1
|
Soria-Chacartegui P, Navares-Gómez M, Molina-Jiménez F, Laserna-Mendieta EJ, Arias-González L, Majano P, Casabona S, Lucendo AJ, Abad-Santos F, Santander C, Zubiaur P. Impact of STAT6 Variants on the Response to Proton Pump Inhibitors and Comorbidities in Patients with Eosinophilic Esophagitis. Int J Mol Sci 2024; 25:3685. [PMID: 38612496 PMCID: PMC11011338 DOI: 10.3390/ijms25073685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Proton pump inhibitors (PPIs) are the first-line drug for eosinophilic esophagitis (EoE), although it is estimated that there is a lack of histological remission in 50% of patients. This research aimed to identify pharmacogenetic biomarkers predictive of PPI effectiveness and to study their association with disease features. Peak eosinophil count (PEC) and the endoscopic reference score (EREFS) were determined before and after an eight-week PPI course in 28 EoE patients. The impact of the signal transducer and activator of transcription 6 (STAT6), CYP2C19, CYP3A4, CYP3A5, and ABCB1 genetic variations on baseline PEC and EREFS, their reduction and histological response, and on EoE symptoms and comorbidities was analyzed. PEC reduction was higher in omeprazole-treated patients (92.5%) compared to other PPIs (57.9%, p = 0.003). STAT6 rs12368672 (g.18453G>C) G/G genotype showed higher baseline PEC values compared to G/C and C/C genotypes (83.2 vs. 52.9, p = 0.027). EREFS reduction in STAT6 rs12368672 G/G and G/C genotypes was higher than in the C/C genotype (36.7% vs. -75.0% p = 0.011). However, significance was lost after Bonferroni correction. Heartburn incidence was higher in STAT6 rs167769 (g.27148G>A) G/G patients compared to G/A (54.55% vs. 11.77%, p = 0.030). STAT6 rs12368672G>C and rs167769G>A variants might have a relevant impact on EoE status and PPI response. Further research is warranted to clarify the clinical relevance of these variants.
Collapse
Affiliation(s)
- Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| | - Marcos Navares-Gómez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| | - Francisca Molina-Jiménez
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
| | - Emilio J. Laserna-Mendieta
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Laura Arias-González
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Pedro Majano
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cellular Biology Department, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sergio Casabona
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfredo J. Lucendo
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cecilio Santander
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| |
Collapse
|
2
|
Xu Z, Forno E, Sun Y, Manni ML, Han YY, Kim S, Yue M, Vonk JM, Kersten ETM, Acosta-Perez E, Canino G, Koppelman GH, Chen W, Celedón JC. Nasal epithelial gene expression and total IgE in children and adolescents with asthma. J Allergy Clin Immunol 2024; 153:122-131. [PMID: 37742934 PMCID: PMC10842443 DOI: 10.1016/j.jaci.2023.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Little is known about nasal epithelial gene expression and total IgE in youth. OBJECTIVE We aimed to identify genes whose nasal epithelial expression differs by total IgE in youth, and group them into modules that could be mapped to airway epithelial cell types. METHODS We conducted a transcriptome-wide association study of total IgE in 469 Puerto Ricans aged 9 to 20 years who participated in the Epigenetic Variation and Childhood Asthma in Puerto Ricans study, separately in all subjects and in those with asthma. We then attempted to replicate top findings for each analysis using data from 3 cohorts. Genes with a Benjamini-Hochberg-adjusted P value of less than .05 in the Epigenetic Variation and Childhood Asthma in Puerto Ricans study and a P value of less than .05 in the same direction of association in 1 or more replication cohort were considered differentially expressed genes (DEGs). DEGs for total IgE in subjects with asthma were further dissected into gene modules using coexpression analysis, and such modules were mapped to specific cell types in airway epithelia using public single-cell RNA-sequencing data. RESULTS A higher number of DEGs for total IgE were identified in subjects with asthma (n = 1179 DEGs) than in all subjects (n = 631 DEGs). In subjects with asthma, DEGs were mapped to 11 gene modules. The top module for positive correlation with total IgE was mapped to myoepithelial and mucus secretory cells in lower airway epithelia and was regulated by IL-4, IL5, IL-13, and IL-33. Within this module, hub genes included CDH26, FETUB, NTRK2, CCBL1, CST1, and CST2. Furthermore, an enrichment analysis showed overrepresentation of genes in signaling pathways for synaptogenesis, IL-13, and ferroptosis, supporting interactions between interleukin- and acetylcholine-induced responses. CONCLUSIONS Our findings for nasal epithelial gene expression support neuroimmune coregulation of total IgE in youth with asthma.
Collapse
Affiliation(s)
- Zhongli Xu
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa; School of Medicine, Tsinghua University, Beijing, China
| | - Erick Forno
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Yidan Sun
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| | - Michelle L Manni
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Yueh Ying Han
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Soyeon Kim
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Molin Yue
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Judith M Vonk
- GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands; Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Elin T M Kersten
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| | - Edna Acosta-Perez
- Behavioral Sciences Research Institute of Puerto Rico, University of Puerto Rico, San Juan, Puerto Rico
| | - Glorisa Canino
- Behavioral Sciences Research Institute of Puerto Rico, University of Puerto Rico, San Juan, Puerto Rico; Department of Pediatrics, Medical Science Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, Groningen, The Netherlands
| | - Wei Chen
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | - Juan C Celedón
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pa; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pa.
| |
Collapse
|
3
|
Zhang Y. From gene identifications to therapeutic targets for asthma: Focus on great potentials of TSLP, ORMDL3, and GSDMB. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:139-147. [PMID: 39171126 PMCID: PMC11332877 DOI: 10.1016/j.pccm.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Indexed: 08/23/2024]
Abstract
Asthma is a chronic respiratory disease, and clinically, asthma exacerbations remain difficult to treat. The disease is caused by combinations of and interactions between genetic and environmental factors. Genomic and genetic approaches identified many novel genes to treat asthma and brought new insights into the disease. The products of the genes have functional roles in regulating physiological or pathophysiological processes in airway structural cells and immune system cells. Genetic factors also interact with environmental factors such as air pollutants, and bacterial and viral infections to trigger the disease. Thymic stromal lymphopoietin (TSLP), orosomucoid-like 3 (ORMDL3), and gasdermin B (GSDMB) are three genes identified by genetic studies to have a great potential as therapeutic targets of asthma. TSLP is an important driver of type 2 inflammation. ORMDL3 mediates cell stress, sphingolipid synthesis, and viral and bacterial infections. GSDMB regulates cell pyroptosis through its N and C terminals and can bind sulfatides to influence inflammatory response. Investigating inhibitors or modulators for these pathways would bring a new landscape for therapeutics of asthma in future.
Collapse
Affiliation(s)
- Youming Zhang
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
4
|
Pushpamithran G, Skoglund C, Olsson F, Méndez-Aranda M, Schön T, Segelmark M, Stendahl O, Gilman RH, Blomgran R. No impact of helminth coinfection in patients with smear positive tuberculosis on immunoglobulin levels using a novel method measuring Mycobacterium tuberculosis-specific antibodies. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:55. [PMID: 37386541 DOI: 10.1186/s13223-023-00808-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/11/2023] [Indexed: 07/01/2023]
Abstract
Helminth/tuberculosis (TB)-coinfection can reduce cell-mediated immunity against Mycobacterium tuberculosis (Mtb) and increase disease severity, although the effects are highly helminth species dependent. Mtb have long been ranked as the number one single infectious agent claiming the most lives. The only licensed vaccine for TB (BCG) offers highly variable protection against TB, and almost no protection against transmission of Mtb. In recent few years the identification of naturally occurring antibodies in humans that are protective during Mtb infection has reignited the interest in adaptive humoral immunity against TB and its possible implementation in novel TB vaccine design. The effects of helminth/TB coinfection on the humoral response against Mtb during active pulmonary TB are however still unclear, and specifically the effect by globally prevalent helminth species such as Ascaris lumbricoides, Strongyloides stercoralis, Ancylostoma duodenale, Trichuris trichiura. Plasma samples from smear positive TB patients were used to measure both total and Mtb-specific antibody responses in a Peruvian endemic setting where these helminths are dominating. Mtb-specific antibodies were detected by a novel approach coating ELISA-plates with a Mtb cell-membrane fraction (CDC1551) that contains a broad range of Mtb surface proteins. Compared to controls without helminths or TB, helminth/TB coinfected patients had high levels of Mtb-specific IgG (including an IgG1 and IgG2 subclass response) and IgM, which were similarly increased in TB patients without helminth infection. These data, indicate that helminth/TB coinfected have a sustained humoral response against Mtb at the level of active TB only. More studies on the species-specific impact of helminths on the adaptive humoral response against Mtb using a larger sample size, and in relation to TB disease severity, are needed.
Collapse
Affiliation(s)
- Giggil Pushpamithran
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden
| | - Camilla Skoglund
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Fanny Olsson
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden
- Laboratorio de Investigación en Enfermedades Infecciosas, LID, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Melissa Méndez-Aranda
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden
- Laboratorio de Investigación en Enfermedades Infecciosas, LID, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Thomas Schön
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden
- Department of Infectious Diseases, County of Östergötland and Kalmar, Linköping University, Linköping, Sweden
| | - Mårten Segelmark
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Sciences, Lund University and Department of Nephrology, Skane University Hospital, Lund, Sweden
| | - Olle Stendahl
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden
| | - Robert H Gilman
- Laboratorio de Investigación en Enfermedades Infecciosas, LID, Universidad Peruana Cayetano Heredia, Lima, Peru
- Department of International Health, Johns Hopkins School of Public Health, Baltimore, Mayland, USA
| | - Robert Blomgran
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University Campus US, Building 420 Floor 12, 581 85, Linköping, SE, Sweden.
| |
Collapse
|
5
|
Recto KA, Huan T, Lee DH, Lee GY, Gereige J, Yao C, Hwang SJ, Joehanes R, Kelly RS, Lasky-Su J, O’Connor G, Levy D. Transcriptome-wide association study of circulating IgE levels identifies novel targets for asthma and allergic diseases. Front Immunol 2023; 14:1080071. [PMID: 36793728 PMCID: PMC9922991 DOI: 10.3389/fimmu.2023.1080071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Measurement of circulating immunoglobulin E (IgE) concentration is helpful for diagnosing and treating asthma and allergic diseases. Identifying gene expression signatures associated with IgE might elucidate novel pathways for IgE regulation. To this end, we performed a discovery transcriptome-wide association study to identify differentially expressed genes associated with circulating IgE levels in whole-blood derived RNA from 5,345 participants in the Framingham Heart Study across 17,873 mRNA gene-level transcripts. We identified 216 significant transcripts at a false discovery rate <0.05. We conducted replication using the meta-analysis of two independent external studies: the Childhood Asthma Management Program (n=610) and the Genetic Epidemiology of Asthma in Costa Rica Study (n=326); we then reversed the discovery and replication cohorts, which revealed 59 significant genes that replicated in both directions. Gene ontology analysis revealed that many of these genes were implicated in immune function pathways, including defense response, inflammatory response, and cytokine production. Mendelian randomization (MR) analysis revealed four genes (CLC, CCDC21, S100A13, and GCNT1) as putatively causal (p<0.05) regulators of IgE levels. GCNT1 (beta=1.5, p=0.01)-which is a top result in the MR analysis of expression in relation to asthma and allergic diseases-plays a role in regulating T helper type 1 cell homing, lymphocyte trafficking, and B cell differentiation. Our findings build upon prior knowledge of IgE regulation and provide a deeper understanding of underlying molecular mechanisms. The IgE-associated genes that we identified-particularly those implicated in MR analysis-can be explored as promising therapeutic targets for asthma and IgE-related diseases.
Collapse
Affiliation(s)
- Kathryn A. Recto
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Tianxiao Huan
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Dong Heon Lee
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Gha Young Lee
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Jessica Gereige
- Pulmonary Center, Boston University School of Medicine, Boston, MA, United States
| | - Chen Yao
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Shih-Jen Hwang
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Roby Joehanes
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| | - Rachel S. Kelly
- Brigham and Women’s Hospital, Channing Division of Network Medicine, Boston, MA, United States
| | - Jessica Lasky-Su
- Brigham and Women’s Hospital, Channing Division of Network Medicine, Boston, MA, United States
| | - George O’Connor
- Pulmonary Center, Boston University School of Medicine, Boston, MA, United States
| | - Daniel Levy
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
- The Framingham Heart Study, Framingham, MA, United States
| |
Collapse
|
6
|
Schoettler N, Dissanayake E, Craven MW, Yee JS, Eliason J, Schauberger EM, Lemanske RF, Ober C, Gern JE. New Insights Relating Gasdermin B to the Onset of Childhood Asthma. Am J Respir Cell Mol Biol 2022; 67:430-437. [PMID: 35580164 PMCID: PMC9564923 DOI: 10.1165/rcmb.2022-0043ps] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
Chromosome 17q12-q21 is the most replicated genetic locus for childhood-onset asthma. Polymorphisms in this locus containing ∼10 genes interact with a variety of environmental exposures in the home and outdoors to modify asthma risk. However, the functional basis for these associations and their linkages to the environment have remained enigmatic. Within this extended region, regulation of GSDMB (gasdermin B) expression in airway epithelial cells has emerged as the primary mechanism underlying the 17q12-q21 genome-wide association study signal. Asthma-associated SNPs influence the abundance of GSDMB transcripts as well as the functional properties of GSDMB protein in airway epithelial cells. GSDMB is a member of the gasdermin family of proteins, which regulate pyroptosis and inflammatory responses to microbial infections. The aims of this review are to synthesize recent studies on the relationship of 17q12-q21 SNPs to childhood asthma and the evidence pointing to GSDMB gene expression or protein function as the underlying mechanism and to explore the potential functions of GSDMB that may influence the risk of developing asthma during childhood.
Collapse
Affiliation(s)
| | | | - Mark W. Craven
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Jeremiah S. Yee
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Joshua Eliason
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | | | | | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Illinois; and
| | | |
Collapse
|
7
|
Kulkarni A, Kediya DA. A Multi-Point View of Genetic Factors Affecting Hereditary Transmissibility of Asthma. Cureus 2022; 14:e28768. [PMID: 36225476 PMCID: PMC9531716 DOI: 10.7759/cureus.28768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/04/2022] [Indexed: 11/05/2022] Open
|
8
|
Akenroye AT, Brunetti T, Romero K, Daya M, Kanchan K, Shankar G, Chavan S, Preethi Boorgula M, Ampleford EA, Fonseca HF, Hawkins GA, Pitangueira Teixeira HM, Campbell M, Rafaels N, Winters A, Bleecker ER, Cruz AA, Barreto ML, Meyers DA, Ortega VE, Figueiredo CA, Barnes KC, Checkley W, Hansel NN, Mathias RA. Genome-wide association study of asthma, total IgE, and lung function in a cohort of Peruvian children. J Allergy Clin Immunol 2021; 148:1493-1504. [PMID: 33713768 PMCID: PMC8429514 DOI: 10.1016/j.jaci.2021.02.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Genetic ancestry plays a role in asthma health disparities. OBJECTIVE Our aim was to evaluate the impact of ancestry on and identify genetic variants associated with asthma, total serum IgE level, and lung function. METHODS A total of 436 Peruvian children (aged 9-19 years) with asthma and 291 without asthma were genotyped by using the Illumina Multi-Ethnic Global Array. Genome-wide proportions of indigenous ancestry populations from continental America (NAT) and European ancestry from the Iberian populations in Spain (IBS) were estimated by using ADMIXTURE. We assessed the relationship between ancestry and the phenotypes and performed a genome-wide association study. RESULTS The mean ancestry proportions were 84.7% NAT (case patients, 84.2%; controls, 85.4%) and 15.3% IBS (15.8%; 14.6%). With adjustment for asthma, NAT was associated with higher total serum IgE levels (P < .001) and IBS was associated with lower total serum IgE levels (P < .001). NAT was associated with higher FEV1 percent predicted values (P < .001), whereas IBS was associated with lower FEV1 values in the controls but not in the case patients. The HLA-DR/DQ region on chromosome 6 (Chr6) was strongly associated with total serum IgE (rs3135348; P = 3.438 × 10-10) and was independent of an association with the haplotype HLA-DQA1∼HLA-DQB1:04.01∼04.02 (P = 1.55 × 10-05). For lung function, we identified a locus (rs4410198; P = 5.536 × 10-11) mapping to Chr19, near a cluster of zinc finger interacting genes that colocalizes to the long noncoding RNA CTD-2537I9.5. This novel locus was replicated in an independent sample of pediatric case patients with asthma with similar admixture from Brazil (P = .005). CONCLUSION This study confirms the role of HLA in atopy, and identifies a novel locus mapping to a long noncoding RNA for lung function that may be specific to children with NAT.
Collapse
Affiliation(s)
- Ayobami T Akenroye
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Tonya Brunetti
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Karina Romero
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Md; A.B. PRISMA, Lima, Peru
| | - Michelle Daya
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Kanika Kanchan
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Gautam Shankar
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Sameer Chavan
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Meher Preethi Boorgula
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Elizabeth A Ampleford
- Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - Gregory A Hawkins
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | | | - Monica Campbell
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Nicholas Rafaels
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - Alexandra Winters
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | | | | | - Mauricio L Barreto
- Centro de Integração de Dados e Conhecimento para Saúde, Fiocruz, Salvador, Brazil
| | | | - Victor E Ortega
- Department of Internal Medicine, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Camila A Figueiredo
- Instituto de Ciências da Saúde, Universidade Federal da Bahia (UFBA), Salvador, Brazil
| | - Kathleen C Barnes
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Denver, Colo
| | - William Checkley
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Md; Department of International Health, Program in Global Disease Epidemiology and Control, Johns Hopkins Bloomberg School of Public Health, Baltimore, Md
| | - Nadia N Hansel
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Md.
| | - Rasika A Mathias
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
9
|
Multiethnic genome-wide and HLA association study of total serum IgE level. J Allergy Clin Immunol 2021; 148:1589-1595. [PMID: 34536413 PMCID: PMC8665111 DOI: 10.1016/j.jaci.2021.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/26/2021] [Accepted: 09/01/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Total serum IgE (tIgE) is an important intermediate phenotype of allergic disease. Whole genome genetic association studies across ancestries may identify important determinants of IgE. OBJECTIVE We aimed to increase understanding of genetic variants affecting tIgE production across the ancestry and allergic disease spectrum by leveraging data from the National Heart, Lung and Blood Institute Trans-Omics for Precision Medicine program; the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA); and the Atopic Dermatitis Research Network (N = 21,901). METHODS We performed genome-wide association within strata of study, disease, and ancestry groups, and we combined results via a meta-regression approach that models heterogeneity attributable to ancestry. We also tested for association between HLA alleles called from whole genome sequence data and tIgE, assessing replication of associations in HLA alleles called from genotype array data. RESULTS We identified 6 loci at genome-wide significance (P < 5 × 10-9), including 4 loci previously reported as genome-wide significant for tIgE, as well as new regions in chr11q13.5 and chr15q22.2, which were also identified in prior genome-wide association studies of atopic dermatitis and asthma. In the HLA allele association study, HLA-A∗02:01 was associated with decreased tIgE level (Pdiscovery = 2 × 10-4; Preplication = 5 × 10-4; Pdiscovery+replication = 4 × 10-7), and HLA-DQB1∗03:02 was strongly associated with decreased tIgE level in Hispanic/Latino ancestry populations (PHispanic/Latino discovery+replication = 8 × 10-8). CONCLUSION We performed the largest genome-wide association study and HLA association study of tIgE focused on ancestrally diverse populations and found several known tIgE and allergic disease loci that are relevant in non-European ancestry populations.
Collapse
|
10
|
Abstract
In the pediatric population, asthma is the most common chronic disease. Asthma is a chronic inflammatory disease consisting of variable respiratory symptoms and airflow limitation. Proper and timely diagnosis remains of utmost importance. Early diagnosis allows for earlier treatment and subsequent reduction of morbidity and mortality. Newer research and medications have changed the treatment paradigm, including the addition of biologic agents for more severe cases and use of inhaled corticosteroid-formoterol inhaler as a rescue treatment.
Collapse
Affiliation(s)
- Kevin W Gray
- Department of Community and Family Medicine, Truman Medical Center-Lakewood, 7900 Lee's Summit Road, Kansas City, MO 64139, USA.
| |
Collapse
|
11
|
Hillary RF, Trejo-Banos D, Kousathanas A, McCartney DL, Harris SE, Stevenson AJ, Patxot M, Ojavee SE, Zhang Q, Liewald DC, Ritchie CW, Evans KL, Tucker-Drob EM, Wray NR, McRae AF, Visscher PM, Deary IJ, Robinson MR, Marioni RE. Multi-method genome- and epigenome-wide studies of inflammatory protein levels in healthy older adults. Genome Med 2020; 12:60. [PMID: 32641083 PMCID: PMC7346642 DOI: 10.1186/s13073-020-00754-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The molecular factors which control circulating levels of inflammatory proteins are not well understood. Furthermore, association studies between molecular probes and human traits are often performed by linear model-based methods which may fail to account for complex structure and interrelationships within molecular datasets. METHODS In this study, we perform genome- and epigenome-wide association studies (GWAS/EWAS) on the levels of 70 plasma-derived inflammatory protein biomarkers in healthy older adults (Lothian Birth Cohort 1936; n = 876; Olink® inflammation panel). We employ a Bayesian framework (BayesR+) which can account for issues pertaining to data structure and unknown confounding variables (with sensitivity analyses using ordinary least squares- (OLS) and mixed model-based approaches). RESULTS We identified 13 SNPs associated with 13 proteins (n = 1 SNP each) concordant across OLS and Bayesian methods. We identified 3 CpG sites spread across 3 proteins (n = 1 CpG each) that were concordant across OLS, mixed-model and Bayesian analyses. Tagged genetic variants accounted for up to 45% of variance in protein levels (for MCP2, 36% of variance alone attributable to 1 polymorphism). Methylation data accounted for up to 46% of variation in protein levels (for CXCL10). Up to 66% of variation in protein levels (for VEGFA) was explained using genetic and epigenetic data combined. We demonstrated putative causal relationships between CD6 and IL18R1 with inflammatory bowel disease and between IL12B and Crohn's disease. CONCLUSIONS Our data may aid understanding of the molecular regulation of the circulating inflammatory proteome as well as causal relationships between inflammatory mediators and disease.
Collapse
Affiliation(s)
- Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Daniel Trejo-Banos
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Athanasios Kousathanas
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Sarah E Harris
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Anna J Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Marion Patxot
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Sven Erik Ojavee
- Department of Computational Biology, University of Lausanne, 1015, Lausanne, Switzerland
| | - Qian Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - David C Liewald
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Craig W Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Elliot M Tucker-Drob
- Department of Psychology, The University of Texas at Austin, Austin, TX, 78712, USA
- Population Research Center, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Naomi R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Lothian Birth Cohorts, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Matthew R Robinson
- Institute of Science and Technology Austria, 3400, Klosterneuburg, Austria.
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK.
| |
Collapse
|
12
|
Vince N, Limou S, Daya M, Morii W, Rafaels N, Geffard E, Douillard V, Walencik A, Boorgula MP, Chavan S, Vergara C, Ortega VE, Wilson JG, Lange LA, Watson H, Nicolae DL, Meyers DA, Hansel NN, Ford JG, Faruque MU, Bleecker ER, Campbell M, Beaty TH, Ruczinski I, Mathias RA, Taub MA, Ober C, Noguchi E, Barnes KC, Torgerson D, Gourraud PA. Association of HLA-DRB1∗09:01 with tIgE levels among African-ancestry individuals with asthma. J Allergy Clin Immunol 2020; 146:147-155. [PMID: 31981624 DOI: 10.1016/j.jaci.2020.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 12/05/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Asthma is a complex chronic inflammatory disease of the airways. Association studies between HLA and asthma were first reported in the 1970s, and yet, the precise role of HLA alleles in asthma is not fully understood. Numerous genome-wide association studies were recently conducted on asthma, but were always limited to simple genetic markers (single nucleotide polymorphisms) and not complex HLA gene polymorphisms (alleles/haplotypes), therefore not capturing the biological relevance of this complex locus for asthma pathogenesis. OBJECTIVE To run the first HLA-centric association study with asthma and specific asthma-related phenotypes in a large cohort of African-ancestry individuals. METHODS We collected high-density genomics data for the Consortium on Asthma among African-ancestry Populations in the Americas (N = 4993) participants. Using computer-intensive machine-learning attribute bagging methods to infer HLA alleles, and Easy-HLA to infer HLA 5-gene haplotypes, we conducted a high-throughput HLA-centric association study of asthma susceptibility and total serum IgE (tIgE) levels in subjects with and without asthma. RESULTS Among the 1607 individuals with asthma, 972 had available tIgE levels, with a mean tIgE level of 198.7 IU/mL. We could not identify any association with asthma susceptibility. However, we showed that HLA-DRB1∗09:01 was associated with increased tIgE levels (P = 8.5 × 10-4; weighted effect size, 0.51 [0.15-0.87]). CONCLUSIONS We identified for the first time an HLA allele associated with tIgE levels in African-ancestry individuals with asthma. Our report emphasizes that by leveraging powerful computational machine-learning methods, specific/extreme phenotypes, and population diversity, we can explore HLA gene polymorphisms in depth and reveal the full extent of complex disease associations.
Collapse
Affiliation(s)
- Nicolas Vince
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France
| | - Sophie Limou
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France; Ecole Centrale de Nantes, Nantes, France
| | - Michelle Daya
- Department of Medicine, University of Colorado Denver, Aurora, Colo
| | - Wataru Morii
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Nicholas Rafaels
- Department of Medicine, University of Colorado Denver, Aurora, Colo
| | - Estelle Geffard
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France
| | - Venceslas Douillard
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France
| | - Alexandre Walencik
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France
| | | | - Sameer Chavan
- Department of Medicine, University of Colorado Denver, Aurora, Colo
| | | | - Victor E Ortega
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Miss
| | - Leslie A Lange
- Department of Medicine, University of Colorado Denver, Aurora, Colo
| | - Harold Watson
- Faculty of Medical Sciences Cave Hill Campus, The University of the West Indies, Bridgetown, Barbados
| | - Dan L Nicolae
- Department of Medicine, University of Chicago, Chicago, Ill
| | - Deborah A Meyers
- Department of Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | - Nadia N Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - Jean G Ford
- Department of Medicine, Einstein Medical Center, Philadelphia, Pa
| | - Mezbah U Faruque
- National Human Genome Center, Howard University College of Medicine, Washington, DC
| | - Eugene R Bleecker
- Department of Medicine, University of Arizona College of Medicine, Tucson, Ariz
| | - Monica Campbell
- Department of Medicine, University of Colorado Denver, Aurora, Colo
| | - Terri H Beaty
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Ingo Ruczinski
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, Md; Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Margaret A Taub
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | | | - Dara Torgerson
- McGill University and Genome Quebec Innovation Centre, Montreal, Quebec, Canada
| | - Pierre-Antoine Gourraud
- Université de Nantes, Centrale Nantes, CHU Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, Nantes, France.
| |
Collapse
|
13
|
Samelko L, Petfield J, McAllister K, Hsu J, Hawkinson M, Jacobs JJ, Hallab NJ. Do Battlefield Injury-acquired Indwelling Metal Fragments Induce Metal Immunogenicity? Clin Orthop Relat Res 2020; 478:752-766. [PMID: 32229747 PMCID: PMC7282599 DOI: 10.1097/corr.0000000000000953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 08/13/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND A battlefield-related injury results in increased local and systemic innate immune inflammatory responses, resulting in wound-specific complications and an increased incidence of osteoarthritis. However, little is known about whether severe injuries affect long-term systemic homeostasis, for example, immune function. Moreover, it also remains unknown whether battlefield-acquired metal fragments retained over the long term result in residual systemic effects such as altered immune reactivity to metals. QUESTIONS/PURPOSES Does a retained metal fragment from a battlefield injury contribute to increased (1) adaptive metal-specific immune responses, (2) systemically elevated metal ion serum levels, and (3) serum immunoglobulin levels compared with combat injuries that did not result in a retained metal fragment? METHODS In this pilot study, we analyzed metal-immunogenicity in injured military personnel and noninjured control participants using lymphocyte transformation testing (LTT, lymphocyte proliferation responses to cobalt, chromium and nickel challenge at 0.001, 0.01 and 0.1-mM concentrations in triplicate for each participant), serum metal ion analysis (ICP-mass spectroscopy), and serum immunoglobulin analysis (IgE, IgG, IgA, and IgM ). Military personnel with a battlefield-sustained injury self-recruited without any exclusion for sex, age, degree of injury. Those with battlefield injury resulting in retained metal fragments (INJ-FRAG, n = 20 male, mean time since injury ± SD was 12 ± 10 years) were compared with those with a battlefield injury but without retained metal fragments (INJ-NO-FRAG, n = 12 male, mean time since injury ± SD was 13 ± 12 years). A control group comprised of male noninjured participants was used to compare measured immunogenicity metrics (n = 11, males were selected to match battlefield injury group demographics). RESULTS Military participants with sustained metal fragments had increased levels of metal-induced lymphocyte responses. The lymphocyte stimulation index among military participants with metal fragments was higher than in those with nonretained metal fragments (stimulation index = 4.2 ± 6.0 versus stimulation index = 2.1 ± 1.2 (mean difference 2.1 ± 1.4 [95% confidence interval 5.1 to 0.8]; p = 0.07) and an average stimulation index = 2 ± 1 in noninjured controls. Four of 20 participants injured with retained fragments had a lymphocyte proliferation index greater than 2 to cobalt compared with 0 in the group without a retained metal fragment or 0 in the control participants. However, with the numbers available, military personnel with retained metal fragments did not have higher serum metal ion levels than military participants without retained metal fragment-related injuries or control participants. Military personnel with retained metal fragments had lower serum immunoglobulin levels (IgG, IgA, and IgM) than military personnel without retained metal fragments and noninjured controls, except for IgE. Individuals who were metal-reactive positive (that is, a stimulation index > 2) with retained metal fragments had higher median IgE serum levels than participants who metal-reactive with nonmetal injuries (1198 ± 383 IU/mL versus 171 ± 67 IU/mL, mean difference 1027 ± 477 IU/mL [95% CI 2029 to 25]; p = 0.02). CONCLUSIONS We found that males with retained metal fragments after a battlefield-related injury had altered adaptive immune responses compared with battlefield-injured military personnel without indwelling metal fragments. Military participants with a retained metal fragment had an increased proportion of group members and increased average lymphocyte reactivity to common implant metals such as nickel and cobalt. Further studies are needed to determine a causal association between exposure to amounts of retained metal fragments, type of injury, personnel demographics and general immune function/reactivity that may affect personal health or future metal implant performance. LEVEL OF EVIDENCE Level IV, therapeutic study.
Collapse
Affiliation(s)
- Lauryn Samelko
- L. Samelko, K. McAllister, J. J. Jacobs, Rush University Medical Center, Chicago, IL, USA
| | - Joseph Petfield
- J. Petfield, M. Hawkinson, San Antonio Medical Center, San Antonio, TX, USA
| | - Kyron McAllister
- L. Samelko, K. McAllister, J. J. Jacobs, Rush University Medical Center, Chicago, IL, USA
| | - Joseph Hsu
- J. Hsu, Carolinas Medical Center, Charlotte, NC, USA
| | - Michael Hawkinson
- J. Petfield, M. Hawkinson, San Antonio Medical Center, San Antonio, TX, USA
| | - Joshua J Jacobs
- L. Samelko, K. McAllister, J. J. Jacobs, Rush University Medical Center, Chicago, IL, USA
| | - Nadim J Hallab
- N. J. Hallab, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
14
|
Gao Y, Li J, Zhang Y, Zhang L. Replication study of susceptibility variants associated with allergic rhinitis and allergy in Han Chinese. Allergy Asthma Clin Immunol 2020; 16:13. [PMID: 32082391 PMCID: PMC7014941 DOI: 10.1186/s13223-020-0411-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/28/2020] [Indexed: 12/12/2022] Open
Abstract
Background Allergic rhinitis (AR) is believed to be a complex genetic disease. The last decade has been marked by the publication of more than 20 genome-wide association studies (GWASs) of AR and associated allergic phenotypes and allergic diseases, which have shown allergic diseases and traits to share a large number of genetic susceptibility loci. The aim of present study was therefore to investigate the highly replicated allergy related genes and variants as candidates for AR in Han Chinese subjects. Methods A total of 762 AR patients and 760 control subjects were recruited, and a total of 58 susceptible variants previously reported to be associated with allergic traits were choose for replication. Results Logistic regression analyses revealed that in the co-dominant-effect model as assessed by the AIC, compared with wild-type carriers, significant AR risk were associated with rs9865818 in LPP (P = 0.029, OR = 1.469 for GG vs. AA); rs6554809 in DNAH5 (P = 0.000, OR = 1.597 for TC vs. CC); rs1438673 in WDR36-CAMK4 loci (P = 0.037, OR = 1.396 for CC vs.TT), rs7775228 in HLA region (P = 0.000, OR = 1.589 for TC vs.TT), rs7203459 in CLEC16A (P = 0.025, OR = 0.731 for TC vs. TT). Conclusion We replicated Han Chinese AR-specific susceptibility loci in LPP, DNAH5, HLA, CLEC16A and WDR36-CAMK4. Further understanding the molecular mechanisms underlying these associations may provide new insights into the etiology of allergic disease.
Collapse
Affiliation(s)
- Yunbo Gao
- 1Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China
| | - Jingyun Li
- 1Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China.,2Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, No. 17, HouGouHuTong, DongCheng District, Beijing, 100005 People's Republic of China
| | - Yuan Zhang
- 1Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China.,2Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, No. 17, HouGouHuTong, DongCheng District, Beijing, 100005 People's Republic of China.,3Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China
| | - Luo Zhang
- 1Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China.,2Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, No. 17, HouGouHuTong, DongCheng District, Beijing, 100005 People's Republic of China.,3Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, 100730 People's Republic of China
| |
Collapse
|
15
|
Lou C, Mitra N, Wubbenhorst B, D'Andrea K, Hoffstad O, Kim BS, Yan A, Zaenglein AL, Fuxench ZC, Nathanson KL, Margolis DJ. Association between fine mapping thymic stromal lymphopoietin and atopic dermatitis onset and persistence. Ann Allergy Asthma Immunol 2019; 123:595-601.e1. [PMID: 31491540 DOI: 10.1016/j.anai.2019.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/20/2019] [Accepted: 08/24/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a common chronic relapsing skin disease. Genetic variants have been associated with skin barrier function and immune regulation. Thymic stromal lymphopoietin (TSLP), an immune regulator, has been previously associated with AD. OBJECTIVE To fine map TSLP and evaluate associations with the onset and persistence of AD. METHODS TSLP variation was determined using targeted massively parallel sequencing in a longitudinal cohort of children with AD. Evaluations included linkage disequilibrium and the persistence of AD for as many as 10 years of follow-up. The association between the presence of AD and rs1898671 variation was evaluated in a second independent cohort. RESULTS The minor variant frequency for rs1898671 was 23.5% (95% CI, 21.4%-25.8%). This variant was not in linkage disequilibrium with other TSLP variants in the longitudinal cohort (n = 741). White children with AD were less likely to have rs1898671 variant (odds ratio [OR], 1.41; 95% CI, 1.20-1.66) than Genome Aggregation Database controls. Children with AD and the rs1898671 variant during follow-up were more likely to have remission than children who were wild type for rs1898671 (OR, 1.56; 95% CI, 1.26-1.91). In the second cohort (n = 585), the rs1898671 variant was less prevalent in those with AD than those without. The protective effect was greater in rs1898671 heterozygotes (OR, 1.91; 95% CI, 1.34-2.75) than homozygotes (OR, 1.28; 95% CI, 0.61-2.70). CONCLUSION TSLP and specifically rs1898671 are important in the pathogenesis of AD and could represent a potential clinical target for the development of therapies to treat individuals with AD.
Collapse
Affiliation(s)
- Carolyn Lou
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Bradley Wubbenhorst
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Kurt D'Andrea
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Ole Hoffstad
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Brian S Kim
- Washington University School of Medicine, St Louis, Missouri
| | - Albert Yan
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Andrea L Zaenglein
- Departments of Dermatology and Pediatrics, Pennsylvania State University/Hershey Medical Center, Hershey, Pennsylvania
| | - Zelma Chiesa Fuxench
- Department of Dermatology, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - Katherine L Nathanson
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perlman School of Medicine, Philadelphia, Pennsylvania
| | - David J Margolis
- Department of Biostatistics, Epidemiology and Informatics, Perlman School of Medicine, Philadelphia, Pennsylvania; Department of Dermatology, Perlman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
16
|
Laing BB, Lim AG, Ferguson LR. A Personalised Dietary Approach-A Way Forward to Manage Nutrient Deficiency, Effects of the Western Diet, and Food Intolerances in Inflammatory Bowel Disease. Nutrients 2019; 11:nu11071532. [PMID: 31284450 PMCID: PMC6683058 DOI: 10.3390/nu11071532] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/29/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
This review discusses the personalised dietary approach with respect to inflammatory bowel disease (IBD). It identifies gene–nutrient interactions associated with the nutritional deficiencies that people with IBD commonly experience, and the role of the Western diet in influencing these. It also discusses food intolerances and how particular genotypes can affect these. It is well established that with respect to food there is no “one size fits all” diet for those with IBD. Gene–nutrient interactions may help explain this variability in response to food that is associated with IBD. Nutrigenomic research, which examines the effects of food and its constituents on gene expression, shows that—like a number of pharmaceutical products—food can have beneficial effects or have adverse (side) effects depending on a person’s genotype. Pharmacogenetic research is identifying gene variants with adverse reactions to drugs, and this is modifying clinical practice and allowing individualised treatment. Nutrigenomic research could enable individualised treatment in persons with IBD and enable more accurate tailoring of food intake, to avoid exacerbating malnutrition and to counter some of the adverse effects of the Western diet. It may also help to establish the dietary pattern that is most protective against IBD.
Collapse
Affiliation(s)
- Bobbi B Laing
- Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
- Nutrition Society of New Zealand, Palmerston North 4444, New Zealand
| | - Anecita Gigi Lim
- Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Lynnette R Ferguson
- Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
17
|
Yao TC, Chung RH, Lin CY, Tsai PC, Chang WC, Yeh KW, Tsai MH, Liao SL, Hua MC, Lai SH, Chen LC, Chang SW, Yu YW, Hsu JY, Chang SC, Cheng WC, Hu D, Hong X, Burchard EG, Wang X, Tzeng JY, Tsai HJ, Huang JL. Genetic loci determining total immunoglobulin E levels from birth through adulthood. Allergy 2019; 74:621-625. [PMID: 30378687 DOI: 10.1111/all.13654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Tsung-Chieh Yao
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
- Chang Gung Immunology Consortium; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
| | - Ren-Hua Chung
- Division of Biostatistics and Bioinformatics; Institute of Population Health Sciences; National Health Research Institutes; Zhunan Taiwan
| | - Chung-Yen Lin
- Division of Biostatistics and Bioinformatics; Institute of Population Health Sciences; National Health Research Institutes; Zhunan Taiwan
- Institute of Information Science; Academia Sinica; Taipei Taiwan
| | - Pei-Chien Tsai
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Department and Graduate Institute of Biomedical Sciences; Chang Gung University; Taoyuan Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy; School of Pharmacy; Taipei Medical University; Taipei Taiwan
- Department of Pharmacy; Wan Fang Hospital; Taipei Medical University; Taipei Taiwan
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics; School of Pharmacy; Taipei Medical University; Taipei Taiwan
- Center for Biomarkers and Biotech Drugs; Kaohsiung Medical University; Kaohsiung Taiwan
| | - Kuo-Wei Yeh
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| | - Ming-Han Tsai
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
- Department of Pediatrics; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| | - Sui-Ling Liao
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
- Department of Pediatrics; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| | - Man-Chin Hua
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
- Department of Pediatrics; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| | - Shen-Hao Lai
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
- Division of Pediatric Pulmonology; Department of Pediatrics; Chang Gung Memorial Hospital; Taoyuan Taiwan
| | - Li-Chen Chen
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| | - Su-Wei Chang
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Clinical Informatics and Medical Statistics Research Center; Chang Gung University College of Medicine; Taoyuan Taiwan
| | - Ya-Wen Yu
- Division of Biostatistics and Bioinformatics; Institute of Population Health Sciences; National Health Research Institutes; Zhunan Taiwan
| | - Jing-Ya Hsu
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
| | - Su-Ching Chang
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
| | - Wen-Chih Cheng
- Division of Biostatistics and Bioinformatics; Institute of Population Health Sciences; National Health Research Institutes; Zhunan Taiwan
| | - Donglei Hu
- Department of Medicine; University of California, San Francisco; San Francisco California
| | - Xiumei Hong
- Department of Population, Family and Reproductive Health; Center on Early Life Origins of Disease; Johns Hopkins University Bloomberg School of Public Health; Baltimore Maryland
| | - Esteban G. Burchard
- Department of Medicine; University of California, San Francisco; San Francisco California
- Department of Bioengineering and Therapeutic Sciences; University of California San Francisco; San Francisco California
| | - Xiaobin Wang
- Department of Population, Family and Reproductive Health; Center on Early Life Origins of Disease; Johns Hopkins University Bloomberg School of Public Health; Baltimore Maryland
| | - Jung-Ying Tzeng
- Department of Statistics; North Carolina State University; Raleigh North Carolina
- Bioinformatics Research Center; North Carolina State University; Raleigh North Carolina
- Institute of Epidemiology and Preventive Medicine; National Taiwan University; Taipei Taiwan
- Department of Statistics; National Cheng-Kung University; Tainan Taiwan
| | - Hui-Ju Tsai
- Division of Biostatistics and Bioinformatics; Institute of Population Health Sciences; National Health Research Institutes; Zhunan Taiwan
- Department of Population, Family and Reproductive Health; Center on Early Life Origins of Disease; Johns Hopkins University Bloomberg School of Public Health; Baltimore Maryland
| | - Jing-Long Huang
- Division of Allergy, Asthma, and Rheumatology; Department of Pediatrics; Chang Gung Memorial Hospital; Chang Gung University College of Medicine; Taoyuan Taiwan
- Community Medicine Research Center; Chang Gung Memorial Hospital at Keelung; Keelung Taiwan
| |
Collapse
|
18
|
Association study in African-admixed populations across the Americas recapitulates asthma risk loci in non-African populations. Nat Commun 2019; 10:880. [PMID: 30787307 PMCID: PMC6382865 DOI: 10.1038/s41467-019-08469-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022] Open
Abstract
Asthma is a complex disease with striking disparities across racial and ethnic groups. Despite its relatively high burden, representation of individuals of African ancestry in asthma genome-wide association studies (GWAS) has been inadequate, and true associations in these underrepresented minority groups have been inconclusive. We report the results of a genome-wide meta-analysis from the Consortium on Asthma among African Ancestry Populations (CAAPA; 7009 asthma cases, 7645 controls). We find strong evidence for association at four previously reported asthma loci whose discovery was driven largely by non-African populations, including the chromosome 17q12-q21 locus and the chr12q13 region, a novel (and not previously replicated) asthma locus recently identified by the Trans-National Asthma Genetic Consortium (TAGC). An additional seven loci reported by TAGC show marginal evidence for association in CAAPA. We also identify two novel loci (8p23 and 8q24) that may be specific to asthma risk in African ancestry populations.
Collapse
|
19
|
Kim KW, Ober C. Lessons Learned From GWAS of Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2019; 11:170-187. [PMID: 30661310 PMCID: PMC6340805 DOI: 10.4168/aair.2019.11.2.170] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/05/2018] [Indexed: 02/05/2023]
Abstract
Asthma is a common complex disease of the airways. Genome-wide association studies (GWASs) of asthma have identified many risk alleles and loci that have been replicated in worldwide populations. Although the risk alleles identified by GWAS have small effects and explain only a small portion of prevalence, the discovery of asthma loci can provide an understanding of its genetic architecture and the molecular pathways involved in disease pathogenesis. These discoveries can translate into advances in clinical care by identifying therapeutic targets, preventive strategies and ultimately approaches for personalized medicine. In this review, we summarize results from GWAS of asthma from the past 10 years and the insights gleaned from these discoveries.
Collapse
Affiliation(s)
- Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| |
Collapse
|
20
|
Sanyal RD, Pavel AB, Glickman J, Chan TC, Zheng X, Zhang N, Cueto I, Peng X, Estrada Y, Fuentes-Duculan J, Alexis AF, Krueger JG, Guttman-Yassky E. Atopic dermatitis in African American patients is T H2/T H22-skewed with T H1/T H17 attenuation. Ann Allergy Asthma Immunol 2019; 122:99-110.e6. [PMID: 30223113 DOI: 10.1016/j.anai.2018.08.024] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 08/28/2018] [Accepted: 08/31/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND African Americans (AA) are disproportionately impacted by atopic dermatitis (AD), with increased prevalence and therapeutic challenges unique to this population. Molecular profiling data informing development of targeted therapeutics for AD are derived primarily from European American (EA) patients. These studies are absent in AA, hindering development of effective treatments for this population. OBJECTIVE We sought to characterize the global molecular profile of AD in the skin of AA patients as compared with that of EA AD and healthy controls. METHODS We performed RNA-Seq with reverse transcription polymerase chain reaction validation and immunohistochemistry studies in lesional and nonlesional skin of AA and EA AD patients vs healthy controls. RESULTS African American AD lesions were characterized by greater infiltration of dendritic cells (DCs) marked by the high-affinity immunoglobulin E (IgE) receptor (FcεR1+) compared with EA AD (P < .05). Both AD cohorts showed similarly robust up-regulation of Th2-related (CCL17/18/26) and Th22-related markers (interleukin [IL]-22, S100A8/9/12), but AA AD featured decreased expression of innate immune (tumor necrosis factor [TNF], IL-1β), Th1-related (interferon gamma [IFN-γ], MX1, IL-12RB1), and Th17-related markers (IL-23p19, IL-36G, CXCL1) vs EA AD (P < .05). The Th2 (IL-13) and Th22-related products (IL-22, S100A8/9/12) and serum IgE were significantly correlated with clinical severity (Scoring of Atopic Dermatitis [SCORAD]) in AA. Fillagrin (FLG) was exclusively down-regulated in EA AD, whereas loricrin (LOR) was down-regulated in both AD cohorts and negatively correlated with SCORAD in AA. CONCLUSION The molecular phenotype of AA AD skin is characterized by attenuated Th1 and Th17 but similar Th2/Th22-skewing to EA AD. Our data encourages a personalized medicine approach accounting for phenotype-specific characteristics in future development of targeted therapeutics and clinical trial design for AD.
Collapse
Affiliation(s)
- Riana D Sanyal
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ana B Pavel
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jacob Glickman
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tom C Chan
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan; The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Xiuzhong Zheng
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Ning Zhang
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Inna Cueto
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Xiangyu Peng
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel Estrada
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Judilyn Fuentes-Duculan
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Andrew F Alexis
- Department of Dermatology, Mount Sinai St. Luke's and Mount Sinai West, New York, New York
| | - James G Krueger
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
21
|
Maglie R, Quintarelli L, Verdelli A, Fabbri P, Antiga E, Caproni M. Specific dermatoses of pregnancy other than pemphigoid gestationis. GIORN ITAL DERMAT V 2018; 154:286-298. [PMID: 30375214 DOI: 10.23736/s0392-0488.18.06159-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Specific dermatoses of pregnancy are skin disorders that occur specifically during or immediately after pregnancy and cannot be found in non-pregnant patients. According to the current consensus, they include atopic eruption of pregnancy (AEP), polymorphic eruption of pregnancy (PEP), pemphigoid gestationis (PG), and intrahepatic cholestasis of pregnancy (ICP). The diagnosis of specific dermatoses of pregnancy can be challenging due to their variation in clinical presentation; moreover, the tests currently available do not always provide the clue for the diagnosis. However, some distinctive features may be helpful to differentiate between such entities. Accordingly, the knowledge of specific dermatoses of pregnancy and of their management is critical, since their early recognition may allow to provide care for the mother and prevent potential increased fetal risk. In fact, while AEP and PEP do not affect maternal and fetal prognosis, PG and, mainly, ICP are associated to maternal complications as well as the risk of fetal loss. In this paper, the epidemiology, pathogenesis, clinical features as well as management of AEP and PEP are reviewed in detail, while PG is described in another article of this issue. Moreover, the main features of ICP, which cannot be considered a primarily skin disease but may be managed first by dermatologists, are reported.
Collapse
Affiliation(s)
- Roberto Maglie
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Lavinia Quintarelli
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Alice Verdelli
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Paolo Fabbri
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Emiliano Antiga
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy -
| | - Marzia Caproni
- Section of Dermatology, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
22
|
Ginkel CD, Pettersson ME, Dubois AEJ, Koppelman GH. Association of STAT6 gene variants with food allergy diagnosed by double-blind placebo-controlled food challenges. Allergy 2018; 73:1337-1341. [PMID: 29457221 PMCID: PMC6032865 DOI: 10.1111/all.13432] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2018] [Indexed: 12/25/2022]
Abstract
This study describes the role of two STAT6 gene variants in food allergy using data of patients and their parents who underwent double‐blind placebo‐controlled food challenges (DBPCFCs). After quality control, 369 trios were analysed including 262 children (71.0%) with food allergy. Associations were tested by the Family based association test. The A alleles of both SNPs were associated with food allergy (P = .036 and P = .013 for rs324015 and rs1059513, respectively). Furthermore, these A alleles were associated with peanut allergy, higher sIgE levels to both peanut and cow's milk, more severe symptoms and higher eliciting doses during peanut and cow's milk DBPCFCs (all P < .05). In silico analysis indicates that the identified risk variants increase STAT6 expression which stimulates the differentiation of CD4 + T cells to the Th2 subset. In conclusion, STAT6 variants may be involved in the pathophysiology of food allergy and their role seems to be independent of the allergenic food.
Collapse
Affiliation(s)
- C. D. Ginkel
- University Medical Center Groningen Department of Paediatric Pulmonology and Paediatric Allergy GRIAC Research Institute University of Groningen Groningen The Netherlands
| | - M. E. Pettersson
- University Medical Center Groningen Department of Paediatric Pulmonology and Paediatric Allergy GRIAC Research Institute University of Groningen Groningen The Netherlands
| | - A. E. J. Dubois
- University Medical Center Groningen Department of Paediatric Pulmonology and Paediatric Allergy GRIAC Research Institute University of Groningen Groningen The Netherlands
| | - G. H. Koppelman
- University Medical Center Groningen Department of Paediatric Pulmonology and Paediatric Allergy GRIAC Research Institute University of Groningen Groningen The Netherlands
| |
Collapse
|
23
|
A decade of research on the 17q12-21 asthma locus: Piecing together the puzzle. J Allergy Clin Immunol 2018; 142:749-764.e3. [PMID: 29307657 PMCID: PMC6172038 DOI: 10.1016/j.jaci.2017.12.974] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/13/2017] [Accepted: 12/16/2017] [Indexed: 12/20/2022]
Abstract
Chromosome 17q12–21 remains the most highly replicated and significant asthma locus. Genotypes in the core region defined by the first genome-wide association study correlate with expression of 2 genes, ORM1-like 3 (ORMDL3) and gasdermin B (GSDMB), making these prime candidate asthma genes, although recent studies have implicated gasdermin A (GSDMA) distal to and post-GPI attachment to proteins 3 (PGAP3) proximal to the core region as independent loci. We review 10 years of studies on the 17q12–21 locus and suggest that genotype-specific risks for asthma at the proximal and distal loci are not specific to early-onset asthma and mediated by PGAP3, ORMDL3, and/or GSDMA expression. We propose that the weak and inconsistent associations of 17q single nucleotide polymorphisms with asthma in African Americans is due to the high frequency of some 17q alleles, the breakdown of linkage disequilibrium on African-derived chromosomes, and possibly different early-life asthma endotypes in these children. Finally, the inconsistent association between asthma and gene expression levels in blood or lung cells from older children and adults suggests that genotype effects may mediate asthma risk or protection during critical developmental windows and/or in response to relevant exposures in early life. Thus studies of young children and ethnically diverse populations are required to fully understand the relationship between genotype and asthma phenotype and the gene regulatory architecture at this locus. (J Allergy Clin Immunol 2018;142:749–64.)
Collapse
|
24
|
Kan M, Shumyatcher M, Himes BE. Using omics approaches to understand pulmonary diseases. Respir Res 2017; 18:149. [PMID: 28774304 PMCID: PMC5543452 DOI: 10.1186/s12931-017-0631-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 07/26/2017] [Indexed: 12/24/2022] Open
Abstract
Omics approaches are high-throughput unbiased technologies that provide snapshots of various aspects of biological systems and include: 1) genomics, the measure of DNA variation; 2) transcriptomics, the measure of RNA expression; 3) epigenomics, the measure of DNA alterations not involving sequence variation that influence RNA expression; 4) proteomics, the measure of protein expression or its chemical modifications; and 5) metabolomics, the measure of metabolite levels. Our understanding of pulmonary diseases has increased as a result of applying these omics approaches to characterize patients, uncover mechanisms underlying drug responsiveness, and identify effects of environmental exposures and interventions. As more tissue- and cell-specific omics data is analyzed and integrated for diverse patients under various conditions, there will be increased identification of key mechanisms that underlie pulmonary biological processes, disease endotypes, and novel therapeutics that are efficacious in select individuals. We provide a synopsis of how omics approaches have advanced our understanding of asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), idiopathic pulmonary fibrosis (IPF), and pulmonary arterial hypertension (PAH), and we highlight ongoing work that will facilitate pulmonary disease precision medicine.
Collapse
Affiliation(s)
- Mengyuan Kan
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| | - Maya Shumyatcher
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, 402 Blockley Hall 423 Guardian Drive, Philadelphia, PA 19104 USA
| |
Collapse
|
25
|
Fitzpatrick AM. Severe Asthma in Children: Lessons Learned and Future Directions. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:11-9; quiz 20-1. [PMID: 26772923 DOI: 10.1016/j.jaip.2015.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 10/14/2015] [Accepted: 10/16/2015] [Indexed: 01/02/2023]
Abstract
Severe asthma in children is a complicated and heterogeneous disorder that is extremely challenging to treat. Although most children with asthma derive clinical benefit from daily administration of low-to-medium-dose inhaled corticosteroid (ICS) therapy, a small subset of children with "severe" or "refractory" asthma require high doses of ICS and even systemic corticosteroids to maintain symptom control. These children with severe asthma are at increased risk for adverse outcomes including medication-related side effects and recurrent and life-threatening exacerbations that significantly impair quality of life. This review highlights findings on severe asthma in school-age children (age 6-17 years) from the National Heart, Lung and Blood Institute's Severe Asthma Research Program (SARP) over a 10-year period, between 2001 and 2011. Although SARP has advanced knowledge of the unique clinical, biological, and molecular attributes of severe asthma in children, considerable gaps remain for which additional studies are needed.
Collapse
Affiliation(s)
- Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Ga; Children's Healthcare of Atlanta Center for Cystic Fibrosis and Airways Disease Research, Atlanta, Ga.
| |
Collapse
|
26
|
Kantor DB, Stenquist N, McDonald MC, Schultz BJ, Hauptman M, Smallwood CD, Nelson KA, Perzanowski MS, Matsui EC, Phipatanakul W, Hirschhorn JN. Rhinovirus and serum IgE are associated with acute asthma exacerbation severity in children. J Allergy Clin Immunol 2016; 138:1467-1471.e9. [PMID: 27474123 DOI: 10.1016/j.jaci.2016.04.044] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 04/06/2016] [Accepted: 04/20/2016] [Indexed: 12/13/2022]
Affiliation(s)
- David B Kantor
- Department of Anesthesiology, Perioperative and Pain Medicine, Division of Critical Care Medicine, Boston Children's Hospital, Boston, Mass; Department of Anaesthesia, Harvard Medical School, Boston, Mass
| | - Nicole Stenquist
- Department of Anesthesiology, Perioperative and Pain Medicine, Division of Critical Care Medicine, Boston Children's Hospital, Boston, Mass
| | - Molly C McDonald
- Clinical Research Center, Boston Children's Hospital, Boston, Mass
| | - Blake J Schultz
- Department of Anesthesiology, Perioperative and Pain Medicine, Division of Critical Care Medicine, Boston Children's Hospital, Boston, Mass
| | - Marissa Hauptman
- Division of General Pediatrics, Boston Children's Hospital, Boston, Mass; Region 1 New England Pediatric Environmental Health Specialty Unit, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Craig D Smallwood
- Department of Anesthesiology, Perioperative and Pain Medicine, Division of Critical Care Medicine, Boston Children's Hospital, Boston, Mass
| | - Kyle A Nelson
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Pediatric Emergency Medicine, Boston Children's Hospital, Boston, Mass
| | - Matthew S Perzanowski
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Elizabeth C Matsui
- Division of Pediatric Allergy and Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Wanda Phipatanakul
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Allergy and Immunology, Boston Children's Hospital, Boston, Mass.
| | - Joel N Hirschhorn
- Division of Endocrinology, Boston Children's Hospital, Boston, Mass; Program in Medical & Population Genetics, Broad Institute of Harvard & MIT, Cambridge, Mass; Department of Genetics, Harvard Medical School, Boston, Mass.
| |
Collapse
|
27
|
Ullemar V, Magnusson PKE, Lundholm C, Zettergren A, Melén E, Lichtenstein P, Almqvist C. Heritability and confirmation of genetic association studies for childhood asthma in twins. Allergy 2016; 71:230-8. [PMID: 26786172 DOI: 10.1111/all.12783] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although the genetics of asthma has been extensively studied using both quantitative and molecular genetic analysis methods, both approaches lack studies specific to the childhood phenotype and including other allergic diseases. This study aimed to give specific estimates for the heritability of childhood asthma and other allergic diseases, to attempt to replicate findings from genomewide association studies (GWAS) for childhood asthma and to test the same variants against other allergic diseases. METHODS In a cohort of 25 306 Swedish twins aged 9 or 12 years, data on asthma were available from parental interviews and population-based registers. The interviews also inquired about wheeze, hay fever, eczema, and food allergy. Through structural equation modeling, the heritability of all phenotypes was calculated. A subset of 10 075 twins was genotyped for 16 single nucleotide polymorphisms (SNPs) selected from previous GWAS; these were first tested for association with asthma and significant findings also against the other allergic diseases. RESULTS The heritability of any childhood asthma was 0.82 (95% CI 0.79-0.85). For the other allergic diseases, the range was approximately 0.60-0.80. Associations for six SNPs with asthma were replicated, including rs2305480 in the GSDMB gene (OR 0.80, 95% CI 0.74-0.86, P = 1.5*10(-8) ; other significant associations all below P = 3.5*10(-4) ). Of these, only rs3771180 in IL1RL1 was associated with any other allergic disease (for hay fever, OR 0.64, 95% CI 0.53-0.77, P = 2.5*10(-6) ). CONCLUSION Asthma and allergic diseases of childhood are highly heritable, and these high-risk genetic variants associated specifically with childhood asthma, except for one SNP shared with hay fever.
Collapse
Affiliation(s)
- V. Ullemar
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Stockholm Sweden
| | - P. K. E. Magnusson
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Stockholm Sweden
| | - C. Lundholm
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Stockholm Sweden
| | - A. Zettergren
- Department of Pharmacology; Institute of Neuroscience and Physiology at the Sahlgrenska Academy; University of Gothenburg; Gothenburg Sweden
- Department of Psychiatry and Neurochemistry; Institute of Neuroscience and Physiology; The Sahlgrenska Academy at University of Gothenburg; Gothenburg Sweden
| | - E. Melén
- Institute of Environmental Medicine; Karolinska Institutet; Stockholm Sweden
- Sachs' Children's Hospital; Stockholm Sweden
| | - P. Lichtenstein
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Stockholm Sweden
| | - C. Almqvist
- Department of Medical Epidemiology and Biostatistics; Karolinska Institutet; Stockholm Sweden
- Astrid Lindgren Children's Hospital; Lung and Allergy Unit; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
28
|
Weidinger S, Kabesch M, Rodriguez E. Genetik und Epigenetik von allergischen Erkrankungen und Asthma. ALLERGOLOGIE 2016. [DOI: 10.1007/978-3-642-37203-2_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Portelli MA, Hodge E, Sayers I. Genetic risk factors for the development of allergic disease identified by genome-wide association. Clin Exp Allergy 2015; 45:21-31. [PMID: 24766371 PMCID: PMC4298800 DOI: 10.1111/cea.12327] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
An increasing proportion of the worldwide population is affected by allergic diseases such as allergic rhinitis (AR), atopic dermatitis (AD) and allergic asthma and improved treatment options are needed particularly for severe, refractory disease. Allergic diseases are complex and development involves both environmental and genetic factors. Although the existence of a genetic component for allergy was first described almost 100 years ago, progress in gene identification has been hindered by lack of high throughput technologies to investigate genetic variation in large numbers of subjects. The development of Genome-Wide Association Studies (GWAS), a hypothesis-free method of interrogating large numbers of common variants spanning the entire genome in disease and non-disease subjects has revolutionised our understanding of the genetics of allergic disease. Susceptibility genes for asthma, AR and AD have now been identified with confidence, suggesting there are common and distinct genetic loci associated with these diseases, providing novel insights into potential disease pathways and mechanisms. Genes involved in both adaptive and innate immune mechanisms have been identified, notably including multiple genes involved in epithelial function/secretion, suggesting that the airway epithelium may be particularly important in asthma. Interestingly, concordance/discordance between the genetic factors driving allergic traits such as IgE levels and disease states such as asthma have further supported the accumulating evidence for heterogeneity in these diseases. While GWAS have been useful and continue to identify novel genes for allergic diseases through increased sample sizes and phenotype refinement, future approaches will integrate analyses of rare variants, epigenetic mechanisms and eQTL approaches, leading to greater insight into the genetic basis of these diseases. Gene identification will improve our understanding of disease mechanisms and generate potential therapeutic opportunities.
Collapse
Affiliation(s)
- M A Portelli
- Division of Respiratory Medicine, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
30
|
Abstract
Over the past three decades, a large number of genetic studies have been aimed at finding genetic variants associated with the risk of asthma, applying various genetic and genomic approaches including linkage analysis, candidate gene polymorphism studies, and genome-wide association studies (GWAS). However, contrary to general expectation, even single nucleotide polymorphisms (SNPs) discovered by GWAS failed to fully explain the heritability of asthma. Thus, application of rare allele polymorphisms in well defined phenotypes and clarification of environmental factors have been suggested to overcome the problem of 'missing' heritability. Such factors include allergens, cigarette smoke, air pollutants, and infectious agents during pre- and post-natal periods. The first and simplest interaction between a gene and the environment is a candidate interaction of both a well known gene and environmental factor in a direct physical or chemical interaction such as between CD14 and endotoxin or between HLA and allergens. Several GWAS have found environmental interactions with occupational asthma, aspirin exacerbated respiratory disease, tobacco smoke-related airway dysfunction, and farm-related atopic diseases. As one of the mechanisms behind gene-environment interaction is epigenetics, a few studies on DNA CpG methylation have been reported on subphenotypes of asthma, pitching the exciting idea that it may be possible to intervene at the junction between the genome and the environment. Epigenetic studies are starting to include data from clinical samples, which will make them another powerful tool for re-search on gene-environment interactions in asthma.
Collapse
Affiliation(s)
- Jong-Uk Lee
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Korea
| | - Jeong Dong Kim
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Korea
| | - Choon-Sik Park
- Genome Research Center and Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| |
Collapse
|
31
|
Pino-Yanes M, Gignoux CR, Galanter JM, Levin AM, Campbell CD, Eng C, Huntsman S, Nishimura KK, Gourraud PA, Mohajeri K, O'Roak BJ, Hu D, Mathias RA, Nguyen EA, Roth LA, Padhukasahasram B, Moreno-Estrada A, Sandoval K, Winkler CA, Lurmann F, Davis A, Farber HJ, Meade K, Avila PC, Serebrisky D, Chapela R, Ford JG, Lenoir MA, Thyne SM, Brigino-Buenaventura E, Borrell LN, Rodriguez-Cintron W, Sen S, Kumar R, Rodriguez-Santana JR, Bustamante CD, Martinez FD, Raby BA, Weiss ST, Nicolae DL, Ober C, Meyers DA, Bleecker ER, Mack SJ, Hernandez RD, Eichler EE, Barnes KC, Williams LK, Torgerson DG, Burchard EG. Genome-wide association study and admixture mapping reveal new loci associated with total IgE levels in Latinos. J Allergy Clin Immunol 2015; 135:1502-10. [PMID: 25488688 PMCID: PMC4458233 DOI: 10.1016/j.jaci.2014.10.033] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 09/06/2014] [Accepted: 10/15/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND IgE is a key mediator of allergic inflammation, and its levels are frequently increased in patients with allergic disorders. OBJECTIVE We sought to identify genetic variants associated with IgE levels in Latinos. METHODS We performed a genome-wide association study and admixture mapping of total IgE levels in 3334 Latinos from the Genes-environments & Admixture in Latino Americans (GALA II) study. Replication was evaluated in 454 Latinos, 1564 European Americans, and 3187 African Americans from independent studies. RESULTS We confirmed associations of 6 genes identified by means of previous genome-wide association studies and identified a novel genome-wide significant association of a polymorphism in the zinc finger protein 365 gene (ZNF365) with total IgE levels (rs200076616, P = 2.3 × 10(-8)). We next identified 4 admixture mapping peaks (6p21.32-p22.1, 13p22-31, 14q23.2, and 22q13.1) at which local African, European, and/or Native American ancestry was significantly associated with IgE levels. The most significant peak was 6p21.32-p22.1, where Native American ancestry was associated with lower IgE levels (P = 4.95 × 10(-8)). All but 22q13.1 were replicated in an independent sample of Latinos, and 2 of the peaks were replicated in African Americans (6p21.32-p22.1 and 14q23.2). Fine mapping of 6p21.32-p22.1 identified 6 genome-wide significant single nucleotide polymorphisms in Latinos, 2 of which replicated in European Americans. Another single nucleotide polymorphism was peak-wide significant within 14q23.2 in African Americans (rs1741099, P = 3.7 × 10(-6)) and replicated in non-African American samples (P = .011). CONCLUSION We confirmed genetic associations at 6 genes and identified novel associations within ZNF365, HLA-DQA1, and 14q23.2. Our results highlight the importance of studying diverse multiethnic populations to uncover novel loci associated with total IgE levels.
Collapse
Affiliation(s)
- Maria Pino-Yanes
- Department of Medicine, University of California, San Francisco, Calif; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| | - Christopher R Gignoux
- Department of Medicine, University of California, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Calif; Department of Genetics, Stanford University, Palo Alto, Calif
| | - Joshua M Galanter
- Department of Medicine, University of California, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Calif
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, Mich
| | | | - Celeste Eng
- Department of Medicine, University of California, San Francisco, Calif
| | - Scott Huntsman
- Department of Medicine, University of California, San Francisco, Calif
| | | | | | - Kiana Mohajeri
- Department of Genome Sciences, University of Washington, Seattle, Wash
| | - Brian J O'Roak
- Department of Genome Sciences, University of Washington, Seattle, Wash; Molecular & Medical Genetics Department, Oregon Health and Science University, Portland, Ore
| | - Donglei Hu
- Department of Medicine, University of California, San Francisco, Calif
| | - Rasika A Mathias
- Division of Allergy & Clinical Immunology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | | | - Lindsey A Roth
- Department of Medicine, University of California, San Francisco, Calif
| | - Badri Padhukasahasram
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Mich
| | | | - Karla Sandoval
- Department of Genetics, Stanford University, Palo Alto, Calif
| | - Cheryl A Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical, Frederick National Laboratory for Cancer Research, Frederick, Md
| | | | - Adam Davis
- Children's Hospital and Research Center Oakland, Oakland, Calif
| | - Harold J Farber
- Department of Pediatrics, Section of Pulmonology, Baylor College of Medicine and Texas Children's Hospital, Houston, Tex
| | - Kelley Meade
- Children's Hospital and Research Center Oakland, Oakland, Calif
| | - Pedro C Avila
- Department of Medicine, Northwestern University, Chicago, Ill
| | | | - Rocio Chapela
- Instituto Nacional de Enfermedades Respiratorias (INER), Mexico City, Mexico
| | - Jean G Ford
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Md
| | | | - Shannon M Thyne
- Department of Pediatrics, University of California San Francisco, San Francisco General Hospital, San Francisco, Calif
| | | | - Luisa N Borrell
- Department of Health Sciences, Graduate Program in Public Health, City University of New York, Bronx, NY
| | | | - Saunak Sen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, Calif
| | - Rajesh Kumar
- Children's Memorial Hospital and the Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | | | | | - Fernando D Martinez
- Arizona Respiratory Center, University of Arizona, Tucson, Ariz; BIO5 Institute, University of Arizona, Tucson, Ariz
| | - Benjamin A Raby
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Dan L Nicolae
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Deborah A Meyers
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
| | - Eugene R Bleecker
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC
| | - Steven J Mack
- Children's Hospital Oakland Research Institute, Oakland, Calif
| | - Ryan D Hernandez
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Calif
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington, Seattle, Wash; Howard Hughes Medical Institute, Seattle, Wash
| | - Kathleen C Barnes
- Division of Allergy & Clinical Immunology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - L Keoki Williams
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Mich; Department of Internal Medicine, Henry Ford Health System, Detroit, Mich
| | - Dara G Torgerson
- Department of Medicine, University of California, San Francisco, Calif
| | - Esteban G Burchard
- Department of Medicine, University of California, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, Calif
| |
Collapse
|
32
|
Zhou JC, Zhu YM, Chen Z, He S, Zheng SJ, Mo JL, Liu XL, Gong CM, Hou B, Yang H. Association of IgE-mediated allergen sensitivity and promoter polymorphisms of chemokine (C–C motif) ligand 5 gene in Han Chinese patients with allergic skin diseases. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0274-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Liang L, Willis-Owen SAG, Laprise C, Wong KCC, Davies GA, Hudson TJ, Binia A, Hopkin JM, Yang IV, Grundberg E, Busche S, Hudson M, Rönnblom L, Pastinen TM, Schwartz DA, Lathrop GM, Moffatt MF, Cookson WOCM. An epigenome-wide association study of total serum immunoglobulin E concentration. Nature 2015; 520:670-674. [PMID: 25707804 PMCID: PMC4416961 DOI: 10.1038/nature14125] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 11/27/2014] [Indexed: 12/19/2022]
Abstract
Immunoglobulin E (IgE) is a central mediator of allergic (atopic) inflammation. Therapies directed against IgE can alleviate hay fever and allergic asthma. Genetic association studies have not yet identified novel therapeutic targets or pathways underlying IgE regulation. We therefore surveyed epigenetic associations between serum IgE concentrations and methylation at loci concentrated in CpG islands genome wide in 95 nuclear pedigrees, using DNA from peripheral blood leukocytes. We validated positive results in additional families and in subjects from the general population. Here we show replicated associations--with a meta-analysis false discovery rate less than 10(-4)--between IgE and low methylation at 36 loci. Genes annotated to these loci encode known eosinophil products, and also implicate phospholipid inflammatory mediators, specific transcription factors and mitochondrial proteins. We confirmed that methylation at these loci differed significantly in isolated eosinophils from subjects with and without asthma and high IgE levels. The top three loci accounted for 13% of IgE variation in the primary subject panel, explaining the tenfold higher variance found compared with that derived from large single-nucleotide polymorphism genome-wide association studies. This study identifies novel therapeutic targets and biomarkers for patient stratification for allergic diseases.
Collapse
Affiliation(s)
- Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, MA 02115
| | | | | | - Kenny C C Wong
- National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
| | - Gwyneth A Davies
- Institute of Life Science, College of Medicine, Swansea University, SA2 8PP, UK
| | - Thomas J Hudson
- Ontario Institute for Cancer Research, Toronto, Ontario Canada, M5G 0A3
- Departments of Medical Biophysics and Molecular Genetics, University of Toronto, Canada ON M5S 1A1
| | - Aristea Binia
- National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
| | - Julian M Hopkin
- Institute of Life Science, College of Medicine, Swansea University, SA2 8PP, UK
| | - Ivana V Yang
- University of Colorado School of Medicine and National Jewish Health, Denver, CO 80206
| | - Elin Grundberg
- Department of Human Genetics, McGill University and Génome Québec Innovation Centre, Montréal, Canada
| | - Stephan Busche
- Department of Human Genetics, McGill University and Génome Québec Innovation Centre, Montréal, Canada
| | - Marie Hudson
- Jewish General Hospital and Lady Davis Research Institute, Montréal, Canada H3T 1E2
| | - Lars Rönnblom
- Department of Medical Sciences, SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Tomi M Pastinen
- Department of Human Genetics, McGill University and Génome Québec Innovation Centre, Montréal, Canada
- Department of Medical Genetics, McGill University Health Centre, Montréal, Canada
| | - David A Schwartz
- University of Colorado School of Medicine and National Jewish Health, Denver, CO 80206
| | - G Mark Lathrop
- Department of Human Genetics, McGill University and Génome Québec Innovation Centre, Montréal, Canada
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College, London SW3 6LY, UK
| | | |
Collapse
|
34
|
Kim KW, Myers RA, Lee JH, Igartua C, Lee KE, Kim YH, Kim EJ, Yoon D, Lee JS, Hirota T, Tamari M, Takahashi A, Kubo M, Choi JM, Kim KE, Nicolae DL, Ober C, Sohn MH. Genome-wide association study of recalcitrant atopic dermatitis in Korean children. J Allergy Clin Immunol 2015; 136:678-684.e4. [PMID: 25935106 DOI: 10.1016/j.jaci.2015.03.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 03/05/2015] [Accepted: 03/13/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a heterogeneous chronic inflammatory skin disease. Most AD during infancy resolves during childhood, but moderate-to-severe AD with allergic sensitization is more likely to persist into adulthood and more often occurs with other allergic diseases. OBJECTIVE We sought to find susceptibility loci by performing the first genome-wide association study (GWAS) of AD in Korean children with recalcitrant AD, which was defined as moderate-to-severe AD with allergic sensitization. METHODS Our study included 246 children with recalcitrant AD and 551 adult control subjects with a negative history of both allergic disease and allergic sensitization. DNA from these subjects was genotyped; sets of common single nucleotide polymorphisms (SNPs) were imputed and used in the GWAS after quality control checks. RESULTS SNPs at a region on 13q21.31 were associated with recalcitrant AD at a genome-wide threshold of significance (P < 2.0 × 10(-8)). These associated SNPs are more than 1 Mb from the closest gene, protocadherin (PCDH)9. SNPs at 4 additional loci had P values of less than 1 × 10(-6), including SNPs at or near the neuroblastoma amplified sequence (NBAS; 2p24.3), thymus-expressed molecule involved in selection (THEMIS; 6q22.33), GATA3 (10p14), and S-phase cyclin A-associated protein in the ER (SCAPER; 15q24.3) genes. Further analysis of total serum IgE levels suggested 13q21.31 might be primarily an IgE locus, and analyses of published data demonstrated that SNPs at the 15q24.3 region are expression quantitative trait loci for 2 nearby genes, ISL2 and proline-serine-threonine phosphatase interacting protein 1 (PSTPIP1), in immune cells. CONCLUSION Our GWAS of recalcitrant AD identified new susceptibility regions containing genes involved in epithelial cell function and immune dysregulation, 2 key features of AD, and potentially extend our understanding of their role in pathogenesis.
Collapse
Affiliation(s)
- Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea; Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Rachel A Myers
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Ji Hyun Lee
- Department of Oral Biology, Yonsei University College of Dentistry, Seoul, Korea
| | | | - Kyung Eun Lee
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Hee Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Eun-Jin Kim
- Division of Allergy and Chronic Respiratory Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Korea
| | - Dankyu Yoon
- Division of Allergy and Chronic Respiratory Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Korea
| | - Joo-Shil Lee
- Division of Allergy and Chronic Respiratory Diseases, Center for Biomedical Sciences, Korea National Institute of Health, Osong, Korea
| | - Tomomitsu Hirota
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Mayumi Tamari
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Tokyo, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Je-Min Choi
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul, Korea
| | - Kyu-Earn Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Dan L Nicolae
- Department of Human Genetics, University of Chicago, Chicago, Ill; Department of Medicine and Statistics, University of Chicago, Chicago, Ill
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
35
|
New Developments in Biomarkers for Atopic Dermatitis. J Clin Med 2015; 4:479-87. [PMID: 26239250 PMCID: PMC4470140 DOI: 10.3390/jcm4030479] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/09/2015] [Accepted: 02/24/2015] [Indexed: 12/24/2022] Open
Abstract
The application of biomarkers in medicine is evolving. Biomarkers do not only give us a better understanding of pathogenesis, but also increase treatment efficacy and safety, further enabling more precise clinical care. This paper focuses on the current use of biomarkers in atopic dermatitis, new developments and future perspectives. Biomarkers can be used for many different purposes, including the objective determination of disease severity, confirmation of clinical diagnosis, and to predict response to treatment. In atopic dermatitis, many biomarkers have been investigated as a marker for disease severity. Currently serum thymus and activation-regulated chemokine (TARC) is the superior biomarker for assessing disease severity. However, we have recently shown that the use of a panel of serum biomarkers is more suitable for assessing disease severity than an individual biomarker. In this overview, we will discuss alternative sources for biomarkers, such as saliva and capillary blood, which can increase the user friendliness of biomarkers in atopic dermatitis (AD). Both methods offer simple, non-invasive and cost effective alternatives to venous blood. This provides great translational and clinical potential. Biomarkers will play an increasingly important role in AD research and personalized medicine. The use of biomarkers will enhance the efficacy of AD treatment by facilitating the individualization of therapy targeting the patients' specific biological signature and also by providing tools for predicting and monitoring of therapeutic response.
Collapse
|
36
|
Peprah E, Xu H, Tekola-Ayele F, Royal CD. Genome-wide association studies in Africans and African Americans: expanding the framework of the genomics of human traits and disease. Public Health Genomics 2014; 18:40-51. [PMID: 25427668 DOI: 10.1159/000367962] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 08/29/2014] [Indexed: 01/11/2023] Open
Abstract
Genomic research is one of the tools for elucidating the pathogenesis of diseases of global health relevance and paving the research dimension to clinical and public health translation. Recent advances in genomic research and technologies have increased our understanding of human diseases, genes associated with these disorders, and the relevant mechanisms. Genome-wide association studies (GWAS) have proliferated since the first studies were published several years ago and have become an important tool in helping researchers comprehend human variation and the role genetic variants play in disease. However, the need to expand the diversity of populations in GWAS has become increasingly apparent as new knowledge is gained about genetic variation. Inclusion of diverse populations in genomic studies is critical to a more complete understanding of human variation and elucidation of the underpinnings of complex diseases. In this review, we summarize the available data on GWAS in recent African ancestry populations within the western hemisphere (i.e. African Americans and peoples of the Caribbean) and continental African populations. Furthermore, we highlight ways in which genomic studies in populations of recent African ancestry have led to advances in the areas of malaria, HIV, prostate cancer, and other diseases. Finally, we discuss the advantages of conducting GWAS in recent African ancestry populations in the context of addressing existing and emerging global health conditions.
Collapse
|
37
|
Abstract
Genome-wide association studies (GWAS) have been employed in the field of allergic disease, and significant associations have been published for nearly 100 asthma genes/loci. An outcome of GWAS in allergic disease has been the formation of national and international collaborations leading to consortia meta-analyses, and an appreciation for the specificity of genetic associations to sub-phenotypes of allergic disease. Molecular genetics has undergone a technological revolution, leading to next-generation sequencing strategies that are increasingly employed to hone in on the causal variants associated with allergic diseases. Unmet needs include the inclusion of diverse cohorts and strategies for managing big data.
Collapse
Affiliation(s)
- Romina A Ortiz
- Department of Medicine, The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3A.62, Baltimore, MD 21224, USA
| | - Kathleen C Barnes
- Department of Medicine, The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, Room 3A.62, Baltimore, MD 21224, USA.
| |
Collapse
|
38
|
Myers RA, Scott NM, Gauderman WJ, Qiu W, Mathias RA, Romieu I, Levin AM, Pino-Yanes M, Graves PE, Villarreal AB, Beaty TH, Carey VJ, Croteau-Chonka DC, del Rio Navarro B, Edlund C, Hernandez-Cadena L, Navarro-Olivos E, Padhukasahasram B, Salam MT, Torgerson DG, Van den Berg DJ, Vora H, Bleecker ER, Meyers DA, Williams LK, Martinez FD, Burchard EG, Barnes KC, Gilliland FD, Weiss ST, London SJ, Raby BA, Ober C, Nicolae DL. Genome-wide interaction studies reveal sex-specific asthma risk alleles. Hum Mol Genet 2014; 23:5251-9. [PMID: 24824216 PMCID: PMC4159149 DOI: 10.1093/hmg/ddu222] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 04/09/2014] [Accepted: 05/06/2014] [Indexed: 11/14/2022] Open
Abstract
Asthma is a complex disease with sex-specific differences in prevalence. Candidate gene studies have suggested that genotype-by-sex interaction effects on asthma risk exist, but this has not yet been explored at a genome-wide level. We aimed to identify sex-specific asthma risk alleles by performing a genome-wide scan for genotype-by-sex interactions in the ethnically diverse participants in the EVE Asthma Genetics Consortium. We performed male- and female-specific genome-wide association studies in 2653 male asthma cases, 2566 female asthma cases and 3830 non-asthma controls from European American, African American, African Caribbean and Latino populations. Association tests were conducted in each study sample, and the results were combined in ancestry-specific and cross-ancestry meta-analyses. Six sex-specific asthma risk loci had P-values < 1 × 10(-6), of which two were male specific and four were female specific; all were ancestry specific. The most significant sex-specific association in European Americans was at the interferon regulatory factor 1 (IRF1) locus on 5q31.1. We also identify a Latino female-specific association in RAP1GAP2. Both of these loci included single-nucleotide polymorphisms that are known expression quantitative trait loci and have been associated with asthma in independent studies. The IRF1 locus is a strong candidate region for male-specific asthma susceptibility due to the association and validation we demonstrate here, the known role of IRF1 in asthma-relevant immune pathways and prior reports of sex-specific differences in interferon responses.
Collapse
Affiliation(s)
| | | | - W James Gauderman
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Weiliang Qiu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rasika A Mathias
- Department of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | - Maria Pino-Yanes
- Department of Medicine and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Penelope E Graves
- BIO5 Institute, Arizona Respiratory Care Center, University of Arizona, Tucson, AZ 85721, USA
| | | | - Terri H Beaty
- Department of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vincent J Carey
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Damien C Croteau-Chonka
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Christopher Edlund
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | | | | | | | - Muhammad T Salam
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Dara G Torgerson
- Department of Medicine and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA
| | - David J Van den Berg
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Hita Vora
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Eugene R Bleecker
- Center for Genomics and Personalized Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Deborah A Meyers
- Center for Genomics and Personalized Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - L Keoki Williams
- Center for Health Policy and Health Services Research Department of Internal Medicine, Henry Ford Health System, Detroit, MI 48202, USA
| | - Fernando D Martinez
- BIO5 Institute, Arizona Respiratory Care Center, University of Arizona, Tucson, AZ 85721, USA
| | - Esteban G Burchard
- Department of Medicine and Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA
| | - Kathleen C Barnes
- Department of Medicine and Epidemiology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Frank D Gilliland
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie J London
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | - Benjamin A Raby
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Dan L Nicolae
- Department of Human Genetics Department of Statistics and Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
39
|
Sharma V, Michel S, Gaertner V, Franke A, Vogelberg C, von Berg A, Bufe A, Heinzmann A, Laub O, Rietschel E, Simma B, Frischer T, Genuneit J, Zeilinger S, Illig T, Schedel M, Potaczek DP, Kabesch M. Fine-mapping of IgE-associated loci 1q23, 5q31, and 12q13 using 1000 Genomes Project data. Allergy 2014; 69:1077-84. [PMID: 24930997 DOI: 10.1111/all.12431] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Genome-wide association studies (GWAS) repeatedly identified 1q23 (FCER1A), 5q31 (RAD50-IL13 and IL4), and 12q13 (STAT6) as major susceptibility loci influencing the regulation of total serum IgE levels. As GWAS may be insufficient to capture causal variants, we performed fine-mapping and re-genotyping of the three loci using 1000 Genomes Project datasets. METHODS Linkage disequilibrium tagging polymorphisms and polymorphisms of putative functional relevance were genotyped by chip technology (24 polymorphisms) or MALDI-TOF-MS (40 polymorphisms) in at least 1303 German children (651 asthmatics). The effect of polymorphisms on total serum IgE, IgE percentiles, and atopic diseases was assessed, and a risk score model was applied for gene-by-gene interaction analyses. Functional effects of putative causal variants from these three loci were studied in silico. RESULTS Associations from GWAS were confirmed and extended. For 1q23 and 5q31, the majority of associations were found with mild to moderately elevated IgE levels, while in the 12q13 locus, single-nucleotide polymorphisms (SNPs) were associated with strongly elevated IgE levels. Gene-by-gene interaction analyses suggested that the presence of mutations in all three loci increases the risk for elevated IgE up to fourfold. CONCLUSION This fine-mapping study confirmed previous associations and identified novel associations of SNPs in 1q23, 5q31, and 12q13 with different levels of serum IgE and their concomitant contribution to IgE regulation.
Collapse
Affiliation(s)
- V. Sharma
- Department of Pediatric Pneumology, Allergy and Neonatology; Hannover Medical School; Hannover Germany
| | - S. Michel
- Department of Pediatric Pneumology, Allergy and Neonatology; Hannover Medical School; Hannover Germany
- Department of Pediatric Pneumology and Allergy; University Children's Hospital Regensburg (KUNO); Regensburg Germany
| | - V. Gaertner
- Department of Pediatric Pneumology and Allergy; University Children's Hospital Regensburg (KUNO); Regensburg Germany
| | - A. Franke
- Institute of Clinical Molecular Biology; Christian-Albrechts-University Kiel; Kiel Germany
| | - C. Vogelberg
- University Children's Hospital; Technical University Dresden; Dresden Germany
| | - A. von Berg
- Children's Department; Research Institute for the Prevention of Allergic Diseases; Marien-Hospital; Wesel Germany
| | - A. Bufe
- Department of Experimental Pneumology; Ruhr-University; Bochum Germany
| | - A. Heinzmann
- University Children's Hospital; Albert Ludwigs University; Freiburg Germany
| | - O. Laub
- Kinder-und Jugendarztpraxis Laub; Rosenheim Germany
| | - E. Rietschel
- University Children's Hospital; University of Cologne; Cologne Germany
| | - B. Simma
- Children's Department; University Teaching Hospital; Landeskrankenhaus Feldkirch; Feldkirch Austria
| | - T. Frischer
- University Children's Hospital Vienna; Vienna Austria
| | - J. Genuneit
- Institute of Epidemiology and Medical Biometry; Ulm University; Ulm Germany
| | - S. Zeilinger
- Research Unit of Molecular Epidemiology; Helmholtz Zentrum Munich; Neuherberg Germany
| | - T. Illig
- Research Unit of Molecular Epidemiology; Helmholtz Zentrum Munich; Neuherberg Germany
- Hannover Unified Biobank; Hannover Medical School; Hannover Germany
| | - M. Schedel
- Division of Cell Biology; Department of Pediatrics; National Jewish Health; Denver CO USA
| | - D. P. Potaczek
- Department of Pediatric Pneumology, Allergy and Neonatology; Hannover Medical School; Hannover Germany
- Institute of Laboratory Medicine; Philipps-Universität Marburg; Marburg Germany
| | - M. Kabesch
- Department of Pediatric Pneumology, Allergy and Neonatology; Hannover Medical School; Hannover Germany
- Department of Pediatric Pneumology and Allergy; University Children's Hospital Regensburg (KUNO); Regensburg Germany
- German Lung Research Center (DZL)
| |
Collapse
|
40
|
Smit LAM, Strachan DP, Vermeulen R, de Bakker PIW, Demenais F, Dumas O, Carsin AE, Cullinan P, Curjuric I, Ghosh RE, Heederik D, Imboden M, Jarvis D, Lathrop M, Le Moual N, Mehta A, Miedinger D, Sigsgaard T, Siroux V, Vernez D, Zock JP, Kauffmann F, Probst-Hensch N, Kogevinas M, Bouzigon E. Human leukocyte antigen class II variants and adult-onset asthma: does occupational allergen exposure play a role? Eur Respir J 2014; 44:1234-42. [PMID: 25034568 DOI: 10.1183/09031936.00068014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recently, a locus centred on rs9273349 in the HLA-DQ region emerged from genome-wide association studies of adult-onset asthma. We aimed to further investigate the role of human leukocyte antigen (HLA) class II in adult-onset asthma and a possible interaction with occupational exposures. We imputed classical HLA-II alleles from 7579 single-nucleotide polymorphisms in 6025 subjects (1202 with adult-onset asthma) from European cohorts: ECRHS, SAPALDIA, EGEA and B58C, and from surveys of bakers and agricultural workers. Based on an asthma-specific job-exposure matrix, 2629 subjects had ever been exposed to high molecular weight (HMW) allergens. We explored associations between 23 common HLA-II alleles and adult-onset asthma, and tested for gene-environment interaction with occupational exposure to HMW allergens. Interaction was also tested for rs9273349. Marginal associations of classical HLA-II alleles and adult-onset asthma were not statistically significant. Interaction was detected between the DPB1*03:01 allele and exposure to HMW allergens (p = 0.009), in particular to latex (p = 0.01). In the unexposed group, the DPB1*03:01 allele was associated with adult-onset asthma (OR 0.67, 95%CI 0.53-0.86). HMW allergen exposures did not modify the association of rs9273349 with adult-onset asthma. Common classical HLA-II alleles were not marginally associated with adult-onset asthma. The association of latex exposure and adult-onset asthma may be modified by DPB1*03:01.
Collapse
Affiliation(s)
| | | | - Roel Vermeulen
- For the authors' affiliations see the Acknowledgements section
| | | | | | - Orianne Dumas
- For the authors' affiliations see the Acknowledgements section
| | | | - Paul Cullinan
- For the authors' affiliations see the Acknowledgements section
| | - Ivan Curjuric
- For the authors' affiliations see the Acknowledgements section
| | - Rebecca E Ghosh
- For the authors' affiliations see the Acknowledgements section
| | - Dick Heederik
- For the authors' affiliations see the Acknowledgements section
| | - Medea Imboden
- For the authors' affiliations see the Acknowledgements section
| | - Deborah Jarvis
- For the authors' affiliations see the Acknowledgements section
| | - Mark Lathrop
- For the authors' affiliations see the Acknowledgements section
| | - Nicole Le Moual
- For the authors' affiliations see the Acknowledgements section
| | - Amar Mehta
- For the authors' affiliations see the Acknowledgements section
| | - David Miedinger
- For the authors' affiliations see the Acknowledgements section
| | | | - Valérie Siroux
- For the authors' affiliations see the Acknowledgements section
| | - David Vernez
- For the authors' affiliations see the Acknowledgements section
| | - Jan Paul Zock
- For the authors' affiliations see the Acknowledgements section
| | | | | | | | | |
Collapse
|
41
|
Genome-wide association studies in asthma; perhaps, the end of the beginning. Curr Opin Allergy Clin Immunol 2014; 13:463-9. [PMID: 23945178 DOI: 10.1097/aci.0b013e328364ea5f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW A large number of genetic loci contribute towards an individual's susceptibility to asthma and other complex diseases. Genome-wide association studies (GWASs) have provided us with a wealth of loci associated with asthma susceptibility, asthma endotypes and responsiveness to asthma medications. The reproducibility of these genetic loci across different studies highlights the interplay of general and population-specific risk alleles in asthma. Although GWASs have been successful in identifying disease-associated loci, there is still large potential for such studies to provide further insights into asthma pathogenesis. RECENT FINDINGS GWASs over the past year have extended study design well beyond the simple case-control and continuous phenotype association formats, for example, including interactions with environmental factors, integrating GWAS data with epigenetic data and GWASs in animal models, incorporating pathway analyses and utilising emerging sequencing technologies. SUMMARY Moving beyond traditional GWAS formats is likely to significantly enhance our understanding of the genetic basis for asthma. This review discusses where we are after half a decade of asthma GWASs, and focuses on advances over the past year that show where the GWAS field is headed in the future.
Collapse
|
42
|
Viktorin A, Frankowiack M, Padyukov L, Chang Z, Melén E, Sääf A, Kull I, Klareskog L, Hammarström L, Magnusson PKE. IgA measurements in over 12 000 Swedish twins reveal sex differential heritability and regulatory locus near CD30L. Hum Mol Genet 2014; 23:4177-84. [PMID: 24676358 DOI: 10.1093/hmg/ddu135] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In a broad attempt to improve the understanding of the genetic regulation of serum IgA levels, the heritability was estimated in over 12 000 Swedish twins, and a genome-wide association study was conducted in a subsample of 9617. Using the classical twin model the heritability was found to be significantly larger among females (61%) compared with males (21%), while contribution from shared environment (20%) was only seen for males. By modeling the genetic relationship matrix with IgA levels, we estimate that a substantial proportion (31%) of variance in IgA levels can ultimately be explained by the investigated SNPs. The genome-wide association study revealed significant association to two loci: (i) rs6928791 located on chromosome 6, 22 kb upstream of the gene SAM and SH3 domain containing 1 (SASH1) and (ii) rs13300483 on chromosome 9, situated 12 kb downstream the CD30 ligand (CD30L) encoding gene. The association to rs13300483 was replicated in two additional independent Swedish materials. The heritability of IgA levels is moderate and can partly be attributable to common variation in the CD30L locus.
Collapse
Affiliation(s)
| | | | | | - Zheng Chang
- Department of Medical Epidemiology and Biostatistics
| | - Erik Melén
- Institute of Environmental Medicine and Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden Sachś Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Annika Sääf
- Institute of Environmental Medicine and Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Inger Kull
- Institute of Environmental Medicine and Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden Sachś Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | | | | | | |
Collapse
|
43
|
Tamari M, Hirota T. Genome-wide association studies of atopic dermatitis. J Dermatol 2014; 41:213-20. [DOI: 10.1111/1346-8138.12321] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 09/19/2013] [Indexed: 12/23/2022]
Affiliation(s)
- Mayumi Tamari
- Laboratory for Respiratory and Allergic Diseases; Center for Integrative Medical Sciences; The Institute of Physical and Chemical Research (RIKEN); Kanagawa Japan
| | - Tomomitsu Hirota
- Laboratory for Respiratory and Allergic Diseases; Center for Integrative Medical Sciences; The Institute of Physical and Chemical Research (RIKEN); Kanagawa Japan
| |
Collapse
|
44
|
Martins TB, Bandhauer ME, Bunker AM, Roberts WL, Hill HR. New childhood and adult reference intervals for total IgE. J Allergy Clin Immunol 2014; 133:589-91. [DOI: 10.1016/j.jaci.2013.08.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 08/13/2013] [Accepted: 08/28/2013] [Indexed: 11/25/2022]
|
45
|
Laing B, Han DY, Ferguson LR. Candidate genes involved in beneficial or adverse responses to commonly eaten brassica vegetables in a New Zealand Crohn's disease cohort. Nutrients 2013; 5:5046-64. [PMID: 24352087 PMCID: PMC3875924 DOI: 10.3390/nu5125046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 11/22/2013] [Accepted: 11/29/2013] [Indexed: 12/20/2022] Open
Abstract
Crohn’s disease (CD) is one of the two manifestations of inflammatory bowel disease. Particular foods are thought with CD to exacerbate their illness. Vegetables, especially Brassicaceae, are often shunned by people with CD because of the negative effects they are alleged to have on their symptoms. Brassicaceae supply key nutrients which are necessary to meet recommended daily intakes. We sought to identify the candidate genes involved in the beneficial or adverse effects of Brassicaceae most commonly eaten, as reported by the New Zealand adults from the “Genes and Diet in Inflammatory Bowel disease Study” based in Auckland. An analysis of associations between the single nucleotide polymorphisms (SNPs) and the beneficial or adverse effects of the ten most commonly eaten Brassicaceae was carried out. A total of 37 SNPs were significantly associated with beneficial effects (p = 0.00097 to 0.0497) and 64 SNPs were identified with adverse effects (p = 0.0000751 to 0.049). After correcting for multiple testing, rs7515322 (DIO1) and rs9469220 (HLA) remained significant. Our findings show that the tolerance of some varieties of Brassicaceae may be shown by analysis of a person’s genotype.
Collapse
Affiliation(s)
- Bobbi Laing
- Discipline of Nutrition, School of Medical Sciences, Auckland University, 85 Park Road, Grafton Campus, Auckland 1142, New Zealand.
| | | | | |
Collapse
|
46
|
Yatagai Y, Sakamoto T, Masuko H, Kaneko Y, Yamada H, Iijima H, Naito T, Noguchi E, Hirota T, Tamari M, Imoto Y, Tokunaga T, Fujieda S, Konno S, Nishimura M, Hizawa N. Genome-wide association study for levels of total serum IgE identifies HLA-C in a Japanese population. PLoS One 2013; 8:e80941. [PMID: 24324648 PMCID: PMC3851760 DOI: 10.1371/journal.pone.0080941] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 10/07/2013] [Indexed: 12/11/2022] Open
Abstract
Most of the previously reported loci for total immunoglobulin E (IgE) levels are related to Th2 cell-dependent pathways. We undertook a genome-wide association study (GWAS) to identify genetic loci responsible for IgE regulation. A total of 479,940 single nucleotide polymorphisms (SNPs) were tested for association with total serum IgE levels in 1180 Japanese adults. Fine-mapping with SNP imputation demonstrated 6 candidate regions: the PYHIN1/IFI16, MHC classes I and II, LEMD2, GRAMD1B, and chr13∶60576338 regions. Replication of these candidate loci in each region was assessed in 2 independent Japanese cohorts (n = 1110 and 1364, respectively). SNP rs3130941 in the HLA-C region was consistently associated with total IgE levels in 3 independent populations, and the meta-analysis yielded genome-wide significance (P = 1.07×10−10). Using our GWAS results, we also assessed the reproducibility of previously reported gene associations with total IgE levels. Nine of 32 candidate genes identified by a literature search were associated with total IgE levels after correction for multiple testing. Our findings demonstrate that SNPs in the HLA-C region are strongly associated with total serum IgE levels in the Japanese population and that some of the previously reported genetic associations are replicated across ethnic groups.
Collapse
Affiliation(s)
- Yohei Yatagai
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tohru Sakamoto
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
- * E-mail:
| | - Hironori Masuko
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yoshiko Kaneko
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hideyasu Yamada
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | | | | | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomomitsu Hirota
- Laboratory for Respiratory Diseases, Center for Genomic Medicine, the Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan
| | - Mayumi Tamari
- Laboratory for Respiratory Diseases, Center for Genomic Medicine, the Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan
| | - Yoshimasa Imoto
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Fukui, Fukui, Japan
| | - Takahiro Tokunaga
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Fukui, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, University of Fukui, Fukui, Japan
| | - Satoshi Konno
- First Department of Medicine, School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Masaharu Nishimura
- First Department of Medicine, School of Medicine, Hokkaido University, Hokkaido, Japan
| | - Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
47
|
Kim JH, Cheong HS, Park JS, Jang AS, Uh ST, Kim YH, Kim MK, Choi IS, Cho SH, Choi BW, Bae JS, Park CS, Shin HD. A genome-wide association study of total serum and mite-specific IgEs in asthma patients. PLoS One 2013; 8:e71958. [PMID: 23967269 PMCID: PMC3742455 DOI: 10.1371/journal.pone.0071958] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/08/2013] [Indexed: 12/21/2022] Open
Abstract
Immunoglobulin E (IgE) is one of the central players in asthma and allergic diseases. Although the serum IgE level, a useful endophenotype, is generally increased in patients with asthma, genetic factors influencing IgE regulation in asthma are still not fully understood. To identify the genetic variations associated with total serum and mite-specific IgEs in asthmatics, a genome-wide association study (GWAS) of 657,366 single nucleotide polymorphisms (SNPs) was performed in 877 Korean asthmatics. This study found that several new genes might be associated with total IgE in asthmatics, such as CRIM1 (rs848512, P = 1.18×10−6; rs711254, P = 6.73×10−6), ZNF71 (rs10404342, P = 7.60×10−6), TLN1 (rs4879926, P = 7.74×10−6), and SYNPO2 (rs1472066, P = 8.36×10−6; rs1038770, P = 8.66×10−6). Regarding the association of specific IgE to house dust mites, it was observed that intergenic SNPs nearby to OPRK1 and LOC730217 might be associated with Dermatophagoides pteronyssinus (D.p.) and Dermatophagoides farinae (D.f.) in asthmatics, respectively. In further pathway analysis, the phosphatidylinositol signaling system and adherens junction pathways were estimated to play a role in the regulation of total IgE levels in asthma. Although functional evaluations and replications of these results in other populations are needed, this GWAS of serum IgE in asthmatics could facilitate improved understanding of the role of the newly identified genetic variants in asthma and its related phenotypes.
Collapse
Affiliation(s)
- Jeong-Hyun Kim
- Department of Life Science, Sogang University, Seoul, Republic of Korea
- Research Institute for Basic Science, Sogang University, Seoul, Republic of Korea
| | - Hyun Sub Cheong
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
| | - Jong Sook Park
- Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - An-Soo Jang
- Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Soo-Taek Uh
- Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Yong-Hoon Kim
- Division of Allergy and Respiratory Disease, Soonchunhyang University Cheonan Hospital, Cheonan, Republic of Korea
| | - Mi-Kyeong Kim
- Division of Internal Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Inseon S. Choi
- Department of Allergy, Chonnam National University, Gwangju, Republic of Korea
| | - Sang Heon Cho
- Department of Internal Medicine and Institute of Allergy and Clinical Immunology, Seoul National University, Seoul, Republic of Korea
| | - Byoung Whui Choi
- Department of Internal Medicine, Chung-Ang University Yongsan Hospital, Seoul, Republic of Korea
| | - Joon Seol Bae
- Department of Life Science, Sogang University, Seoul, Republic of Korea
| | - Choon-Sik Park
- Division of Allergy and Respiratory Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
- * E-mail: (HDS); (CSP)
| | - Hyoung Doo Shin
- Department of Life Science, Sogang University, Seoul, Republic of Korea
- Department of Genetic Epidemiology, SNP Genetics Inc., Seoul, Republic of Korea
- Research Institute for Basic Science, Sogang University, Seoul, Republic of Korea
- * E-mail: (HDS); (CSP)
| |
Collapse
|
48
|
Liao M, Shi D, Wang Y, Zhang K, Chen X, Gao Y, Tan A, Xuan Q, Yang X, Hu Y, Qin X, Zhang H, Mo Z. Genome-wide scan on total serum IgE levels identifies no common variants in a healthy Chinese male population. Immunogenetics 2013; 65:561-8. [PMID: 23661040 DOI: 10.1007/s00251-013-0706-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 04/18/2013] [Indexed: 12/30/2022]
Abstract
Immunoglobulin E (IgE) provides important information on the humoral immune status, and the IgE level is routinely detected in clinical practice. There are many diseases associated with IgE, such as atopic disease, autoimmune diseases, and so on. IgE is a genetically complex trait, but comprehensive genetic assessment of the variability in serum IgE levels is lacking. Previous genome-wide association studies (GWAS) on total serum IgE levels have identified FCER1A as the susceptibility locus; however, the candidate gene association study in southern Chinese patients reported no association. Given the genetic difference in different populations, we firstly conducted this two-stage GWAS in a Chinese population of 3,495 men, including 1,999 unrelated subjects in the first stage and 1,496 independent individuals replicated in the second stage. In the first stage, we totally identified three single nucleotide polymorphisms (SNPs) which reached a P value of 1.0 × 10⁻⁵. Rs17090302 on chromosome 3 and Rs28708846 on chromosome 13 are intergenic. Rs432085 from chromosome 3p28 is located in the gene CCDC50. When the two-stage data was combined, none of the SNPs reached the genome-wide significant level. Collectively, we did not identify novel loci associated with the serum IgE level in Chinese males, but we hypothesized that CCDC50 was a candidate gene in regulation on IgE level.
Collapse
Affiliation(s)
- Ming Liao
- Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, No. 22 Shuangyong Road, 530021, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|