1
|
Li JX, Huang XZ, Fu WP, Zhang XH, Mauki DH, Zhang J, Sun C, Dai LM, Zhong L, Yu L, Zhang YP. Remote regulation of rs80245547 and rs72673891 mediated by transcription factors C-Jun and CREB1 affect GSTCD expression. iScience 2023; 26:107383. [PMID: 37609638 PMCID: PMC10440715 DOI: 10.1016/j.isci.2023.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/30/2022] [Accepted: 07/11/2023] [Indexed: 08/24/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD), the third leading cause of death worldwide, is influenced by genetic factors. The genetic signal rs10516526 in the glutathione S-transferase C-terminal domain containing (GSTCD) gene is a highly significant and reproducible signal associated with lung function and COPD on chromosome 4q24. In this study, comprehensive bioinformatics analyses and experimental verifications were detailly implemented to explore the regulation mechanism of rs10516526 and GSTCD in COPD. The results suggested that low expression of GSTCD was associated with COPD (p = 0.010). And C-Jun and CREB1 transcription factors were found to be essential for the regulation of GSTCD by rs80245547 and rs72673891. Moreover, rs80245547T and rs72673891G had a stronger binding ability to these transcription factors, which may promote the allele-specific long-range enhancer-promoter interactions on GSTCD, thus making COPD less susceptible. Our study provides a new insight into the relationship between rs10516526, GSTCD, and COPD.
Collapse
Affiliation(s)
- Jin-Xiu Li
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650000, China
| | - Xue-Zhen Huang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - Wei-ping Fu
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Xiao-hua Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - David H. Mauki
- Faculty of Pharmaceutical Sciences, Institute of Biomedicine and Biotechnology, Center for Cancer Immunology, Chinese Academy of Sciences, Shenzhen Institute of Advanced Technology, Shenzhen 518000, Guangdong China
| | - Jing Zhang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Chang Sun
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- College of Life Sciences, Shaanxi Normal University, Xi’an 710000, China
| | - Lu-Ming Dai
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650000, China
| | - Li Zhong
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- College of Life Sciences, Shaanxi Normal University, Xi’an 710000, China
- Provincial Demonstration Center for Experimental Biology Education, Shaanxi Normal University, Xi’an 710000, China
| | - Li Yu
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
| | - Ya-ping Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resource in Yunnan, School of Life Sciences, Yunnan University, Kunming 650000, China
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650000, China
| |
Collapse
|
2
|
HLA-G in asthma and its potential as an effective therapeutic agent. Allergol Immunopathol (Madr) 2023; 51:22-29. [PMID: 36617818 DOI: 10.15586/aei.v51i1.650] [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: 04/13/2022] [Accepted: 07/06/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Asthma is a heterogeneous disease. Severity of asthma and sensitivity to medications vary across asthma subtypes. Human leukocyte antigen (HLA)-G has a wide range of functions in normal and pathological physiology. Due to its powerful immune function, HLA-G participates in the pathogenesis of different asthma phenotypes by regulating the activity and function of various immune cells. The mechanism of HLA-G in asthma is not fully clear, and there is no consensus on its mechanism in asthma. Further studies are needed to explore the role of HLA-G in different phenotypes of human asthma. METHODS Observational study. RESULTS HLA-G is an important immunomodulatory factor in asthma. Studies have found different levels of HLA-G in patients with different asthma subtypes and healthy controls, but other studies have come to the opposite conclusion. CONCLUSION We speculate that further study on the mechanism of HLA-G in asthma pheno-types may explain some of the contradictions in current studies. Findings should provide information regarding the potential of HLA-G as a novel target for asthma diagnosis and treatment.
Collapse
|
3
|
Fiouane S, Chebbo M, Beley S, Paganini J, Picard C, D'Journo X, Thomas P, Chiaroni J, Chanez P, Gras D, Di Cristofaro J. Mobilisation of HLA-F on the surface of bronchial epithelial cells and platelets in asthmatic patients. HLA 2022; 100:491-499. [PMID: 35988034 PMCID: PMC9804204 DOI: 10.1111/tan.14782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/26/2022] [Accepted: 08/16/2022] [Indexed: 01/05/2023]
Abstract
Uncontrolled inflammation of the airways in chronic obstructive lung diseases leads to exacerbation, accelerated lung dysfunction and respiratory insufficiency. Among these diseases, asthma affects 358 million people worldwide. Human bronchial epithelium cells (HBEC) express both anti-inflammatory and activating molecules, and their deregulated expression contribute to immune cell recruitment and activation, especially platelets (PLT) particularly involved in lung tissue inflammation in asthma context. Previous results supported that HLA-G dysregulation in lung tissue is associated with immune cell activation. We investigated here HLA-F expression, reported to be mobilised on immune cell surface upon activation and displaying its highest affinity for the KIR3DS1-activating NK receptor. We explored HLA-F transcriptional expression in HBEC; HLA-F total expression in PBMC and HBEC collected from healthy individuals at rest and upon chemical activation and HLA-F membrane expression in PBMC, HBEC and PLT collected from healthy individuals at rest and upon chemical activation. We compared HLA-F transcriptional expression in HBEC from healthy individuals and asthmatic patients and its surface expression in HBEC and PLT from healthy individuals and asthmatic patients. Our results support that HLA-F is expressed by HBEC and PLT under healthy physiological conditions and is retained in cytoplasm, barely expressed on the surface, as previously reported in immune cells. In both cell types, HLA-F reaches the surface in the inflammatory asthma context whereas no effect is observed at the transcriptional level. Our study suggests that HLA-F surface expression is a ubiquitous post-transcriptional process in activated cells. It may be of therapeutic interest in controlling lung inflammation.
Collapse
Affiliation(s)
- Sabrina Fiouane
- CNRS, EFS, ADES, UMR7268Aix Marseille UniversityMarseilleFrance,Etablissement Français du Sang PACA CorseMarseilleFrance
| | - Mohamad Chebbo
- INSERM 1263, INRAE 1260, C2VNAix Marseille UniversityMarseilleFrance
| | - Sophie Beley
- CNRS, EFS, ADES, UMR7268Aix Marseille UniversityMarseilleFrance,Etablissement Français du Sang PACA CorseMarseilleFrance
| | | | - Christophe Picard
- CNRS, EFS, ADES, UMR7268Aix Marseille UniversityMarseilleFrance,Etablissement Français du Sang PACA CorseMarseilleFrance
| | - Xavier‐Benoît D'Journo
- Department of Thoracic Surgery, North HospitalAix‐Marseille University and Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
| | - Pascal‐Alexandre Thomas
- Department of Thoracic Surgery, North HospitalAix‐Marseille University and Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
| | - Jacques Chiaroni
- CNRS, EFS, ADES, UMR7268Aix Marseille UniversityMarseilleFrance,Etablissement Français du Sang PACA CorseMarseilleFrance
| | - Pascal Chanez
- INSERM 1263, INRAE 1260, C2VNAix Marseille UniversityMarseilleFrance,Clinique des Bronches, Allergies et SommeilNorth Hospital, Assistance Publique‐Hôpitaux de MarseilleMarseilleFrance
| | - Delphine Gras
- INSERM 1263, INRAE 1260, C2VNAix Marseille UniversityMarseilleFrance
| | - Julie Di Cristofaro
- CNRS, EFS, ADES, UMR7268Aix Marseille UniversityMarseilleFrance,Etablissement Français du Sang PACA CorseMarseilleFrance
| |
Collapse
|
4
|
Lin NW, Liu C, Yang IV, Maier LA, DeMeo DL, Wood C, Ye S, Cruse MH, Smith VL, Vyhlidal CA, Kechris K, Sharma S. Sex-Specific Differences in MicroRNA Expression During Human Fetal Lung Development. Front Genet 2022; 13:762834. [PMID: 35480332 PMCID: PMC9037032 DOI: 10.3389/fgene.2022.762834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/05/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Sex-specific differences in fetal lung maturation have been well described; however, little is known about the sex-specific differences in microRNA (miRNA) expression during human fetal lung development. Interestingly, many adult chronic lung diseases also demonstrate sex-specific differences in prevalence. The developmental origins of health and disease hypothesis suggests that these sex-specific differences in fetal lung development may influence disease susceptibility later in life. In this study, we performed miRNA sequencing on human fetal lung tissue samples to investigate differential expression of miRNAs between males and females in the pseudoglandular stage of lung development. We hypothesized that differences in miRNA expression are present between sexes in early human lung development and may contribute to the sex-specific differences seen in pulmonary diseases later in life. Methods: RNA was isolated from human fetal lung tissue samples for miRNA sequencing. The count of each miRNA was modeled by sex using negative binomial regression models in DESeq2, adjusting for post-conception age, age2, smoke exposure, batch, and RUV factors. We tested for differential expression of miRNAs by sex, and for the presence of sex-by-age interactions to determine if miRNA expression levels by age were distinct between males and females. Results: miRNA expression profiles were generated on 298 samples (166 males and 132 females). Of the 809 miRNAs expressed in human fetal lung tissue during the pseudoglandular stage of lung development, we identified 93 autosomal miRNAs that were significantly differentially expressed by sex and 129 miRNAs with a sex-specific pattern of miRNA expression across the course of the pseudoglandular period. Conclusion: Our study demonstrates differential expression of numerous autosomal miRNAs between the male and female developing human lung. Additionally, the expression of some miRNAs are modified by age across the pseudoglandular stage in a sex-specific way. Some of these differences in miRNA expression may impact susceptibility to pulmonary disease later in life. Our results suggest that sex-specific miRNA expression during human lung development may be a potential mechanism to explain sex-specific differences in lung development and may impact subsequent disease susceptibility.
Collapse
Affiliation(s)
- Nancy W. Lin
- Division of Environmental and Occupational Health, National Jewish Health, Denver, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Cuining Liu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Ivana V. Yang
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lisa A. Maier
- Division of Environmental and Occupational Health, National Jewish Health, Denver, CO, United States
- Environmental and Occupational Health, Colorado School of Public Health, Aurora, CO, United States
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, United States
| | - Cheyret Wood
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Shuyu Ye
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Margaret H. Cruse
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | - Vong L. Smith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| | | | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
5
|
Portelli MA, Rakkar K, Hu S, Guo Y, Adcock IM, Sayers I. Translational Analysis of Moderate to Severe Asthma GWAS Signals Into Candidate Causal Genes and Their Functional, Tissue-Dependent and Disease-Related Associations. FRONTIERS IN ALLERGY 2022; 2:738741. [PMID: 35386986 PMCID: PMC8974692 DOI: 10.3389/falgy.2021.738741] [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: 07/09/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Asthma affects more than 300 million people globally and is both under diagnosed and under treated. The most recent and largest genome-wide association study investigating moderate to severe asthma to date was carried out in 2019 and identified 25 independent signals. However, as new and in-depth downstream databases become available, the translational analysis of these signals into target genes and pathways is timely. In this study, unique (U-BIOPRED) and publicly available datasets (HaploReg, Open Target Genetics and GTEx) were investigated for the 25 GWAS signals to identify 37 candidate causal genes. Additional traits associated with these signals were identified through PheWAS using the UK Biobank resource, with asthma and eosinophilic traits amongst the strongest associated. Gene expression omnibus dataset examination identified 13 candidate genes with altered expression profiles in the airways and blood of asthmatic subjects, including MUC5AC and STAT6. Gene expression analysis through publicly available datasets highlighted lung tissue cell specific expression, with both MUC5AC and SLC22A4 genes showing enriched expression in ciliated cells. Gene enrichment pathway and interaction analysis highlighted the dominance of the HLA-DQA1/A2/B1/B2 gene cluster across many immunological diseases including asthma, type I diabetes, and rheumatoid arthritis. Interaction and prediction analyses found IL33 and IL18R1 to be key co-localization partners for other genes, predicted that CD274 forms co-expression relationships with 13 other genes, including the HLA-DQA1/A2/B1/B2 gene cluster and that MUC5AC and IL37 are co-expressed. Drug interaction analysis revealed that 11 of the candidate genes have an interaction with available therapeutics. This study provides significant insight into these GWAS signals in the context of cell expression, function, and disease relationship with the view of informing future research and drug development efforts for moderate-severe asthma.
Collapse
Affiliation(s)
- Michael A Portelli
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Kamini Rakkar
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Sile Hu
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Yike Guo
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- The National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian Sayers
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, National Institute for Health Research Nottingham Biomedical Research Centre, Nottingham University Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
6
|
Hsu CG, Fazal F, Rahman A, Berk BC, Yan C. Phosphodiesterase 10A Is a Key Mediator of Lung Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:3010-3020. [PMID: 34117108 PMCID: PMC8664899 DOI: 10.4049/jimmunol.2001026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 04/08/2021] [Indexed: 11/19/2022]
Abstract
Cyclic nucleotides cAMP and cGMP are important regulators of immune cell functions. Phosphodiesterases (PDEs) hydrolyze cAMP and/or cGMP and, thus, play crucial roles in cyclic nucleotide homeostasis. Abnormal alterations of PDE expression have been implicated in several diseases. To understand the function of PDEs in macrophages, we screened for all PDE genes in both peritoneal and alveolar macrophages from C57BL/6J mice and found that PDE4B and PDE10A are highly induced by LPS. A number of PDE4 inhibitors have been used clinically for the treatment of inflammatory lung diseases. However, the role of PDE10A in inflammation is still poorly understood. We therefore investigated the role of PDE10A in macrophage inflammatory response in vitro and acute lung inflammation in vivo. We found that LPS induces a sustained PDE10A expression in macrophages, which is different from a transient induction by PDE4B. PDE10A inhibition blocked LPS-induced MCP-1 expression, but not TNF-α, whereas PDE4B inhibition blocked LPS-induced TNF-α expression, but not MCP-1. In addition, PDE10A inhibition or deficiency decreased LPS-induced HIF-1α protein expression and subsequently suppressed MCP-1 expression. In vivo, PDE10A expression was also elevated in lung tissue after LPS exposure. Global PDE10A knockout or systemic administration of the PDE10A inhibitor TP-10 in mice significantly suppressed inflammatory molecule levels in the lung tissue and bronchoalveolar lavage fluid as well as inflammatory cell infiltration. These findings show that PDE10A plays a critical role in lung inflammation by promoting the activation of resident macrophages and infiltration of neutrophils.
Collapse
Affiliation(s)
- Chia George Hsu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY; and
| | - Fabeha Fazal
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Arshad Rahman
- Department of Pediatrics, Lung Biology and Disease Program, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Bradford C Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY; and
| | - Chen Yan
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY; and
| |
Collapse
|
7
|
Huang N, Li C, Sun W, Wu J, Xiao F. Long non-coding RNA TUG1 participates in LPS-induced periodontitis by regulating miR-498/RORA pathway. Oral Dis 2020; 27:600-610. [PMID: 32762066 DOI: 10.1111/odi.13590] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/27/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
Abstract
AIM This study was aimed to investigate the role of TUG1 in LPS-stimulated hPDLCs and to evaluate the potential functions of TUG1 in the pathogenesis of periodontitis. METHODS LPS-stimulated hPDLCs were established as the cell model. CCK-8 assay was performed to assess cell proliferation ability. Flow cytometry was performed to detect cell cycle distribution, and quantitative RT-PCR and Western blotting were conducted to measure gene expressions. ELISA kits were used to evaluate the production of inflammatory cytokines. The putative binding site between TUG1 and miR-498 was verified using luciferase reporter and RNA immunoprecipitation assays. RESULTS TUG1 was downregulated upon LPS stimulation in hPDLCs. TUG1 overexpression promoted cell proliferation through regulating the cell cycle distribution, along with the decreased expression of p21 and increased expression of CDK2 and cyclin D1. Besides, TUG1 overexpression decreased the production of inflammatory cytokines. The effects were opposite upon TUG1 knockdown. TUG1 negatively regulated its target miR-498, and influenced the expression of RORA, the direct target of miR-498. Simultaneous TUG1 overexpression and miR-498 reversed the effect of TUG1 overexpression alone on alleviating LPS-induced cell injury and inhibition of Wnt/β-catenin signaling, which was further changeover after co-overexpression with RORA. CONCLUSION Therefore, TUG1 could protect against periodontitis via regulating miR-498/RORA mediated Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Nannan Huang
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Chanxiu Li
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Wenjuan Sun
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Jian Wu
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| | - Feng Xiao
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
8
|
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
|
9
|
Gaertner VD, Michel S, Curtin JA, Pulkkinen V, Acevedo N, Söderhäll C, von Berg A, Bufe A, Laub O, Rietschel E, Heinzmann A, Simma B, Vogelberg C, Pershagen G, Melén E, Simpson A, Custovic A, Kere J, Kabesch M. Nocturnal asthma is affected by genetic interactions between RORA and NPSR1. Pediatr Pulmonol 2019; 54:847-857. [PMID: 30927345 DOI: 10.1002/ppul.24292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/17/2018] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Neuropeptide S Receptor 1 ( NPSR1) and Retinoid Acid Receptor-Related Orphan Receptor Alpha (RORA ) interact biologically, are both known candidate genes for asthma, and are involved in controlling circadian rhythm. Thus, we assessed (1) whether interactions between RORA and NPSR1 specifically affect the nocturnal asthma phenotype and (2) how this may differ from other asthma phenotypes. METHODS Interaction effects between 24 single-nucleotide polymorphisms (SNPs) in RORA and 35 SNPs in NPSR1 on asthma and nocturnal asthma symptoms were determined in 1432 subjects (763 asthmatics [192 with nocturnal asthma symptoms]; 669 controls) from the Multicenter Asthma Genetic in Childhood/International Study of Asthma and Allergies in Childhood studies. The results were validated and extended in children from the Manchester Asthma and Allergy Study (N = 723) and the Children Allergy Milieu Stockholm and Epidemiological cohort (N = 1646). RESULTS RORA* NPSR1 interactions seemed to affect both asthma and nocturnal asthma. In stratified analyses, however, interactions mainly affected nocturnal asthma and less so asthma without nocturnal symptoms or asthma severity. Results were replicated in two independent cohorts and seemed to remain constant over time throughout youth. CONCLUSION RORA* NPSR1 interactions appear to be involved in mechanisms specific for nocturnal asthma. In contrast to previous studies focusing on the role of beta 2 receptor polymorphisms in nocturnal asthma as a feature of asthma control or severity in general, our data suggest that changes in circadian rhythm control are associated with nighttime asthma symptoms.
Collapse
Affiliation(s)
- Vincent D Gaertner
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - Sven Michel
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
| | - John A Curtin
- Division of Infection Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, UK
| | - Ville Pulkkinen
- Heart and Lung Center, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Nathalie Acevedo
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm, Sweden.,Institute for Immunological Research, University of Cartagena, Cartagena, Colombia
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Department of Women´s and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Andrea von Berg
- Children's Department, Research Institute for the Prevention of Allergic Diseases, Marien-Hospital, Wesel, Germany
| | - Albrecht Bufe
- Department of Experimental Pneumology, Ruhr-University, Bochum, Germany
| | - Otto Laub
- Kinder- und Jugendarztpraxis Laub, Rosenheim, Germany
| | - Ernst Rietschel
- Faculty of Medicine, University Children's Hospital, University of Cologne, Cologne, Germany
| | - Andrea Heinzmann
- Center for Pediatrics, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Burkhard Simma
- Children's Department, University Teaching Hospital, Landeskrankenhaus Feldkirch, Feldkirch, Austria
| | - Christian Vogelberg
- University Children's Hospital, Technical University Dresden, Dresden, Germany
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet and Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden.,Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Angela Simpson
- Division of Infection Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, UK
| | | | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Research Programs Unit, Program for Molecular Neurology, University of Helsinki, Folkhälsän Institute of Genetics, Helsinki, Finland
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany.,School of Basic & Medical Biosciences, King's College London, London, England
| |
Collapse
|
10
|
Yang S, Lee S, Kim H, Kim H, Leem J, Yang H, Kwon H, Seo J, Cho H, Yoon J, Lee E, Jung Y, Kim Y, Jung S, Kwon H, Hong S. Prenatal particulate matter affects new asthma via airway hyperresponsiveness in schoolchildren. Allergy 2019; 74:675-684. [PMID: 30372532 DOI: 10.1111/all.13649] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/04/2018] [Accepted: 10/12/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND The most relevant time of PM10 exposure to affect airway hyperresponsiveness (AHR) and new development of asthma in school-aged children is unclear. The aims of this study were to investigate the most critical time of PM10 exposure to affect AHR and new diagnosis of asthma from AHR in school-aged children. METHODS Elementary schoolchildren (n = 3570) have been enrolled in a nationwide prospective 4-year follow-up survey in Korea from 2005 to 2006. Individual annual PM10 exposure was estimated by using an ordinary kriging method from the prenatal period to 7 years of age. AHR at 7 years was defined by a methacholine PC20 ≤8 mg/mL. RESULTS PM10 exposure during pregnancy and at 1 year of age showed significant effects on AHR (aOR: 1.694, 95% CI: 1.298-2.209; and aOR: 1.750, 95% CI: 1.343-2.282, respectively). PM10 exposure during pregnancy was associated with the risk of a new diagnosis of asthma (aOR: 2.056, 95% CI: 1.240-3.409), with the highest risk in children with AHR at age 7 (aOR: 6.080, 95% CI: 2.150-17.195). PM10 exposure in the second trimester was associated with the highest risk of a new diagnosis of asthma in children with AHR at age 7 (aOR: 4.136, 95% CI: 1.657-10.326). CONCLUSIONS Prenatal PM10 exposure in the second trimester is associated with an increased risk of a new diagnosis of asthma in school-aged children with AHR at 7 years. This study suggests that PM10 exposure during a specific trimester in utero may affect the onset of childhood asthma via AHR.
Collapse
Affiliation(s)
- Song‐I Yang
- Department of Pediatrics Hallym University Sacred Heart Hospital Hallym University College of Medicine Anyang Korea
| | - So‐Yeon Lee
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Hyo‐Bin Kim
- Department of Pediatrics Inje University Sanggye Paik Hospital Inje University College of Medicine Seoul Korea
| | - Hwan‐Cheol Kim
- Departments of Occupational and Environmental Medicine School of Medicine Inha University Incheon Korea
| | - Jong‐Han Leem
- Departments of Occupational and Environmental Medicine School of Medicine Inha University Incheon Korea
| | - Hyeon‐Jong Yang
- Department of Pediatrics Soonchunhyang University College of Medicine Seoul Korea
| | - Hyeok Kwon
- Asan Institute for Life Science Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Ju‐Hee Seo
- Department of Pediatrics Dankook University Hospital Cheonan Korea
| | - Hyun‐Ju Cho
- Department of Pediatrics International St. Mary's hospital Catholic Kwandong University Incheon Korea
| | - Jisun Yoon
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Eun Lee
- Department of Pediatrics Chonnam National University Hospital Chonnam National University Medical School Gwangju Korea
| | - Young‐Ho Jung
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Yeongho Kim
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Sungsu Jung
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| | - Ho‐Jang Kwon
- Department of Preventive Medicine Dankook University College of Medicine Cheonan Korea
| | - Soo‐Jong Hong
- Department of Pediatrics Childhood Asthma Atopy Center Environmental Health Center Asan Medical Center University of Ulsan College of Medicine Seoul Korea
| |
Collapse
|
11
|
Neville MDC, Choi J, Lieberman J, Duan QL. Identification of deleterious and regulatory genomic variations in known asthma loci. Respir Res 2018; 19:248. [PMID: 30541564 PMCID: PMC6292105 DOI: 10.1186/s12931-018-0953-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 11/23/2018] [Indexed: 11/25/2022] Open
Abstract
Background Candidate gene and genome-wide association studies have identified hundreds of asthma risk loci. The majority of associated variants, however, are not known to have any biological function and are believed to represent markers rather than true causative mutations. We hypothesized that many of these associated markers are in linkage disequilibrium (LD) with the elusive causative variants. Methods We compiled a comprehensive list of 449 asthma-associated variants previously reported in candidate gene and genome-wide association studies. Next, we identified all sequence variants located within the 305 unique genes using whole-genome sequencing data from the 1000 Genomes Project. Then, we calculated the LD between known asthma variants and the sequence variants within each gene. LD variants identified were then annotated to determine those that are potentially deleterious and/or functional (i.e. coding or regulatory effects on the encoded transcript or protein). Results We identified 10,130 variants in LD (r2 > 0.6) with known asthma variants. Annotations of these LD variants revealed that several have potentially deleterious effects including frameshift, alternate splice site, stop-lost, and missense. Moreover, 24 of the LD variants have been reported to regulate gene expression as expression quantitative trait loci (eQTLs). Conclusions This study is proof of concept that many of the genetic loci previously associated with complex diseases such as asthma are not causative but represent markers of disease, which are in LD with the elusive causative variants. We hereby report a number of potentially deleterious and regulatory variants that are in LD with the reported asthma loci. These reported LD variants could account for the original association signals with asthma and represent the true causative mutations at these loci. Electronic supplementary material The online version of this article (10.1186/s12931-018-0953-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthew D C Neville
- Department of Biomedical and Molecular Sciences, Queen's University, Botterell Hall, Room 530 - 18 Stuart St, Kingston, ON, K7L3N6, Canada
| | - Jihoon Choi
- Department of Biomedical and Molecular Sciences, Queen's University, Botterell Hall, Room 530 - 18 Stuart St, Kingston, ON, K7L3N6, Canada
| | - Jonathan Lieberman
- Department of Biomedical and Molecular Sciences, Queen's University, Botterell Hall, Room 530 - 18 Stuart St, Kingston, ON, K7L3N6, Canada
| | - Qing Ling Duan
- Department of Biomedical and Molecular Sciences, Queen's University, Botterell Hall, Room 530 - 18 Stuart St, Kingston, ON, K7L3N6, Canada. .,School of Computing, Queen's University, 557 Goodwin Hall, Room 531, Kingston, ON, K7L 2N8, Canada.
| |
Collapse
|
12
|
Zonoobi E, Saeedfar K, Pourdowlat G, Masjedi MR, Behmanesh M. The Study of IL-10 and IL-17A Genes Expression in Patients with Different Stages of Asthma: a Case-Control Study. TANAFFOS 2018; 17:146-154. [PMID: 30915130 PMCID: PMC6428381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
BACKGROUND Asthma is considered as a complex disorder in which genetics and environment play crucial role in its susceptibility. In addition to the huge financial costs that significantly reduce the quality of life of the patients and their families, it causes high prevalence of lung diseases. Finding contributing new genetic factors involved in early diagnosis or progression of asthma can provide novel approaches for treatment or managing of asthma. In the present study, the potential role of two key cytokines of IL-10 and IL-17A was investigated in asthma pathogenesis. MATERIALS AND METHODS Using real-time PCR technique, we analyzed the expression levels of target genes in two groups of mild and severe asthma patient in comparison with healthy individuals. RESULTS In comparison with control population, obtained data showed 4 and 7-fold down-regulation of IL-17A in the group of mild and severe asthma, respectively. Down-regulation of IL-17A showed a significant correlation with progression of asthma severity. While IL-10 showed up to 10-fold down-regulation in the group of severe asthma, its expression level was not correlated with severity of asthma. CONCLUSION Obtained data revealed that deregulation IL-10 and IL-17A have potential to play crucial role in pathogenesis and prognosis of asthma. Observed down-regulation of these cytokines in blood cells suggests their usefulness as a marker in diagnosis of asthmatic types in patients.
Collapse
Affiliation(s)
- Elham Zonoobi
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Kayvan Saeedfar
- Tracheal Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Guitti Pourdowlat
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mehrdad Behmanesh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran,Correspondence to: Behmanesh M, Address: Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran, Email address:
| |
Collapse
|
13
|
Cai X, Lin M, Cao S, Liu Y, Lin N. The association of RAR-related orphan receptor A (RORA) gene polymorphisms with the risk of asthma. Ann Hum Genet 2017; 82:158-164. [PMID: 29282706 DOI: 10.1111/ahg.12238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 11/03/2017] [Accepted: 11/20/2017] [Indexed: 12/29/2022]
Abstract
Asthma is a common, heterogeneous chronic respiratory disease characterized by chronic inflammation of the airway, airway hyperreactivity, and airway remodeling. The RAR-related orphan receptor A (RORA) gene has been identified for the pathogenesis of asthma. The purpose of this research was to investigate the relationship between RORA gene polymorphisms and asthma susceptibility in the Chinese Zhuang population. This was a case-control study including 231 children with asthma and 343 healthy controls. The RORA gene polymorphisms were measured by the polymerase chain reaction-ligase detection reaction genotyping assays and confirmed by sequencing. The distribution of the genotype frequencies of the RORA rs11071559 C>T was significantly different in the group of cases and the healthy children (P < 0.05). By haplotype analyses, the haplotype TT (rs7164773/rs11071559) was statistically significant between asthmatics and nonasthmatics, but the association was not significant after correction for multiple comparisons. Our results provided evidence that the RORA rs11071559C>T polymorphism was associated with an elevated susceptibility to pediatric asthma in the Chinese Zhuang population.
Collapse
Affiliation(s)
- Xulong Cai
- Youjiang Medical University for Nationalities, Baise, China
| | - Mali Lin
- Youjiang Medical University for Nationalities, Baise, China
| | - Shan Cao
- Youjiang Medical University for Nationalities, Baise, China
| | - Yunguang Liu
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Na Lin
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
14
|
Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3226-3242. [PMID: 28866134 DOI: 10.1016/j.bbadis.2017.08.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/29/2017] [Indexed: 12/23/2022]
Abstract
Embryonic lung development requires reciprocal endodermal-mesodermal interactions; mediated by various signaling proteins. Wnt/β-catenin is a signaling protein that exhibits the pivotal role in lung development, injury and repair while aberrant expression of Wnt/β-catenin signaling leads to asthmatic airway remodeling: characterized by hyperplasia and hypertrophy of airway smooth muscle cells, alveolar and vascular damage goblet cells metaplasia, and deposition of extracellular matrix; resulting in decreased lung compliance and increased airway resistance. The substantial evidence suggests that Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Here, we summarized the recent advances related to the mechanistic role of Wnt/β-catenin signaling in lung development, consequences of aberrant expression or deletion of Wnt/β-catenin signaling in expansion and progression of asthmatic airway remodeling, and linking early-impaired pulmonary development and airway remodeling later in life. Finally, we emphasized all possible recent potential therapeutic significance and future prospectives, that are adaptable for therapeutic intervention to treat asthmatic airway remodeling.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China.
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| |
Collapse
|
15
|
Kleyman M, Sefer E, Nicola T, Espinoza C, Chhabra D, Hagood JS, Kaminski N, Ambalavanan N, Bar-Joseph Z. Selecting the most appropriate time points to profile in high-throughput studies. eLife 2017; 6:e18541. [PMID: 28124972 PMCID: PMC5319842 DOI: 10.7554/elife.18541] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 01/23/2017] [Indexed: 12/25/2022] Open
Abstract
Biological systems are increasingly being studied by high throughput profiling of molecular data over time. Determining the set of time points to sample in studies that profile several different types of molecular data is still challenging. Here we present the Time Point Selection (TPS) method that solves this combinatorial problem in a principled and practical way. TPS utilizes expression data from a small set of genes sampled at a high rate. As we show by applying TPS to study mouse lung development, the points selected by TPS can be used to reconstruct an accurate representation for the expression values of the non selected points. Further, even though the selection is only based on gene expression, these points are also appropriate for representing a much larger set of protein, miRNA and DNA methylation changes over time. TPS can thus serve as a key design strategy for high throughput time series experiments. Supporting Website: www.sb.cs.cmu.edu/TPS.
Collapse
Affiliation(s)
- Michael Kleyman
- Machine Learning and Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, United States
| | - Emre Sefer
- Machine Learning and Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, United States
| | - Teodora Nicola
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, United States
| | - Celia Espinoza
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, United States.,CARady Children's Hospital San Diego, San Diego, United States
| | - Divya Chhabra
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, United States.,CARady Children's Hospital San Diego, San Diego, United States
| | - James S Hagood
- Division of Respiratory Medicine, Department of Pediatrics, University of California, San Diego, United States.,CARady Children's Hospital San Diego, San Diego, United States
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, School of Medicine, Yale University, New Haven, United States
| | - Namasivayam Ambalavanan
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, United States
| | - Ziv Bar-Joseph
- Machine Learning and Computational Biology, School of Computer Science, Carnegie Mellon University, Pittsburgh, United States
| |
Collapse
|
16
|
Carlini F, Picard C, Garulli C, Piquemal D, Roubertoux P, Chiaroni J, Chanez P, Gras D, Di Cristofaro J. Bronchial Epithelial Cells from Asthmatic Patients Display Less Functional HLA-G Isoform Expression. Front Immunol 2017; 8:6. [PMID: 28303134 PMCID: PMC5333864 DOI: 10.3389/fimmu.2017.00006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/03/2017] [Indexed: 11/13/2022] Open
Abstract
Not all asthmatic patients adequately respond to current available treatments, such as inhaled corticosteroids or omalizumab®. New treatments will aim to target the bronchial epithelium-immune response interaction using different pathways. HLA-G is involved in immunomodulation and may promote epithelial cell differentiation and proliferation. HLA-G protein has several isoforms generated by alternative splicing that might have differential functionalities. HLA-G protein expression and genetic polymorphisms have been reported to be associated with asthma. Our hypothesis is that bronchial epithelium from asthmatic patients displays less functional HLA-G isoforms. HLA-G transcriptional isoforms were quantified by real-time PCR in human bronchial epithelium cells (HBEC) grown in air-liquid interface culture obtained from five healthy controls (HC), seven patients with mild asthma (MA), and seven patients with severe asthma (SA). They were re-differentiated, and IL-13 exposure was used as a proxy for a pro-inflammatory cytokine. HLA-G protein expression was assessed by western blot analysis. HLA-G allele was typed by direct sequencing. Our results showed that both MA and SA display less functional HLA-G isoforms than HC (p < 0.05); in vitro HBEC re-differentiation from SA displays a particular isoform expression profile compared to MA and HC (p = 0.03); HLA-G*01:06 frequency in MA and SA was significantly higher than in the healthy population (p = 0.03 and p < 0.001, respectively); and IL-13 exposure had no impact on HLA-G expression. Our results support that an impaired expression of HLA-G isoforms in asthmatic patients could contribute to the loss of inflammation control and epithelium structural remodeling. Therefore, HLA-G might be an interesting alternative target for asthmatic patients not adequately responding to current drugs.
Collapse
Affiliation(s)
- Federico Carlini
- Etablissement Français du Sang Alpes Méditerranée , Marseille , France
| | - Christophe Picard
- Etablissement Français du Sang Alpes Méditerranée, Marseille, France; Aix-Marseille Univ, CNRS, EFS, ADES, "Biologie des Groupes Sanguins", Marseille, France
| | - Céline Garulli
- Aix-Marseille Univ, INSERM U1067 CNRS UMR 7333 , Marseille , France
| | | | - Pierre Roubertoux
- INSERM U491, Génétique Médicale et Développement, Aix-Marseille Université , Marseille , France
| | - Jacques Chiaroni
- Etablissement Français du Sang Alpes Méditerranée, Marseille, France; Aix-Marseille Univ, CNRS, EFS, ADES, "Biologie des Groupes Sanguins", Marseille, France
| | - Pascal Chanez
- Aix-Marseille Univ, INSERM U1067 CNRS UMR 7333, Marseille, France; Clinique des Bronches, Allergie et Sommeil, AP-HM Hôpital Nord, Marseille, France
| | - Delphine Gras
- Aix-Marseille Univ, INSERM U1067 CNRS UMR 7333 , Marseille , France
| | - Julie Di Cristofaro
- Etablissement Français du Sang Alpes Méditerranée, Marseille, France; Aix-Marseille Univ, CNRS, EFS, ADES, "Biologie des Groupes Sanguins", Marseille, France
| |
Collapse
|
17
|
Miller S, Henry AP, Hodge E, Kheirallah AK, Billington CK, Rimington TL, Bhaker SK, Obeidat M, Melén E, Merid SK, Swan C, Gowland C, Nelson CP, Stewart CE, Bolton CE, Kilty I, Malarstig A, Parker SG, Moffatt MF, Wardlaw AJ, Hall IP, Sayers I. The Ser82 RAGE Variant Affects Lung Function and Serum RAGE in Smokers and sRAGE Production In Vitro. PLoS One 2016; 11:e0164041. [PMID: 27755550 PMCID: PMC5068780 DOI: 10.1371/journal.pone.0164041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 09/19/2016] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Genome-Wide Association Studies have identified associations between lung function measures and Chronic Obstructive Pulmonary Disease (COPD) and chromosome region 6p21 containing the gene for the Advanced Glycation End Product Receptor (AGER, encoding RAGE). We aimed to (i) characterise RAGE expression in the lung, (ii) identify AGER transcripts, (iii) ascertain if SNP rs2070600 (Gly82Ser C/T) is associated with lung function and serum sRAGE levels and (iv) identify whether the Gly82Ser variant is functionally important in altering sRAGE levels in an airway epithelial cell model. METHODS Immunohistochemistry was used to identify RAGE protein expression in 26 human tissues and qPCR was used to quantify AGER mRNA in lung cells. Gene expression array data was used to identify AGER expression during lung development in 38 fetal lung samples. RNA-Seq was used to identify AGER transcripts in lung cells. sRAGE levels were assessed in cells and patient serum by ELISA. BEAS2B-R1 cells were transfected to overexpress RAGE protein with either the Gly82 or Ser82 variant and sRAGE levels identified. RESULTS Immunohistochemical assessment of 6 adult lung samples identified high RAGE expression in the alveoli of healthy adults and individuals with COPD. AGER/RAGE expression increased across developmental stages in human fetal lung at both the mRNA (38 samples) and protein levels (20 samples). Extensive AGER splicing was identified. The rs2070600T (Ser82) allele is associated with higher FEV1, FEV1/FVC and lower serum sRAGE levels in UK smokers. Using an airway epithelium model overexpressing the Gly82 or Ser82 variants we found that HMGB1 activation of the RAGE-Ser82 receptor results in lower sRAGE production. CONCLUSIONS This study provides new information regarding the expression profile and potential role of RAGE in the human lung and shows a functional role of the Gly82Ser variant. These findings advance our understanding of the potential mechanisms underlying COPD particularly for carriers of this AGER polymorphism.
Collapse
Affiliation(s)
- Suzanne Miller
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
- * E-mail:
| | - Amanda P. Henry
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | | | | | - Tracy L. Rimington
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sangita K. Bhaker
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ma’en Obeidat
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Simon K. Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Swan
- Department of Biology, University of York, York, United Kingdom
| | - Catherine Gowland
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Carl P. Nelson
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ceri E. Stewart
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte E. Bolton
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Iain Kilty
- Pfizer Worldwide Research & Development, Cambridge, Massachusetts, United States of America
| | - Anders Malarstig
- Pfizer Worldwide Research & Development, Cambridge, United Kingdom
| | - Stuart G. Parker
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Miriam F. Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew J. Wardlaw
- Institute for Lung Health, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Ian P. Hall
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
18
|
Association between obesity and asthma - epidemiology, pathophysiology and clinical profile. Nutr Res Rev 2016; 29:194-201. [PMID: 27514726 DOI: 10.1017/s0954422416000111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Obesity is a risk factor for asthma, and obese asthmatics have lower disease control and increased symptom severity. Several putative links have been proposed, including genetics, mechanical restriction of the chest and the intake of corticosteroids. The most consistent evidence, however, comes from studies of cytokines produced by the adipose tissue called adipokines. Adipokine imbalance is associated with both proinflammatory status and asthma. Although reverse causation has been proposed, it is now acknowledged that obesity precedes asthma symptoms. Nevertheless, prenatal origins of both conditions complicate the search for causality. There is a confirmed role of neuro-immune cross-talk mediating obesity-induced asthma, with leptin playing a key role in these processes. Obesity-induced asthma is now considered a distinct asthma phenotype. In fact, it is one of the most important determinants of asthma phenotypes. Two main subphenotypes have been distinguished. The first phenotype, which affects adult women, is characterised by later onset and is more likely to be non-atopic. The childhood obesity-induced asthma phenotype is characterised by primary and predominantly atopic asthma. In obesity-induced asthma, the immune responses are shifted towards T helper (Th) 1 polarisation rather than the typical atopic Th2 immunological profile. Moreover, obese asthmatics might respond differently to environmental triggers. The high cost of treatment of obesity-related asthma, and the burden it causes for the patients and their families call for urgent intervention. Phenotype-specific approaches seem to be crucial for the success of prevention and treatment.
Collapse
|
19
|
Miller S, Melén E, Merid SK, Hall IP, Sayers I. Genes associated with polymorphic variants predicting lung function are differentially expressed during human lung development. Respir Res 2016; 17:95. [PMID: 27473260 PMCID: PMC4966770 DOI: 10.1186/s12931-016-0410-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/20/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Recent meta-analyses of genome-wide association studies have identified single nucleotide polymorphisms (SNPs) within/near 54 genes associated with lung function measures. Current understanding of the contribution of these genes to human lung development is limited. We set out to further define i) the expression profile of these genes during human lung development using a unique set of resources to examine both mRNA and protein expression and ii) the link between key polymorphisms and genes using expression quantitative trait loci (eQTL) approaches. METHODS The mRNA expression profile of lung function associated genes across pseudoglandular and canalicular stages of lung development were determined using expression array data of 38 human fetal lungs. eQTLs were investigated for selected genes using blood cell and lung tissue data. Immunohistochemistry of the top 5 candidates was performed in a panel of 24 fetal lung samples. RESULTS Twenty-nine lung function associated genes were differentially expressed during lung development at the mRNA level. The greatest magnitude of effect was observed for 5 genes; TMEM163, FAM13A and HHIP which had increasing expression and CDC123 and PTCH1 with decreased expression across developmental stages. Focussed eQTL analyses investigating SNPs in these five loci identified several cis-eQTL's. Protein expression of TMEM163 increased and CDC123 decreased with fetal lung age in agreement with mRNA data. Protein expression in FAM13A, HHIP and PTCH1 remained relatively constant throughout lung development. CONCLUSIONS We have identified that > 50 % of lung function associated genes show evidence of differential expression during lung development and we show that in particular TMEM163 and CDC123 are differentially expressed at both the mRNA and protein levels. Our data provides a systematic evaluation of lung function associated genes in this context and offers some insight into the potential role of several of these genes in contributing to human lung development.
Collapse
Affiliation(s)
- S Miller
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
| | - E Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, Stockholm, Sweden
| | - S K Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - I P Hall
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - I Sayers
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| |
Collapse
|
20
|
Rizzo R, Bergamini G, Bortolotti D, Leal T, D'Orazio C, Pintani E, Melchiorri L, Zavatti E, Assael BM, Sorio C, Melotti P. HLA-G expression and regulation during Pseudomonas aeruginosa infection in cystic fibrosis patients. Future Microbiol 2016; 11:363-73. [PMID: 26934639 DOI: 10.2217/fmb.15.143] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Deregulated immune response fails to control biofilm-forming bacteria, as Pseudomonas aeruginosa, in the lungs of cystic fibrosis (CF) patients. HLA-G is an immune-modulatory molecule involved in respiratory diseases and infections. MATERIALS & METHODS HLA-G mRNA and protein were analyzed in plasma and exhaled breath condensate from CF patients undergoing intravenous antibiotic treatment, CF cell line and murine model. RESULTS Therapy normalizes HLA-G plasmatic in CF patients suggesting a systemic anti-inflammatory role while in CF airway system, higher expression of HLA-G is associated with P. aeruginosa infection. CF cell line and murine model expressed higher HLA-G molecules in the presence of P. aeruginosa. CONCLUSION Plasmatic and lung HLA-G expression suggest a role in reducing systemic inflammation and supporting P. aeruginosa infection.
Collapse
Affiliation(s)
- Roberta Rizzo
- Section of Microbiology & Medical Genetics, Medical Sciences Department, Ferrara University, 44121 Ferrara FE, Italy
| | - Gabriella Bergamini
- Section of Microbiology & Medical Genetics, Medical Sciences Department, Ferrara University, 44121 Ferrara FE, Italy.,Department of Medicine, Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," University of Verona, Verona, Italy
| | - Daria Bortolotti
- Section of Microbiology & Medical Genetics, Medical Sciences Department, Ferrara University, 44121 Ferrara FE, Italy
| | - Teresinha Leal
- Louvain Centre for Toxicology & Applied Pharmacology, Université Catholique de Louvain, Place de l'Université 1, 1348, Belgium
| | - Ciro D'Orazio
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata di Verona, 37122 Verona VR, Italy
| | - Emily Pintani
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata di Verona, 37122 Verona VR, Italy
| | - Loredana Melchiorri
- Section of Microbiology & Medical Genetics, Medical Sciences Department, Ferrara University, 44121 Ferrara FE, Italy
| | - Eleonora Zavatti
- Service Planning and Management Control, University Hospital of Ferrara, Ferrara, Italy
| | - Baroukh M Assael
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata di Verona, 37122 Verona VR, Italy
| | - Claudio Sorio
- Department of Medicine, Cystic Fibrosis Translational Research Laboratory "Daniele Lissandrini," University of Verona, Verona, Italy
| | - Paola Melotti
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata di Verona, 37122 Verona VR, Italy
| |
Collapse
|
21
|
Translating Lung Function Genome-Wide Association Study (GWAS) Findings. ADVANCES IN GENETICS 2016; 93:57-145. [DOI: 10.1016/bs.adgen.2015.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Gene expression analysis in Fmr1KO mice identifies an immunological signature in brain tissue and mGluR5-related signaling in primary neuronal cultures. Mol Autism 2015; 6:66. [PMID: 26697163 PMCID: PMC4687343 DOI: 10.1186/s13229-015-0061-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/10/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Fragile X syndrome (FXS) is a neurodevelopmental disorder whose biochemical manifestations involve dysregulation of mGluR5-dependent pathways, which are widely modeled using cultured neurons. In vitro phenotypes in cultured neurons using standard morphological, functional, and chemical approaches have demonstrated considerable variability. Here, we study transcriptomes obtained in situ in the intact brain tissues of a murine model of FXS to see how they reflect the in vitro state. METHODS We used genome-wide mRNA expression profiling as a robust characterization tool for studying differentially expressed pathways in fragile X mental retardation 1 (Fmr1) knockout (KO) and wild-type (WT) murine primary neuronal cultures and in embryonic hippocampal and cortical murine tissue. To study the developmental trajectory and to relate mouse model data to human data, we used an expression map of human development to plot murine differentially expressed genes in KO/WT cultures and brain. RESULTS We found that transcriptomes from cell cultures showed a stronger signature of Fmr1KO than whole tissue transcriptomes. We observed an over-representation of immunological signaling pathways in embryonic Fmr1KO cortical and hippocampal tissues and over-represented mGluR5-downstream signaling pathways in Fmr1KO cortical and hippocampal primary cultures. Genes whose expression was up-regulated in Fmr1KO murine cultures tended to peak early in human development, whereas differentially expressed genes in embryonic cortical and hippocampal tissues clustered with genes expressed later in human development. CONCLUSIONS The transcriptional profile in brain tissues primarily centered on immunological mechanisms, whereas the profiles from cell cultures showed defects in neuronal activity. We speculate that the isolation and culturing of neurons caused a shift in neurological transcriptome towards a "juvenile" or "de-differentiated" state. Moreover, cultured neurons lack the close coupling with glia that might be responsible for the immunological phenotype in the intact brain. Our results suggest that cultured cells may recapitulate an early phase of the disease, which is also less obscured with a consequent "immunological" phenotype and in vivo compensatory mechanisms observed in the embryonic brain. Together, these results suggest that the transcriptome of cultured primary neuronal cells, in comparison to whole brain tissue, more robustly demonstrated the difference between Fmr1KO and WT mice and might reveal a molecular phenotype, which is typically hidden by compensatory mechanisms present in vivo. Moreover, cultures might be useful for investigating the perturbed pathways in early human brain development and genes previously implicated in autism.
Collapse
|
23
|
Cook DN, Kang HS, Jetten AM. Retinoic Acid-Related Orphan Receptors (RORs): Regulatory Functions in Immunity, Development, Circadian Rhythm, and Metabolism. NUCLEAR RECEPTOR RESEARCH 2015; 2. [PMID: 26878025 PMCID: PMC4750502 DOI: 10.11131/2015/101185] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In this overview, we provide an update on recent progress made in understanding the mechanisms of action, physiological functions, and roles in disease of retinoic acid related orphan receptors (RORs). We are particularly focusing on their roles in the regulation of adaptive and innate immunity, brain function, retinal development, cancer, glucose and lipid metabolism, circadian rhythm, metabolic and inflammatory diseases and neuropsychiatric disorders. We also summarize the current status of ROR agonists and inverse agonists, including their regulation of ROR activity and their therapeutic potential for management of various diseases in which RORs have been implicated.
Collapse
Affiliation(s)
- Donald N Cook
- Immunogenetics Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
24
|
Artigas MS, Wain LV, Miller S, Kheirallah AK, Huffman JE, Ntalla I, Shrine N, Obeidat M, Trochet H, McArdle WL, Alves AC, Hui J, Zhao JH, Joshi PK, Teumer A, Albrecht E, Imboden M, Rawal R, Lopez LM, Marten J, Enroth S, Surakka I, Polasek O, Lyytikäinen LP, Granell R, Hysi PG, Flexeder C, Mahajan A, Beilby J, Bossé Y, Brandsma CA, Campbell H, Gieger C, Gläser S, González JR, Grallert H, Hammond CJ, Harris SE, Hartikainen AL, Heliövaara M, Henderson J, Hocking L, Horikoshi M, Hutri-Kähönen N, Ingelsson E, Johansson Å, Kemp JP, Kolcic I, Kumar A, Lind L, Melén E, Musk AW, Navarro P, Nickle DC, Padmanabhan S, Raitakari OT, Ried JS, Ripatti S, Schulz H, Scott RA, Sin DD, Starr JM, Viñuela A, Völzke H, Wild SH, Wright AF, Zemunik T, Jarvis DL, Spector TD, Evans DM, Lehtimäki T, Vitart V, Kähönen M, Gyllensten U, Rudan I, Deary IJ, Karrasch S, Probst-Hensch NM, Heinrich J, Stubbe B, Wilson JF, Wareham NJ, James AL, Morris AP, Jarvelin MR, Hayward C, Sayers I, Strachan DP, Hall IP, Tobin MD. Sixteen new lung function signals identified through 1000 Genomes Project reference panel imputation. Nat Commun 2015; 6:8658. [PMID: 26635082 PMCID: PMC4686825 DOI: 10.1038/ncomms9658] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 09/17/2015] [Indexed: 01/11/2023] Open
Abstract
Lung function measures are used in the diagnosis of chronic obstructive pulmonary disease. In 38,199 European ancestry individuals, we studied genome-wide association of forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC) and FEV1/FVC with 1000 Genomes Project (phase 1)-imputed genotypes and followed up top associations in 54,550 Europeans. We identify 14 novel loci (P<5 × 10(-8)) in or near ENSA, RNU5F-1, KCNS3, AK097794, ASTN2, LHX3, CCDC91, TBX3, TRIP11, RIN3, TEKT5, LTBP4, MN1 and AP1S2, and two novel signals at known loci NPNT and GPR126, providing a basis for new understanding of the genetic determinants of these traits and pulmonary diseases in which they are altered.
Collapse
Affiliation(s)
- María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Suzanne Miller
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Abdul Kader Kheirallah
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jennifer E. Huffman
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Ioanna Ntalla
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Nick Shrine
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Ma'en Obeidat
- University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada V6Z 1Y6
| | - Holly Trochet
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
| | - Wendy L. McArdle
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Alexessander Couto Alves
- Department of Epidemiology and Biostatistics, MRC -PHE Centre for Environment & Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
| | - Jennie Hui
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- PathWest Laboratory Medicine WA, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Population Health, The University of Western Australia, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, Western Australia 6009, Australia
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Peter K. Joshi
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Alexander Teumer
- University Medicine Greifswald, Community Medicine, SHIP—Clinical Epidemiological Research, Greifswald 17489, Germany
- Department for Genetics and Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17489, Germany
| | - Eva Albrecht
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Rajesh Rawal
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Lorna M. Lopez
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Department of Psychology, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Jonathan Marten
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Stefan Enroth
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
| | - Ida Surakka
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FI-00014, Finland
- The National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - Ozren Polasek
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
- Department of Public Health, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Raquel Granell
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Pirro G. Hysi
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Claudia Flexeder
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - John Beilby
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- PathWest Laboratory Medicine WA, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Pathology and Laboratory Medicine, The University of Western Australia, Western Australia 6009, Australia
| | - Yohan Bossé
- Department of Molecular Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec, Canada G1V 0A6
| | - Corry-Anke Brandsma
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen 9700, The Netherlands
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Sven Gläser
- Department of Internal Medicine B, Pneumology, Cardiology, Intensive Care, Weaning, Field of Research: Pneumological Epidemiology, University Medicine Greifswald, Greifswald 17489, Germany
| | - Juan R. González
- Centre for Research in Environmental Epidemiology (CREAL), Barcelona E-08003, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid 28029, Spain
- Pompeu Fabra University (UPF), Barcelona 08002, Catalonia, Spain
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Chris J. Hammond
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Sarah E. Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Anna-Liisa Hartikainen
- Department of Obstetrics and Gynecology of Oulu University Hospital ,MRC of Oulu University, Oulu 90220, Finland
| | - Markku Heliövaara
- The National Institute for Health and Welfare (THL), Helsinki FI-00271, Finland
| | - John Henderson
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
| | - Lynne Hocking
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
- Division of Applied Health Sciences, University of Aberdeen, Aberdeen, Scotland AB24 3FX, UK
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX1 2JD, UK
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere 33521, Finland
- Department of Pediatrics, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Erik Ingelsson
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala 751 23, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Åsa Johansson
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
- Uppsala Clinical Research Centre, Uppsala University, Uppsala 751 23, Sweden
| | - John P. Kemp
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland QLD 4072, Australia
- MRC Integrative Epidemiology Unit, Bristol BS8 1TH, UK
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Ashish Kumar
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm SE-171 7, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala 751 23, Sweden
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs' Children's Hospital, Stockholm SE-171 7, Sweden
| | - Arthur W. Musk
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Western Australia 6009, Australia
| | - Pau Navarro
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - David C. Nickle
- Genetics and Pharmacogenomics, Merck Research Labs, Boston, Massachusetts 02115, USA
| | - Sandosh Padmanabhan
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
- Division of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, Scotland, UK
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20520, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20014, Finland
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg D-85764, Germany
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki FI-00014, Finland
- Department of Public Health, University of Helsinki, Helsinki FI-00014, Finland
- Department of Human Genomics, Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Holger Schulz
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich 85764, Germany
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Don D. Sin
- University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, British Columbia, Canada V6Z 1Y6
- Respiratory Division, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z4
| | - John M. Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Alzheimer Scotland Research Centre, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Ana Viñuela
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Henry Völzke
- University Medicine Greifswald, Community Medicine, SHIP—Clinical Epidemiological Research, Greifswald 17489, Germany
| | - Sarah H. Wild
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Alan F. Wright
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Tatijana Zemunik
- Department of Medical Biology, Faculty of Medicine, University of Split, Split 21000, Croatia
| | - Deborah L. Jarvis
- Respiratory Epidemiology and Public Health, Imperial College London, London SW7 2AZ, UK
- MRC Health Protection Agency (HPA) Centre for Environment and Health, Imperial College London, London SW7 2AZ, UK
| | - Tim D. Spector
- KCL Department of Twins Research and Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - David M. Evans
- School of Social and Community Medicine, University of Bristol, Bristol BS8 1TH, UK
- Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Queensland QLD 4072, Australia
- MRC Integrative Epidemiology Unit, Bristol BS8 1TH, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere FI-33101, Finland
- Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere FI-33520, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, Tampere 33521, Finland
| | - Ulf Gyllensten
- Department of Immunology, Genetics, and Pathology, Biomedical Center, SciLifeLab Uppsala, Uppsala University, Uppsala 751 23, Sweden
| | - Igor Rudan
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
- Centre for Population Health Sciences, Medical School, University of Edinburgh, Edinburgh EH8 9AD, Scotland, UK
| | - Ian J. Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9AD, UK
- Department of Psychology, University of Edinburgh, Edinburgh EH8 9AD, UK
| | - Stefan Karrasch
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Institute of General Practice, University Hospital Klinikum rechts der Isar, Technische Universität München, Munich D - 81675, Germany
- Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilians-Universität, Munich 80539, Germany
| | - Nicole M. Probst-Hensch
- Swiss Tropical and Public Health Institute, Basel 4051, Switzerland
- University of Basel, Basel 4001, Switzerland
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research, Munich 85764, Germany
- University Hospital Munich, Institute and Outpatient Clinic for Occupational, Social and Environmental Medicine, Ludwig-Maximilian University Munich, Munich 80539, Germany
| | - Beate Stubbe
- Department of Internal Medicine B, Pneumology, Cardiology, Intensive Care, Weaning, Field of Research: Pneumological Epidemiology, University Medicine Greifswald, Greifswald 17489, Germany
| | - James F. Wilson
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH8 9AD, UK
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AD, Scotland, UK
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge CB2 0SL, UK
| | - Alan L. James
- Busselton Population Medical Research Institute, Busselton, Western Australia 6280, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Western Australia 6009, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Western Australia 6009, Australia
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 7ZX, UK
- Estonian Genome Centre, University of Tartu, Tartu 50090, Estonia
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC -PHE Centre for Environment & Health, School of Public Health, Imperial College London, London SW7 2AZ, UK
- Center for Life Course Epidemiology, Faculty of Medicine, P.O.Box 5000, FI-90014 University of Oulu, Oulu FI-01051, Finland
- Biocenter Oulu, P.O.Box 5000, Aapistie 5A, FI-90014 University of Oulu, Oulu FI-01051, Finland
- Unit of Primary Care, Oulu University Hospital, Kajaanintie 50, P.O.Box 20, FI-90220 Oulu, 90029 OYS, Finland
| | - Caroline Hayward
- Generation Scotland, A Collaboration between the University Medical Schools and NHS, Aberdeen, Dundee, Edinburgh, Glasgow EH4 2XU, UK
| | - Ian Sayers
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - David P. Strachan
- Population Health Research Institute, St George's, University of London, Cranmer Terrace, London WC1B 5DN, UK
| | - Ian P. Hall
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| |
Collapse
|
25
|
Costa GNO, Dudbridge F, Fiaccone RL, da Silva TM, Conceição JS, Strina A, Figueiredo CA, Magalhães WCS, Rodrigues MR, Gouveia MH, Kehdy FSG, Horimoto ARVR, Horta B, Burchard EG, Pino-Yanes M, Del Rio Navarro B, Romieu I, Hancock DB, London S, Lima-Costa MF, Pereira AC, Tarazona E, Rodrigues LC, Barreto ML. A genome-wide association study of asthma symptoms in Latin American children. BMC Genet 2015; 16:141. [PMID: 26635092 PMCID: PMC4669662 DOI: 10.1186/s12863-015-0296-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/17/2015] [Indexed: 01/14/2023] Open
Abstract
Background Asthma is a chronic disease of the airways and, despite the advances in the knowledge of associated genetic regions in recent years, their mechanisms have yet to be explored. Several genome-wide association studies have been carried out in recent years, but none of these have involved Latin American populations with a high level of miscegenation, as is seen in the Brazilian population. Methods 1246 children were recruited from a longitudinal cohort study in Salvador, Brazil. Asthma symptoms were identified in accordance with an International Study of Asthma and Allergies in Childhood (ISAAC) questionnaire. Following quality control, 1 877 526 autosomal SNPs were tested for association with childhood asthma symptoms by logistic regression using an additive genetic model. We complemented the analysis with an estimate of the phenotypic variance explained by common genetic variants. Replications were investigated in independent Mexican and US Latino samples. Results Two chromosomal regions reached genome-wide significance level for childhood asthma symptoms: the 14q11 region flanking the DAD1 and OXA1L genes (rs1999071, MAF 0.32, OR 1.78, 95 % CI 1.45–2.18, p-value 2.83 × 10−8) and 15q22 region flanking the FOXB1 gene (rs10519031, MAF 0.04, OR 3.0, 95 % CI 2.02–4.49, p-value 6.68 × 10−8 and rs8029377, MAF 0.03, OR 2.49, 95 % CI 1.76–3.53, p-value 2.45 × 10−7). eQTL analysis suggests that rs1999071 regulates the expression of OXA1L gene. However, the original findings were not replicated in the Mexican or US Latino samples. Conclusions We conclude that the 14q11 and 15q22 regions may be associated with asthma symptoms in childhood. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0296-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gustavo N O Costa
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Frank Dudbridge
- Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | | | - Thiago M da Silva
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | | | - Agostino Strina
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil.
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.
| | - Wagner C S Magalhães
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Maira R Rodrigues
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Mateus H Gouveia
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Fernanda S G Kehdy
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | | | - Bernardo Horta
- Programa de Pós Graduação em Epidemiologia, Universidade Federal de Pelotas, Pelotas, Brazil.
| | | | - Maria Pino-Yanes
- Department of Medicine, University of California, San Francisco, USA.
| | - Blanca Del Rio Navarro
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Dana B Hancock
- Behavioral and Urban Health Program, Research Triangle Institute (RTI) International, Research Triangle Park, North Carolina, USA.
| | - Stephanie London
- Department of Health and Human Services, Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.
| | | | - Alexandre C Pereira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.
| | - Eduardo Tarazona
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Mauricio L Barreto
- Instituto de Saúde Coletiva, Universidade Federal da Bahia, Salvador, Brazil. .,Centro de Pesquisa Gonçalo Muniz, Fundação Osvaldo Cruz, Salvador, Brazil.
| |
Collapse
|
26
|
Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK Biobank. THE LANCET. RESPIRATORY MEDICINE 2015; 3:769-81. [PMID: 26423011 PMCID: PMC4593935 DOI: 10.1016/s2213-2600(15)00283-0] [Citation(s) in RCA: 281] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/07/2015] [Accepted: 07/09/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND Understanding the genetic basis of airflow obstruction and smoking behaviour is key to determining the pathophysiology of chronic obstructive pulmonary disease (COPD). We used UK Biobank data to study the genetic causes of smoking behaviour and lung health. METHODS We sampled individuals of European ancestry from UK Biobank, from the middle and extremes of the forced expiratory volume in 1 s (FEV1) distribution among heavy smokers (mean 35 pack-years) and never smokers. We developed a custom array for UK Biobank to provide optimum genome-wide coverage of common and low-frequency variants, dense coverage of genomic regions already implicated in lung health and disease, and to assay rare coding variants relevant to the UK population. We investigated whether there were shared genetic causes between different phenotypes defined by extremes of FEV1. We also looked for novel variants associated with extremes of FEV1 and smoking behaviour and assessed regions of the genome that had already shown evidence for a role in lung health and disease. We set genome-wide significance at p<5 × 10(-8). FINDINGS UK Biobank participants were recruited from March 15, 2006, to July 7, 2010. Sample selection for the UK BiLEVE study started on Nov 22, 2012, and was completed on Dec 20, 2012. We selected 50,008 unique samples: 10,002 individuals with low FEV1, 10,000 with average FEV1, and 5002 with high FEV1 from each of the heavy smoker and never smoker groups. We noted a substantial sharing of genetic causes of low FEV1 between heavy smokers and never smokers (p=2.29 × 10(-16)) and between individuals with and without doctor-diagnosed asthma (p=6.06 × 10(-11)). We discovered six novel genome-wide significant signals of association with extremes of FEV1, including signals at four novel loci (KANSL1, TSEN54, TET2, and RBM19/TBX5) and independent signals at two previously reported loci (NPNT and HLA-DQB1/HLA-DQA2). These variants also showed association with COPD, including in individuals with no history of smoking. The number of copies of a 150 kb region containing the 5' end of KANSL1, a gene that is important for epigenetic gene regulation, was associated with extremes of FEV1. We also discovered five new genome-wide significant signals for smoking behaviour, including a variant in NCAM1 (chromosome 11) and a variant on chromosome 2 (between TEX41 and PABPC1P2) that has a trans effect on expression of NCAM1 in brain tissue. INTERPRETATION By sampling from the extremes of the lung function distribution in UK Biobank, we identified novel genetic causes of lung function and smoking behaviour. These results provide new insight into the specific mechanisms underlying airflow obstruction, COPD, and tobacco addiction, and show substantial shared genetic architecture underlying airflow obstruction across individuals, irrespective of smoking behaviour and other airway disease. FUNDING Medical Research Council.
Collapse
|
27
|
DNA methylation, bacteria and airway inflammation: latest insights. Curr Opin Allergy Clin Immunol 2015; 15:27-32. [PMID: 25479316 DOI: 10.1097/aci.0000000000000130] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW DNA methylation is an epigenetic mechanism that has been implicated in the pathogenesis of chronic inflammatory diseases by regulating differentiation, proliferation, apoptosis, and activation of immune cells. Changes in the methylation status of relevant genes have been linked to the origin, perpetuation, and severity of airway diseases. The DNA methylation profile can be also modified by the action of viral and bacterial colonization. Bacteria and specially Staphylococcus aureus toxins are recognized inflammatory amplifying factors in both lower and upper airway chronic diseases. This review summarizes the existent knowledge about the role of DNA methylation changes in chronic airway diseases and the contribution of bacterial infection on this event. RECENT FINDINGS It has been demonstrated that changes in DNA methylation, either intrinsic or induced by allergen or infection, may be linked to the pathogenesis of asthma and allergy. These changes in methylation may suppress the production of anti-inflammatory mediators and increase the survival and activation of pro-inflammatory cells, as well as modify the immune response in response to bacterial infection, increasing their survival and pathogenicity within the infected organism. SUMMARY Understanding the intrinsic epigenetic mechanisms, as well as the effect of environment -for example, bacterial infection in the pathogenesis of airways diseases - will greatly improve the management and the diagnosis of these diseases.
Collapse
|
28
|
Bousquet J, Anto JM, Wickman M, Keil T, Valenta R, Haahtela T, Lodrup Carlsen K, van Hage M, Akdis C, Bachert C, Akdis M, Auffray C, Annesi-Maesano I, Bindslev-Jensen C, Cambon-Thomsen A, Carlsen KH, Chatzi L, Forastiere F, Garcia-Aymerich J, Gehrig U, Guerra S, Heinrich J, Koppelman GH, Kowalski ML, Lambrecht B, Lupinek C, Maier D, Melén E, Momas I, Palkonen S, Pinart M, Postma D, Siroux V, Smit HA, Sunyer J, Wright J, Zuberbier T, Arshad SH, Nadif R, Thijs C, Andersson N, Asarnoj A, Ballardini N, Ballereau S, Bedbrook A, Benet M, Bergstrom A, Brunekreef B, Burte E, Calderon M, De Carlo G, Demoly P, Eller E, Fantini MP, Hammad H, Hohman C, Just J, Kerkhof M, Kogevinas M, Kull I, Lau S, Lemonnier N, Mommers M, Nawijn M, Neubauer A, Oddie S, Pellet J, Pin I, Porta D, Saes Y, Skrindo I, Tischer CG, Torrent M, von Hertzen L. Are allergic multimorbidities and IgE polysensitization associated with the persistence or re-occurrence of foetal type 2 signalling? The MeDALL hypothesis. Allergy 2015; 70:1062-78. [PMID: 25913421 DOI: 10.1111/all.12637] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2015] [Indexed: 12/22/2022]
Abstract
Allergic diseases [asthma, rhinitis and atopic dermatitis (AD)] are complex. They are associated with allergen-specific IgE and nonallergic mechanisms that may coexist in the same patient. In addition, these diseases tend to cluster and patients present concomitant or consecutive diseases (multimorbidity). IgE sensitization should be considered as a quantitative trait. Important clinical and immunological differences exist between mono- and polysensitized subjects. Multimorbidities of allergic diseases share common causal mechanisms that are only partly IgE-mediated. Persistence of allergic diseases over time is associated with multimorbidity and/or IgE polysensitization. The importance of the family history of allergy may decrease with age. This review puts forward the hypothesis that allergic multimorbidities and IgE polysensitization are associated and related to the persistence or re-occurrence of foetal type 2 signalling. Asthma, rhinitis and AD are manifestations of a common systemic immune imbalance (mesodermal origin) with specific patterns of remodelling (ectodermal or endodermal origin). This study proposes a new classification of IgE-mediated allergic diseases that allows the definition of novel phenotypes to (i) better understand genetic and epigenetic mechanisms, (ii) better stratify allergic preschool children for prognosis and (iii) propose novel strategies of treatment and prevention.
Collapse
Affiliation(s)
- J. Bousquet
- University Hospital; Montpellier France
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - J. M. Anto
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - M. Wickman
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - T. Keil
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
- Institute for Clinical Epidemiology and Biometry; University of Wuerzburg; Wuerzburg Germany
| | - R. Valenta
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - T. Haahtela
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| | - K. Lodrup Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - M. van Hage
- Clinical Immunology and Allergy Unit; Department of Medicine Solna; Karolinska Institutet and University Hospital; Stockholm Sweden
| | - C. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Bachert
- ENT Department; Ghent University Hospital; Gent Belgium
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zurich; Davos Switzerland
| | - C. Auffray
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Annesi-Maesano
- EPAR U707 INSERM; Paris France
- EPAR UMR-S UPMC; Paris VI; Paris France
| | - C. Bindslev-Jensen
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - A. Cambon-Thomsen
- UMR Inserm U1027; Université de Toulouse III Paul Sabatier; Toulouse France
| | - K. H. Carlsen
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- University of Oslo; Oslo Norway
| | - L. Chatzi
- Department of Social Medicine; Faculty of Medicine; University of Crete; Heraklion Crete Greece
| | - F. Forastiere
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - J. Garcia-Aymerich
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - U. Gehrig
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - S. Guerra
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - J. Heinrich
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - G. H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. L. Kowalski
- Department of Immunology, Rheumatology and Allergy; Medical University of Lodz; Lodz Poland
| | - B. Lambrecht
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Lupinek
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | | | - E. Melén
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - I. Momas
- Department of Public Health and Biostatistics, EA 4064; Paris Descartes University; Paris France
- Paris Municipal Department of Social Action, Childhood, and Health; Paris France
| | - S. Palkonen
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - M. Pinart
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - D. Postma
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - H. A. Smit
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - J. Sunyer
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - J. Wright
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - T. Zuberbier
- Allergy-Centre-Charité at the Department of Dermatology; Charité - Universitätsmedizin Berlin; Berlin Germany
- Secretary General of the Global Allergy and Asthma European Network (GA2LEN); Berlin Germany
| | - S. H. Arshad
- David Hide Asthma and Allergy Research Centre; Isle of Wight UK
| | - R. Nadif
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - C. Thijs
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - N. Andersson
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - A. Asarnoj
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - N. Ballardini
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Ballereau
- European Institute for Systems Biology and Medicine; Lyon France
| | - A. Bedbrook
- MACVIA-LR; Contre les MAladies Chroniques pour un VIeillissement Actif en Languedoc-Roussillon; European Innovation Partnership on Active and Healthy Ageing Reference Site; Paris France
| | - M. Benet
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
| | - A. Bergstrom
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - B. Brunekreef
- Julius Center of Health Sciences and Primary Care; University Medical Center Utrecht; University of Utrecht; Utrecht the Netherlands
| | - E. Burte
- INSERM; VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, U1168; Paris France
- UVSQ; UMR-S 1168; Université Versailles St-Quentin-en-Yvelines; Versailles France
| | - M. Calderon
- National Heart and Lung Institute; Imperial College London; Royal Brompton Hospital NHS; London UK
| | - G. De Carlo
- EFA European Federation of Allergy and Airways Diseases Patients' Associations; Brussels Belgium
| | - P. Demoly
- Department of Respiratory Diseases; Montpellier University Hospital; Montpellier France
| | - E. Eller
- Department of Dermatology and Allergy Centre; Odense University Hospital; Odense Denmark
| | - M. P. Fantini
- Department of Medicine and Public Health; Alma Mater Studiorum - University of Bologna; Bologna Italy
| | - H. Hammad
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - C. Hohman
- Institute of Social Medicine, Epidemiology and Health Economics; Charité - Universitätsmedizin Berlin; Berlin Germany
| | - J. Just
- Allergology Department; Centre de l'Asthme et des Allergies; Hôpital d'Enfants Armand-Trousseau (APHP); Paris France
- Institut Pierre Louis d'Epidémiologie et de Santé Publique; Equipe EPAR; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 1136; Paris France
| | - M. Kerkhof
- Department of Respiratory Medicine; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | - M. Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Hospital del Mar Research Institute (IMIM); Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP); Barcelona Spain
- Department of Experimental and Health Sciences; University of Pompeu Fabra (UPF); Barcelona Spain
| | - I. Kull
- Sachs’ Children's Hospital; Stockholm Sweden
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
| | - S. Lau
- Department for Pediatric Pneumology and Immunology; Charité Medical University; Berlin Germany
| | - N. Lemonnier
- European Institute for Systems Biology and Medicine; Lyon France
| | - M. Mommers
- Department of Epidemiology; CAPHRI School of Public Health and Primary Care; Maastricht University; Maastricht the Netherlands
| | - M. Nawijn
- Department of Pediatric Pulmonology and Pediatric Allergology; GRIAC Research Institute; University Medical Center Groningen; Beatrix Children's Hospital; University of Groningen; Groningen the Netherlands
| | | | - S. Oddie
- Bradford Institute for Health Research; Bradford Royal Infirmary; Bradford UK
| | - J. Pellet
- European Institute for Systems Biology and Medicine; Lyon France
| | - I. Pin
- Département de pédiatrie; CHU de Grenoble; Grenoble Cedex 9 France
| | - D. Porta
- Department of Epidemiology; Regional Health Service Lazio Region; Rome Italy
| | - Y. Saes
- VIB Inflammation Research Center; Ghent University; Ghent Belgium
| | - I. Skrindo
- Department of Paediatrics; Oslo University Hospital; Oslo Norway
- Faculty of Medicine; Institute of Clinical Medicine; University of Oslo; Oslo Norway
| | - C. G. Tischer
- Institute of Epidemiology; German Research Centre for Environmental Health; Helmholtz Zentrum München; Neuherberg Germany
| | - M. Torrent
- Centre for Research in Environmental Epidemiology (CREAL); Barcelona Spain
- Area de Salut de Menorca, ib-salut; Illes Balears Spain
| | - L. von Hertzen
- Skin and Allergy Hospital; Helsinki University Hospital; Helsinki Finland
| |
Collapse
|
29
|
Cook DN, Kang HS, Jetten AM. Retinoic Acid-Related Orphan Receptors (RORs): Regulatory Functions in Immunity, Development, Circadian Rhythm, and Metabolism. NUCLEAR RECEPTOR RESEARCH 2015. [PMID: 26878025 DOI: 10.1038/nbt.3121.chip-nexus] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023] Open
Abstract
In this overview, we provide an update on recent progress made in understanding the mechanisms of action, physiological functions, and roles in disease of retinoic acid related orphan receptors (RORs). We are particularly focusing on their roles in the regulation of adaptive and innate immunity, brain function, retinal development, cancer, glucose and lipid metabolism, circadian rhythm, metabolic and inflammatory diseases and neuropsychiatric disorders. We also summarize the current status of ROR agonists and inverse agonists, including their regulation of ROR activity and their therapeutic potential for management of various diseases in which RORs have been implicated.
Collapse
Affiliation(s)
- Donald N Cook
- Immunogenetics Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Hong Soon Kang
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Anton M Jetten
- Cell Biology Section, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
30
|
Ather JL, Martin RA, Ckless K, Poynter ME. Inflammasome Activity in Non-Microbial Lung Inflammation. JOURNAL OF ENVIRONMENTAL IMMUNOLOGY AND TOXICOLOGY 2014; 1:108-117. [PMID: 25642415 PMCID: PMC4308734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The understanding of interleukin-1 (IL-1) family cytokines in inflammatory disease has rapidly developed, due in part to the discovery and characterization of inflammasomes, which are multi-subunit intracellular protein scaffolds principally enabling recognition of a myriad of cellular stimuli, leading to the activation of caspase-1 and the processing of IL-1β and IL-18. Studies continue to elucidate the role of inflammasomes in immune responses induced by both microbes and environmental factors. This review focuses on the current understanding of inflammasome activity in the lung, with particular focus on the non-microbial instigators of inflammasome activation, including inhaled antigens, oxidants, cigarette smoke, diesel exhaust particles, mineral fibers, and engineered nanomaterials, as well as exposure to trauma and pre-existing inflammatory conditions such as metabolic syndrome. Inflammasome activity in these sterile inflammatory states contribute to diseases including asthma, chronic obstructive disease, acute lung injury, ventilator-induced lung injury, pulmonary fibrosis, and lung cancer.
Collapse
Affiliation(s)
- Jennifer L. Ather
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| | - Rebecca A. Martin
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| | - Karina Ckless
- Chemistry Department, State University of New York at Plattsburgh, Plattsburgh, USA
| | - Matthew E. Poynter
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, USA
| |
Collapse
|
31
|
Gunawardhana LP, Gibson PG, Simpson JL, Benton MC, Lea RA, Baines KJ. Characteristic DNA methylation profiles in peripheral blood monocytes are associated with inflammatory phenotypes of asthma. Epigenetics 2014; 9:1302-16. [PMID: 25147914 DOI: 10.4161/epi.33066] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epigenetic changes including DNA methylation caused by environmental exposures may contribute to the heterogeneous inflammatory response in asthma. Here we investigate alterations in DNA methylation of purified blood monocytes that are associated with inflammatory phenotypes of asthma. Peripheral blood was collected from adults with eosinophilic asthma (EA; n = 21), paucigranulocytic asthma (PGA; n = 22), neutrophilic asthma (NA; n = 9), and healthy controls (n = 10). Blood monocytes were isolated using ficoll density gradient and immuno-magnetic cell separation. Bisulfite converted genomic DNA was hybridized to Illumina Infinium Methylation27 arrays and analyzed for differential methylation using R/Bioconductor packages; networks of gene interactions were identified using the STRING database. Compared with healthy controls, differentially methylated CpG loci were identified in EA (n = 413), PGA (n = 495), and NA (n = 89). We found that 223, 237, and 72 loci were significantly hypermethylated in EA, PGA, and NA, respectively. Nine genes were common to all three phenotypes and showed increased methylation in asthma. Three pathway networks were identified in EA, involved in purine metabolism, calcium signaling, and ECM-receptor interaction. In PGA, two networks were identified, involved in neuroactive ligand-receptor interaction and ubiquitin mediated proteolysis. In NA, one network was identified involving sFRP1 as a key node, over representing the Wnt signaling pathway. We have identified characteristic alterations in DNA methylation that are associated with inflammatory phenotypes of asthma and may contribute to the disease mechanisms. This network-based characterization may help in the development of epigenetic biomarkers and therapeutic targets for asthma.
Collapse
Affiliation(s)
- Lakshitha P Gunawardhana
- Priority Research Centre for Asthma and Respiratory Disease; Hunter Medical Research Institute; The University of Newcastle; Newcastle, NSW Australia; Department of Respiratory & Sleep Medicine; HMRI; John Hunter Hospital; New Lambton, NSW Australia
| | - Peter G Gibson
- Priority Research Centre for Asthma and Respiratory Disease; Hunter Medical Research Institute; The University of Newcastle; Newcastle, NSW Australia; Department of Respiratory & Sleep Medicine; HMRI; John Hunter Hospital; New Lambton, NSW Australia
| | - Jodie L Simpson
- Priority Research Centre for Asthma and Respiratory Disease; Hunter Medical Research Institute; The University of Newcastle; Newcastle, NSW Australia; Department of Respiratory & Sleep Medicine; HMRI; John Hunter Hospital; New Lambton, NSW Australia
| | - Miles C Benton
- Genomics Research Centre; Institute of Health and Biomedical Innovation; Queensland Institute of Technology; Brisbane, QLD Australia
| | - Rodney A Lea
- Genomics Research Centre; Institute of Health and Biomedical Innovation; Queensland Institute of Technology; Brisbane, QLD Australia
| | - Katherine J Baines
- Priority Research Centre for Asthma and Respiratory Disease; Hunter Medical Research Institute; The University of Newcastle; Newcastle, NSW Australia; Department of Respiratory & Sleep Medicine; HMRI; John Hunter Hospital; New Lambton, NSW Australia
| |
Collapse
|
32
|
Zhong B, Yang X, Sun Q, Liu L, Lan X, Tian J, He Q, Hou W, Liu H, Jiang C, Gao N, Lu S. Pdcd4 modulates markers of macrophage alternative activation and airway remodeling in antigen-induced pulmonary inflammation. J Leukoc Biol 2014; 96:1065-75. [PMID: 25097194 DOI: 10.1189/jlb.3a0313-136rrr] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pdcd4 has been known as a tumor-suppressor gene initially and is up-regulated during apoptosis. Surprisingly, we found that Pdcd4 was differentially expressed in the lung from E3 rats with AIPI, an animal model for asthma, but the precise role of Pdcd4 in AIPI still remained to be defined. In the present study, we first evaluated the expression of Pdcd4 in lung from control and AIPI rats with RT-qPCR, Western blot, and immunohistochemistry. Then, we investigated the effects of intervention of Pdcd4 on markers of macrophage alternative activation and airway remodeling. Upon challenging E3 rats with OVA, Pdcd4 was up-regulated in lung tissue with AIPI. Immunohistochemistry results showed that alveolar macrophages and airway epithelia expressed Pdcd4 protein. Overexpression of Pdcd4 in the rat alveolar macrophage cell line, NR8383 cells, increased the mRNA expression of arginase-1 and TGF-β1, which are markers of macrophage alternative activation. In response to Pdcd4 RNAi in NR8383 cells, the mRNA expression of markers Fizz1, Ym1/2, arginase-1, and TGF-β1 was decreased significantly. In addition, Pdcd4 RNAi in AIPI rats led to a decrease of the mRNA expression of Fizz1, Ym1/2, arginase-1, and TGF-β1 in BALF cells. Finally, knockdown of Pdcd4 suppressed airway eosinophil infiltration, bronchus collagen deposition, and mucus production. Overall, these results suggest that Pdcd4 may be worthy of further investigation as a target for macrophage alternative activation and airway remodeling in allergic pulmonary inflammation.
Collapse
Affiliation(s)
- Bo Zhong
- Department of Genetics and Molecular Biology, Departments of Pediatrics and
| | - Xudong Yang
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Qingzhu Sun
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Li Liu
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Xi Lan
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Jia Tian
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Qirui He
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Wei Hou
- Departments of Pediatrics and
| | | | - Congshan Jiang
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Ning Gao
- Clinical Laboratory, the Second Affiliated Hospital, and
| | - Shemin Lu
- Department of Genetics and Molecular Biology, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China Department of Epidemiology and Health Statistics, School of Public Health, Xi'an Jiaotong University College of Medicine, and
| |
Collapse
|
33
|
Bérubé JC, Bossé Y. Future clinical implications emerging from recent genome-wide expression studies in asthma. Expert Rev Clin Immunol 2014; 10:985-1004. [PMID: 25001610 DOI: 10.1586/1744666x.2014.932249] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Host susceptibility to environmental triggers is the most likely explanation for the development of asthma. Quantifying gene expression levels in disease-relevant tissues and cell types using fast evolving genomic technologies have generated new hypotheses about the pathogenesis of asthma and identified new therapeutic targets to treat asthma and asthma-exacerbations. New biomarkers and distinct transcriptomic phenotypes in blood, sputum and other tissues were also identified and proved effective to refine asthma classification and guide targeted therapies. The wealth of information provided by transcriptomic studies in asthma is yet to be fully exploited, but discoveries in this field may soon be implemented in clinical settings to improve diagnosis and treatment of patients afflicted with this common disease.
Collapse
Affiliation(s)
- Jean-Christophe Bérubé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Pavillon Marguerite-d'Youville, Y4190, 2725 Chemin Ste-Foy, Quebec, Canada, G1V 4G5
| | | |
Collapse
|
34
|
Genome-wide association analysis identifies six new loci associated with forced vital capacity. Nat Genet 2014; 46:669-77. [PMID: 24929828 DOI: 10.1038/ng.3011] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Abstract
Forced vital capacity (FVC), a spirometric measure of pulmonary function, reflects lung volume and is used to diagnose and monitor lung diseases. We performed genome-wide association study meta-analysis of FVC in 52,253 individuals from 26 studies and followed up the top associations in 32,917 additional individuals of European ancestry. We found six new regions associated at genome-wide significance (P < 5 × 10(-8)) with FVC in or near EFEMP1, BMP6, MIR129-2-HSD17B12, PRDM11, WWOX and KCNJ2. Two loci previously associated with spirometric measures (GSTCD and PTCH1) were related to FVC. Newly implicated regions were followed up in samples from African-American, Korean, Chinese and Hispanic individuals. We detected transcripts for all six newly implicated genes in human lung tissue. The new loci may inform mechanisms involved in lung development and the pathogenesis of restrictive lung disease.
Collapse
|
35
|
Sordillo J, Raby BA. Gene expression profiling in asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 795:157-81. [PMID: 24162908 DOI: 10.1007/978-1-4614-8603-9_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Joanne Sordillo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 181 Longwood Avenue, Boston, MA, 02115, USA,
| | | |
Collapse
|
36
|
Bortolotti D, Gentili V, Rotola A, Cassai E, Rizzo R, Luca DD. Impact of HLA-G analysis in prevention, diagnosis and treatment of pathological conditions. World J Methodol 2014; 4:11-25. [PMID: 25237627 PMCID: PMC4145573 DOI: 10.5662/wjm.v4.i1.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 12/28/2013] [Accepted: 01/16/2014] [Indexed: 02/06/2023] Open
Abstract
Human leukocyte antigen-G (HLA-G) is a non-classical HLA class I molecule that differs from classical HLA class I molecules by low polymorphism and tissue distribution. HLA-G is a tolerogenic molecule with an immune-modulatory and anti-inflammatory function on both innate and adaptative immunity. This peculiar characteristic of HLA-G has led to investigations of its role in pathological conditions in order to define possible uses in diagnosis, prevention and treatment. In recent years, HLA-G has been shown to have an important implication in different inflammatory and autoimmune diseases, pregnancy complications, tumor development and aggressiveness, and susceptibility to viral infections. In fact, HLA-G molecules have been reported to alternate at both genetic and protein level in different disease situations, supporting its crucial role in pathological conditions. Specific pathologies show altered levels of soluble (s)HLA-G and different HLA-G gene polymorphisms seem to correlate with disease. This review aims to update scientific knowledge on the contribution of HLA-G in managing pathological conditions.
Collapse
|
37
|
Sharma S, Chhabra D, Kho AT, Hayden LP, Tantisira KG, Weiss ST. The genomic origins of asthma. Thorax 2014; 69:481-7. [PMID: 24668408 DOI: 10.1136/thoraxjnl-2014-205166] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lung function tracks from the earliest age that it can be reliably measured. Genome wide association studies suggest that most variants identified for common complex traits are regulatory in function and active during fetal development. Fetal programming of gene expression during development is critical to the formation of a normal lung. An understanding of how fetal developmental genes related to diseases of the lungs and airways is a critical area for research. This review article considers the developmental origins hypothesis, the stages of normal lung development and a variety of environmental exposures that might influence the developmental process: in utero cigarette smoke exposure, vitamin D and folate. We conclude with some information on developmental genes and asthma.
Collapse
Affiliation(s)
- Sunita Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, , Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
38
|
Obeidat M, Miller S, Probert K, Billington CK, Henry AP, Hodge E, Nelson CP, Stewart CE, Swan C, Wain LV, Artigas MS, Melén E, Ushey K, Hao K, Lamontagne M, Bossé Y, Postma DS, Tobin MD, Sayers I, Hall IP. GSTCD and INTS12 regulation and expression in the human lung. PLoS One 2013; 8:e74630. [PMID: 24058608 PMCID: PMC3776747 DOI: 10.1371/journal.pone.0074630] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/05/2013] [Indexed: 12/29/2022] Open
Abstract
Genome-Wide Association Study (GWAS) meta-analyses have identified a strong association signal for lung function, which maps to a region on 4q24 containing two oppositely transcribed genes: glutathione S-transferase, C-terminal domain containing (GSTCD) and integrator complex subunit 12 (INTS12). Both genes were found to be expressed in a range of human airway cell types. The promoter regions and transcription start sites were determined in mRNA from human lung and a novel splice variant was identified for each gene. We obtained the following evidence for GSTCD and INTS12 co-regulation and expression: (i) correlated mRNA expression was observed both via Q-PCR and in a lung expression quantitative trait loci (eQTL) study, (ii) induction of both GSTCD and INTS12 mRNA expression in human airway smooth muscle cells was seen in response to TGFβ1, (iii) a lung eQTL study revealed that both GSTCD and INTS12 mRNA levels positively correlate with percent predicted FEV1, and (iv) FEV1 GWAS associated SNPs in 4q24 were found to act as an eQTL for INTS12 in a number of tissues. In fixed sections of human lung tissue, GSTCD protein expression was ubiquitous, whereas INTS12 expression was predominantly in epithelial cells and pneumocytes. During human fetal lung development, GSTCD protein expression was observed to be highest at the earlier pseudoglandular stage (10-12 weeks) compared with the later canalicular stage (17-19 weeks), whereas INTS12 expression levels did not alter throughout these stages. Knowledge of the transcriptional and translational regulation and expression of GSTCD and INTS12 provides important insights into the potential role of these genes in determining lung function. Future work is warranted to fully define the functions of INTS12 and GSTCD.
Collapse
Affiliation(s)
- Ma’en Obeidat
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
- James Hogg Research Centre, Institute for Heart and Lung Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanne Miller
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Kelly Probert
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Charlotte K. Billington
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Amanda P. Henry
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Carl P. Nelson
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Ceri E. Stewart
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Caroline Swan
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Louise V. Wain
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - María Soler Artigas
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs’ Children’s Hospital, Stockholm, Sweden
| | - Kevin Ushey
- James Hogg Research Centre, Institute for Heart and Lung Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Maxime Lamontagne
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Canada
| | - Yohan Bossé
- Department of Molecular Medicine, Laval University, Québec City, Canada
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Québec City, Canada
| | - Dirkje S. Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin D. Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, United Kingdom
- National Institute for Health Research (NIHR) Leicester Respiratory Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| | - Ian P. Hall
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Center, Nottingham, United Kingdom
| |
Collapse
|
39
|
Melén E, Granell R, Kogevinas M, Strachan D, Gonzalez JR, Wjst M, Jarvis D, Ege M, Braun-Fahrländer C, Genuneit J, Horak E, Bouzigon E, Demenais F, Kauffmann F, Siroux V, Michel S, von Berg A, Heinzmann A, Kabesch M, Probst-Hensch NM, Curjuric I, Imboden M, Rochat T, Henderson J, Sterne JAC, McArdle WL, Hui J, James AL, William Musk A, Palmer LJ, Becker A, Kozyrskyj AL, Chan-Young M, Park JE, Leung A, Daley D, Freidin MB, Deev IA, Ogorodova LM, Puzyrev VP, Celedón JC, Brehm JM, Cloutier MM, Canino G, Acosta-Pérez E, Soto-Quiros M, Avila L, Bergström A, Magnusson J, Söderhäll C, Kull I, Scholtens S, Marike Boezen H, Koppelman GH, Wijga AH, Marenholz I, Esparza-Gordillo J, Lau S, Lee YA, Standl M, Tiesler CMT, Flexeder C, Heinrich J, Myers RA, Ober C, Nicolae DL, Farrall M, Kumar A, Moffatt MF, Cookson WOCM, Lasky-Su J. Genome-wide association study of body mass index in 23 000 individuals with and without asthma. Clin Exp Allergy 2013; 43:463-74. [PMID: 23517042 DOI: 10.1111/cea.12054] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 09/28/2012] [Accepted: 10/22/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND Both asthma and obesity are complex disorders that are influenced by environmental and genetic factors. Shared genetic factors between asthma and obesity have been proposed to partly explain epidemiological findings of co-morbidity between these conditions. OBJECTIVE To identify genetic variants that are associated with body mass index (BMI) in asthmatic children and adults, and to evaluate if there are differences between the genetics of BMI in asthmatics and healthy individuals. METHODS In total, 19 studies contributed with genome-wide analysis study (GWAS) data from more than 23 000 individuals with predominantly European descent, of whom 8165 are asthmatics. RESULTS We report associations between several DENND1B variants (P = 2.2 × 10(-7) for rs4915551) on chromosome 1q31 and BMI from a meta-analysis of GWAS data using 2691 asthmatic children (screening data). The top DENND1B single nucleotide polymorphisms(SNPs) were next evaluated in seven independent replication data sets comprising 2014 asthmatics, and rs4915551 was nominally replicated (P < 0.05) in two of the seven studies and of borderline significance in one (P = 0.059). However, strong evidence of effect heterogeneity was observed and overall, the association between rs4915551 and BMI was not significant in the total replication data set, P = 0.71. Using a random effects model, BMI was overall estimated to increase by 0.30 kg/m(2) (P = 0.01 for combined screening and replication data sets, N = 4705) per additional G allele of this DENND1BSNP. FTO was confirmed as an important gene for adult and childhood BMI regardless of asthma status. CONCLUSIONS AND CLINICAL RELEVANCE DENND1B was recently identified as an asthma susceptibility gene in a GWAS on children, and here, we find evidence that DENND1B variants may also be associated with BMI in asthmatic children. However, the association was overall not replicated in the independent data sets and the heterogeneous effect of DENND1B points to complex associations with the studied diseases that deserve further study.
Collapse
Affiliation(s)
- E Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fabbro A, Sucapane A, Toma FM, Calura E, Rizzetto L, Carrieri C, Roncaglia P, Martinelli V, Scaini D, Masten L, Turco A, Gustincich S, Prato M, Ballerini L. Adhesion to carbon nanotube conductive scaffolds forces action-potential appearance in immature rat spinal neurons. PLoS One 2013; 8:e73621. [PMID: 23951361 PMCID: PMC3741175 DOI: 10.1371/journal.pone.0073621] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 07/29/2013] [Indexed: 12/19/2022] Open
Abstract
In the last decade, carbon nanotube growth substrates have been used to investigate neurons and neuronal networks formation in vitro when guided by artificial nano-scaled cues. Besides, nanotube-based interfaces are being developed, such as prosthesis for monitoring brain activity. We recently described how carbon nanotube substrates alter the electrophysiological and synaptic responses of hippocampal neurons in culture. This observation highlighted the exceptional ability of this material in interfering with nerve tissue growth. Here we test the hypothesis that carbon nanotube scaffolds promote the development of immature neurons isolated from the neonatal rat spinal cord, and maintained in vitro. To address this issue we performed electrophysiological studies associated to gene expression analysis. Our results indicate that spinal neurons plated on electro-conductive carbon nanotubes show a facilitated development. Spinal neurons anticipate the expression of functional markers of maturation, such as the generation of voltage dependent currents or action potentials. These changes are accompanied by a selective modulation of gene expression, involving neuronal and non-neuronal components. Our microarray experiments suggest that carbon nanotube platforms trigger reparative activities involving microglia, in the absence of reactive gliosis. Hence, future tissue scaffolds blended with conductive nanotubes may be exploited to promote cell differentiation and reparative pathways in neural regeneration strategies.
Collapse
Affiliation(s)
| | | | - Francesca Maria Toma
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Enrica Calura
- Department of Biology, University of Padua, Padova, Italy
| | - Lisa Rizzetto
- Department of Neuroscience, Psychology, Drug Research and Child's Health, University of Florence, Florence, Italy
- Innovation and Research Center, Fondazione Edmund Mach, San Michele all’Adige, Trento, Italy
| | - Claudia Carrieri
- European Molecular Biology Laboratory, Mouse Biology Unit, Monterotondo (Rome), Italy
| | - Paola Roncaglia
- International School for Advanced Studies (SISSA), Trieste, Italy
- European Bioinformatics Institute (EMBL-EBI), Hinxton, United Kingdom
| | - Valentina Martinelli
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Denis Scaini
- Life Science Department, University of Trieste, Trieste, Italy
- SENIL, ELETTRA Synchrotron Light Source, Trieste, Italy
| | - Lara Masten
- Life Science Department, University of Trieste, Trieste, Italy
| | - Antonio Turco
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | | | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy
| | - Laura Ballerini
- Life Science Department, University of Trieste, Trieste, Italy
| |
Collapse
|
41
|
Hodge E, Nelson CP, Miller S, Billington CK, Stewart CE, Swan C, Malarstig A, Henry AP, Gowland C, Melén E, Hall IP, Sayers I. HTR4 gene structure and altered expression in the developing lung. Respir Res 2013; 14:77. [PMID: 23890215 PMCID: PMC3750317 DOI: 10.1186/1465-9921-14-77] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/23/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Meta-analyses of genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) spanning the 5-hydroxytryptamine receptor 4 (5-HT₄R) gene (HTR4) associated with lung function. The aims of this study were to i) investigate the expression profile of HTR4 in adult and fetal lung tissue and cultured airway cells, ii) further define HTR4 gene structure and iii) explore the potential functional implications of key SNPs using a bioinformatic approach. METHODS Following reverse transcription (RT)-PCR in human brain, 5' rapid amplification of cDNA ends (5' RACE) was used to examine the exonic structure of HTR4 at the 5' end. Quantitative (Q)-PCR was used to quantify HTR4 mRNA expression in total RNA from cultured airway cells and whole lung tissue. Publically available gene microarray data on fetal samples of estimated gestational age 7-22 weeks were mined for HTR4 expression. Immunohistochemistry (IHC; in adult and fetal lung tissue) and a radioligand binding assay (in cultured airway cells) were used to analyze 5-HT₄R protein expression. RESULTS IHC in adult lung, irrespective of the presence of chronic obstructive pulmonary disease (COPD), suggested low level expression of 5-HT₄R protein, which was most prominent in alveolar pneumocytes. There was evidence of differential 5-HT₄R protein levels during gestation in fetal lung, which was also evident in gene expression microarray data. HTR4 mRNA expression, assessed by Q-PCR, was <0.5% relative to brain in total adult lung tissue and in human airway smooth muscle (HASM) and bronchial epithelial cells (HBEC) derived from adult donors. Radioligand binding experiments also indicated that HBEC and HASM cells did not express a significant 5-HT₄R population. 5' RACE in brain identified a novel N-terminal variant, containing an extended N-terminal sequence. The functional significance of key HTR4 SNPs was investigated using the encyclopedia of DNA elements consortium (ENCODE) dataset. These analyses identified multiple alterations in regulatory motifs for transcription factors implicated in lung development, including Foxp1. CONCLUSIONS Taken together, these data suggest a role for HTR4 in lung development, which may at least in part explain the genetic association with lung function.
Collapse
Affiliation(s)
- Emily Hodge
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Carl P Nelson
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Suzanne Miller
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Charlotte K Billington
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Ceri E Stewart
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Caroline Swan
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Anders Malarstig
- Precision Medicine Unit, Pfizer Global Research and Development, Cambridge, UK
| | - Amanda P Henry
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Catherine Gowland
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet and Sachs’ Children’s Hospital, Stockholm, Sweden
| | - Ian P Hall
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Ian Sayers
- Division of Respiratory Medicine, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
42
|
Martin RA, Ather JL, Lundblad LKA, Suratt BT, Boyson JE, Budd RC, Alcorn JF, Flavell RA, Eisenbarth SC, Poynter ME. Interleukin-1 receptor and caspase-1 are required for the Th17 response in nitrogen dioxide-promoted allergic airway disease. Am J Respir Cell Mol Biol 2013; 48:655-64. [PMID: 23371061 DOI: 10.1165/rcmb.2012-0423oc] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nitrogen dioxide (NO2) is an environmental pollutant and endogenously generated oxidant associated with the development, severity, and exacerbation of asthma. NO2 exposure is capable of allergically sensitizing mice to the innocuous inhaled antigen ovalbumin (OVA), promoting neutrophil and eosinophil recruitment, and a mixed Th2/Th17 response upon antigen challenge that is reminiscent of severe asthma. However, the identity of IL-17A-producing cells and the mechanisms governing their ontogeny in NO2-promoted allergic airway disease remain unstudied. We measured the kinetics of lung inflammation after antigen challenge in NO2-promoted allergic airway disease, including inflammatory cells in bronchoalveolar lavage and antigen-specific IL-17A production from the lung. We determined that IL-17A(+) cells were predominately CD4(+)T cell receptor (TCR)β(+) Th17 cells, and that a functional IL-1 receptor was required for Th17, but not Th2, cytokine production after in vitro antigen restimulation of lung cells. The absence of natural killer T cells, γδ T cells, or the inflammasome scaffold nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain (Nlrp)3 did not affect the development of NO2-promoted allergic inflammation or IL-17A production. Similarly, neutrophil depletion or the neutralization of IL-1α during sensitization exerted no effect on these parameters. However, the absence of caspase-1 significantly reduced IL-17A production from lung cells without affecting Th2 cytokines or lung inflammation. Finally, the intranasal administration of IL-1β and the inhalation of antigen promoted allergic sensitization that was reflected by neutrophilic airway inflammation and IL-17A production from CD4(+)TCRβ(+) Th17 cells subsequent to antigen challenge. These data implicate a role for caspase-1 and IL-1β in the IL-1 receptor-dependent Th17 response manifest in NO2-promoted allergic airway disease.
Collapse
Affiliation(s)
- Rebecca A Martin
- Vermont Lung Center, Division of Pulmonary Disease and Critical Care, Department of Medicine, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Interaction between retinoid acid receptor-related orphan receptor alpha (RORA) and neuropeptide S receptor 1 (NPSR1) in asthma. PLoS One 2013; 8:e60111. [PMID: 23565190 PMCID: PMC3615072 DOI: 10.1371/journal.pone.0060111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 02/21/2013] [Indexed: 12/31/2022] Open
Abstract
Retinoid acid receptor-related Orphan Receptor Alpha (RORA) was recently identified as a susceptibility gene for asthma in a genome-wide association study. To investigate the impact of RORA on asthma susceptibility, we performed a genetic association study between RORA single nucleotide polymorphisms (SNPs) in the vicinity of the asthma-associated SNP (rs11071559) and asthma-related traits. Because the regulatory region of a previously implicated asthma susceptibility gene, Neuropeptide S receptor 1 (NPSR1), has predicted elements for RORA binding, we hypothesized that RORA may interact biologically and genetically with NPSR1. 37 RORA SNPs and eight NPSR1 SNPs were genotyped in the Swedish birth cohort BAMSE (2033 children) and the European cross-sectional PARSIFAL study (1120 children). Seven RORA SNPs confined into a 49 kb region were significantly associated with physician-diagnosed childhood asthma. The most significant association with rs7164773 (T/C) was driven by the CC genotype in asthma cases (OR = 2.0, 95%CI 1.36-2.93, p = 0.0003 in BAMSE; and 1.61, 1.18-2.19, p = 0.002 in the combined BAMSE-PARSIFAL datasets, respectively), and strikingly, the risk effect was dependent on the Gln344Arg mutation in NPSR1. In cell models, stimulation of NPSR1 activated a pathway including RORA and other circadian clock genes. Over-expression of RORA decreased NPSR1 promoter activity further suggesting a regulatory loop between these genes. In addition, Rora mRNA expression was lower in the lung tissue of Npsr1 deficient mice compared to wildtype littermates during the early hours of the light period. We conclude that RORA SNPs are associated with childhood asthma and show epistasis with NPSR1, and the interaction between RORA and NPSR1 may be of biological relevance. Combinations of common susceptibility alleles and less common functional polymorphisms may modify the joint risk effects on asthma susceptibility.
Collapse
|
44
|
Abstract
A greater understanding of the regulatory processes contributing to lung development could be helpful to identify strategies to ameliorate morbidity and mortality in premature infants and to identify individuals at risk for congenital and/or chronic lung diseases. Over the past decade, genomics technologies have enabled the production of rich gene expression databases providing information for all genes across developmental time or in diseased tissue. These data sets facilitate systems biology approaches for identifying underlying biological modules and programs contributing to the complex processes of normal development and those that may be associated with disease states. The next decade will undoubtedly see rapid and significant advances in redefining both lung development and disease at the systems level.
Collapse
Affiliation(s)
- Soumyaroop Bhattacharya
- Division of Neonatology and Program in Pediatric Molecular and Personalized Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | | |
Collapse
|
45
|
Nicodemus-Johnson J, Laxman B, Stern RK, Sudi J, Tierney CN, Norwick L, Hogarth DK, McConville JF, Naureckas ET, Sperling AI, Solway J, Krishnan JA, Nicolae DL, White SR, Ober C. Maternal asthma and microRNA regulation of soluble HLA-G in the airway. J Allergy Clin Immunol 2013; 131:1496-503. [PMID: 23534973 DOI: 10.1016/j.jaci.2013.01.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/17/2013] [Accepted: 01/25/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND We previously reported an interaction between maternal asthma and the child's HLA-G genotype on the child's subsequent risk for asthma. The implicated single nucleotide polymorphism at +3142 disrupted a target site for the microRNA (miR)-152 family. We hypothesized that the interaction effect might be mediated by these miRs. OBJECTIVE The objective of this study was to test this hypothesis in adults with asthma who are a subset of the same subjects who participated in our earlier family-based studies. METHODS We measured soluble HLA-G (sHLA-G) concentrations in bronchoalveolar lavage fluid (n = 36) and plasma (n = 57) from adult asthmatic subjects with and without a mother with asthma, and HLA-G and miR-152 family (miR-148a, miR-148b, and miR-152) transcript levels in airway epithelial cells from the same subjects. RESULTS miR-148b levels were significantly increased in airway epithelial cells from asthmatic subjects with an asthmatic mother compared with those seen in asthmatic subjects without an asthmatic mother, and +3142 genotypes were associated with sHLA-G concentrations in bronchoalveolar lavage fluid among asthmatic subjects with an asthmatic mother but not among those with a nonasthmatic mother. Neither effect was observed in the plasma (sHLA-G) or white blood cells (miRNA). CONCLUSION These combined results are consistent with +3142 allele-specific targeting of HLA-G by the miR-152 family and support our hypothesis that miRNA regulation of sHLA-G in the airway is influenced by both the asthma status of the subject's mother and the subject's genotype. Moreover, we demonstrate that the effects of maternal asthma on the gene regulatory landscape in the airways of the mother's children persist into adulthood.
Collapse
|
46
|
Baitaluk M, Kozhenkov S, Ponomarenko J. An integrative approach to inferring gene regulatory module networks. PLoS One 2012; 7:e52836. [PMID: 23285197 PMCID: PMC3527610 DOI: 10.1371/journal.pone.0052836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 11/22/2012] [Indexed: 12/31/2022] Open
Abstract
Background Gene regulatory networks (GRNs) provide insight into the mechanisms of differential gene expression at a system level. However, the methods for inference, functional analysis and visualization of gene regulatory modules and GRNs require the user to collect heterogeneous data from many sources using numerous bioinformatics tools. This makes the analysis expensive and time-consuming. Results In this work, the BiologicalNetworks application–the data integration and network based research environment–was extended with tools for inference and analysis of gene regulatory modules and networks. The backend database of the application integrates public data on gene expression, pathways, transcription factor binding sites, gene and protein sequences, and functional annotations. Thus, all data essential for the gene regulation analysis can be mined publicly. In addition, the user’s data can either be integrated in the database and become public, or kept private within the application. The capabilities to analyze multiple gene expression experiments are also provided. Conclusion The generated modular networks, regulatory modules and binding sites can be visualized and further analyzed within this same application. The developed tools were applied to the mouse model of asthma and the OCT4 regulatory network in embryonic stem cells. Developed methods and data are available through the Java application from BiologicalNetworks program at http://www.biologicalnetworks.org.
Collapse
Affiliation(s)
- Michael Baitaluk
- San Diego Supercomputer Center, University of California San Diego, La Jolla, California, United States of America
| | - Sergey Kozhenkov
- San Diego Supercomputer Center, University of California San Diego, La Jolla, California, United States of America
| | - Julia Ponomarenko
- San Diego Supercomputer Center, University of California San Diego, La Jolla, California, United States of America
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Affiliation(s)
- Erik Melén
- Institute of Environmental Medicine and Centre for Allergy Research, Karolinska Institutet, Stockholm,
| | | |
Collapse
|
48
|
Genome-wide association studies of asthma in population-based cohorts confirm known and suggested loci and identify an additional association near HLA. PLoS One 2012; 7:e44008. [PMID: 23028483 PMCID: PMC3461045 DOI: 10.1371/journal.pone.0044008] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 07/27/2012] [Indexed: 01/03/2023] Open
Abstract
RATIONALE Asthma has substantial morbidity and mortality and a strong genetic component, but identification of genetic risk factors is limited by availability of suitable studies. OBJECTIVES To test if population-based cohorts with self-reported physician-diagnosed asthma and genome-wide association (GWA) data could be used to validate known associations with asthma and identify novel associations. METHODS The APCAT (Analysis in Population-based Cohorts of Asthma Traits) consortium consists of 1,716 individuals with asthma and 16,888 healthy controls from six European-descent population-based cohorts. We examined associations in APCAT of thirteen variants previously reported as genome-wide significant (P<5 x 10(-8)) and three variants reported as suggestive (P<5× 10(-7)). We also searched for novel associations in APCAT (Stage 1) and followed-up the most promising variants in 4,035 asthmatics and 11,251 healthy controls (Stage 2). Finally, we conducted the first genome-wide screen for interactions with smoking or hay fever. MAIN RESULTS We observed association in the same direction for all thirteen previously reported variants and nominally replicated ten of them. One variant that was previously suggestive, rs11071559 in RORA, now reaches genome-wide significance when combined with our data (P = 2.4 × 10(-9)). We also identified two genome-wide significant associations: rs13408661 near IL1RL1/IL18R1 (P(Stage1+Stage2) = 1.1x10(-9)), which is correlated with a variant recently shown to be associated with asthma (rs3771180), and rs9268516 in the HLA region (P(Stage1+Stage2) = 1.1x10(-8)), which appears to be independent of previously reported associations in this locus. Finally, we found no strong evidence for gene-environment interactions with smoking or hay fever status. CONCLUSIONS Population-based cohorts with simple asthma phenotypes represent a valuable and largely untapped resource for genetic studies of asthma.
Collapse
|
49
|
Melén E, Pershagen G. Pathophysiology of asthma: lessons from genetic research with particular focus on severe asthma. J Intern Med 2012; 272:108-20. [PMID: 22632610 DOI: 10.1111/j.1365-2796.2012.02555.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is good evidence that both inherited and environmental factors influence the risk of developing asthma. Only recently, large well-designed studies have been undertaken with the power to identify the genetic causes for asthma, and methods developed in parallel with the Human Genome Project, such as gene expression and epigenetic studies, have made large-scale analyses of functional genetics possible. In this review, we discuss the recent findings from genetic and genomic research studies of asthma, particularly severe asthma, and highlight specific genes for which there are multiple lines of evidence for involvement in asthma pathogenesis. Bio-ontologic enrichment analyses of the most recently identified asthma-related genes point to attributes such as 'molecular and signal transducer activity' and 'immune system processes', which indicates the importance of immunoregulation and inflammatory response in the pathogenesis of asthma. Finally, we discuss how genetic and environmental factors jointly influence asthma susceptibility and summarize how the results may increase understanding of the pathophysiology of asthma-related diseases.
Collapse
Affiliation(s)
- E Melén
- Institute of Environmental Medicine and Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Agriculture represents a major industry worldwide, and despite protection against the development of IgE-mediated diseases, chronic exposure to agriculture-related organic dusts is associated with an increased risk of developing respiratory disease. This article will review the literature regarding new knowledge of important etiologic agents in the dusts and focus on the immunologic responses following acute and repetitive organic dust exposures. RECENT FINDINGS Although endotoxin remains important, there is an emerging role of nonendotoxin components such as peptidoglycans from Gram-positive bacteria. Pattern recognition receptors including Toll-like receptor 4 (TLR4), TLR2 and intracellular nucleotide oligomerization domain-like receptors are partially responsible for mediating the inflammatory consequences. Repeated organic dust exposures modulate innate and adaptive immune function with a resultant adaptation-like response. However, repetitive exposures cause lung parenchymal inflammation, chronic disease, and lung function decline over time. SUMMARY The immunological consequences of organic dust exposure in the farming industry are likely explained by the diversity of microbial motifs in dust that can elicit differing innate immune receptor signaling pathways. Whereas initial activation results in a robust inflammatory response, repetitive dust exposures modulate immunity. This can result in low-grade, chronic inflammation, and/or protection against allergic disease.
Collapse
|