrosis (CF), often precipitated by infection. Timely detection is critical to minimise morbidity and lung function declines associated with acute inflammation during AE. Based on our previous observations that airway protein Short Palate Lung Nasal epithelium Clone 1 (SPLUNC1) is regulated by inflammatory signals, we investigated the use of SPLUNC1 fluctuations to diagnose and predict AE in CF.We enrolled CF participants from two independent cohorts to measure AE markers of inflammation in sputum and recorded clinical outcomes for a 1-year follow-up period.SPLUNC1 levels were high in healthy controls (n=9, 10.7 μg mL-1), and significantly decreased in CF participants without AE (n=30, 5.7 μg mL-1, p=0.016). SPLUNC1 levels were 71.9% lower during AE (n=14, 1.6 μg mL-1, p=0.0034) regardless of age, sex, CF-causing mutation, or microbiology findings. Cytokines Il-1β and TNFα were also increased in AE, whereas lung function did not consistently decrease. Stable CF participants with lower SPLUNC1 levels were much more likely to have an AE at 60 days (HR: 11.49, Standard Error: 0.83, p=0.0033). Low-SPLUNC1 stable participants remained at higher AE risk even one year after sputum collection (HR: 3.21, Standard Error: 0.47, p=0.0125). SPLUNC1 was downregulated by inflammatory cytokines and proteases increased in sputum during AE.In acute CF care, low SPLUNC1 levels could support a decision to increase airway clearance or to initiate pharmacological interventions. In asymptomatic, stable patients, low SPLUNC1 levels could inform changes in clinical management to improve long-term disease control and clinical outcomes in CF.
Collapse
Affiliation(s)
- Sara Khanal
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Megan Webster
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Naiqian Niu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jana Zielonka
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Myra Nunez
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Martin D Slade
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert Tarran
- Department of Cell Biology & Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marie Egan
- Division of Pediatric Pulmonology, Allergy, Immunology, and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Theresa Laguna
- Division of Pediatric Respiratory Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Clemente J Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
14
|
Chen A, Diaz-Soto MP, Sanmamed MF, Adams T, Schupp JC, Gupta A, Britto C, Sauler M, Yan X, Liu Q, Nino G, Cruz CSD, Chupp GL, Gomez JL. Single-cell characterization of a model of poly I:C-stimulated peripheral blood mononuclear cells in severe asthma. Respir Res 2021; 22:122. [PMID: 33902571 PMCID: PMC8074196 DOI: 10.1186/s12931-021-01709-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 04/08/2021] [Indexed: 11/22/2022] Open
Abstract
Background Asthma has been associated with impaired interferon response. Multiple cell types have been implicated in such response impairment and may be responsible for asthma immunopathology. However, existing models to study the immune response in asthma are limited by bulk profiling of cells. Our objective was to Characterize a model of peripheral blood mononuclear cells (PBMCs) of patients with severe asthma (SA) and its response to the TLR3 agonist Poly I:C using two single-cell methods. Methods Two complementary single-cell methods, DropSeq for single-cell RNA sequencing (scRNA-Seq) and mass cytometry (CyTOF), were used to profile PBMCs of SA patients and healthy controls (HC). Poly I:C-stimulated and unstimulated cells were analyzed in this study. Results PBMCs (n = 9414) from five SA (n = 6099) and three HC (n = 3315) were profiled using scRNA-Seq. Six main cell subsets, namely CD4 + T cells, CD8 + T cells, natural killer (NK) cells, B cells, dendritic cells (DCs), and monocytes, were identified. CD4 + T cells were the main cell type in SA and demonstrated a pro-inflammatory profile characterized by increased JAK1 expression. Following Poly I:C stimulation, PBMCs from SA had a robust induction of interferon pathways compared with HC. CyTOF profiling of Poly I:C stimulated and unstimulated PBMCs (n = 160,000) from the same individuals (SA = 5; HC = 3) demonstrated higher CD8 + and CD8 + effector T cells in SA at baseline, followed by a decrease of CD8 + effector T cells after poly I:C stimulation. Conclusions Single-cell profiling of an in vitro model using PBMCs in patients with SA identified activation of pro-inflammatory pathways at baseline and strong response to Poly I:C, as well as quantitative changes in CD8 + effector cells. Thus, transcriptomic and cell quantitative changes are associated with immune cell heterogeneity in this model to evaluate interferon responses in severe asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-021-01709-9.
Collapse
Affiliation(s)
- Ailu Chen
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Maria P Diaz-Soto
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Miguel F Sanmamed
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.,Program of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Taylor Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Jonas C Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Amolika Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Clemente Britto
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Maor Sauler
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Qing Liu
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Gustavo Nino
- Division of Pediatric Pulmonary and Sleep Medicine, Children's National Hospital, Department of Pediatrics, George Washington University, Washington, DC, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street (S419 TAC), New Haven, CT, 06520-8057, USA.
| |
Collapse
|
15
|
Perez MF, Atuegwu NC, Mortensen EM, Oncken C. The inflammatory biomarker YKL-40 is elevated in the serum, but not the sputum, of E-cigarette users. Exp Lung Res 2021; 47:55-66. [PMID: 33200966 PMCID: PMC8168626 DOI: 10.1080/01902148.2020.1847216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 10/23/2022]
Abstract
METHODS We conducted a cross-sectional study of adults between 18 and 55 years old. Inclusion criteria were: exclusive e-cigarette use or cigarette smoking for ≥ 1 year or no history of tobacco use. Participants with a history of pulmonary illness, atopy, medications (except birth control pills), marijuana, and illegal substance use were excluded. Custom Multiplex ELISA was used to measure YKL-40 and other biomarker levels in the serum and induced sputum of the participants. Multivariable linear regression was used to compare the levels of YLK-40 in healthy participants, e-cigarette, and cigarette users after adjusting for age, sex, and BMI. RESULTS We recruited 20 healthy controls, 23 cigarette smokers, and 22 exclusive e-cigarette users. Serum YKL-40 (ng/ml) was significantly higher in e-cigarette users (Median 21.2 [IQR 12.1-24.0] ng/ml) when compared to controls (12.2 [IQR 8.7-18.1] ng/ml, p = 0.016) but comparable to cigarette smokers (21.6 [IQR 11.62-51.7] ng/ml, p = 0.31). No significant differences were found in the serum or sputum of the other biomarkers tested. CONCLUSION The inflammatory biomarker, YKL-40 is elevated in the serum but not the sputum of e-cigarette users with no reported pulmonary disease. Further research is necessary to characterize this association.
Collapse
Affiliation(s)
- Mario F Perez
- Pulmonary, Critical Care and Sleep Medicine, Deparment of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Nkiruka C Atuegwu
- Pulmonary, Critical Care and Sleep Medicine, Deparment of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Eric M Mortensen
- Pulmonary, Critical Care and Sleep Medicine, Deparment of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Cheryl Oncken
- Pulmonary, Critical Care and Sleep Medicine, Deparment of Medicine, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
16
|
Abstract
The community of cells lining our airways plays a collaborative role in the preservation of immune homeostasis in the lung and provides protection from the pathogens and pollutants in the air we breathe. In addition to its structural attributes that provide effective mucociliary clearance of the lower airspace, the airway epithelium is an immunologically active barrier surface that senses changes in the airway environment and interacts with resident and recruited immune cells. Single-cell RNA-sequencing is illuminating the cellular heterogeneity that exists in the airway wall and has identified novel cell populations with unique molecular signatures, trajectories of differentiation and diverse functions in health and disease. In this Review, we discuss how our view of the airway epithelial landscape has evolved with the advent of transcriptomic approaches to cellular phenotyping, with a focus on epithelial interactions with the local neuronal and immune systems.
Collapse
|
17
|
Jakobi M, Kiefer A, Mirzakhani H, Rauh M, Zimmermann T, Xepapadaki P, Stanic B, Akdis M, Papadopoulos NG, Raby BA, Weiss ST, Finotto S. Role of nuclear factor of activated T cells 2 (NFATc2) in allergic asthma. Immun Inflamm Dis 2020; 8:704-712. [PMID: 33079489 PMCID: PMC7654396 DOI: 10.1002/iid3.360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND We recently described increased NFATc1, IRF4, and NIP45 messenger RNA (mRNA) expression in peripheral blood mononuclear cells (PBMCs) of asthmatic children and adults with multiple allergies. OBJECTIVE NFATc2 has been described to associate with IRF4 to induce interleukin-4, and to be inhibited by T-bet. Here, we analyzed the role of NFATc2 in asthmatic children and adults. METHODS PBMCs were isolated from the blood of control of asthmatics subjects. Some PBMCs were analyzed untreated and some cultured with and without phytohemagglutinin. Then, RNA was extracted from the cells and cytokines were measured in the supernatants via enzyme-linked immunosorbent assay or multiplex analysis. RNA was then reverse-transcribed and NFATc1, NFATC2, IRF4, and T-bet mRNA were analyzed by real-time polymerase chain reaction. In addition, in peripheral blood cells, NFATc2 expression was analyzed, in a population of asthmatic children and adults from the Asthma BRIDGE study. RESULTS In addition to NFATc1 and NIP45, also NFATc2 was found upregulated in PBMCs and peripheral blood cells from asthmatic children and adults with allergic asthma. Moreover, NFATc1 directly correlated with lymphocytes number whereas NFATc2 correlated with peripheral eosinophilia in asthma. CONCLUSIONS In addition to NFATc1 and NIP45, NFATc2 was found upregulated in asthma. Moreover, NFATc1 mRNA correlated with lymphocytes both in control and asthma, and NFATC1 and NFATc2 mRNA showed a direct correlation with eosinophils in controls but not in asthma, indicating that NFATc1 is associated with lymphocytes and not eosinophils in asthma. CLINICAL SIGNIFICANCE Targeting NFATc2 in T lymphocytes might ameliorate the allergic phenotype in asthmatic subjects.
Collapse
Affiliation(s)
- Marielena Jakobi
- Department of Molecular Pneumology, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| | - Alexander Kiefer
- Department of Allergy and Pneumology, Children's Hospital, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Manfred Rauh
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Theodor Zimmermann
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Paraskevi Xepapadaki
- Department of Allergy, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
| | - Barbara Stanic
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavos WolfgangSwitzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavos WolfgangSwitzerland
| | - Nikolaos G. Papadopoulos
- Department of Allergy, 2nd Pediatric ClinicNational and Kapodistrian University of AthensAthensGreece
- Centre for Respiratory Medicine and AllergyUniversity of ManchesterUK
| | - Benjamin A. Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Susetta Finotto
- Department of Molecular Pneumology, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität Erlangen‐NürnbergErlangenGermany
| |
Collapse
|
18
|
Groth EE, Weber M, Bahmer T, Pedersen F, Kirsten A, Börnigen D, Rabe KF, Watz H, Ammerpohl O, Goldmann T. Exploration of the sputum methylome and omics deconvolution by quadratic programming in molecular profiling of asthma and COPD: the road to sputum omics 2.0. Respir Res 2020; 21:274. [PMID: 33076907 PMCID: PMC7574293 DOI: 10.1186/s12931-020-01544-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/11/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND To date, most studies involving high-throughput analyses of sputum in asthma and COPD have focused on identifying transcriptomic signatures of disease. No whole-genome methylation analysis of sputum cells has been performed yet. In this context, the highly variable cellular composition of sputum has potential to confound the molecular analyses. METHODS Whole-genome transcription (Agilent Human 4 × 44 k array) and methylation (Illumina 450 k BeadChip) analyses were performed on sputum samples of 9 asthmatics, 10 healthy and 10 COPD subjects. RNA integrity was checked by capillary electrophoresis and used to correct in silico for bias conferred by RNA degradation during biobank sample storage. Estimates of cell type-specific molecular profiles were derived via regression by quadratic programming based on sputum differential cell counts. All analyses were conducted using the open-source R/Bioconductor software framework. RESULTS A linear regression step was found to perform well in removing RNA degradation-related bias among the main principal components of the gene expression data, increasing the number of genes detectable as differentially expressed in asthma and COPD sputa (compared to controls). We observed a strong influence of the cellular composition on the results of mixed-cell sputum analyses. Exemplarily, upregulated genes derived from mixed-cell data in asthma were dominated by genes predominantly expressed in eosinophils after deconvolution. The deconvolution, however, allowed to perform differential expression and methylation analyses on the level of individual cell types and, though we only analyzed a limited number of biological replicates, was found to provide good estimates compared to previously published data about gene expression in lung eosinophils in asthma. Analysis of the sputum methylome indicated presence of differential methylation in genomic regions of interest, e.g. mapping to a number of human leukocyte antigen (HLA) genes related to both major histocompatibility complex (MHC) class I and II molecules in asthma and COPD macrophages. Furthermore, we found the SMAD3 (SMAD family member 3) gene, among others, to lie within differentially methylated regions which has been previously reported in the context of asthma. CONCLUSIONS In this methodology-oriented study, we show that methylation profiling can be easily integrated into sputum analysis workflows and exhibits a strong potential to contribute to the profiling and understanding of pulmonary inflammation. Wherever RNA degradation is of concern, in silico correction can be effective in improving both sensitivity and specificity of downstream analyses. We suggest that deconvolution methods should be integrated in sputum omics analysis workflows whenever possible in order to facilitate the unbiased discovery and interpretation of molecular patterns of inflammation.
Collapse
Affiliation(s)
- Espen E Groth
- LungenClinic Grosshansdorf, Großhansdorf, Germany. .,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany. .,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany. .,Department of Oncology, Hematology and BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Melanie Weber
- Program in Applied and Computational Mathematics, Princeton University, Princeton, NJ, USA
| | - Thomas Bahmer
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Frauke Pedersen
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Anne Kirsten
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Daniela Börnigen
- Bioinformatics Core Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus F Rabe
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Henrik Watz
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Ole Ammerpohl
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Institute of Human Genetics, University Medical Center Ulm, Ulm, Germany
| | - Torsten Goldmann
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Research Center Borstel, Pathology, Borstel, Germany
| |
Collapse
|
19
|
Latent-space embedding of expression data identifies gene signatures from sputum samples of asthmatic patients. BMC Bioinformatics 2020; 21:457. [PMID: 33059594 PMCID: PMC7560063 DOI: 10.1186/s12859-020-03785-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 09/28/2020] [Indexed: 11/30/2022] Open
Abstract
Background The pathogenesis of asthma is a complex process involving multiple genes and pathways. Identifying biomarkers from asthma datasets, especially those that include heterogeneous subpopulations, is challenging. Potentially, autoencoders provide ideal frameworks for such tasks as they can embed complex, noisy high-dimensional gene expression data into a low-dimensional latent space in an unsupervised fashion, enabling us to extract distinguishing features from expression data. Results Here, we developed a framework combining a denoising autoencoder and a supervised learning classifier to identify gene signatures related to asthma severity. Using the trained autoencoder with 50 hidden units, we found that hierarchical clustering on the low-dimensional embedding corresponds well with previously defined and clinically relevant clusters of patients. Moreover, each hidden unit has contributions from each of the genes, and pathway analysis of these contributions shows that the hidden units are significantly enriched in known asthma-related pathways. We then used genes that contribute most to the hidden units to develop a secondary random-forest classifier for directly predicting asthma severity. The feature importance metric from this classifier identified a signature based on 50 key genes, which are associated with severity. Furthermore, we can use these key genes to successfully estimate FEV1/FVC ratios across patients, via support-vector-machine regression. Conclusion We found that the denoising autoencoder framework can extract meaningful patterns corresponding to functional gene groups and patient clusters from the gene expression of asthma patients.
Collapse
|
20
|
Stewart E, Wang X, Chupp GL, Montgomery RR. Profiling cellular heterogeneity in asthma with single cell multiparameter CyTOF. J Leukoc Biol 2020; 108:1555-1564. [PMID: 32911570 DOI: 10.1002/jlb.5ma0720-770rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/13/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the airways that afflicts over 30 million individuals in the United States and over 300 million individuals worldwide. The inflammatory response in the airways is often characterized by the analysis of sputum, which contains multiple types of cells including neutrophils, macrophages, lymphocytes, and rare bronchial epithelial cells. Subtyping patients using microscopy of the sputum has identified both neutrophilic and eosinophilic infiltrates in airway inflammation. However, with the extensive heterogeneity among these cell types, a higher resolution understanding of the inflammatory cell types present in the sputum is needed to dissect the heterogeneity of disease. Improved recognition of the distinct phenotypes and sources of inflammation in asthmatic granulocytes may identify relevant pathways for clinical management or investigation of novel therapeutic mediators. Here, we employed mass cytometry or cytometry by time-of-flight to quantify frequency and define functional status of sputum derived airway cells in asthmatic patients and healthy controls. This in-depth single cell analysis method identified multiple distinct subtypes of airway immune cells, especially in neutrophils. Significance was discovered by statistical analysis as well as a data-driven unbiased clustering approach. Our multidimensional assessment method identifies differences in cellular function and supports identification of cellular status that may contribute to diverse clinical responses. This technical advance is relevant for studies of pathogenesis and may provide meaningful insights to advance our knowledge of asthmatic inflammation.
Collapse
Affiliation(s)
- Emma Stewart
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xiaomei Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Geoffrey L Chupp
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
21
|
Baines KJ, Negewo NA, Gibson PG, Fu JJ, Simpson JL, Wark PAB, Fricker M, McDonald VM. A Sputum 6 Gene Expression Signature Predicts Inflammatory Phenotypes and Future Exacerbations of COPD. Int J Chron Obstruct Pulmon Dis 2020; 15:1577-1590. [PMID: 32669843 PMCID: PMC7337431 DOI: 10.2147/copd.s245519] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/24/2020] [Indexed: 02/05/2023] Open
Abstract
Background The 6 gene expression signature (6GS) predicts inflammatory phenotype, exacerbation risk, and corticosteroid responsiveness in asthma. In COPD, patterns of airway inflammation are similar, suggesting the 6GS may be useful. This study determines the diagnostic and prognostic ability of 6GS in predicting inflammatory phenotypes and exacerbation risk in COPD. Methods We performed 2 studies: a cross-sectional phenotype prediction study in stable COPD (total N=132; n=34 eosinophilic (E)-COPD, n=42 neutrophilic (N)-COPD, n=39 paucigranulocytic (PG)-COPD, n=17 mixed-granulocytic (MG)-COPD) that assessed 6GS ability to discriminate phenotypes (eosinophilia≥3%; neutrophilia≥61%); and a prospective cohort study (total n=54, n=8 E-COPD; n=18 N-COPD; n=20 PG-COPD; n=8 MG-COPD, n=21 exacerbation prone (≥2/year)) that investigated phenotype and exacerbation prediction utility. 6GS was measured by qPCR and evaluated using multiple logistic regression and area under the curve (AUC). Short-term reproducibility (intra-class correlation) and phenotyping method agreement (κ statistic) were assessed. Results In the phenotype prediction study, 6GS could accurately identify and discriminate patients with E-COPD from N-COPD (AUC=96.4%; p<0.0001), PG-COPD (AUC=88.2%; p<0.0001) or MG-COPD (AUC=86.2%; p=0.0001), as well as N-COPD from PG-COPD (AUC=83.6%; p<0.0001) or MG-COPD (AUC=87.4%; p<0.0001) and was reproducible. In the prospective cohort study, 6GS had substantial agreement for neutrophilic inflammation (82%, κ=0.63, p<0.001) and moderate agreement for eosinophilic inflammation (78%, κ=0.42, p<0.001). 6GS could significantly discriminate exacerbation prone patients (AUC=77.2%; p=0.034). Higher IL1B levels were associated with poorer lung function and increased COPD severity. Conclusion 6GS can significantly and reproducibly discriminate COPD inflammatory phenotypes and predict exacerbation prone patients and may become a useful molecular diagnostic tool assisting COPD management.
Collapse
Affiliation(s)
- Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Netsanet A Negewo
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Peter G Gibson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Juan-Juan Fu
- Respiratory Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People's Republic of China
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Michael Fricker
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia
| | - Vanessa M McDonald
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Callaghan, NSW, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia.,School of Nursing and Midwifery, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
22
|
Ray A, Camiolo M, Fitzpatrick A, Gauthier M, Wenzel SE. Are We Meeting the Promise of Endotypes and Precision Medicine in Asthma? Physiol Rev 2020; 100:983-1017. [PMID: 31917651 PMCID: PMC7474260 DOI: 10.1152/physrev.00023.2019] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
While the term asthma has long been known to describe heterogeneous groupings of patients, only recently have data evolved which enable a molecular understanding of the clinical differences. The evolution of transcriptomics (and other 'omics platforms) and improved statistical analyses in combination with large clinical cohorts opened the door for molecular characterization of pathobiologic processes associated with a range of asthma patients. When linked with data from animal models and clinical trials of targeted biologic therapies, emerging distinctions arose between patients with and without elevations in type 2 immune and inflammatory pathways, leading to the confirmation of a broad categorization of type 2-Hi asthma. Differences in the ratios, sources, and location of type 2 cytokines and their relation to additional immune pathway activation appear to distinguish several different (sub)molecular phenotypes, and perhaps endotypes of type 2-Hi asthma, which respond differently to broad and targeted anti-inflammatory therapies. Asthma in the absence of type 2 inflammation is much less well defined, without clear biomarkers, but is generally linked with poor responses to corticosteroids. Integration of "big data" from large cohorts, over time, using machine learning approaches, combined with validation and iterative learning in animal (and human) model systems is needed to identify the biomarkers and tightly defined molecular phenotypes/endotypes required to fulfill the promise of precision medicine.
Collapse
Affiliation(s)
- Anuradha Ray
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary Allergy Critical Care Medicine, Departments of Medicine and of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Matthew Camiolo
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary Allergy Critical Care Medicine, Departments of Medicine and of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Anne Fitzpatrick
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary Allergy Critical Care Medicine, Departments of Medicine and of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Marc Gauthier
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary Allergy Critical Care Medicine, Departments of Medicine and of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Sally E Wenzel
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania; Pulmonary Allergy Critical Care Medicine, Departments of Medicine and of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania; and Department of Pediatrics, Emory University, Atlanta, Georgia
| |
Collapse
|
23
|
Gomez JL, Chen A, Diaz MP, Zirn N, Gupta A, Britto C, Sauler M, Yan X, Stewart E, Santerian K, Grant N, Liu Q, Fry R, Rager J, Cohn L, Alexis N, Chupp GL. A Network of Sputum MicroRNAs Is Associated with Neutrophilic Airway Inflammation in Asthma. Am J Respir Crit Care Med 2020; 202:51-64. [PMID: 32255668 PMCID: PMC7328332 DOI: 10.1164/rccm.201912-2360oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/06/2020] [Indexed: 01/06/2023] Open
Abstract
Rationale: MicroRNAs are potent regulators of biologic systems that are critical to tissue homeostasis. Individual microRNAs have been identified in airway samples. However, a systems analysis of the microRNA-mRNA networks present in the sputum that contribute to airway inflammation in asthma has not been published.Objectives: Identify microRNA and mRNA networks in the sputum of patients with asthma.Methods: We conducted a genome-wide analysis of microRNA and mRNA in the sputum from patients with asthma and correlated expression with clinical phenotypes. Weighted gene correlation network analysis was implemented to identify microRNA networks (modules) that significantly correlate with clinical features of asthma and mRNA expression networks. MicroRNA expression in peripheral blood neutrophils and lymphocytes and in situ hybridization of the sputum were used to identify the cellular sources of microRNAs. MicroRNA expression obtained before and after ozone exposure was also used to identify changes associated with neutrophil counts in the airway.Measurements and Main Results: Six microRNA modules were associated with clinical features of asthma. A single module (nely) was associated with a history of hospitalizations, lung function impairment, and numbers of neutrophils and lymphocytes in the sputum. Of the 12 microRNAs in the nely module, hsa-miR-223-3p was the highest expressed microRNA in neutrophils and was associated with increased neutrophil counts in the sputum in response to ozone exposure. Multiple microRNAs in the nely module correlated with two mRNA modules enriched for TLR (Toll-like receptor) and T-helper cell type 17 (Th17) signaling and endoplasmic reticulum stress. hsa-miR-223-3p was a key regulator of the TLR and Th17 pathways in the sputum of subjects with asthma.Conclusions: This study of sputum microRNA and mRNA expression from patients with asthma demonstrates the existence of microRNA networks and genes that are associated with features of asthma severity. Among these, hsa-miR-223-3p, a neutrophil-derived microRNA, regulates TLR/Th17 signaling and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Jose L. Gomez
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Ailu Chen
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Maria Paula Diaz
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Nicholas Zirn
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Amolika Gupta
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Clemente Britto
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Xiting Yan
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Emma Stewart
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kyle Santerian
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Nicole Grant
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Qing Liu
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina; and
| | - Julia Rager
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina; and
| | - Lauren Cohn
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Neil Alexis
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Geoffrey L. Chupp
- Pulmonary, Critical Care and Sleep Medicine, Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Spakowicz D, Lou S, Barron B, Gomez JL, Li T, Liu Q, Grant N, Yan X, Hoyd R, Weinstock G, Chupp GL, Gerstein M. Approaches for integrating heterogeneous RNA-seq data reveal cross-talk between microbes and genes in asthmatic patients. Genome Biol 2020; 21:150. [PMID: 32571363 PMCID: PMC7310008 DOI: 10.1186/s13059-020-02033-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 04/30/2020] [Indexed: 11/16/2022] Open
Abstract
Sputum induction is a non-invasive method to evaluate the airway environment, particularly for asthma. RNA sequencing (RNA-seq) of sputum samples can be challenging to interpret due to the complex and heterogeneous mixtures of human cells and exogenous (microbial) material. In this study, we develop a pipeline that integrates dimensionality reduction and statistical modeling to grapple with the heterogeneity. LDA(Latent Dirichlet allocation)-link connects microbes to genes using reduced-dimensionality LDA topics. We validate our method with single-cell RNA-seq and microscopy and then apply it to the sputum of asthmatic patients to find known and novel relationships between microbes and genes.
Collapse
Affiliation(s)
- Daniel Spakowicz
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Division of Medical Oncology, Ohio State University College of Medicine, Columbus, OH, USA
- Department of Biomedical Informatics, Ohio State University College of Medicine, Columbus, OH, USA
| | - Shaoke Lou
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Brian Barron
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Jose L Gomez
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tianxiao Li
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Qing Liu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Nicole Grant
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiting Yan
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Ohio State University College of Medicine, Columbus, OH, USA
| | - George Weinstock
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Geoffrey L Chupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Mark Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA.
- Department of Computer Science, Yale University, New Haven, CT, USA.
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA.
| |
Collapse
|
25
|
Phenotypes and endotypes of adult asthma: Moving toward precision medicine. J Allergy Clin Immunol 2020; 144:1-12. [PMID: 31277742 DOI: 10.1016/j.jaci.2019.05.031] [Citation(s) in RCA: 286] [Impact Index Per Article: 57.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic inflammatory disease of the airways that is challenging to dissect into subgroups because of the heterogeneity present across the spectrum of the disease. Efforts to subclassify asthma using advanced computational methods have identified a number of different phenotypes that suggest that multiple pathobiologically driven clusters of disease exist. The main phenotypes that have been identified include (1) early-onset allergic asthma, (2) early-onset allergic moderate-to-severe remodeled asthma, (3) late-onset nonallergic eosinophilic asthma, and (4) late-onset nonallergic noneosinophilic asthma. Subgroups of these phenotypes also exist but have not been as consistently identified. Advances in our understanding of the diverse immunologic perturbations that drive airway inflammation are consistent with clinical traits associated with these phenotypes and their response to biologic therapies. This has improved the clinician's approach to characterizing asthmatic patients in the clinic. Being able to define asthma endotypes using clinical characteristics and biomarkers will move physicians toward even more personalized management of asthma and precision-based care in the future. Here we will review the most prominent phenotypes and immunologic advances that suggest these disease subtypes represent asthma endotypes.
Collapse
|
26
|
Korde A, Ahangari F, Haslip M, Zhang X, Liu Q, Cohn L, Gomez JL, Chupp G, Pober JS, Gonzalez A, Takyar SS. An endothelial microRNA-1-regulated network controls eosinophil trafficking in asthma and chronic rhinosinusitis. J Allergy Clin Immunol 2020; 145:550-562. [PMID: 32035607 PMCID: PMC8440091 DOI: 10.1016/j.jaci.2019.10.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 09/25/2019] [Accepted: 10/16/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Airway eosinophilia is a prominent feature of asthma and chronic rhinosinusitis (CRS), and the endothelium plays a key role in eosinophil trafficking. To date, microRNA-1 (miR-1) is the only microRNA known to be regulated in the lung endothelium in asthma models. OBJECTIVE We sought to determine the role of endothelial miR-1 in allergic airway inflammation. METHODS We measured microRNA and mRNA expression using quantitative RT-PCR. We used ovalbumin and house dust mite models of asthma. Endothelium-specific overexpression of miR-1 was achieved through lentiviral vector delivery or induction of a transgene. Tissue eosinophilia was quantified by using Congo red and anti-eosinophil peroxidase staining. We measured eosinophil binding with a Sykes-Moore adhesion chamber. Target recruitment to RNA-induced silencing complex was assessed by using anti-Argonaute2 RNA immunoprecipitation. Surface P-selectin levels were measured by using flow cytometry. RESULTS Serum miR-1 levels had inverse correlations with sputum eosinophilia, airway obstruction, and number of hospitalizations in asthmatic patients and sinonasal tissue eosinophilia in patients with CRS. IL-13 stimulation decreased miR-1 levels in human lung endothelium. Endothelium-specific overexpression of miR-1 reduced airway eosinophilia and asthma phenotypes in murine models and inhibited IL-13-induced eosinophil binding to endothelial cells. miR-1 recruited P-selectin, thymic stromal lymphopoietin, eotaxin-3, and thrombopoietin receptor to the RNA-induced silencing complex; downregulated these genes in the lung endothelium; and reduced surface P-selectin levels in IL-13-stimulated endothelial cells. In our asthma and CRS cohorts, miR-1 levels correlated inversely with its target genes. CONCLUSION Endothelial miR-1 regulates eosinophil trafficking in the setting of allergic airway inflammation. miR-1 has therapeutic potential in asthmatic patients and patients with CRS.
Collapse
Affiliation(s)
- Asawari Korde
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Farida Ahangari
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Maria Haslip
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn; Yale School of Nursing, Orange, Conn
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Conn
| | - Qing Liu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Lauren Cohn
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Jose L Gomez
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Geoffrey Chupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn
| | - Jordan S Pober
- Department of Immunobiology, Yale School of Medicine, New Haven, Conn
| | | | - Shervin S Takyar
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Conn.
| |
Collapse
|
27
|
Hodge S. Sputum transcriptomics: A tool for identifying gene expression underlying mechanistic pathways in severe asthma. Respirology 2020; 25:668-669. [PMID: 31971331 DOI: 10.1111/resp.13769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 01/13/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Sandra Hodge
- Department of Medicine, University of Adelaide, Adelaide, SA, Australia.,Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
28
|
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
|
29
|
Willis-Owen SAG, Cookson WOC, Moffatt MF. The Genetics and Genomics of Asthma. Annu Rev Genomics Hum Genet 2019; 19:223-246. [PMID: 30169121 DOI: 10.1146/annurev-genom-083117-021651] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Asthma is a common, clinically heterogeneous disease with strong evidence of heritability. Progress in defining the genetic underpinnings of asthma, however, has been slow and hampered by issues of inconsistency. Recent advances in the tools available for analysis-assaying transcription, sequence variation, and epigenetic marks on a genome-wide scale-have substantially altered this landscape. Applications of such approaches are consistent with heterogeneity at the level of causation and specify patterns of commonality with a wide range of alternative disease traits. Looking beyond the individual as the unit of study, advances in technology have also fostered comprehensive analysis of the human microbiome and its varied roles in health and disease. In this article, we consider the implications of these technological advances for our current understanding of the genetics and genomics of asthma.
Collapse
Affiliation(s)
- Saffron A G Willis-Owen
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| | - William O C Cookson
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom; , ,
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Target therapy is the necessary step towards personalized medicine. The definition of asthma phenotypes and underlying mechanisms (endotypes) represent a key point in the development of new asthma treatments. Big data analysis, biomarker research and the availability of monoclonal antibodies, targeting specific cytokines is leading to the rapid evolution of knowledge. In this review, we sought to outline many of the recent advances in the field. RECENT FINDINGS Several attempts have been made to identify asthma phenotypes, sometimes with contrasting results. More success has been obtained concerning the pathogenetic mechanism of specific asthma patterns with the consequent identification of biomarkers and development of effective ad hoc treatment. SUMMARY We are in the middle of an extraordinary revolution of our mode of thinking about and approaching asthma. All the effort in the identification of clusters of patients with different disease clinical patterns, prognosis and response to treatment is closely linked to the identification of endotypes (Th2-low and Th2-high). This approach has allowed the development of the specific treatments (anti IgE, Anti IL5 and IL5R) that are now available and is leading to new ones.
Collapse
|
31
|
A sputum 6-gene signature predicts future exacerbations of poorly controlled asthma. J Allergy Clin Immunol 2019; 144:51-60.e11. [PMID: 30682452 DOI: 10.1016/j.jaci.2018.12.1020] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/17/2018] [Accepted: 12/26/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Improved diagnostic tools for predicting future exacerbation frequency in asthmatic patients are required. A sputum gene expression signature of 6 biomarkers (6-gene signature [6GS], including Charcot-Leyden crystal galectin [CLC]; carboxypeptidase 3 [CPA3]; deoxyribonuclease 1-like 3 [DNASE1L3]; alkaline phosphatase, liver/bone/kidney [ALPL]; CXCR2; and IL1B) predicts inflammatory and treatment response phenotypes in patients with stable asthma. Recently, we demonstrated that azithromycin (AZM) add-on treatment in patients with uncontrolled moderate-to-severe asthma significantly reduced asthma exacerbations (AMAZES clinical trial). OBJECTIVES We sought to test whether the 6GS predicts future exacerbation and inflammatory phenotypes in a subpopulation of AMAZES and to test the effect of AZM therapy on 6GS expression and prognostic capacity. METHODS One hundred forty-two patients (73 placebo-treated and 69 AZM-treated patients) had sputum stored for quantitative PCR of 6GS markers at baseline and after 48 weeks of treatment. Logistic regression and receiver operating characteristic and area under the curve (AUC) determination were performed on baseline measures, and in an exploratory analysis the predictive value of the 6GS was compared with conventional biomarkers for exacerbation and inflammatory phenotypes. RESULTS The 6GS significantly predicted all future exacerbation phenotypes tested. Calculated AUCs for the 6GS were significantly greater than AUCs for peripheral blood eosinophil counts, sputum neutrophil counts, and combined sputum eosinophil and neutrophil counts. 6GS AUCs were also numerically but not significantly greater than those for fractional exhaled nitric oxide values and sputum eosinophil counts. AZM treatment altered neither 6GS expression nor the predictive capacity of the 6GS for future exacerbation phenotypes. The 6GS was a significant predictor of airway inflammatory phenotype in this population. CONCLUSION We demonstrate that a sputum gene signature can predict future exacerbation phenotypes of asthma, with the greatest biomarker performance in identifying those who would experience frequent severe exacerbations. AZM therapy did not modify 6GS expression or biomarker performance, suggesting the therapeutic action of AZM is independent of 6GS-related inflammatory pathways.
Collapse
|
32
|
Hernandez-Pacheco N, Pino-Yanes M, Flores C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front Pediatr 2019; 7:6. [PMID: 30805318 PMCID: PMC6370703 DOI: 10.3389/fped.2019.00006] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
Asthma is a complex respiratory disease considered as the most common chronic condition in children. A large genetic contribution to asthma susceptibility is predicted by the clustering of asthma and allergy symptoms among relatives and the large disease heritability estimated from twin studies, ranging from 55 to 90%. Genetic basis of asthma has been extensively investigated in the past 40 years using linkage analysis and candidate-gene association studies. However, the development of dense arrays for polymorphism genotyping has enabled the transition toward genome-wide association studies (GWAS), which have led the discovery of several unanticipated asthma genes in the last 11 years. Despite this, currently known risk variants identified using many thousand samples from distinct ethnicities only explain a small proportion of asthma heritability. This review examines the main findings of the last 2 years in genomic studies of asthma using GWAS and admixture mapping studies, as well as the direction of studies fostering integrative perspectives involving omics data. Additionally, we discuss the need for assessing the whole spectrum of genetic variation in association studies of asthma susceptibility, severity, and treatment response in order to further improve our knowledge of asthma genes and predictive biomarkers. Leveraging the individual's genetic information will allow a better understanding of asthma pathogenesis and will facilitate the transition toward a more precise diagnosis and treatment.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Maria Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| |
Collapse
|
33
|
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
|
34
|
Implication of fraction of exhaled nitric oxide and blood eosinophil count in severe asthma. Allergol Int 2018; 67S:S3-S11. [PMID: 29754974 DOI: 10.1016/j.alit.2018.04.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/04/2018] [Accepted: 04/07/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Severe asthma is a complex disease with heterogeneous features and involves type 2 airway inflammation, including eosinophil accumulation. Surrogate biomarkers, fraction of exhaled nitric oxide (FeNO) and blood eosinophil count (b-EOS), may predict eosinophilic airway inflammation. Here we investigated clinical characteristics of severe asthma phenotype using a combined analysis of FeNO and b-EOS. METHODS This retrospective study examined clinical data of patients with severe asthma (N = 107; median age, 64 years) treated at Saitama Medical University Hospital from 2009 to 2016. Thresholds of FeNO and b-EOS for sputum eosinophil ratio ≥2% were determined using receiver operating characteristic curve (ROC) analysis. Clinical characteristics were analyzed after classifying patients into four subgroups according to these thresholds. RESULTS Of 39 induced sputum samples examined, ROC area under the curve for predicting sputum eosinophilia was 82.0% (p = 0.001) for b-EOS and 77.0% (p = 0.006) for FeNO at optimal cut-off values of ≥300/μL and ≥25 ppb, respectively. The number of sensitized allergens was higher in the high FeNO/low b-EOS and high FeNO/high b-EOS subgroups (p < 0.05). The prevalence of chronic sinusitis was higher in the low FeNO/high b-EOS and high FeNO/high b-EOS subgroups (p = 0.04). The high FeNO/high b-EOS subgroup included the largest proportion (approximately 40%) of patients experiencing frequent severe exacerbations. Both low FeNO/low b-EOS and high FeNO/low b-EOS subgroups showed less severe exacerbations. CONCLUSIONS Combined evaluation of FeNO and b-EOS can identify patients with frequent exacerbations and stratify the appropriate therapy for type 2 inflammation-predominant severe asthma.
Collapse
|
35
|
Yeh YL, Su MW, Chiang BL, Yang YH, Tsai CH, Lee YL. Genetic profiles of transcriptomic clusters of childhood asthma determine specific severe subtype. Clin Exp Allergy 2018; 48:1164-1172. [PMID: 29758111 DOI: 10.1111/cea.13175] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 04/24/2018] [Accepted: 05/02/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Previous studies have defined transcriptomic subtypes of adult asthma using samples of induced sputum and bronchial epithelium; however, those procedures are not readily applicable in the clinic, especially for childhood asthma. OBJECTIVE We aim to dissect the transcriptomic clusters of childhood asthma using highly variably expressed genes of peripheral blood mononuclear cells (PBMC) among patients. METHODS Gene expression of PBMC from 133 asthmatic children and 11 healthy controls was measured with Illumina microarrays. We applied the k-means clustering algorithm of 2048 genes to assign asthmatic children into clusters. Genes with differential expression between asthma clusters and healthy controls were used to investigate whether they could identify severe asthma of children and adults. RESULTS We identified 3 asthma clusters with distinct inflammatory profiles in peripheral blood. Cluster 1 had the highest eosinophil count. Cluster 2 showed lower counts of both eosinophils and neutrophils. Cluster 3 had the highest neutrophil count and the poorest treatment control. Compared with other patients, Cluster 3 exhibited a unique gene expression pattern which was associated with changes in the glucocorticoid signalling and activation of the T helper 1/T helper 17 (TH 1/TH 17) immune pathways. In the validation studies, an 84-gene signature could identify severe asthma in children on leucocytes, as well as severe asthma in adults on CD8+ T cells. CONCLUSIONS AND CLINICAL RELEVANCE Gene expression profiling of PBMC is useful for the identification of TH 1/TH 17-mediated asthma with poor treatment control. PBMC and CD8+ T cells could be important targets for the investigation and identification of severe asthma.
Collapse
Affiliation(s)
- Y-L Yeh
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - M-W Su
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - B-L Chiang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Y-H Yang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - C-H Tsai
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Y L Lee
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
36
|
Burg D, Schofield JPR, Brandsma J, Staykova D, Folisi C, Bansal A, Nicholas B, Xian Y, Rowe A, Corfield J, Wilson S, Ward J, Lutter R, Fleming L, Shaw DE, Bakke PS, Caruso M, Dahlen SE, Fowler SJ, Hashimoto S, Horváth I, Howarth P, Krug N, Montuschi P, Sanak M, Sandström T, Singer F, Sun K, Pandis I, Auffray C, Sousa AR, Adcock IM, Chung KF, Sterk PJ, Djukanović R, Skipp PJ, The U-Biopred Study Group. Large-Scale Label-Free Quantitative Mapping of the Sputum Proteome. J Proteome Res 2018; 17:2072-2091. [PMID: 29737851 DOI: 10.1021/acs.jproteome.8b00018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Analysis of induced sputum supernatant is a minimally invasive approach to study the epithelial lining fluid and, thereby, provide insight into normal lung biology and the pathobiology of lung diseases. We present here a novel proteomics approach to sputum analysis developed within the U-BIOPRED (unbiased biomarkers predictive of respiratory disease outcomes) international project. We present practical and analytical techniques to optimize the detection of robust biomarkers in proteomic studies. The normal sputum proteome was derived using data-independent HDMSE applied to 40 healthy nonsmoking participants, which provides an essential baseline from which to compare modulation of protein expression in respiratory diseases. The "core" sputum proteome (proteins detected in ≥40% of participants) was composed of 284 proteins, and the extended proteome (proteins detected in ≥3 participants) contained 1666 proteins. Quality control procedures were developed to optimize the accuracy and consistency of measurement of sputum proteins and analyze the distribution of sputum proteins in the healthy population. The analysis showed that quantitation of proteins by HDMSE is influenced by several factors, with some proteins being measured in all participants' samples and with low measurement variance between samples from the same patient. The measurement of some proteins is highly variable between repeat analyses, susceptible to sample processing effects, or difficult to accurately quantify by mass spectrometry. Other proteins show high interindividual variance. We also highlight that the sputum proteome of healthy individuals is related to sputum neutrophil levels, but not gender or allergic sensitization. We illustrate the importance of design and interpretation of disease biomarker studies considering such protein population and technical measurement variance.
Collapse
Affiliation(s)
- Dominic Burg
- Centre for Proteomic Research, Biological Sciences , University of Southampton , Southampton SO17 1BJ , U.K.,NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - James P R Schofield
- Centre for Proteomic Research, Biological Sciences , University of Southampton , Southampton SO17 1BJ , U.K.,NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Joost Brandsma
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Doroteya Staykova
- Centre for Proteomic Research, Biological Sciences , University of Southampton , Southampton SO17 1BJ , U.K
| | - Caterina Folisi
- Centre for Proteomic Research, Biological Sciences , University of Southampton , Southampton SO17 1BJ , U.K
| | | | - Ben Nicholas
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Yang Xian
- Data Science Institute , Imperial College London , London SW7 2AZ , U.K
| | - Anthony Rowe
- Janssen Research & Development , Buckinghamshire HP12 4DP , U.K
| | | | - Susan Wilson
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Jonathan Ward
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Rene Lutter
- AMC, Department of Experimental Immunology , University of Amsterdam , 1012 WX Amsterdam , The Netherlands.,AMC, Department of Respiratory Medicine , University of Amsterdam , 1012 WX Amsterdam , The Netherlands
| | - Louise Fleming
- Airways Disease , National Heart and Lung Institute, Imperial College, London & Royal Brompton NIHR Biomedical Research Unit , London SW7 2AZ , United Kingdom
| | - Dominick E Shaw
- Respiratory Research Unit , University of Nottingham , Nottingham NG7 2RD , U.K
| | - Per S Bakke
- Institute of Medicine , University of Bergen , 5007 Bergen , Norway
| | - Massimo Caruso
- Department of Clinical and Experimental Medicine Hospital University , University of Catania , 95124 Catania , Italy
| | - Sven-Erik Dahlen
- The Centre for Allergy Research , The Institute of Environmental Medicine, Karolinska Institutet , SE-171 77 Stockholm , Sweden
| | - Stephen J Fowler
- Respiratory and Allergy Research Group , University of Manchester , Manchester M13 9PL , U.K
| | - Simone Hashimoto
- Department of Respiratory Medicine, Academic Medical Centre , University of Amsterdam , 1012 WX Amsterdam , The Netherlands
| | - Ildikó Horváth
- Department of Pulmonology , Semmelweis University , Budapest 1085 , Hungary
| | - Peter Howarth
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine Hannover , 30625 Hannover , Germany
| | - Paolo Montuschi
- Faculty of Medicine , Catholic University of the Sacred Heart , 00168 Rome , Italy
| | - Marek Sanak
- Laboratory of Molecular Biology and Clinical Genetics, Medical College , Jagiellonian University , 31-007 Krakow , Poland
| | - Thomas Sandström
- Department of Medicine, Department of Public Health and Clinical Medicine Respiratory Medicine Unit , Umeå University , 901 87 Umeå , Sweden
| | - Florian Singer
- University Children's Hospital Zurich , 8032 Zurich , Switzerland
| | - Kai Sun
- Data Science Institute , Imperial College London , London SW7 2AZ , U.K
| | - Ioannis Pandis
- Data Science Institute , Imperial College London , London SW7 2AZ , U.K
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM , Université de Lyon , 69007 Lyon , France
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GSK , Stockley Park , Uxbridge UB11 1BT , U.K
| | - Ian M Adcock
- Cell and Molecular Biology Group, Airways Disease Section , National Heart and Lung Institute, Imperial College London , Dovehouse Street , London SW3 6LR , U.K
| | - Kian Fan Chung
- Airways Disease , National Heart and Lung Institute, Imperial College, London & Royal Brompton NIHR Biomedical Research Unit , London SW7 2AZ , United Kingdom
| | - Peter J Sterk
- AMC, Department of Experimental Immunology , University of Amsterdam , 1012 WX Amsterdam , The Netherlands
| | - Ratko Djukanović
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine , University of Southampton , Southampton SO16 6YD , U.K
| | - Paul J Skipp
- Centre for Proteomic Research, Biological Sciences , University of Southampton , Southampton SO17 1BJ , U.K
| | | |
Collapse
|
37
|
Jones AC, Troy NM, White E, Hollams EM, Gout AM, Ling KM, Kicic A, Stick SM, Sly PD, Holt PG, Hall GL, Bosco A. Persistent activation of interlinked type 2 airway epithelial gene networks in sputum-derived cells from aeroallergen-sensitized symptomatic asthmatics. Sci Rep 2018; 8:1511. [PMID: 29367592 PMCID: PMC5784090 DOI: 10.1038/s41598-018-19837-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/04/2018] [Indexed: 02/08/2023] Open
Abstract
Atopic asthma is a persistent disease characterized by intermittent wheeze and progressive loss of lung function. The disease is thought to be driven primarily by chronic aeroallergen-induced type 2-associated inflammation. However, the vast majority of atopics do not develop asthma despite ongoing aeroallergen exposure, suggesting additional mechanisms operate in conjunction with type 2 immunity to drive asthma pathogenesis. We employed RNA-Seq profiling of sputum-derived cells to identify gene networks operative at baseline in house dust mite-sensitized (HDMS) subjects with/without wheezing history that are characteristic of the ongoing asthmatic state. The expression of type 2 effectors (IL-5, IL-13) was equivalent in both cohorts of subjects. However, in HDMS-wheezers they were associated with upregulation of two coexpression modules comprising multiple type 2- and epithelial-associated genes. The first module was interlinked by the hubs EGFR, ERBB2, CDH1 and IL-13. The second module was associated with CDHR3 and mucociliary clearance genes. Our findings provide new insight into the molecular mechanisms operative at baseline in the airway mucosa in atopic asthmatics undergoing natural aeroallergen exposure, and suggest that susceptibility to asthma amongst these subjects involves complex interactions between type 2- and epithelial-associated gene networks, which are not operative in equivalently sensitized/exposed atopic non-asthmatics.
Collapse
Affiliation(s)
- Anya C Jones
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia
| | - Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia
| | - Elisha White
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Elysia M Hollams
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Alexander M Gout
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Kak-Ming Ling
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Anthony Kicic
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Stephen M Stick
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, Australia
| | - Peter D Sly
- Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Patrick G Holt
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Graham L Hall
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Physiotherapy and Exercise Science, Curtin University, Perth, Australia.,Centre of Child Health Research, The University of Western Australia, Perth, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.
| |
Collapse
|
38
|
Affiliation(s)
- Klaus F Rabe
- 1 LungenClinic Grosshansdorf and.,2 Department of Medicine, Christian Albrechts University Kiel, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| |
Collapse
|
39
|
Gomez JL, Kaminski N. Toward Precision Medicine of Symptom Control in Asthma. Am J Respir Crit Care Med 2017; 195:147-148. [PMID: 28084831 DOI: 10.1164/rccm.201608-1600ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jose L Gomez
- 1 Department of Internal Medicine Yale University School of Medicine New Haven, Connecticut
| | - Naftali Kaminski
- 1 Department of Internal Medicine Yale University School of Medicine New Haven, Connecticut
| |
Collapse
|
40
|
Svenningsen S, Nair P. Asthma Endotypes and an Overview of Targeted Therapy for Asthma. Front Med (Lausanne) 2017; 4:158. [PMID: 29018800 PMCID: PMC5622943 DOI: 10.3389/fmed.2017.00158] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/12/2017] [Indexed: 12/04/2022] Open
Abstract
Guidelines for the management of severe asthma do not emphasize the measurement of the inflammatory component of airway disease to indicate appropriate treatments or to monitor response to treatment. Inflammation is a central component of asthma and contributes to symptoms, physiological, and structural abnormalities. It can be assessed by a number of endotyping strategies based on “omics” technology such as proteomics, transcriptomics, and metabolomics. It can also be assessed using simple cellular responses by quantitative cytometry in sputum. Bronchitis may be eosinophilic, neutrophilic, mixed-granulocytic, or paucigranulocytic (eosinophils and neutrophils not elevated). Eosinophilic bronchitis is usually a Type 2 (T2)-driven process and therefore a sputum eosinophilia of greater than 3% usually indicates a response to treatment with corticosteroids or novel therapies directed against T2 cytokines such as IL-4, IL-5, and IL-13. Neutrophilic bronchitis represents a non-T2-driven disease, which is generally a predictor of response to antibiotics and may be a predictor to therapies targeted at pathways that lead to neutrophil recruitment such as TNF, IL-1, IL-6, IL-8, IL-23, and IL-17. Paucigranulocytic disease may not warrant anti-inflammatory therapy. These patients, whose symptoms may be driven largely by airway hyper-responsiveness may benefit from smooth muscle-directed therapies such as bronchial thermoplasty or mast-cell directed therapies. This review will briefly summarize the current knowledge regarding “omics-based signatures” and cellular endotyping of severe asthma and give an overview of segmentation of asthma therapeutics guided by the endotype.
Collapse
Affiliation(s)
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,St Joseph's Healthcare, Hamilton, ON, Canada
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW The aims of the present review were to describe the heterogeneous nature of near-fatal asthma (NFA) and to summarize the distinctive phenotypes identified in this subtype of asthma. RECENT FINDINGS Clinical, physiological, and histological studies have shown a large number of triggers, pathological mechanisms, and risk factors associated with NFA. Based on the demographic and clinical characteristics of the patients, the circumstances surrounding the asthma exacerbation and some distinctive features of the disease, several clinical profiles of asthma patients with NFA have been described. Recent data show new associations between some gene expression patterns and fatal asthma, as well as with some biological markers related to inflammatory or immunologic mechanisms in the airways. Also, the use of statistical methods, such as cluster analysis, allowed identifying and confirming different phenotypes of life-threatening asthma patients. SUMMARY NFA is a heterogeneous clinical entity, in which different patients' clinical profiles may coexist [e.g. rapid-onset NFA, NFA in patients with dyspnea hypoperception or sensitized to certain pneumoallergens (Alternaria alternata, soybean), NFA related to the menstrual cycle, brittle asthma]. Knowledge of these phenotypes as well as adequate and specific management strategies can reduce morbidity and mortality in patients suffering from NFA.
Collapse
|
42
|
Gaurav R, Varasteh JT, Weaver MR, Jacobson SR, Hernandez-Lagunas L, Liu Q, Nozik-Grayck E, Chu HW, Alam R, Nordestgaard BG, Kobylecki CJ, Afzal S, Chupp GL, Bowler RP. The R213G polymorphism in SOD3 protects against allergic airway inflammation. JCI Insight 2017; 2:95072. [PMID: 28878123 DOI: 10.1172/jci.insight.95072] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/03/2017] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress is important in the pathogenesis of allergic asthma. Extracellular superoxide dismutase (EC-SOD; SOD3) is the major antioxidant in lungs, but its role in allergic asthma is unknown. Here we report that asthmatics have increased SOD3 transcript levels in sputum and that a single nucleotide polymorphism (SNP) in SOD3 (R213G; rs1799895) changes lung distribution of EC-SOD, and decreases likelihood of asthma-related symptoms. Knockin mice analogous to the human R213G SNP had lower airway hyperresponsiveness, inflammation, and mucus hypersecretion with decreased interleukin-33 (IL-33) in bronchoalveolar lavage fluid and reduced type II innate lymphoid cells (ILC2s) in lungs. SOD mimetic (Mn (III) tetrakis (N-ethylpyridinium-2-yl) porphyrin) attenuated Alternaria-induced expression of IL-33 and IL-8 release in BEAS-2B cells. These results suggest that R213G SNP potentially benefits its carriers by resulting in high EC-SOD in airway-lining fluid, which ameliorates allergic airway inflammation by dampening the innate immune response, including IL-33/ST2-mediated changes in ILC2s.
Collapse
Affiliation(s)
- Rohit Gaurav
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Jason T Varasteh
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Michael R Weaver
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Sean R Jacobson
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Laura Hernandez-Lagunas
- Cardiovascular Pulmonary Research Laboratories and Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Qing Liu
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | - Eva Nozik-Grayck
- Cardiovascular Pulmonary Research Laboratories and Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Rafeul Alam
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, and.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Shoaib Afzal
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, and.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Geoffrey L Chupp
- Department of Internal Medicine, Yale University, New Haven, Connecticut, USA
| | - Russell P Bowler
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
43
|
Kuo CHS, Pavlidis S, Loza M, Baribaud F, Rowe A, Pandis I, Hoda U, Rossios C, Sousa A, Wilson SJ, Howarth P, Dahlen B, Dahlen SE, Chanez P, Shaw D, Krug N, Sandstrӧm T, De Meulder B, Lefaudeux D, Fowler S, Fleming L, Corfield J, Auffray C, Sterk PJ, Djukanovic R, Guo Y, Adcock IM, Chung KF. A Transcriptome-driven Analysis of Epithelial Brushings and Bronchial Biopsies to Define Asthma Phenotypes in U-BIOPRED. Am J Respir Crit Care Med 2017; 195:443-455. [PMID: 27580351 DOI: 10.1164/rccm.201512-2452oc] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
RATIONALE Asthma is a heterogeneous disease driven by diverse immunologic and inflammatory mechanisms. OBJECTIVES Using transcriptomic profiling of airway tissues, we sought to define the molecular phenotypes of severe asthma. METHODS The transcriptome derived from bronchial biopsies and epithelial brushings of 107 subjects with moderate to severe asthma were annotated by gene set variation analysis using 42 gene signatures relevant to asthma, inflammation, and immune function. Topological data analysis of clinical and histologic data was performed to derive clusters, and the nearest shrunken centroid algorithm was used for signature refinement. MEASUREMENTS AND MAIN RESULTS Nine gene set variation analysis signatures expressed in bronchial biopsies and airway epithelial brushings distinguished two distinct asthma subtypes associated with high expression of T-helper cell type 2 cytokines and lack of corticosteroid response (group 1 and group 3). Group 1 had the highest submucosal eosinophils, as well as high fractional exhaled nitric oxide levels, exacerbation rates, and oral corticosteroid use, whereas group 3 patients showed the highest levels of sputum eosinophils and had a high body mass index. In contrast, group 2 and group 4 patients had an 86% and 64% probability, respectively, of having noneosinophilic inflammation. Using machine learning tools, we describe an inference scheme using the currently available inflammatory biomarkers sputum eosinophilia and fractional exhaled nitric oxide levels, along with oral corticosteroid use, that could predict the subtypes of gene expression within bronchial biopsies and epithelial cells with good sensitivity and specificity. CONCLUSIONS This analysis demonstrates the usefulness of a transcriptomics-driven approach to phenotyping that segments patients who may benefit the most from specific agents that target T-helper cell type 2-mediated inflammation and/or corticosteroid insensitivity.
Collapse
Affiliation(s)
- Chih-Hsi Scott Kuo
- 1 Department of Computing.,2 Data Science Institute, and.,3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stelios Pavlidis
- 1 Department of Computing.,2 Data Science Institute, and.,4 Janssen Research and Development, High Wycombe, United Kingdom
| | - Matthew Loza
- 4 Janssen Research and Development, High Wycombe, United Kingdom
| | - Fred Baribaud
- 4 Janssen Research and Development, High Wycombe, United Kingdom
| | - Anthony Rowe
- 4 Janssen Research and Development, High Wycombe, United Kingdom
| | - Ioannis Pandis
- 1 Department of Computing.,2 Data Science Institute, and
| | - Uruj Hoda
- 3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,5 Biomedical Research Unit, Royal Brompton & Harefield National Health Service Trust, London, United Kingdom
| | - Christos Rossios
- 3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ana Sousa
- 6 Respiratory Therapeutic Unit, GlaxoSmithKline, Stockley Park, United Kingdom
| | - Susan J Wilson
- 7 Faculty of Medicine, Southampton University, Southampton, United Kingdom
| | - Peter Howarth
- 7 Faculty of Medicine, Southampton University, Southampton, United Kingdom
| | - Barbro Dahlen
- 8 Centre for Allergy Research, Karolinska Institute, Stockholm, Sweden
| | - Sven-Erik Dahlen
- 8 Centre for Allergy Research, Karolinska Institute, Stockholm, Sweden
| | | | - Dominick Shaw
- 10 Centre for Respiratory Research, University of Nottingham, Nottingham, United Kingdom
| | - Norbert Krug
- 11 Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Thomas Sandstrӧm
- 12 Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bertrand De Meulder
- 13 European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, University of Lyon, Lyon, France
| | - Diane Lefaudeux
- 13 European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, University of Lyon, Lyon, France
| | - Stephen Fowler
- 14 Centre for Respiratory Medicine and Allergy, University of Manchester, Manchester, United Kingdom
| | - Louise Fleming
- 3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,5 Biomedical Research Unit, Royal Brompton & Harefield National Health Service Trust, London, United Kingdom
| | - Julie Corfield
- 15 AstraZeneca R&D, Molndal, Sweden.,16 Areteva R&D, Nottingham, United Kingdom; and
| | - Charles Auffray
- 13 European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL, University of Lyon, Lyon, France
| | - Peter J Sterk
- 17 Faculty of Medicine, University of Amsterdam, Amsterdam, the Netherlands
| | - Ratko Djukanovic
- 7 Faculty of Medicine, Southampton University, Southampton, United Kingdom
| | - Yike Guo
- 1 Department of Computing.,2 Data Science Institute, and
| | - Ian M Adcock
- 3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,5 Biomedical Research Unit, Royal Brompton & Harefield National Health Service Trust, London, United Kingdom
| | - Kian Fan Chung
- 3 Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,5 Biomedical Research Unit, Royal Brompton & Harefield National Health Service Trust, London, United Kingdom
| | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Asthma is heterogeneous with different endotypes/phenotypes. Response to corticosteroids is variable and novel biological therapies are proving useful. Biomarkers allow individualization of treatment. This review provides an update on available data regarding asthma biomarkers with focus on their utility for prediction of response to steroidal and new biological therapies. RECENT FINDINGS Blood eosinophils are a biomarker with acceptable accuracy as a surrogate for sputum eosinophilia, are associated with relevant outcomes, and are more readily measureable. New evidence supports fraction of exhaled nitric oxide (FENO)-based treatment algorithms for cost-effective maintenance of asthma control/quality of life. Serum and sputum-derived periostin are biomarkers of lung function decline and associated with eosinophilic airway inflammation. Transcriptomics show promise for endotyping; their role in management remains to be determined. Biomarker panels may improve predictive value as shown for the combination of FENO/urinary bromotyrosine in prediction of steroid responsiveness. Novel biological therapies are proving effective in biomarker-selected populations. SUMMARY Biomarkers including blood eosinophils and FENO are proving to have utility for the effective administration of steroidal and novel biological therapies in asthma, allowing individualized treatment. Transcriptomics can discriminate subtypes of asthma and may have a role in delivery of individualized therapy.
Collapse
|
45
|
The Reemergence of the Asthma-COPD Overlap Syndrome: Characterizing a Syndrome in the Precision Medicine Era. Curr Allergy Asthma Rep 2017; 16:81. [PMID: 27796796 DOI: 10.1007/s11882-016-0660-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE OF REVIEW The asthma-COPD overlap syndrome (ACOS) has reemerged in the medical literature. This review addresses our current understanding of ACOS as a clinical and biological entity and how new and existing therapies may be targeted to this group. RECENT FINDINGS Many studies suggest that ACOS is common and associated with more morbidity than asthma and COPD in general. However, there is no consensus on an ACOS definition, likely due to the heterogeneity of the disease. Variable definitions have led to variable results in ACOS studies. Given this clinical variability, biomarkers (e.g., eosinophils and type 2 inflammatory markers) are increasingly being used to identify an ACOS molecular phenotype which appears to be more responsive to inhaled corticosteroids. Although ACOS has become a popular diagnosis, it is unclear whether identifying ACOS as a separate disease entity is clinically useful. Future studies should focus on identifying key clinical features and biomarkers that characterize vulnerable and treatment-responsive patients.
Collapse
|
46
|
Peters MC, Nguyen MLT, Dunican EM. Biomarkers of Airway Type-2 Inflammation and Integrating Complex Phenotypes to Endotypes in Asthma. Curr Allergy Asthma Rep 2017; 16:71. [PMID: 27613654 DOI: 10.1007/s11882-016-0651-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Over the past decade, the most important advance in the field of asthma has been the widespread recognition that asthma is a heterogeneous disease driven by multiple molecular processes. RECENT FINDINGS The most well-established molecular mechanism in asthma is increased airway type-2 inflammation, and consequently, non-invasive biomarkers of increased airway type-2 inflammation, such as blood eosinophil counts or blood periostin levels, have proven important in stratifying asthma patients in clinical trials of type-2 cytokine inhibitors. However, it remains ambiguous how well these non-invasive biomarkers represent airway measures of type-2 inflammation in asthma. As a result, the utility of these biomarkers to assist with asthma management or as research tools to better understand asthma pathogenesis remains unclear. This article reviews primary data assessing biomarkers of airway type-2 inflammation in asthma and describes how the use of biomarkers can advance a precision medicine approach to asthma treatment.
Collapse
Affiliation(s)
- Michael C Peters
- The Airway Clinical Research Center, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA. .,Division of Pulmonary and Critical Care Medicine, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA. .,Cardiovascular Research Institute, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA.
| | | | - Eleanor M Dunican
- The Airway Clinical Research Center, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA.,Division of Pulmonary and Critical Care Medicine, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA.,Cardiovascular Research Institute, University of California, Box 0130, 505 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
47
|
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
|
48
|
Yan X, Liang A, Gomez J, Cohn L, Zhao H, Chupp GL. A novel pathway-based distance score enhances assessment of disease heterogeneity in gene expression. BMC Bioinformatics 2017. [PMID: 28637421 PMCID: PMC5480187 DOI: 10.1186/s12859-017-1727-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Distance based unsupervised clustering of gene expression data is commonly used to identify heterogeneity in biologic samples. However, high noise levels in gene expression data and relatively high correlation between genes are often encountered, so traditional distances such as Euclidean distance may not be effective at discriminating the biological differences between samples. An alternative method to examine disease phenotypes is to use pre-defined biological pathways. These pathways have been shown to be perturbed in different ways in different subjects who have similar clinical features. We hypothesize that differences in the expressions of genes in a given pathway are more predictive of differences in biological differences compared to standard approaches and if integrated into clustering analysis will enhance the robustness and accuracy of the clustering method. To examine this hypothesis, we developed a novel computational method to assess the biological differences between samples using gene expression data by assuming that ontologically defined biological pathways in biologically similar samples have similar behavior. RESULTS Pre-defined biological pathways were downloaded and genes in each pathway were used to cluster samples using the Gaussian mixture model. The clustering results across different pathways were then summarized to calculate the pathway-based distance score between samples. This method was applied to both simulated and real data sets and compared to the traditional Euclidean distance and another pathway-based clustering method, Pathifier. The results show that the pathway-based distance score performs significantly better than the Euclidean distance, especially when the heterogeneity is low and genes in the same pathways are correlated. Compared to Pathifier, we demonstrated that our approach achieves higher accuracy and robustness for small pathways. When the pathway size is large, by downsampling the pathways into smaller pathways, our approach was able to achieve comparable performance. CONCLUSIONS We have developed a novel distance score that represents the biological differences between samples using gene expression data and pre-defined biological pathway information. Application of this distance score results in more accurate, robust, and biologically meaningful clustering results in both simulated data and real data when compared to traditional methods. It also has comparable or better performance compared to Pathifier.
Collapse
Affiliation(s)
- Xiting Yan
- Center for Pulmonary Personalized Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA. .,Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA.
| | - Anqi Liang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA
| | - Jose Gomez
- Center for Pulmonary Personalized Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Lauren Cohn
- Center for Pulmonary Personalized Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Hongyu Zhao
- Center for Pulmonary Personalized Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA.,Department of Biostatistics, Yale School of Public Health, New Haven, CT, 06520, USA.,Department of Genetics, Yale School of Medicine, New Haven, CT, 06520, USA.,Computational Biology and Bioinformatics Program, Yale School of Medicine, New Haven, CT, 06520, USA
| | - Geoffrey L Chupp
- Center for Pulmonary Personalized Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06520, USA
| |
Collapse
|
49
|
Lefaudeux D, De Meulder B, Loza MJ, Peffer N, Rowe A, Baribaud F, Bansal AT, Lutter R, Sousa AR, Corfield J, Pandis I, Bakke PS, Caruso M, Chanez P, Dahlén SE, Fleming LJ, Fowler SJ, Horvath I, Krug N, Montuschi P, Sanak M, Sandstrom T, Shaw DE, Singer F, Sterk PJ, Roberts G, Adcock IM, Djukanovic R, Auffray C, Chung KF, Adriaens N, Ahmed H, Aliprantis A, Alving K, Badorek P, Balgoma D, Barber C, Bautmans A, Behndig AF, Bel E, Beleta J, Berglind A, Berton A, Bigler J, Bisgaard H, Bochenek G, Boedigheimer MJ, Bøonnelykke K, Brandsma J, Braun A, Brinkman P, Burg D, Campagna D, Carayannopoulos L, Carvalho da Purfição Rocha JP, Chaiboonchoe A, Chaleckis R, Coleman C, Compton C, D'Amico A, Dahlén B, De Alba J, de Boer P, De Lepeleire I, Dekker T, Delin I, Dennison P, Dijkhuis A, Draper A, Edwards J, Emma R, Ericsson M, Erpenbeck V, Erzen D, Faulenbach C, Fichtner K, Fitch N, Flood B, Frey U, Gahlemann M, Galffy G, Gallart H, Garret T, Geiser T, Gent J, Gerhardsson de Verdier M, Gibeon D, Gomez C, Gove K, Gozzard N, Guo YK, Hashimoto S, Haughney J, Hedlin G, Hekking PP, Henriksson E, Hewitt L, Higgenbottam T, Hoda U, Hohlfeld J, et alLefaudeux D, De Meulder B, Loza MJ, Peffer N, Rowe A, Baribaud F, Bansal AT, Lutter R, Sousa AR, Corfield J, Pandis I, Bakke PS, Caruso M, Chanez P, Dahlén SE, Fleming LJ, Fowler SJ, Horvath I, Krug N, Montuschi P, Sanak M, Sandstrom T, Shaw DE, Singer F, Sterk PJ, Roberts G, Adcock IM, Djukanovic R, Auffray C, Chung KF, Adriaens N, Ahmed H, Aliprantis A, Alving K, Badorek P, Balgoma D, Barber C, Bautmans A, Behndig AF, Bel E, Beleta J, Berglind A, Berton A, Bigler J, Bisgaard H, Bochenek G, Boedigheimer MJ, Bøonnelykke K, Brandsma J, Braun A, Brinkman P, Burg D, Campagna D, Carayannopoulos L, Carvalho da Purfição Rocha JP, Chaiboonchoe A, Chaleckis R, Coleman C, Compton C, D'Amico A, Dahlén B, De Alba J, de Boer P, De Lepeleire I, Dekker T, Delin I, Dennison P, Dijkhuis A, Draper A, Edwards J, Emma R, Ericsson M, Erpenbeck V, Erzen D, Faulenbach C, Fichtner K, Fitch N, Flood B, Frey U, Gahlemann M, Galffy G, Gallart H, Garret T, Geiser T, Gent J, Gerhardsson de Verdier M, Gibeon D, Gomez C, Gove K, Gozzard N, Guo YK, Hashimoto S, Haughney J, Hedlin G, Hekking PP, Henriksson E, Hewitt L, Higgenbottam T, Hoda U, Hohlfeld J, Holweg C, Howarth P, Hu R, Hu S, Hu X, Hudson V, James AJ, Kamphuis J, Kennington EJ, Kerry D, Klüglich M, Knobel H, Knowles R, Knox A, Kolmert J, Konradsen J, Kots M, Krueger L, Kuo S, Kupczyk M, Lambrecht B, Lantz AS, Larsson L, Lazarinis N, Lone-Satif S, Marouzet L, Martin J, Masefield S, Mathon C, Matthews JG, Mazein A, Meah S, Maiser A, Menzies-Gow A, Metcalf L, Middelveld R, Mikus M, Miralpeix M, Monk P, Mores N, Murray CS, Musial J, Myles D, Naz S, Nething K, Nicholas B, Nihlen U, Nilsson P, Nordlund B, Östling J, Pacino A, Pahus L, Palkonnen S, Pavlidis S, Pennazza G, Petrén A, Pink S, Postle A, Powel P, Rahman-Amin M, Rao N, Ravanetti L, Ray E, Reinke S, Reynolds L, Riemann K, Riley J, Robberechts M, Roberts A, Rossios C, Russell K, Rutgers M, Santini G, Sentoninco M, Schoelch C, Schofield JP, Seibold W, Sigmund R, Sjödin M, Skipp PJ, Smids B, Smith C, Smith J, Smith KM, Söderman P, Sogbesan A, Staykova D, Strandberg K, Sun K, Supple D, Szentkereszty M, Tamasi L, Tariq K, Thörngren JO, Thornton B, Thorsen J, Valente S, van Aalderenm W, van de Pol M, van Drunen K, van Geest M, Versnel J, Vestbo J, Vink A, Vissing N, von Garnier C, Wagerner A, Wagers S, Wald F, Walker S, Ward J, Weiszhart Z, Wetzel K, Wheelock CE, Wiegman C, Williams S, Wilson SJ, Woosdcock A, Yang X, Yeyashingham E, Yu W, Zetterquist W, Zwinderman K. U-BIOPRED clinical adult asthma clusters linked to a subset of sputum omics. J Allergy Clin Immunol 2017; 139:1797-1807. [DOI: 10.1016/j.jaci.2016.08.048] [Show More Authors] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 07/23/2016] [Accepted: 08/08/2016] [Indexed: 01/20/2023]
|
50
|
Croteau-Chonka DC, Qiu W, Martinez FD, Strunk RC, Lemanske RF, Liu AH, Gilliland FD, Millstein J, Gauderman WJ, Ober C, Krishnan JA, White SR, Naureckas ET, Nicolae DL, Barnes KC, London SJ, Barraza-Villarreal A, Carey VJ, Weiss ST, Raby BA. Gene Expression Profiling in Blood Provides Reproducible Molecular Insights into Asthma Control. Am J Respir Crit Care Med 2017; 195:179-188. [PMID: 27494826 PMCID: PMC5394783 DOI: 10.1164/rccm.201601-0107oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Maintaining optimal symptom control remains the primary objective of asthma treatment. Better understanding of the biologic underpinnings of asthma control may lead to the development of improved clinical and pharmaceutical approaches. OBJECTIVES To identify molecular pathways and interrelated genes whose differential expression was associated with asthma control. METHODS We performed gene set enrichment analyses of asthma control in 1,170 adults with asthma, each with gene expression data derived from either whole blood (WB) or unstimulated CD4+ T lymphocytes (CD4), and a self-reported asthma control score representing either the preceding 6 months (chronic) or 7 days (acute). Our study comprised a discovery WB cohort (n = 245, chronic) and three independent, nonoverlapping replication cohorts: a second WB set (n = 448, acute) and two CD4 sets (n = 300, chronic; n = 77, acute). MEASUREMENTS AND MAIN RESULTS In the WB discovery cohort, we found significant overrepresentation of genes associated with asthma control in 1,106 gene sets from the Molecular Signatures Database (false discovery rate, <5%). Of these, 583 (53%) replicated in at least one replication cohort (false discovery rate, <25%). Suboptimal control was associated with signatures of eosinophilic and granulocytic inflammatory signals, whereas optimal control signatures were enriched for immature lymphocytic patterns. These signatures included two related biologic processes related to activation by TREM-1 (triggering receptor expressed on myeloid cells 1) and lipopolysaccharide. CONCLUSIONS Together, these results demonstrate the existence of specific, reproducible transcriptomic components in blood that vary with degree of asthma control and implicate a novel biologic target (TREM-1).
Collapse
Affiliation(s)
| | | | - Fernando D. Martinez
- Arizona Respiratory Center and BIO5 Institute, University of Arizona, Tucson, Arizona
| | - Robert C. Strunk
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Robert F. Lemanske
- University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Andrew H. Liu
- Division of Allergy and Clinical Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado
| | | | - Joshua Millstein
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - W. James Gauderman
- Division of Biostatistics, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Jerry A. Krishnan
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Steven R. White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | - Dan L. Nicolae
- Department of Human Genetics
- Section of Genetic Medicine, Department of Medicine, and
- Department of Statistics, University of Chicago, Chicago, Illinois
| | - Kathleen C. Barnes
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Stephanie J. London
- Division of Intramural Research, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | | | | | - Scott T. Weiss
- Channing Division of Network Medicine and
- Partners HealthCare Personalized Medicine, Partners Health Care, Boston, Massachusetts
| | - Benjamin A. Raby
- Channing Division of Network Medicine and
- Pulmonary Genetics Center and Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|