1
|
Lee YH, Kim M, Ku CH, Lee HS, Lee JH, Kwon DH, Lee EJ. Association between poly-sensitisation and sensitisation to staphylococcal enterotoxin A and B affecting allergic severity in children. Eur Arch Otorhinolaryngol 2023; 280:4121-4129. [PMID: 37039894 DOI: 10.1007/s00405-023-07968-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/03/2023] [Indexed: 04/12/2023]
Abstract
PURPOSE Accumulating evidence suggests that Staphylococcus aureus plays a significant role as a disease modifier in upper and lower airway diseases. We aimed to assess the association between staphylococcal enterotoxins (SEs) with allergic diseases and the degree of allergen sensitisation in children, which remains unclear. METHODS We retrospectively reviewed the medical records of 455 patients aged 3-18 years between March 2018 and March 2022. Clinical history and demographic data were obtained. The baseline study included paranasal sinus X-ray scan, multiple allergen simultaneous test, and ImmunoCAP® for measuring serum total and specific immunoglobulin E (IgE) levels to allergens and staphylococcal enterotoxin A and B (SEA and SEB). RESULTS The mean age was 9.77 ± 4.3 years. 133 patients (29.2%) were sensitised to one inhalant allergen, and 188 patients (41.3%) showed polysensitisation. Patients sensitised to SEs showed higher total and specific IgE levels and total eosinophil counts compared to non-SE-sensitised patients. Sensitisation to SEs is closely associated with polysensitisation to inhalant allergens and allergic multimorbidity. When the SE-IgE value was 0.35 or higher, the odds ratio for allergen polysensitisation was significantly higher than when the SE-IgE value was lower than 0.35. CONCLUSIONS Association between polysensitisation and sensitisation to SEs in children shows the higher the specific IgE levels for SEs, the higher the likelihood of polysensitisation. Considering the relationship between polysensitisation, high IgE levels, and the severity of allergic morbidity, sensitisation to SEs is thought to be related to allergy severity.
Collapse
Affiliation(s)
- Young-Ha Lee
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Minheon Kim
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Cheol Hyo Ku
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Hyun Su Lee
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Joo Hyung Lee
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Dong Hwan Kwon
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea
| | - Eun Jung Lee
- Department of Otorhinolaryngology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, Gangwon-do, Republic of Korea.
| |
Collapse
|
2
|
Watanabe N. The effect of inheritance of IgE responsiveness on the susceptibility of mice to Trichinella spiralis infection. Front Immunol 2023; 14:1185094. [PMID: 37283738 PMCID: PMC10240059 DOI: 10.3389/fimmu.2023.1185094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023] Open
Abstract
IgE antibodies are likely involved in host protection. Trichinella spiralis is a helminth that induces protection through IgE antibodies. The present study examined T. spiralis susceptibility in high and low IgE responder mice, with a specific focus on the inheritance of IgE responsiveness, which controls IgE production specific for the IgE isotype and non-specific for antigens. Furthermore, low IgE responsiveness is inherited as a recessive trait under a single gene, which is not linked to the H-2 gene. This study determined the total IgE and anti-T. spiralis IgE antibody levels after T. spiralis infection in low IgE responder SJL/J mice were several times lower than those in high IgE responders, such as the BALB/c mice. An IgE-dependent susceptibility to T. spiralis, evaluated in mice treated with anti-IgE antibodies and in control mice, was observed in high IgE responder mice but not in low IgE responder mice. The inheritance of IgE responsiveness and susceptibility to T. spiralis was investigated using crosses of SJL/J with high IgE responders. All of the (BALB/c × SJL/J) F1 and half of the (BALB/c × SJL/J) F1 × SJL backcross progenies were high IgE responders after T. spiralis infection. Total IgE and antigen-specific IgE antibody levels were correlated and not linked to H-2. It should be noted that high IgE responders always exhibited low susceptibility, suggesting that the trait of IgE responsiveness functions as a trait of susceptibility to T. spiralis.
Collapse
|
3
|
Genetic predisposition of alopecia areata in jordanians: A case-control study. Heliyon 2022; 8:e09184. [PMID: 35392398 PMCID: PMC8980757 DOI: 10.1016/j.heliyon.2022.e09184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/30/2021] [Accepted: 03/17/2022] [Indexed: 11/29/2022] Open
Abstract
Alopecia areata (AA) is a common non-scarring hair loss disease of defined patterns with varied patches size and body sites. The etiology of AA has a complex basis of autoimmunity, environment, and genetic variations. The latter factor is found to play a crucial role in AA risk. Thus, this study aimed to investigate the potential impact of specific immune-related gene polymorphisms among a cohort of Jordanian patients, which was previously reported in other populations. Blood samples of AA patients and control subjects were collected for genomic DNA (gDNA) extraction. Targeted single nucleotide polymorphisms (SNPs) of MASP2, TLR1, CTLA4, and C11orf30 were genotyped in duplicate using the Sequenom MassARRAY® system (iPLEX GOLD). Genotype and allele analysis reveals statistical differences in TLR1 rs4833095 (allele C, P = 0.044), MASP2 rs2273346 (genotype AA, P = 0.0026), and C11orf30 rs2155219 (genotype GG, P = 0.0069) distribution. These findings present the significant contribution of genetic variations in AA susceptibility in the Jordanian population, which is infrequently studied.
Collapse
|
4
|
Racca F, Pellegatta G, Cataldo G, Vespa E, Carlani E, Pelaia C, Paoletti G, Messina MR, Nappi E, Canonica GW, Repici A, Heffler E. Type 2 Inflammation in Eosinophilic Esophagitis: From Pathophysiology to Therapeutic Targets. Front Physiol 2022; 12:815842. [PMID: 35095572 PMCID: PMC8790151 DOI: 10.3389/fphys.2021.815842] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated disease of the esophagus characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation, whose incidence is rising. It significantly affects patients’ quality of life and, if left untreated, results in fibrotic complications. Although broad consensus has been achieved on first-line therapy, a subset of patients remains non-responder to standard therapy. The pathogenesis of EoE is multifactorial and results from the complex, still mostly undefined, interaction between genetics and intrinsic factors, environment, and antigenic stimuli. A deep understanding of the pathophysiology of this disease is pivotal for the development of new therapies. This review provides a comprehensive description of the pathophysiology of EoE, starting from major pathogenic mechanisms (genetics, type 2 inflammation, epithelial barrier dysfunction, gastroesophageal reflux, allergens, infections and microbiota) and subsequently focusing on the single protagonists of type 2 inflammation (involved cells, cytokines, soluble effectors, surface proteins and transcription factors) that could represent present and future therapeutic targets, while summarizing previous therapeutic approaches in literature.
Collapse
Affiliation(s)
- Francesca Racca
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- *Correspondence: Francesca Racca,
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giuseppe Cataldo
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Edoardo Vespa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Carlani
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Maria Rita Messina
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
5
|
Choi BY, Han M, Kwak JW, Kim TH. Genetics and Epigenetics in Allergic Rhinitis. Genes (Basel) 2021; 12:2004. [PMID: 34946955 PMCID: PMC8700872 DOI: 10.3390/genes12122004] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of allergic rhinitis is associated with genetic, environmental, and epigenetic factors. Genotyping of single nucleotide polymorphisms (SNPs) is an advanced technique in the field of molecular genetics that is closely correlated with genome-wide association studies (GWASs) in large population groups with allergic diseases. Many recent studies have paid attention to the role of epigenetics, including alteration of DNA methylation, histone acetylation, and miRNA levels in the pathogenesis of allergic rhinitis. In this review article, genetics and epigenetics of allergic rhinitis, including information regarding functions and significance of previously known and newly-discovered genes, are summarized. Directions for future genetic and epigenetic studies of allergic rhinitis are also proposed.
Collapse
Affiliation(s)
| | | | | | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea; (B.Y.C.); (M.H.); (J.W.K.)
| |
Collapse
|
6
|
Laulajainen‐Hongisto A, Lyly A, Hanif T, Dhaygude K, Kankainen M, Renkonen R, Donner K, Mattila P, Jartti T, Bousquet J, Kauppi P, Toppila‐Salmi S. Genomics of asthma, allergy and chronic rhinosinusitis: novel concepts and relevance in airway mucosa. Clin Transl Allergy 2020; 10:45. [PMID: 33133517 PMCID: PMC7592594 DOI: 10.1186/s13601-020-00347-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/25/2020] [Indexed: 12/14/2022] Open
Abstract
Genome wide association studies (GWASs) have revealed several airway disease-associated risk loci. Their role in the onset of asthma, allergic rhinitis (AR) or chronic rhinosinusitis (CRS), however, is not yet fully understood. The aim of this review is to evaluate the airway relevance of loci and genes identified in GWAS studies. GWASs were searched from databases, and a list of loci associating significantly (p < 10-8) with asthma, AR and CRS was created. This yielded a total of 267 significantly asthma/AR-associated loci from 31 GWASs. No significant CRS -associated loci were found in this search. A total of 170 protein coding genes were connected to these loci. Of these, 76/170 (44%) showed bronchial epithelial protein expression in stained microscopic figures of Human Protein Atlas (HPA), and 61/170 (36%) had a literature report of having airway epithelial function. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation analyses were performed, and 19 functional protein categories were found as significantly (p < 0.05) enriched among these genes. These were related to cytokine production, cell activation and adaptive immune response, and all were strongly connected in network analysis. We also identified 15 protein pathways that were significantly (p < 0.05) enriched in these genes, related to T-helper cell differentiation, virus infection, JAK-STAT signaling pathway, and asthma. A third of GWAS-level risk loci genes of asthma or AR seemed to have airway epithelial functions according to our database and literature searches. In addition, many of the risk loci genes were immunity related. Some risk loci genes also related to metabolism, neuro-musculoskeletal or other functions. Functions overlapped and formed a strong network in our pathway analyses and are worth future studies of biomarker and therapeutics.
Collapse
Affiliation(s)
- Anu Laulajainen‐Hongisto
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Laboratory of Cellular and Molecular ImmunologyInstitute of Microbiology of the Czech Academy of SciencesPragueCzech Republic
| | - Annina Lyly
- Department of Otorhinolaryngology–Head and Neck SurgeryUniversity of Helsinki and Helsinki University HospitalP.O.Box 263Kasarmikatu 11‐1300029 HUSHelsinkiFinland
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | | | | | - Matti Kankainen
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
- Translational Immunology Research Program and Department of Clinical ChemistryUniversity of HelsinkiHelsinkiFinland
| | - Risto Renkonen
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- HUS Diagnostic CenterHelsinki University HospitalHelsinkiFinland
| | - Kati Donner
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Pirkko Mattila
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
- Hematology Research Unit HelsinkiDepartment of HematologyHelsinki University Hospital Comprehensive Cancer CenterHelsinkiFinland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent MedicineTurku University Hospital and University of TurkuTurkuFinland
| | - Jean Bousquet
- Université MontpellierMontpellierFrance
- MACVIA‐FranceMontpellierFrance
- Corporate Member of Freie Universität BerlinHumboldt‐Universität Zu BerlinBerlin Institute of HealthComprehensive Allergy CenterDepartment of Dermatology and AllergyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Paula Kauppi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Sanna Toppila‐Salmi
- Skin and Allergy HospitalUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Haartman InstituteUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
7
|
Chen J, Chen Q, Wu C, Jin Y. Genetic variants of the C11orf30-LRRC32 region are associated with childhood asthma in the Chinese population. Allergol Immunopathol (Madr) 2020; 48:390-394. [PMID: 31812328 DOI: 10.1016/j.aller.2019.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION AND OBJECTIVES To investigate whether genetic polymorphisms of C11orf30-LRRC32 region are associated with the development of childhood asthma in the Chinese population. METHODS A total of 732 asthma children and 824 age-matched healthy controls were included in the study. Blood samples were collected from the subjects for total IgE analysis, DNA extraction and RNA extraction. Three previously reported asthma-related SNPs were genotyped, including rs7936070 (G/T), rs7927894 (A/G), and rs6592657 (A/G). Blood samples from 50 patients and 50 controls were randomly selected to detect the mRNA expression levels of C11orf30 and LRRC32 in serum. RESULTS There were significantly different genotype frequencies between the two groups in terms of rs7936070 and rs7927894. Compared with controls, patients were found to have remarkably higher risk allele frequencies of rs7936070 and rs7927894. Genotype GG of rs7936070 was indicative of remarkably elevated total IgE level as compared with genotype TT and genotype GT. Similarly, genotype AA of rs7927894 was also associated with significantly elevated total IgE level. The serum expression of C11orf30 was significantly lower in the patients than in the controls. The C11orf30 expression was significantly correlated with the total IgE level (r = -0.463, p = 0.01). CONCLUSIONS Variants of C11orf30 were associated with the risk of childhood asthma in the Chinese population. Besides, abnormally decreased expression of C11orf30 was detected in the serum of patients, which was correlated with the total IgE level. The C11orf30 might play a role in asthma via biological pathways involving the regulation of total serum IgE level.
Collapse
Affiliation(s)
- J Chen
- Department of Paediatrics, Jiangsu Taizhou People's Hospital, Hailing South Road No. 399, Taizhou, China
| | - Q Chen
- Department of Paediatrics, Jiangsu Taizhou People's Hospital, Hailing South Road No. 399, Taizhou, China
| | - C Wu
- Department of Paediatrics, Jiangsu Taizhou People's Hospital, Hailing South Road No. 399, Taizhou, China
| | - Y Jin
- Department of Paediatrics, Jiangsu Taizhou People's Hospital, Hailing South Road No. 399, Taizhou, China.
| |
Collapse
|
8
|
Mari A, Tsoukali E, Yaccob A. Eosinophilic Esophagitis in Adults: A Concise Overview of an Evolving Disease. Korean J Fam Med 2020; 41:75-83. [PMID: 32062959 PMCID: PMC7093678 DOI: 10.4082/kjfm.18.0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 10/15/2018] [Indexed: 12/19/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disease that encompasses esophageal symptoms along with eosinophilic infiltration of the esophageal epithelium. EoE is an evolving disease that has been a subject of interest to many researchers since the first studies recognized this condition as a new and distinct clinicopathological entity 25 years ago. Clinical presentation in adult patients may include dysphagia, food impaction, vomiting, and reflux symptoms. The diagnosis of EoE is based on the combination of clinical history suggestive of esophageal dysfunction, endoscopic features indicative of the disease, and histology revealing eosinophilic infiltration of the esophageal epithelium that persists after a trial of proton pump inhibitor therapy along with the exclusion of other disorders that may be associated with esophageal tissue eosinophilia. The interplay between EoE and gastroesophageal reflux disease (GERD) is complex, and differentiating these two conditions continues to be difficult and challenging in clinical practice. The mainstay treatment includes dietary modification, topical steroids, and/or endoscopic dilation. The primary care physician (PCP) plays an important role in improving patient care and quality of life by ensuring early referral and participating in management and follow-up. This article provides an overview of the current knowledge base regarding the disease including epidemiology, genetics, pathogenesis, common clinical presentations, the interplay between EoE and GERD, diagnostic approaches, and therapeutic options available to the PCP.
Collapse
Affiliation(s)
- Amir Mari
- Gastroenterology Institute, Nazareth EMMS Hospital, Nazareth, Israel.,The Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
| | - Emmanouela Tsoukali
- Gastroenterology and Hepatology Department, Evangelismos General Hospital of Athens, Athens, Greece
| | - Afif Yaccob
- Gastroenterology and Liver Disease Department, Rambam Healthcare Campus, Haifa, Israel
| |
Collapse
|
9
|
Elias MS, Wright SC, Remenyi J, Abbott JC, Bray SE, Cole C, Edwards S, Gierlinski M, Glok M, McGrath JA, Nicholson WV, Paternoster L, Prescott AR, Have ST, Whitfield PD, Lamond AI, Brown SJ. EMSY expression affects multiple components of the skin barrier with relevance to atopic dermatitis. J Allergy Clin Immunol 2019; 144:470-481. [PMID: 31158401 PMCID: PMC6683598 DOI: 10.1016/j.jaci.2019.05.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022]
Abstract
Background Atopic dermatitis (AD) is a common, complex, and highly heritable inflammatory skin disease. Genome-wide association studies offer opportunities to identify molecular targets for drug development. A risk locus on chromosome 11q13.5 lies between 2 candidate genes, EMSY and LRRC32 (leucine-rich repeat-containing 32) but the functional mechanisms affecting risk of AD remain unclear. Objectives We sought to apply a combination of genomic and molecular analytic techniques to investigate which genes are responsible for genetic risk at this locus and to define mechanisms contributing to atopic skin disease. Methods We used interrogation of available genomic and chromosome conformation data in keratinocytes, small interfering RNA (siRNA)–mediated knockdown in skin organotypic culture and functional assessment of barrier parameters, mass spectrometric global proteomic analysis and quantitative lipid analysis, electron microscopy of organotypic skin, and immunohistochemistry of human skin samples. Results Genomic data indicate active promoters in the genome-wide association study locus and upstream of EMSY; EMSY, LRRC32, and intergenic variants all appear to be within a single topologically associating domain. siRNA-knockdown of EMSY in organotypic culture leads to enhanced development of barrier function, reflecting increased expression of structural and functional proteins, including filaggrin and filaggrin-2, as well as long-chain ceramides. Conversely, overexpression of EMSY in keratinocytes leads to a reduction in markers of barrier formation. Skin biopsy samples from patients with AD show greater EMSY staining in the nucleus, which is consistent with an increased functional effect of this transcriptional control protein. Conclusion Our findings demonstrate an important role for EMSY in transcriptional regulation and skin barrier formation, supporting EMSY inhibition as a therapeutic approach.
Collapse
Affiliation(s)
- Martina S Elias
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom.
| | - Sheila C Wright
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Judit Remenyi
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - James C Abbott
- Data Analysis/Bioinformatics Group, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Susan E Bray
- NHS Research Scotland Biorepository Tayside, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Christian Cole
- Data Analysis/Bioinformatics Group, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sharon Edwards
- Department of Pathology, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Marek Gierlinski
- Data Analysis/Bioinformatics Group, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Mateusz Glok
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, King's College London (Guy's Campus), London, United Kingdom
| | - William V Nicholson
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sara Ten Have
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Phillip D Whitfield
- Lipidomics Research Facility, Division of Biomedical Sciences, University of the Highlands and Islands, Inverness, United Kingdom
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Sara J Brown
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom; Department of Dermatology, Ninewells Hospital, Dundee, United Kingdom.
| |
Collapse
|
10
|
Ruffner MA, Kennedy K, Cianferoni A. Pathophysiology of eosinophilic esophagitis: recent advances and their clinical implications. Expert Rev Clin Immunol 2019; 15:83-95. [PMID: 30394139 PMCID: PMC9044497 DOI: 10.1080/1744666x.2019.1544893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023]
Abstract
Introduction: Diagnostic and therapeutic strategies in eosinophilic esophagitis (EoE) are evolving. New knowledge regarding the pathophysiology of EoE has been the foundation for updated diagnostic recommendations and new therapeutic trials. Areas covered: We performed structured literature searches in Medline and PubMed, Cochrane meta-analyses, and abstracts of international congresses to review therapeutic approaches for EoE in July 2018. Additional articles were obtained by perusing the references of articles identified in the original PubMed search. Articles were excluded if they did not focus on the mechanism of disease, diagnosis, or treatment of humans with EoE. Expert commentary: Recent advances in the understanding of mechanisms underlying the pathology of EoE have resulted in significant change in the diagnostic algorithm for EoE, and are identifying promising potential targets for personalized medicine. There is a clinical need for improved targeted therapy for EoE, and better understanding the underlying pathophysiology of EoE will help to determine therapeutic targets. In this review, we highlight key mechanisms in the pathophysiology of EoE and how they are being utilized to change therapy in EoE.
Collapse
Affiliation(s)
- Melanie A Ruffner
- Division of Allergy & Immunology, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perlman School of Medicine, USA
| | - Katie Kennedy
- Division of Allergy & Immunology, Children’s Hospital of Philadelphia, PA, USA
| | - Antonella Cianferoni
- Division of Allergy & Immunology, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perlman School of Medicine, USA
| |
Collapse
|
11
|
Fahey LM, Guzek R, Ruffner MA, Sullivan KE, Spergel J, Cianferoni A. EMSY is increased and activates TSLP & CCL5 expression in eosinophilic esophagitis. Pediatr Allergy Immunol 2018; 29:565-568. [PMID: 29663593 DOI: 10.1111/pai.12907] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Lisa M Fahey
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Guzek
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Melanie A Ruffner
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathleen E Sullivan
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan Spergel
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Antonella Cianferoni
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
12
|
Zhu Z, Lee PH, Chaffin MD, Chung W, Loh PR, Lu Q, Christiani DC, Liang L. A genome-wide cross-trait analysis from UK Biobank highlights the shared genetic architecture of asthma and allergic diseases. Nat Genet 2018; 50:857-864. [PMID: 29785011 PMCID: PMC5980765 DOI: 10.1038/s41588-018-0121-0] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/27/2018] [Indexed: 01/10/2023]
Abstract
Clinical and epidemiological data suggest that asthma and allergic
diseases are associated and may share a common genetic etiology. We analyzed
genome-wide single-nucleotide polymorphism (SNP) data for asthma and allergic
diseases in 33,593 cases and 76,768 controls of European ancestry from the UK
Biobank. Two publicly available independent genome wide association studies
(GWAS) were used for replication. We have found a strong genome-wide genetic
correlation between asthma and allergic diseases (rg
= 0.75, P =
6.84×10−62). Cross trait analysis identified 38
genome-wide significant loci, including 7 novel shared loci. Computational
analysis showed that shared genetic loci are enriched in immune/inflammatory
systems and tissues with epithelium cells. Our work identifies common genetic
architectures shared between asthma and allergy and will help to advance our
understanding of the molecular mechanisms underlying co-morbid asthma and
allergic diseases.
Collapse
Affiliation(s)
- Zhaozhong Zhu
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Phil H Lee
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mark D Chaffin
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Wonil Chung
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Po-Ru Loh
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA. .,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.
| |
Collapse
|
13
|
Clayton F, Peterson K. Eosinophilic Esophagitis: Pathophysiology and Definition. Gastrointest Endosc Clin N Am 2018; 28:1-14. [PMID: 29129294 DOI: 10.1016/j.giec.2017.07.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Eosinophilic esophagitis is an adaptive immune response to patient-specific antigens, mostly foods. Eosinophilic esophagitis is not solely IgE-mediated and is likely characterized by Th2 lymphocytes with an impaired esophageal barrier function. The key cytokines and chemokines are thymic stromal lymphopoeitin, interleukin-13, CCL26/eotaxin-3, and transforming growth factor-β, all involved in eosinophil recruitment and remodeling. Chronic food dysphagia and food impactions, the feared late complications, are related in part to dense subepithelial fibrosis, likely induced by interleukin-13 and transforming growth factor-β.
Collapse
Affiliation(s)
- Frederic Clayton
- Department of Pathology, The University of Utah, Huntsman Cancer Hospital, 1950 Circle of Hope, Room N3100, Salt Lake City, UT 84112, USA
| | - Kathryn Peterson
- Division of Gastroenterology, The University of Utah, 30 North 1900 East SOM 4R118, Salt Lake City, UT 84132, USA.
| |
Collapse
|
14
|
Amat F, Louha M, Benet M, Guiddir T, Bourgoin-Heck M, Saint-Pierre P, Paluel-Marmont C, Fontaine C, Lambert N, Couderc R, Gonzalez JR, Just J. The IL-4 rs2070874 polymorphism may be associated with the severity of recurrent viral-induced wheeze. Pediatr Pulmonol 2017; 52:1435-1442. [PMID: 28950434 DOI: 10.1002/ppul.23834] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/10/2017] [Accepted: 09/01/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND Childhood recurrent wheezing and consequently asthma corresponds to various phenotypes. Our aim was to link genetic variants of asthma candidate genes to the phenotypes of early onset wheezing. STUDY DESIGN We included very young consecutive children presenting with recurrent wheezing who had been evaluated for the severity of wheezing, associated atopic comorbidities, and tested for biomarkers of atopy and inflammation. All were genotyped for 16 single nucleotide polymorphisms (SNPs) linked with asthma or atopy. An unsupervised hierarchical bottom-up method was used for clustering the phenotypes and a multinomial logistic regression was performed for each individual SNP. RESULTS We replicated the three phenotypes previously described Trousseau Asthma Program in 317 children aged 21.5 ± 7.9 months: cluster 1 (nonatopic uncontrolled severe wheeze), n = 207, a severe viral-induced wheeze, cluster 2 (atopic multiple trigger wheeze), n = 61, with multiple allergic comorbidities, and cluster 3 (episodic viral wheeze), n = 49, a mild viral-induced wheeze. The TT-genotype of the IL-4 rs2070874 polymorphism was significantly associated with the nonatopic uncontrolled severe wheeze compared to the episodic viral wheeze (OR 7.9; CI95% [2.5-25.3]; P = 0.001). CONCLUSION Association between the TT-genotype of IL-4 rs2070874 polymorphism and a severe phenotype of viral-induced wheeze further underlines the role IL-4 plays in the inflammation pathway leading to viral respiratory infections.
Collapse
Affiliation(s)
- Flore Amat
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France.,Equipe EPAR, Institut Pierre Louis d'Epidémiologie et de Santé Publique, UMR_S1136, INSERM, Faculté de Médecine Saint-Antoine, rue de Chaligny, Paris, France.,ISGlobal, Barcelona Institute for Global Health-Campus MAR, Barcelona Biomedical Research Park (PRBB) Doctor Aiguader, 88, 08003, Barcelona, Spain
| | - Malek Louha
- Laboratory of Biochemistry and Molecular Biology-Laboratoire de Biochimie et Biologie Moléculaire, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marta Benet
- ISGlobal, Barcelona Institute for Global Health-Campus MAR, Barcelona Biomedical Research Park (PRBB) Doctor Aiguader, 88, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Tamazoust Guiddir
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mélisande Bourgoin-Heck
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France
| | - Philippe Saint-Pierre
- Institut de Mathématiques de Toulouse, Université Toulouse III Paul Sabatier, Toulouse, France
| | - Colombe Paluel-Marmont
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France
| | - Cécile Fontaine
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathalie Lambert
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Rémy Couderc
- Laboratory of Biochemistry and Molecular Biology-Laboratoire de Biochimie et Biologie Moléculaire, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Juan-Ramon Gonzalez
- ISGlobal, Barcelona Institute for Global Health-Campus MAR, Barcelona Biomedical Research Park (PRBB) Doctor Aiguader, 88, 08003, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Jocelyne Just
- Department of Allergology-Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand Trousseau, Assistance Publique-Hôpitaux de Paris, Paris, France.,UPMC Univ Paris 06, Sorbonne Universités, Paris, France.,Equipe EPAR, Institut Pierre Louis d'Epidémiologie et de Santé Publique, UMR_S1136, INSERM, Faculté de Médecine Saint-Antoine, rue de Chaligny, Paris, France
| |
Collapse
|
15
|
Genome-wide association study and meta-analysis in multiple populations identifies new loci for peanut allergy and establishes C11orf30/EMSY as a genetic risk factor for food allergy. J Allergy Clin Immunol 2017; 141:991-1001. [PMID: 29030101 DOI: 10.1016/j.jaci.2017.09.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 09/05/2017] [Accepted: 09/19/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Peanut allergy (PA) is a complex disease with both environmental and genetic risk factors. Previously, PA loci were identified in filaggrin (FLG) and HLA in candidate gene studies, and loci in HLA were identified in a genome-wide association study and meta-analysis. OBJECTIVE We sought to investigate genetic susceptibility to PA. METHODS Eight hundred fifty cases and 926 hyper-control subjects and more than 7.8 million genotyped and imputed single nucleotide polymorphisms (SNPs) were analyzed in a genome-wide association study to identify susceptibility variants for PA in the Canadian population. A meta-analysis of 2 phenotypes (PA and food allergy) was conducted by using 7 studies from the Canadian, American (n = 2), Australian, German, and Dutch (n = 2) populations. RESULTS An SNP near integrin α6 (ITGA6) reached genome-wide significance with PA (P = 1.80 × 10-8), whereas SNPs associated with Src kinase-associated phosphoprotein 1 (SKAP1), matrix metallopeptidase 12 (MMP12)/MMP13, catenin α3 (CTNNA3), rho GTPase-activating protein 24 (ARHGAP24), angiopoietin 4 (ANGPT4), chromosome 11 open reading frame (C11orf30/EMSY), and exocyst complex component 4 (EXOC4) reached a threshold suggestive of association (P ≤ 1.49 × 10-6). In the meta-analysis of PA, loci in or near ITGA6, ANGPT4, MMP12/MMP13, C11orf30, and EXOC4 were significant (P ≤ 1.49 × 10-6). When a phenotype of any food allergy was used for meta-analysis, the C11orf30 locus reached genome-wide significance (P = 7.50 × 10-11), whereas SNPs associated with ITGA6, ANGPT4, MMP12/MMP13, and EXOC4 and additional C11orf30 SNPs were suggestive (P ≤ 1.49 × 10-6). Functional annotation indicated that SKAP1 regulates expression of CBX1, which colocalizes with the EMSY protein coded by C11orf30. CONCLUSION This study identifies multiple novel loci as risk factors for PA and food allergy and establishes C11orf30 as a risk locus for both PA and food allergy. Multiple genes (C11orf30/EMSY, SKAP1, and CTNNA3) identified by this study are involved in epigenetic regulation of gene expression.
Collapse
|
16
|
Bousquet J, Anto JM, Akdis M, Auffray C, Keil T, Momas I, Postma D, Valenta R, Wickman M, Cambon‐Thomsen A, Haahtela T, Lambrecht BN, Lodrup Carlsen KC, Koppelman GH, Sunyer J, Zuberbier T, Annesi‐Maesano I, Arno A, Bindslev‐Jensen C, De Carlo G, Forastiere F, Heinrich J, Kowalski ML, Maier D, Melén E, Palkonen S, Smit HA, Standl M, Wright J, Asarnoj A, Benet M, Ballardini N, Garcia‐Aymerich J, Gehring U, Guerra S, Hohman C, Kull I, Lupinek C, Pinart M, Skrindo I, Westman M, Smagghe D, Akdis C, Albang R, Anastasova V, Anderson N, Bachert C, Ballereau S, Ballester F, Basagana X, Bedbrook A, Bergstrom A, Berg A, Brunekreef B, Burte E, Carlsen KH, Chatzi L, Coquet JM, Curin M, Demoly P, Eller E, Fantini MP, Gerhard B, Hammad H, Hertzen L, Hovland V, Jacquemin B, Just J, Keller T, Kerkhof M, Kiss R, Kogevinas M, Koletzko S, Lau S, Lehmann I, Lemonnier N, McEachan R, Mäkelä M, Mestres J, Minina E, Mowinckel P, Nadif R, Nawijn M, Oddie S, Pellet J, Pin I, Porta D, Rancière F, Rial‐Sebbag A, Saeys Y, Schuijs MJ, Siroux V, Tischer CG, Torrent M, Varraso R, De Vocht J, Wenger K, Wieser S, Xu C. Paving the way of systems biology and precision medicine in allergic diseases: the MeDALL success story: Mechanisms of the Development of ALLergy; EU FP7-CP-IP; Project No: 261357; 2010-2015. Allergy 2016; 71:1513-1525. [PMID: 26970340 PMCID: PMC5248602 DOI: 10.1111/all.12880] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2016] [Indexed: 01/06/2023]
Abstract
MeDALL (Mechanisms of the Development of ALLergy; EU FP7-CP-IP; Project No: 261357; 2010-2015) has proposed an innovative approach to develop early indicators for the prediction, diagnosis, prevention and targets for therapy. MeDALL has linked epidemiological, clinical and basic research using a stepwise, large-scale and integrative approach: MeDALL data of precisely phenotyped children followed in 14 birth cohorts spread across Europe were combined with systems biology (omics, IgE measurement using microarrays) and environmental data. Multimorbidity in the same child is more common than expected by chance alone, suggesting that these diseases share causal mechanisms irrespective of IgE sensitization. IgE sensitization should be considered differently in monosensitized and polysensitized individuals. Allergic multimorbidities and IgE polysensitization are often associated with the persistence or severity of allergic diseases. Environmental exposures are relevant for the development of allergy-related diseases. To complement the population-based studies in children, MeDALL included mechanistic experimental animal studies and in vitro studies in humans. The integration of multimorbidities and polysensitization has resulted in a new classification framework of allergic diseases that could help to improve the understanding of genetic and epigenetic mechanisms of allergy as well as to better manage allergic diseases. Ethics and gender were considered. MeDALL has deployed translational activities within the EU agenda.
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 France
- INSERM VIMA: Ageing and Chronic Diseases, Epidemiological and Public Health Approaches UVSQ Université Versailles St‐Quentin‐en‐Yvelines Paris France
| | - J. M. Anto
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
- IMIM (Hospital del Mar Research Institute) Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP) Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - C. Auffray
- European Institute for Systems Biology and Medicine CNRS‐ENS‐UCBL Université de Lyon Lyon France
| | - 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
| | - I. Momas
- Department of Public Health and Health Products Paris Descartes University‐Sorbonne Paris Cité Paris France
- Paris Municipal Department of Social Action, Childhood, and Health Paris France
| | - D.S. Postma
- Department of Pulmonary Medicine and Tuberculosis GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen the Netherlands
| | - R. Valenta
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - M. Wickman
- Sachs’ Children and Youth Hospital, Södersjukhuset Stockholm and Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | - A. Cambon‐Thomsen
- UMR Inserm U1027 and Université de Toulouse III Paul Sabatier Toulouse France
| | - T. Haahtela
- Skin and Allergy Hospital Helsinki University Hospital Helsinki Finland
| | - B. N. Lambrecht
- VIB Inflammation Research Center Ghent University Ghent Belgium
| | - K. C. Lodrup Carlsen
- Department of Paediatrics Faculty of Medicine Institute of Clinical Medicine Oslo University Hospital University of Oslo Oslo Norway
| | - G. H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology Beatrix Children's Hospital GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen the Netherlands
| | - J. Sunyer
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
- IMIM (Hospital del Mar Research Institute) Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP) Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
| | - T. Zuberbier
- Secretary General of the Global Allergy and Asthma European Network (GALEN) Allergy‐Centre‐Charité at the Department of Dermatology Charité–Universitätsmedizin Berlin Berlin Germany
| | | | - A. Arno
- Onmedic Networks Barcelona Spain
| | - C. Bindslev‐Jensen
- Department of Dermatology and Allergy Centre Odense University Hospital Odense Denmark
| | - G. De Carlo
- EFA European Federation of Allergy and Airways Diseases Patients’ Associations Brussels Belgium
| | - F. Forastiere
- Department of Epidemiology Regional Health Service Lazio Region Rome Italy
| | - J. Heinrich
- Institute of Epidemiology I German Research Centre for Environmental Health Helmholtz Zentrum München Neuherberg Germany
| | - M. L. Kowalski
- Department of Immunology, Rheumatology and Allergy Medical University of Lodz Lodz Poland
| | - D. Maier
- Biomax Informatics AG Munich Germany
| | - E. Melén
- Department of Pulmonary Medicine and Tuberculosis GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen the Netherlands
- Stockholm County Council Centre for Occupational and Environmental Medicine Stockholm Sweden
| | - S. Palkonen
- EFA European Federation of Allergy and Airways Diseases Patients’ Associations Brussels Belgium
| | - H. A. Smit
- Julius Center of Health Sciences and Primary Care University Medical Center Utrecht University of Utrecht Utrecht the Netherlands
| | - M. Standl
- Institute of Epidemiology I German Research Centre for Environmental Health Helmholtz Zentrum München Neuherberg Germany
| | - J. Wright
- Bradford Institute for Health Research Bradford Royal Infirmary Bradford UK
| | - A. Asarnoj
- Clinical Immunology and Allergy Unit Department of Medicine Solna Karolinska Institutet Stockholm Sweden
- Astrid Lindgren Children's Hospital Department of Pediatric Pulmonology and Allergy Karolinska University Hospital Stockholm Sweden
| | - M. Benet
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
| | - N. Ballardini
- Sachs’ Children and Youth Hospital, Södersjukhuset Stockholm and Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- St John's Institute of Dermatology King's College London London UK
| | - J. Garcia‐Aymerich
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
- IMIM (Hospital del Mar Research Institute) Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP) Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
| | - U. Gehring
- Institute for Risk Assessment Sciences Utrecht University Utrecht the Netherlands
| | - S. Guerra
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
| | - C. Hohman
- Institute of Social Medicine, Epidemiology and Health Economics Charité–Universitätsmedizin Berlin Germany
| | - I. Kull
- Sachs’ Children and Youth Hospital, Södersjukhuset Stockholm and Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- Department of Clinical Science and Education, Södersjukhuset Karolinska InstitutetStockholm Sweden
| | - C. Lupinek
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - M. Pinart
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
| | - I. Skrindo
- Department of Paediatrics Faculty of Medicine Institute of Clinical Medicine Oslo University Hospital University of Oslo Oslo Norway
| | - M. Westman
- Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
- Department of ENT Diseases Karolinska University Hospital Stockholm Sweden
| | | | - C. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - R. Albang
- Biomax Informatics AG Munich Germany
| | - V. Anastasova
- UMR Inserm U1027 and Université de Toulouse III Paul Sabatier Toulouse France
| | - N. Anderson
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | - C. Bachert
- ENT Department Ghent University Hospital Gent Belgium
| | - S. Ballereau
- European Institute for Systems Biology and Medicine CNRS‐ENS‐UCBL Université de Lyon Lyon France
| | - F. Ballester
- Environment and Health Area Centre for Public Health Research (CSISP) CIBERESP Department of Nursing University of Valencia Valencia Spain
| | - X. Basagana
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
| | - 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 France
| | - A. Bergstrom
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | - A. Berg
- Research Institute Department of Pediatrics Marien‐Hospital Wesel Germany
| | - 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 UVSQ Université Versailles St‐Quentin‐en‐Yvelines Paris France
| | - K. H. Carlsen
- Department of Paediatrics Oslo University Hospital University of Oslo Oslo Norway
| | - L. Chatzi
- Department of Social Medicine Faculty of Medicine University of Crete Heraklion Crete Greece
| | - J. M. Coquet
- VIB Inflammation Research Center Ghent University Ghent Belgium
| | - M. Curin
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - P. Demoly
- Department of Respiratory Diseases Montpellier University Hospital 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
| | - L. Hertzen
- Skin and Allergy Hospital Helsinki University Hospital Helsinki Finland
| | - V. Hovland
- Department of Paediatrics Oslo University Hospital University of Oslo Oslo Norway
| | - B. Jacquemin
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
| | - J. Just
- Allergology Department Centre de l'Asthme et des Allergies Hôpital d'Enfants Armand‐Trousseau (APHP) Sorbonne Universités Institut Pierre Louis d'Epidémiologie et de Santé Publique Paris France
| | - T. Keller
- Institute of Social Medicine, Epidemiology and Health Economics Charité–Universitätsmedizin Berlin Germany
| | - M. Kerkhof
- Department of Pulmonary Medicine and Tuberculosis GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen the Netherlands
| | - R. Kiss
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - M. Kogevinas
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
- IMIM (Hospital del Mar Research Institute) Barcelona Spain
- CIBER Epidemiología y Salud Pública (CIBERESP) Barcelona Spain
- Universitat Pompeu Fabra (UPF) Barcelona Spain
| | - S. Koletzko
- Division of Paediatric Gastroenterology and Hepatology Ludwig Maximilians University of Munich Munich Germany
| | - S. Lau
- Department for Pediatric Pneumology and Immunology Charité Medical University Berlin Germany
| | - I. Lehmann
- Department of Environmental Immunology/Core Facility Studies Helmholtz Centre for Environmental Research, UFZ Leipzig Germany
| | - N. Lemonnier
- European Institute for Systems Biology and Medicine CNRS‐ENS‐UCBL Université de Lyon Lyon France
| | - R. McEachan
- Bradford Institute for Health Research Bradford Royal Infirmary Bradford UK
| | - M. Mäkelä
- Skin and Allergy Hospital Helsinki University Hospital Helsinki Finland
| | - J. Mestres
- Chemotargets SL and Chemogenomics Laboratory GRIB Unit IMIM‐Hospital del Mar and University Pompeu Fabra Barcelona Catalonia Spain
| | - E. Minina
- Biomax Informatics AG Munich Germany
| | - P. Mowinckel
- Department of Paediatrics Oslo University Hospital University of Oslo Oslo Norway
| | - R. Nadif
- INSERM VIMA: Ageing and Chronic Diseases, Epidemiological and Public Health Approaches UVSQ Université Versailles St‐Quentin‐en‐Yvelines Paris France
| | - M. Nawijn
- Department of Pediatric Pulmonology and Pediatric Allergology Beatrix Children's Hospital GRIAC Research Institute University Medical Center Groningen 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 CNRS‐ENS‐UCBL Université de Lyon 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
| | - F. Rancière
- Department of Public Health and Health Products Paris Descartes University‐Sorbonne Paris Cité Paris France
| | - A. Rial‐Sebbag
- UMR Inserm U1027 and Université de Toulouse III Paul Sabatier Toulouse France
| | - Y. Saeys
- VIB Inflammation Research Center Ghent University Ghent Belgium
| | - M. J. Schuijs
- VIB Inflammation Research Center Ghent University Ghent Belgium
| | | | - C. G. Tischer
- Institute of Epidemiology I German Research Centre for Environmental Health Helmholtz Zentrum München Neuherberg Germany
| | - M. Torrent
- Centre for Research in Environmental Epidemiology (CREAL) ISGLoBAL Barcelona Spain
- ib‐salut Area de Salut de Menorca Spain
| | - R. Varraso
- INSERM VIMA: Ageing and Chronic Diseases, Epidemiological and Public Health Approaches UVSQ Université Versailles St‐Quentin‐en‐Yvelines Paris France
| | - J. De Vocht
- EFA European Federation of Allergy and Airways Diseases Patients’ Associations Brussels Belgium
| | - K. Wenger
- Biomax Informatics AG Munich Germany
| | - S. Wieser
- Division of Immunopathology Department of Pathophysiology and Allergy Research Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - C. Xu
- Department of Pulmonary Medicine and Tuberculosis GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen the Netherlands
| |
Collapse
|
17
|
Stemmler S, Hoffjan S. Trying to understand the genetics of atopic dermatitis. Mol Cell Probes 2016; 30:374-385. [PMID: 27725295 DOI: 10.1016/j.mcp.2016.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is a common and complex skin disease associated with both genetic and environmental factors. Loss-of-function mutations in the filaggrin gene, encoding a structural protein with an important role in epidermal barrier function, constitutes a well recognised susceptibility locus for AD. Further, genome-wide association studies (GWAS), including large meta-analyses, have discovered 38 additional susceptibility loci with genome-wide significance. However, the reported variations only explain a fraction of the overall heritability of AD. Here, we summarize the current knowledge of the role of filaggrin and the epidermal differentiation complex as well as the results of GWAS, with an emphasis on novel findings and observations made in the past two years. Additionally, we present first results of exome sequencing for AD and discuss novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Sabine Hoffjan
- Department of Human Genetics, Ruhr-University, Bochum, Germany
| |
Collapse
|
18
|
Ng CL, Wang DY. Latest developments in allergic rhinitis in Allergy for clinicians and researchers. Allergy 2015; 70:1521-30. [PMID: 26443244 DOI: 10.1111/all.12782] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2015] [Indexed: 02/06/2023]
Abstract
Research efforts in allergic rhinitis have always been intense. Over the past 3 years, numerous breakthroughs in basic science and clinical research have been made, augmenting our understanding of this condition that afflicts a significant proportion of the global population. New epidemiological findings, novel insights into the molecular and cellular mechanisms of allergy, enhancement of current developmental theories, new concepts of the goals and endpoints of management, and latest therapeutic modalities that includes the harnessing of information technology and big data are some areas where important advances were made. We attempt to bring you a summary of the key research advances made in the field of allergic rhinitis from 2013 to 2015.
Collapse
Affiliation(s)
- C. L. Ng
- Department of Otolaryngology; Yong Loo Lin School of Medicine; National University of Singapore; National University Health System; Singapore City Singapore
| | - D. Y. Wang
- Department of Otolaryngology; Yong Loo Lin School of Medicine; National University of Singapore; National University Health System; Singapore City Singapore
| |
Collapse
|
19
|
Sleiman PMA, March M, Hakonarson H. The genetic basis of eosinophilic esophagitis. Best Pract Res Clin Gastroenterol 2015; 29:701-707. [PMID: 26552769 DOI: 10.1016/j.bpg.2015.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/02/2015] [Indexed: 01/31/2023]
Abstract
Eosinophilic esophagitis is characterized by destructive responses of the immune system to environmental allergens, including food, on the human esophagus. EoE is now reported as a major cause of upper gastrointestinal morbidity in children and adults and the incidence is reported to be on the increase. It is known that EoE has a high degree of heritability, with a majority of the phenotypic variation believed to be genetic in origin as shown by genetic epidemiology studies of twins and families. Prior to 2010, there were no known genetic risk factors for the disease. Three GWAS have since been published identifying 5 loci which influence risk for EoE in both children and adults. The information gained from GWAS has been of value in elucidating the pathways involved, such as IL4/STAT6, and more unexpected pathways such as epithelial apical transport and wound healing. We will review the results of the EoE GWAS and the known associated genes, concluding with a discussion of some future directions for genetic studies in EoE.
Collapse
Affiliation(s)
- Patrick M A Sleiman
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, PA, USA; Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael March
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, PA, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, The Children's Hospital of Philadelphia, PA, USA; Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
20
|
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: 8.6] [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
|