1
|
Carewicz A, Michalska-Radź OA, Tomasiak-Łozowska MM, Moniuszko M, Eljaszewicz A, Carewicz P. Epigenetics changes during allergen immunotherapy - Review of available literature. Gene 2025; 960:149535. [PMID: 40316066 DOI: 10.1016/j.gene.2025.149535] [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: 02/02/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
Allergic diseases are very broad ranging conditions and affect the health and comfort of patients in different manners. To date, the only effective treatment is allergen immunotherapy. Unfortunately, apart from observations of drug use or the severity of symptoms, there is no direct method of determining the effectiveness or ineffectiveness of the medical intervention that has been provided. The hope of gaining insight into the adaptive processes occurring in the body during immunotherapy is offered by epigenetics. By observing such areas of biological activity as DNA methylation, modification of histone proteins, or studies of non-coding RNA fragments, physicians will gain reliable tools for determining the effectiveness of the undertaken therapy. They can link the data acquired to the expression levels of specific genes that activate or suppress the immune response, perhaps even before it is initiated. This paper describes the basic epigenetic mechanisms, their known links to specific diseases and therapies, and potential areas for further research.
Collapse
Affiliation(s)
- Adrianna Carewicz
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland.
| | | | | | - Marcin Moniuszko
- Department of Allergology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland; Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland.
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland.
| | - Paweł Carewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
2
|
Akenroye A, Boyce JA, Kita H. Targeting alarmins in asthma: From bench to clinic. J Allergy Clin Immunol 2025; 155:1133-1148. [PMID: 39855362 DOI: 10.1016/j.jaci.2025.01.017] [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: 09/20/2024] [Revised: 12/24/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Over the past 2 decades, mechanistic studies of allergic and type 2 (T2)-mediated airway inflammation have led to multiple approved therapies for the treatment of moderate-to-severe asthma. The approval and availability of these monoclonal antibodies targeting IgE, a T2 cytokine (IL-5) and/or cytokine receptors (IL-5Rα, IL-4Rα) has been central to the progresses made in the management of moderate-to-severe asthma over this period. However, there are persistent gaps in clinician's ability to provide precise care, given that many patients with T2-high asthma do not respond to IgE- or T2 cytokine-targeting therapies and that patients with T2-low asthma have few therapeutic options. The new frontier of precision medicine in asthma, as well as in other allergic diseases, includes the targeting of epithelium-derived cytokines known as alarmins, including thymic stromal lymphopoietin, IL-25, IL-33, and their receptors. The effects of these alarmins, which can act upstream of immune cells, involve both the innate and adaptive systems and hold potential for the treatment of both T2-high and -low disease. Tezepelumab, an anti-thymic stromal lymphopoietin antibody, has already been approved for the treatment of severe asthma. In this review, we discuss our current understanding of alarmin biology with a primary focus on allergic airway diseases. We link the mechanistic corollaries to the clinical implications and advances in drug development targeting alarmins, with a particular focus on currently approved treatments, those under study, and future potential targets in alarmin signaling pathways.
Collapse
Affiliation(s)
- Ayobami Akenroye
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| | - Joshua A Boyce
- Jeff and Penny Vinik Immunology Center, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Hirohito Kita
- Division of Allergy, Asthma and Clinical Immunology, the Department of Medicine, and the Department of Immunology, Mayo Clinic Arizona, Scottsdale, Ariz; Department of Immunology, Mayo Clinic Rochester, Rochester, Minn
| |
Collapse
|
3
|
Noguchi E, Morii W, Kitazawa H, Hirota T, Sonehara K, Masuko H, Okada Y, Hizawa N. A genome-wide meta-analysis reveals shared and population-specific variants for allergic sensitization. J Allergy Clin Immunol 2025; 155:1321-1332. [PMID: 39644933 DOI: 10.1016/j.jaci.2024.11.033] [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: 01/17/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Allergic diseases are major causes of morbidity in both developed and developing countries and represent a global burden on health care systems. Allergic sensitization is defined as the production of IgE specific to common environmental allergens and is an important indicator in the assessment of allergic diseases. OBJECTIVE We sought to clarify the genetic basis of allergic sensitization. METHODS We performed a genome-wide association study (GWAS) of allergic sensitization in the Japanese population followed by a cross-ancestry meta-analysis with a European population including 20,492 cases and 23,342 controls for Japanese and 8,246 cases and 16,786 controls for Europeans. We also performed a polysensitization GWAS of a Japanese population including 4,923 cases and 17,009 controls. RESULTS Allergic sensitization GWAS identified 18 susceptibility loci for Japanese only and 23 loci for the cross-ancestry population, among which 4 loci were novel. Polysensitization GWAS identified 8 significant loci. Expression quantitative trait locus colocalization analysis revealed polysensitization GWAS significant variants affecting both the phenotype and the expression of the CD28, LPP, and LRCC32 genes. Cross-population genetic correlation analysis of allergic sensitization suggested that heterogeneity exists in allergic sensitization between Europeans and Japanese, indicating that more genetic heterogeneity may exist in allergic sensitization than allergic diseases. CONCLUSIONS Our investigation provides new insights into the molecular mechanism of allergic sensitization that could enhance current understanding of allergy and allergic diseases.
Collapse
Affiliation(s)
- Emiko Noguchi
- Department of Medical Genetics, Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
| | - Wataru Morii
- Department of Medical Genetics, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Haruna Kitazawa
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tomomitsu Hirota
- Division of Molecular Genetics, Jikei University School of Medicine, Research Center for Medical Science, Tokyo, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Hironori Masuko
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan; Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita, Japan; Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Nobuyuki Hizawa
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
4
|
Lawson LP, Parameswaran S, Panganiban RA, Constantine GM, Weirauch MT, Kottyan LC. Update on the genetics of allergic diseases. J Allergy Clin Immunol 2025:S0091-6749(25)00327-6. [PMID: 40139464 DOI: 10.1016/j.jaci.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
The field of genetic etiology of allergic diseases has advanced significantly in recent years. Shared risk loci reflect the contribution of genetic factors to the sequential development of allergic conditions across the atopic march, while unique risk loci provide opportunities to understand tissue specific manifestations of allergic disease. Most identified risk variants are noncoding, indicating that they likely influence gene expression through gene regulatory mechanisms. Despite recent advances, challenges persist, particularly regarding the need for increased ancestral diversity in research populations. Further, while polygenic risk scores show promise for identifying individuals at higher genetic risk for allergic diseases, their predictive accuracy varies across different ancestries and can be difficult to translate to an individual's absolute risk of developing a disease. Methodologies, including "nearest gene," 3D chromatin interaction analysis, expression quantitative trait locus analysis, experimental screens, and integrative bioinformatic models, have established connections between genetic variants and their regulatory targets, enhancing our understanding of disease risk and phenotypic variability. In this review, we focus on the state of knowledge of allergic sensitization and 5 allergic diseases: asthma, atopic dermatitis, allergic rhinitis, food allergy, and eosinophilic esophagitis. We summarize recent progress and highlight opportunities for advancing our understanding of their genetic etiology.
Collapse
Affiliation(s)
- Lucinda P Lawson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ronald A Panganiban
- Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Gregory M Constantine
- Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Md
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
5
|
Campos-Bayardo TI, Román-Rojas D, García-Sánchez A, Cardona-Muñoz EG, Sánchez-Lozano DI, Totsuka-Sutto S, Gómez-Hermosillo LF, Casillas-Moreno J, Andrade-Sierra J, Pazarín-Villaseñor L, Campos-Pérez W, Martínez-López E, Miranda-Díaz AG. The Role of TLRs in Obesity and Its Related Metabolic Disorders. Int J Mol Sci 2025; 26:2229. [PMID: 40076851 PMCID: PMC11900219 DOI: 10.3390/ijms26052229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Obesity affects the adaptability of adipose tissue (AT), impairing its ability to regulate energy and metabolism. Obesity is associated with many metabolic disorders, including dyslipidemia, hypertension, sleep disorders, non-alcoholic liver disease, and some types of cancer. Toll-like receptors (TLRs) are important in obesity and related metabolic disorders. TLRs are pattern-recognizing receptors (PRRs) involved in the innate immune system and recognize pathogen-associated molecular patterns (PAMPs) and endogenous ligands. TLRs, especially TLR2 and TLR4, are activated by fatty acids, endotoxins, and other ligands. TLR2 and TLR4 activation triggers inflammatory responses. Chronic inflammation driven by TLR activation is a hallmark of obesity and metabolic diseases. The inflammatory response triggered by TLR activation alters insulin signaling, contributing to insulin resistance, a key feature of metabolic syndrome and type 2 diabetes. Modulation of TLR activity through lifestyle changes (diet and exercise), obesity surgery, and pharmacological agents is under study as a possible therapeutic approach to controlling obesity and its complications.
Collapse
Affiliation(s)
- Tannia Isabel Campos-Bayardo
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniel Román-Rojas
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Andrés García-Sánchez
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Ernesto Germán Cardona-Muñoz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Daniela Itzel Sánchez-Lozano
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Sylvia Totsuka-Sutto
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| | - Luis Francisco Gómez-Hermosillo
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Casillas-Moreno
- Department of Laparoscopic Surgery, Hospital Civil de Guadalajara, “Juan I Menchaca”, Guadalajara 44360, Jalisco, Mexico; (L.F.G.-H.); (J.C.-M.)
| | - Jorge Andrade-Sierra
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Leonardo Pazarín-Villaseñor
- Department of Nephrology, National Medical Center of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico; (J.A.-S.); (L.P.-V.)
| | - Wendy Campos-Pérez
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Erika Martínez-López
- Department of Molecular Biology and Genomics, Institute of Nutrigenetics and Translational Nutrigenomics, University of Guadalajara, Guadalajara 44340, Jalisco, Mexico; (W.C.-P.); (E.M.-L.)
| | - Alejandra Guillermina Miranda-Díaz
- Department of Physiology, University Center of Health Sciences, University of Guadalajara, Guadalajara 44360, Jalisco, Mexico; (T.I.C.-B.); (D.R.-R.); (A.G.-S.); (E.G.C.-M.); (D.I.S.-L.); (S.T.-S.)
| |
Collapse
|
6
|
Hao Y, Yang Y, Zhao H, Chen Y, Zuo T, Zhang Y, Yu H, Cui L, Song X. Multi-omics in Allergic Rhinitis: Mechanism Dissection and Precision Medicine. Clin Rev Allergy Immunol 2025; 68:19. [PMID: 39964644 PMCID: PMC11836232 DOI: 10.1007/s12016-025-09028-3] [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] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Allergic rhinitis (AR) is a common chronic inflammatory airway disease caused by inhaled allergens, and its prevalence has increased in recent decades. AR not only causes nasal leakage, itchy nose, nasal congestion, sneezing, and allergic conjunctivitis but also induces asthma, as well as sleep disorders, anxiety, depression, memory loss, and other phenomena that seriously affect the patient's ability to study and work, lower their quality of life, and burden society. The current methods used to diagnose and treat AR are still far from ideal. Multi-omics technology can be used to comprehensively and systematically analyze the differentially expressed DNA, RNA, proteins, and metabolites and their biological functions in patients with AR. These capabilities allow for an in-depth understanding of the intrinsic pathogenic mechanism of AR, the ability to explore key cells and molecules that drive its progression, and to design personalized treatment for AR. This article summarizes the progress made in studying AR by use of genomics, epigenomics, transcriptomics, proteomics, metabolomics, and microbiomics in order to illustrate the important role of multi-omics technologies in facilitating the precise diagnosis and treatment of AR.
Collapse
Affiliation(s)
- Yan Hao
- Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Yujuan Yang
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Hongfei Zhao
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Ying Chen
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- The 2Nd Medical College of Binzhou Medical University, Yantai, 264000, Shandong, China
| | - Ting Zuo
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- The 2Nd Medical College of Binzhou Medical University, Yantai, 264000, Shandong, China
| | - Yu Zhang
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Hang Yu
- Qingdao Medical College, Qingdao University, Qingdao, 266000, Shandong, China
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China
| | - Limei Cui
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
| | - Xicheng Song
- Department of Otolaryngology Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, Shandong, China.
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, 264000, Shandong, China.
| |
Collapse
|
7
|
Wang Q, He W, Zhou Y, Liu Y, Li X, Wang Y, Wang J, Han X, Zhang X. Improvement of glucocorticoid sensitivity and attenuation of pulmonary allergic reactions by exogenous supplementation with betaine in HDM and LPS-induced allergic mouse model. Clin Transl Allergy 2025; 15:e70039. [PMID: 39921638 PMCID: PMC11806522 DOI: 10.1002/clt2.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Childhood asthma is a heterogeneous disease that exhibits different characteristics and varying severity; however, the metabolite alterations underlying the difference in asthma severity, especially in severe asthma, are not well understood. The aim of this study was to identify the plasma metabolic profile of children with different asthma severity and explore the potential intervention targets in severe asthma and glucocorticoid resistance. METHODS Untargeted liquid chromatography mass spectrometry was utilized to analyze plasma metabolites in 54 children with mild-to-moderate asthma, 50 children with severe asthma and 39 healthy controls. Multivariate statistical analyses were used to explore plasma metabolic alterations that were strongly associated with asthma severity. Meanwhile, the severe allergic airway inflammation mice with glucocorticoid resistance were constructed to validate the potential therapeutic capacity of metabolites. RESULTS The plasma metabolic profiles of children with mild to moderate asthma and severe asthma exhibited significant alterations. The distinct plasma metabolite shifts were accompanied by functional alterations in lipid metabolism, particularly choline metabolism, glycerophospholipids and sphingolipid metabolism. 11-cis-retinol, LysoPC (20:4 [8Z,11Z,14Z,17Z]), and glycerophosphatidylcholine were associated with exacerbated airway inflammation and lung function. Furthermore, 2-Hydroxyestradiol, LysoPC (18:3 [6Z,9Z,12Z]), zeaxanthin, and betaine were shifted exclusively in the severe asthma group and may serve as potential biomarkers. Subsequent in vivo studies demonstrated that betaine supplementation partially improved glucocorticoid resistance. CONCLUSIONS Overall, children with different asthma severity displayed distinct plasma metabolic patterns. These may contribute to the difference in response to glucocorticoids in childhood asthma and could be potential targets and interventions.
Collapse
Affiliation(s)
- Qing Wang
- Department of Respiratory MedicineChildren's Hospital of Fudan UniversityShanghaiChina
| | - Wen He
- Department of Respiratory MedicineChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yufeng Zhou
- International Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitute of PediatricsChildren's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical EpigeneticsInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- National Health Commission (NHC) Key Laboratory of Neonatal DiseasesFudan UniversityChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular MedicineDepartment of Biochemistry and Molecular BiologySchool of Basic Medical Sciences and Shanghai Xuhui Central HospitalFudan UniversityShanghaiChina
| | - Xiaoling Li
- Department of Respiratory MedicineChildren's Hospital of Fudan UniversityShanghaiChina
| | - Yingwen Wang
- Department of NursingChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jiayu Wang
- National Health Commission (NHC) Key Laboratory of Neonatal DiseasesFudan UniversityChildren's Hospital of Fudan UniversityShanghaiChina
| | - Xiao Han
- Guangzhou Women and Children's Medical CentreInstitute of PediatricsGuangzhou Medical UniversityGuangzhouChina
| | - Xiaobo Zhang
- Department of Respiratory MedicineChildren's Hospital of Fudan UniversityShanghaiChina
- Center for Pediatric Clinical Quality Control of ShanghaiShanghaiChina
| |
Collapse
|
8
|
Cao Z, Zhao S, Hu S, Wu T, Sun F, Shi LI. Screening COPD-Related Biomarkers and Traditional Chinese Medicine Prediction Based on Bioinformatics and Machine Learning. Int J Chron Obstruct Pulmon Dis 2024; 19:2073-2095. [PMID: 39346628 PMCID: PMC11438478 DOI: 10.2147/copd.s476808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Purpose To employ bioinformatics and machine learning to predict the characteristics of immune cells and genes associated with the inflammatory response and ferroptosis in chronic obstructive pulmonary disease (COPD) patients and to aid in the development of targeted traditional Chinese medicine (TCM). Mendelian randomization analysis elucidates the causal relationships among immune cells, genes, and COPD, offering novel insights for the early diagnosis, prevention, and treatment of COPD. This approach also provides a fresh perspective on the use of traditional Chinese medicine for treating COPD. Methods R software was used to extract COPD-related data from the Gene Expression Omnibus (GEO) database, differentially expressed genes were identified for enrichment analysis, and WGCNA was used to pinpoint genes within relevant modules associated with COPD. This analysis included determining genes linked to the inflammatory response in COPD patients and analyzing their correlation with ferroptosis. Further steps involved filtering core genes, constructing TF-miRNA‒mRNA network diagrams, and employing three types of machine learning to predict the core miRNAs, key immune cells, and characteristic genes of COPD patients. This process also delves into their correlations, single-gene GSEA, and diagnostic model predictions. Reverse inference complemented by molecular docking was used to predict compounds and traditional Chinese medicines for treating COPD; Mendelian randomization was applied to explore the causal relationships among immune cells, genes, and COPD. Results We identified 2443 differential genes associated with COPD through the GEO database, along with 8435 genes relevant to WGCNA and 1226 inflammation-related genes. A total of 141 genes related to the inflammatory response in COPD patients were identified, and 37 core genes related to ferroptosis were selected for further enrichment analysis and analysis. The core miRNAs predicted for COPD include hsa-miR-543, hsa-miR-181c, and hsa-miR-200a, among others. The key immune cells identified were plasma cells, activated memory CD4 T cells, gamma delta T cells, activated NK cells, M2 macrophages, and eosinophils. Characteristic genes included EGF, PLG, PTPN22, and NR4A1. A total of 78 compounds and 437 traditional Chinese medicines were predicted. Mendelian randomization analysis revealed a causal relationship between 36 types of immune cells and COPD, whereas no causal relationship was found between the core genes and COPD. Conclusion A definitive causal relationship exists between immune cells and COPD, while the prediction of core miRNAs, key immune cells, characteristic genes, and targeted traditional Chinese medicines offers novel insights for the early diagnosis, prevention, and treatment of COPD.
Collapse
Affiliation(s)
- Zhenghua Cao
- Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Shengkun Zhao
- Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Shaodan Hu
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Tong Wu
- Geriatric Department, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, Jiangsu, People's Republic of China
| | - Feng Sun
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - L I Shi
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People's Republic of China
| |
Collapse
|
9
|
DuToit G, Smith P, Muraro A, Fox AT, Roberts G, Ring J, Worm M. Identifying patients at risk of anaphylaxis. World Allergy Organ J 2024; 17:100904. [PMID: 38966605 PMCID: PMC11223123 DOI: 10.1016/j.waojou.2024.100904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 03/31/2024] [Accepted: 04/05/2024] [Indexed: 07/06/2024] Open
Abstract
Anaphylaxis is an acute, potentially fatal, systemic hypersensitivity reaction that warrants prompt diagnosis and management. It continues to be challenging to anticipate who may be at risk of a severe, life-threatening allergic reaction. Anaphylaxis can be caused by a range of allergens, such as certain foods, medications, latex, insect stings, etc. Cofactors that augment the severity of clinical symptoms and increase the risk of poor outcomes include exercise, stress, infectious diseases, underlying mast cell disease, active allergic disease such as asthma, advanced age, intake of certain medications, history of previous anaphylaxis, and delayed or missed administration of adrenaline. According to the European Anaphylaxis Registry, food is the major elicitor of anaphylaxis, especially eggs, cow milk, and nuts, in children and adolescents. Reaction to insect venom has also been noted in young adulthood. Early recognition of signs and symptoms and prompt treatment are crucial in anaphylaxis management to avoid serious and even fatal outcomes. It is crucial for both individuals and clinicians to identify the cause of anaphylaxis. Biomarkers of anaphylaxis, such as histamine, tryptase, platelet activation factor (PAF), chymase, carboxypeptidase A3, dipeptidyl peptidase I (DPPI), basogranulin, CCL-2, hsa-miR-451a, may be useful in diagnosis and management. The purpose of this review article is to present a comprehensive overview of current evidence and expert opinions regarding the risk factors that predispose individuals to anaphylaxis. Additionally, it provides insights into potential biomarkers and genetic markers for accurate diagnosis and management. This review underscores the significance of expert guidance in enhancing patient outcomes and enabling self-management of anaphylactic episodes.
Collapse
Affiliation(s)
- George DuToit
- Pediatric Allergy King's College London and Guy's and St Thomas', London, United Kingdom
| | - Pete Smith
- Clinical School of Medicine, Griffith University, Southport, Queensland, Australia
| | - Antonella Muraro
- Food Allergy Referral Centre, Department of Woman and Child Health, Padua University Hospital, Padua, Italy
| | - Adam T. Fox
- Children's Allergy Service, Guy's and St Thomas' Hospitals NHS Foundation Trust, Westminster Bridge, London, United Kingdom
| | - Graham Roberts
- University of Southampton, Pediatric Allergy & Respiratory Medicine, Tremona Road, Southampton, United Kingdom
| | - Johannes Ring
- Technical University Munich (TUM), Dept Dermatology Allergology Biederstein, Germany
| | - Margitta Worm
- Allergologie und Immunologie, Klinik für Dermatologie, Venerologie und Allergologie, Campus Charité Mitte, Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
10
|
Wise SK, Hamzavi-Abedi Y, Hannikainen PA, Anand MP, Pitt T, Savoure M, Toskala E. Rhinitis Disease Burden and the Impact of Social Determinants of Health. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1449-1461.e1. [PMID: 38570070 DOI: 10.1016/j.jaip.2024.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Social determinants of health (SDHs) have a substantial impact on patient care and outcomes globally, both in low- to middle-income countries and in high-income countries. In the clinic, lack of availability of diagnostic tools, inequities in access to care, and challenges obtaining and adhering to prescribed treatment plans may further compound these issues. This article addresses a case of rhinitis in the context of SDHs and inequities in care that may affect various communities and populations around the world. SDHs may include various aspects of one's financial means, education, access to medical care, environment and living situation, and community factors, each of which could play a role in the rhinitis disease manifestations, diagnosis, and management. Allergic and nonallergic rhinitis are considered from this perspective. Rhinitis epidemiology, disease burden, and risk factors are broadly addressed. Patient evaluation, diagnostic tests, and management options are also reviewed, and issues related to SDHs are noted. Finally, inequities in care, knowledge gaps, and unmet needs are highlighted. It is critical to consider SDHs and care inequities when evaluating and treating patients for rhinitis and other allergic conditions.
Collapse
Affiliation(s)
- Sarah K Wise
- Department of Otolaryngology-Head and Neck Surgery, Emory University, Atlanta, Ga.
| | - Yasmin Hamzavi-Abedi
- Departments of Pediatrics and Medicine, Division of Allergy and Immunology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY
| | | | - Mahesh Padukudru Anand
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysore, Karnataka, India
| | - Tracy Pitt
- Department of Paediatrics, Humber River Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Marine Savoure
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elina Toskala
- Department of Otolaryngology-Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pa
| |
Collapse
|
11
|
Kennedy KV, Muir AB, Ruffner MA. Pathophysiology of Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:119-128. [PMID: 38575212 DOI: 10.1016/j.iac.2023.12.001] [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] [Indexed: 04/06/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, progressive immune-mediated disease associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Research over the last 2 decades has dramatically furthered our understanding of the complex interplay between genetics, environmental exposures, and cellular and molecular interactions involved in EoE. This review provides an overview of our current understanding of EoE pathogenesis.
Collapse
Affiliation(s)
- Kanak V Kennedy
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Melanie A Ruffner
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Division of Pediatric Allergy and Immunology, Department of Pediatrics, The Children's Hospital of Philadelphia
| |
Collapse
|
12
|
Glatthardt T, van Tilburg Bernardes E, Arrieta MC. The mycobiome in atopic diseases: Inducers and triggers. J Allergy Clin Immunol 2023; 152:1368-1375. [PMID: 37865199 DOI: 10.1016/j.jaci.2023.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/03/2023] [Accepted: 10/17/2023] [Indexed: 10/23/2023]
Abstract
Atopic diseases are characterized by type 2 inflammation, with high levels of allergen-specific TH2 cell immune responses and elevated production of IgE. These common disorders have increased in incidence around the world, which is partly explained by detrimental disturbances to the early-life intestinal microbiome. Although most studies have focused exclusively on bacterial members of the microbiome, intestinal fungi have started to be recognized for their impact on host immune development and atopy pathogenesis. From this perspective, we review recent findings demonstrating the strong interactions between members of the mycobiome and the host immune system early in life, leading to immune tolerance during eubiosis or inducing sensitization and overt TH2 cell responses during dysbiosis. Current evidence places intestinal fungi as central players in the development of allergic diseases and potential targets for atopy prevention and treatments.
Collapse
Affiliation(s)
- Thais Glatthardt
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary
| | - Erik van Tilburg Bernardes
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary
| | - Marie-Claire Arrieta
- the Department of Physiology and Pharmacology, University of Calgary, Calgary; the International Microbiome Centre, Snyder Institute for Chronic Diseases, University of Calgary, Calgary; the Department of Pediatrics, Alberta Children Hospital Research Institute, University of Calgary, Calgary.
| |
Collapse
|
13
|
Wenger M, Grosse-Kathoefer S, Kraiem A, Pelamatti E, Nunes N, Pointner L, Aglas L. When the allergy alarm bells toll: The role of Toll-like receptors in allergic diseases and treatment. Front Mol Biosci 2023; 10:1204025. [PMID: 37426425 PMCID: PMC10325731 DOI: 10.3389/fmolb.2023.1204025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Toll-like receptors of the human immune system are specialized pathogen detectors able to link innate and adaptive immune responses. TLR ligands include among others bacteria-, mycoplasma- or virus-derived compounds such as lipids, lipo- and glycoproteins and nucleic acids. Not only are genetic variations in TLR-related genes associated with the pathogenesis of allergic diseases, including asthma and allergic rhinitis, their expression also differs between allergic and non-allergic individuals. Due to a complex interplay of genes, environmental factors, and allergen sources the interpretation of TLRs involved in immunoglobulin E-mediated diseases remains challenging. Therefore, it is imperative to dissect the role of TLRs in allergies. In this review, we discuss i) the expression of TLRs in organs and cell types involved in the allergic immune response, ii) their involvement in modulating allergy-associated or -protective immune responses, and iii) how differential activation of TLRs by environmental factors, such as microbial, viral or air pollutant exposure, results in allergy development. However, we focus on iv) allergen sources interacting with TLRs, and v) how targeting TLRs could be employed in novel therapeutic strategies. Understanding the contributions of TLRs to allergy development allow the identification of knowledge gaps, provide guidance for ongoing research efforts, and built the foundation for future exploitation of TLRs in vaccine design.
Collapse
|
14
|
Schwab AD, Poole JA. Mechanistic and Therapeutic Approaches to Occupational Exposure-Associated Allergic and Non-Allergic Asthmatic Disease. Curr Allergy Asthma Rep 2023; 23:313-324. [PMID: 37154874 PMCID: PMC10896074 DOI: 10.1007/s11882-023-01079-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE OF REVIEW Occupational lung disease, including asthma, is a significant cause of disability worldwide. The dose, exposure frequency, and nature of the causal agent influence the inflammatory pathomechanisms that inform asthma disease phenotype and progression. While surveillance, systems engineering, and exposure mitigation strategies are essential preventative considerations, no targeted medical therapies are currently available to ameliorate lung injury post-exposure and prevent chronic airway disease development. RECENT FINDINGS This article reviews contemporary understanding of allergic and non-allergic occupational asthma mechanisms. In addition, we discuss the available therapeutic options, patient-specific susceptibility and prevention measures, and recent scientific advances in post-exposure treatment conception. The course of occupational lung disease that follows exposure is informed by individual predisposition, immunobiologic response, agent identity, overall environmental risk, and preventative workplace practices. When protective strategies fail, knowledge of underlying disease mechanisms is necessary to inform targeted therapy development to lessen occupational asthma disease severity and occurrence.
Collapse
Affiliation(s)
- Aaron D Schwab
- Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Jill A Poole
- Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
15
|
Sharma M, Leung D, Momenilandi M, Jones LC, Pacillo L, James AE, Murrell JR, Delafontaine S, Maimaris J, Vaseghi-Shanjani M, Del Bel KL, Lu HY, Chua GT, Di Cesare S, Fornes O, Liu Z, Di Matteo G, Fu MP, Amodio D, Tam IYS, Chan GSW, Sharma AA, Dalmann J, van der Lee R, Blanchard-Rohner G, Lin S, Philippot Q, Richmond PA, Lee JJ, Matthews A, Seear M, Turvey AK, Philips RL, Brown-Whitehorn TF, Gray CJ, Izumi K, Treat JR, Wood KH, Lack J, Khleborodova A, Niemela JE, Yang X, Liang R, Kui L, Wong CSM, Poon GWK, Hoischen A, van der Made CI, Yang J, Chan KW, Rosa Duque JSD, Lee PPW, Ho MHK, Chung BHY, Le HTM, Yang W, Rohani P, Fouladvand A, Rokni-Zadeh H, Changi-Ashtiani M, Miryounesi M, Puel A, Shahrooei M, Finocchi A, Rossi P, Rivalta B, Cifaldi C, Novelli A, Passarelli C, Arasi S, Bullens D, Sauer K, Claeys T, Biggs CM, Morris EC, Rosenzweig SD, O’Shea JJ, Wasserman WW, Bedford HM, van Karnebeek CD, Palma P, Burns SO, Meyts I, Casanova JL, Lyons JJ, Parvaneh N, Nguyen ATV, Cancrini C, Heimall J, Ahmed H, McKinnon ML, Lau YL, Béziat V, Turvey SE. Human germline heterozygous gain-of-function STAT6 variants cause severe allergic disease. J Exp Med 2023; 220:e20221755. [PMID: 36884218 PMCID: PMC10037107 DOI: 10.1084/jem.20221755] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/12/2022] [Accepted: 02/09/2023] [Indexed: 03/09/2023] Open
Abstract
STAT6 (signal transducer and activator of transcription 6) is a transcription factor that plays a central role in the pathophysiology of allergic inflammation. We have identified 16 patients from 10 families spanning three continents with a profound phenotype of early-life onset allergic immune dysregulation, widespread treatment-resistant atopic dermatitis, hypereosinophilia with esosinophilic gastrointestinal disease, asthma, elevated serum IgE, IgE-mediated food allergies, and anaphylaxis. The cases were either sporadic (seven kindreds) or followed an autosomal dominant inheritance pattern (three kindreds). All patients carried monoallelic rare variants in STAT6 and functional studies established their gain-of-function (GOF) phenotype with sustained STAT6 phosphorylation, increased STAT6 target gene expression, and TH2 skewing. Precision treatment with the anti-IL-4Rα antibody, dupilumab, was highly effective improving both clinical manifestations and immunological biomarkers. This study identifies heterozygous GOF variants in STAT6 as a novel autosomal dominant allergic disorder. We anticipate that our discovery of multiple kindreds with germline STAT6 GOF variants will facilitate the recognition of more affected individuals and the full definition of this new primary atopic disorder.
Collapse
Affiliation(s)
- Mehul Sharma
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Daniel Leung
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Mana Momenilandi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
| | - Lauren C.W. Jones
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Lucia Pacillo
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Academic Dept. of Pediatrics (DPUO), Unit of Clinical Immunology and Vaccinology, IRCCS Bambin Gesù Children Hospital, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Alyssa E. James
- Translational Allergic Immunopathology Unit, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jill R. Murrell
- Pathology and Laboratory Medicine, Division of Genomic Diagnostics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Selket Delafontaine
- Dept. of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium
- Dept. of Pediatrics, Pediatric Immunodeficiencies Division, University Hospitals Leuven, Leuven, Belgium
| | - Jesmeen Maimaris
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Dept. of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Maryam Vaseghi-Shanjani
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Kate L. Del Bel
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Henry Y. Lu
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Dept. of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Gilbert T. Chua
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Allergy Centre, Union Hospital, Hong Kong, China
| | - Silvia Di Cesare
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Oriol Fornes
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
- Dept. of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Zhongyi Liu
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Gigliola Di Matteo
- Academic Dept. of Pediatrics (DPUO), Unit of Clinical Immunology and Vaccinology, IRCCS Bambin Gesù Children Hospital, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Maggie P. Fu
- Dept. of Medical Genetics, The University of British Columbia, Vancouver, Canada
- Genome Science and Technology Program, Faculty of Science, The University of British Columbia, Vancouver, Canada
| | - Donato Amodio
- Academic Dept. of Pediatrics (DPUO), Unit of Clinical Immunology and Vaccinology, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Issan Yee San Tam
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | | | | | - Joshua Dalmann
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Robin van der Lee
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
- Dept. of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Géraldine Blanchard-Rohner
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
- Unit of Immunology and Vaccinology, Division of General Pediatrics, Dept. of Woman, Child, and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Susan Lin
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
| | - Phillip A. Richmond
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Jessica J. Lee
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, Canada
| | - Allison Matthews
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
- Dept. of Paediatrics, University of Toronto, Toronto, Canada
| | - Michael Seear
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Alexandra K. Turvey
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Rachael L. Philips
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Terri F. Brown-Whitehorn
- Dept. of Pediatrics, Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christopher J. Gray
- Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kosuke Izumi
- Pediatrics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James R. Treat
- Pediatrics, Division of Pediatric Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kathleen H. Wood
- Pathology and Laboratory Medicine, Division of Genomic Diagnostics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, NIAID, NIH, Bethesda, MD, USA
| | - Asya Khleborodova
- NIAID Collaborative Bioinformatics Resource, NIAID, NIH, Bethesda, MD, USA
| | | | - Xingtian Yang
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Rui Liang
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Lin Kui
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Dept. of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Christina Sze Man Wong
- Dept. of Medicine, Divison of Dermatology, The University of Hong Kong, Hong Kong, China
| | - Grace Wing Kit Poon
- Dept. of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, China
| | - Alexander Hoischen
- Dept. of Human Genetics, Radboud University Medical Center, Nijmegen, Netherlands
| | | | - Jing Yang
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Koon Wing Chan
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Jaime Sou Da Rosa Duque
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Pamela Pui Wah Lee
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Marco Hok Kung Ho
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Virtus Medical, Hong Kong, China
| | - Brian Hon Yin Chung
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Huong Thi Minh Le
- Pediatric Center, Vinmec Times City International General Hospital, Hanoi, Vietnam
| | - Wanling Yang
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Pejman Rohani
- Pediatrics, Pediatric Gastroenterology and Hepatology Research Center, Pediatrics Centre of Excellence, Children’s Medical Center, University of Medical Sciences, Tehran, Iran
| | - Ali Fouladvand
- Pediatrics, Allergy and Clinical Immunology, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Hassan Rokni-Zadeh
- Dept. of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Mohammad Miryounesi
- Dept. of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Mohammad Shahrooei
- Microbiology and Immunology, Laboratory of Clinical Bacteriology and Mycology, KU Leuven, Leuven, Belgium
| | - Andrea Finocchi
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Paolo Rossi
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- DPUO, Research Unit of Infectivology and Pediatrics Drugs Development, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Beatrice Rivalta
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Academic Dept. of Pediatrics (DPUO), Unit of Clinical Immunology and Vaccinology, IRCCS Bambin Gesù Children Hospital, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Cristina Cifaldi
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Chiara Passarelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Stefania Arasi
- Allergy Unit, Area of Translational Research in Pediatric Specialities, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Dominique Bullens
- Dept. of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, KU Leuven, Leuven, Belgium
- Dept. of Pediatrics, Pediatric Allergy Division, University Hospitals Leuven, Leuven, Belgium
| | - Kate Sauer
- Dept. of Pediatrics, Pediatric Pulmonology Division, AZ Sint-Jan Brugge, Brugge, Belgium
- Dept. of Pediatrics, Pediatric Pulmonology Division, University Hospitals Leuven, Leuven, Belgium
| | - Tania Claeys
- Dept. of Pediatrics, Pediatric Gastroenterology Division, AZ Sint-Jan Brugge, Brugge, Belgium
| | - Catherine M. Biggs
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| | - Emma C. Morris
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Dept. of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | | | - John J. O’Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, NIH, Bethesda, MD, USA
| | - Wyeth W. Wasserman
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
| | - H. Melanie Bedford
- Dept. of Paediatrics, University of Toronto, Toronto, Canada
- Genetics Program, North York General Hospital, Toronto, Canada
| | - Clara D.M. van Karnebeek
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Vancouver, Canada
- Depts. of Pediatrics and Clinical Genetics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Paolo Palma
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Academic Dept. of Pediatrics (DPUO), Unit of Clinical Immunology and Vaccinology, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Siobhan O. Burns
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Dept. of Immunology, Royal Free London NHS Foundation Trust, London, UK
| | - Isabelle Meyts
- Dept. of Microbiology, Immunology and Transplantation, Laboratory for Inborn Errors of Immunity, KU Leuven, Leuven, Belgium
- Dept. of Pediatrics, Pediatric Immunodeficiencies Division, University Hospitals Leuven, Leuven, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jonathan J. Lyons
- Translational Allergic Immunopathology Unit, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Nima Parvaneh
- Department of Pediatrics, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Anh Thi Van Nguyen
- Dept. of Immunology, Allergy and Rheumatology, Division of Primary Immunodeficiency, Vietnam National Children’s Hospital, Hanoi, Vietnam
| | - Caterina Cancrini
- Dept. of System Medicine, Pediatric Chair, University of Tor Vergata, Rome, Italy
- Research Unit of Primary Immunodeficiency, IRCCS Bambin Gesù Children Hospital, Rome, Italy
| | - Jennifer Heimall
- Dept. of Pediatrics, Division of Allergy and Immunology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hanan Ahmed
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | | | - Yu Lung Lau
- Dept. of Paediatrics and Adolescent Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris-Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Stuart E. Turvey
- Dept. of Pediatrics, BC Children’s Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
16
|
Morii W, Kasai K, Nakamura T, Hayashi D, Hara M, Naito T, Sonehara K, Fukuie T, Saito-Abe M, Yang L, Yamamoto-Hanada K, Narita M, Maruo K, Okada Y, Noguchi E, Ohya Y. A genome-wide association study for allergen component sensitizations identifies allergen component-specific and allergen protein group-specific associations. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100086. [PMID: 37780799 PMCID: PMC10509904 DOI: 10.1016/j.jacig.2023.100086] [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: 07/05/2022] [Revised: 10/03/2022] [Accepted: 11/04/2022] [Indexed: 10/03/2023]
Abstract
Background Allergic diseases are some of the most common diseases worldwide. Genome-wide association studies (GWASs) have been conducted to elucidate the genetic factors of allergic diseases. However, no GWASs for allergen component sensitization have been performed. Objective We sought to detect genetic variants associated with differences in immune responsiveness against allergen components. Methods The participants of the present study were recruited from the Tokyo Children's Health, Illness, and Development study, and allergen component-specific IgE level at age 9 years was measured by means of allergen microarray immunoassays. We performed GWASs for allergen component sensitization against each allergen (single allergen component sensitization, number of allergen components analyzed, n = 31), as well as against allergen protein families (allergen protein group sensitization, number of protein groups analyzed, n = 16). Results We performed GWAS on 564 participants of the Tokyo Children's Health, Illness, and Development study and found associations between Amb a 1 sensitization and the immunoglobulin heavy-chain variable gene on chromosome 14 and between Phl p 1 sensitization and the HLA class II region on chromosome 6 (P < 5.0 × 10-8). A GWAS-significant association was also observed between the HLA class II region and profilin sensitization (P < 5.0 × 10-8). Conclusions Our data provide the first demonstration of genetic risk for allergen component sensitization and show that this genetic risk is related to immune response genes including immunoglobulin heavy-chain variable gene and HLA.
Collapse
Affiliation(s)
- Wataru Morii
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Koki Kasai
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takako Nakamura
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Daisuke Hayashi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Monami Hara
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tatsuhiko Naito
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Neurology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
| | - Tatsuki Fukuie
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Mayako Saito-Abe
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Limin Yang
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | | | - Masami Narita
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
- Department of Pediatrics, School of Medicine, Kyorin University, Tokyo, Japan
| | - Kazushi Maruo
- Department of Biostatistics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Japan
| | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
17
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
18
|
Wang J, Zhou Y, Zhang H, Hu L, Liu J, Wang L, Wang T, Zhang H, Cong L, Wang Q. Pathogenesis of allergic diseases and implications for therapeutic interventions. Signal Transduct Target Ther 2023; 8:138. [PMID: 36964157 PMCID: PMC10039055 DOI: 10.1038/s41392-023-01344-4] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 03/26/2023] Open
Abstract
Allergic diseases such as allergic rhinitis (AR), allergic asthma (AAS), atopic dermatitis (AD), food allergy (FA), and eczema are systemic diseases caused by an impaired immune system. Accompanied by high recurrence rates, the steadily rising incidence rates of these diseases are attracting increasing attention. The pathogenesis of allergic diseases is complex and involves many factors, including maternal-fetal environment, living environment, genetics, epigenetics, and the body's immune status. The pathogenesis of allergic diseases exhibits a marked heterogeneity, with phenotype and endotype defining visible features and associated molecular mechanisms, respectively. With the rapid development of immunology, molecular biology, and biotechnology, many new biological drugs have been designed for the treatment of allergic diseases, including anti-immunoglobulin E (IgE), anti-interleukin (IL)-5, and anti-thymic stromal lymphopoietin (TSLP)/IL-4, to control symptoms. For doctors and scientists, it is becoming more and more important to understand the influencing factors, pathogenesis, and treatment progress of allergic diseases. This review aimed to assess the epidemiology, pathogenesis, and therapeutic interventions of allergic diseases, including AR, AAS, AD, and FA. We hope to help doctors and scientists understand allergic diseases systematically.
Collapse
Affiliation(s)
- Ji Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Yumei Zhou
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Honglei Zhang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Linhan Hu
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Juntong Liu
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Lei Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 1000210, China
| | - Tianyi Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Haiyun Zhang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Linpeng Cong
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China
| | - Qi Wang
- National Institute of TCM constitution and Preventive Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, P.R. China.
| |
Collapse
|
19
|
Hanif T, Ivaska LE, Ahmad F, Tan G, Mikola E, Puhakka T, Palomares O, Akdis CA, Toppila-Salmi S, Jartti T. Tonsillar transcriptional profiles in atopic and non-atopic subjects. Allergy 2023; 78:522-536. [PMID: 35899482 PMCID: PMC10087516 DOI: 10.1111/all.15458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/29/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Emerging research suggests that local lymphatic tissue such as tonsils have important role in regulating the immune responses. However, allergen sensitization-induced alterations in transcriptome of tonsils are not known. OBJECTIVES To examine the key differences in tonsillar gene expression between atopic and non-atopic subjects and further by type of sensitization. METHODS RNA-sequencing was performed on 52 tonsillar samples from atopic and non-atopic tonsillectomy patients. Sensitization to common food- and aero-allergen was defined by allergen specific IgE. Following groups were studied: (1) aero- and food-allergen sensitized (AS+FS) versus non-sensitized (NS), (2) aeroallergen-sensitized (AS) versus food-allergen sensitized (FS), (3) AS versus NS, (4) FS versus NS. Bioinformatics analysis was done using DESeq2(v3.10.2), WGCNA and GATK pipeline in R software (v3.3.1). Protein-protein interaction network was made from String database. RESULTS We studied 13 aeroallergen-sensitized, 6 food-allergen sensitized, 4 both food-and aero-allergen-sensitized and 29 non-sensitized tonsillectomy patients. Overall, 697 unique differentially expressed genes (DEGs) were detected in all sensitized subgroups including chemokines (CXCL2, CXCL8, CXCL10, CXCL11), IL-20RA, MUC1 and MUC20. When comparing different groups, the gene expression profiles overlapped except the AS versus FS group comparison, suggesting significantly different gene expression between the two sensitization subgroups. Furthermore, aeroallergen-sensitized subjects had more prominent immune responses compared with non-sensitized and food-allergen sensitized subjects including gene expression for IL-17 pathway and Toll-like receptor signalling pathway. CONCLUSION Allergic sensitization is associated with extensive tonsillar transcriptomic alterations and changes in immune related genes and pathways. Distinct differences were found between aero-allergen and food-allergen sensitization.
Collapse
Affiliation(s)
- Tanzeela Hanif
- Department of Pediatrics and Adolescent Medicine, University of Turku, Turku, Finland.,Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Lotta E Ivaska
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Freed Ahmad
- Department of Biology University of Turku, Turku, Finland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zürich/University of Zürich, Zurich, Switzerland.,Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Emilia Mikola
- Department of Otorhinolaryngology, Satakunta Central Hospital, Pori, Finland
| | - Tuomo Puhakka
- Department of Otorhinolaryngology - Head and Neck Surgery, Turku University Hospital and University of Turku, Turku, Finland
| | - Oscar Palomares
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.,Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research, University of Zürich, Davos, Switzerland.,Christine Kuhne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Sanna Toppila-Salmi
- Haartman Institute, University of Helsinki & Skin and Allergy Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine, University of Turku, Turku, Finland.,PEDEGO Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
20
|
Margolis DJ, Mitra N, Hoffstad OJ, Berna R, Kim BS, Chopra A, Phillips EJ. Association of KIR2DL5, KIR2DS5, and KIR2DS1 allelic variation and atopic dermatitis. Sci Rep 2023; 13:1730. [PMID: 36720995 PMCID: PMC9889380 DOI: 10.1038/s41598-023-28847-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Natural killer cells (NK) have been associated with the pathophysiology of atopic dermatitis (AD). NK function is regulated by killer cell Ig-like receptor family (KIR) receptors that interact with HLA ligands. The study goal was to focus on allelic variation in genes KIR2DL5, KIR2DS5, and KIR2DS1 with respect to AD. This was a case-control study of individuals with (n = 313) and without (n = 176) AD. Associations were estimated using logistic regression. The prevalence of KIR2DL5 was 52.5% (95% CI 48.0,57.0), KIR2DS5 was 33.0% (28.8,37.3), and KIR2DS1 was 33.6% (29.4,38.0). The presence of the KIR2DL5*001:01 increased the odds of having AD by about 86% (odds ratio (OR): 1.86(1.23,2.82) p = 0.003). The risk for individuals homozygous for KIR2DL5*001:01 was even greater (OR: 2.16 (95% CI 1.31,3.53) p = 0.0023). The odds of having AD with KIR2DL5*001:01 was similar in Whites and Blacks. Allelic variation in KIR2DS5 and KIR2DS1 was not associated with AD. There is no known HLA binding ligand for KIR2DL5. The effect of KIR2DL5*001:01 increased in the presence of HLA-B*-21TT leader sequence (2.46(1.37,4.41) p = 0.0025) and the HLA-C2 ligand (2.07 (1.37,4.41, p = 0.000002). Our study shows an independent association of the KIR2DL5*001:01 with AD and is the first study to associate AD with KIR allelic variation.
Collapse
Affiliation(s)
- David J Margolis
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA, USA. .,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 901 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA.
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, PA, USA
| | - Ole J Hoffstad
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 901 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Ronald Berna
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, 901 Blockley Hall, 423 Guardian Drive, Philadelphia, PA, 19104, USA
| | - Brian S Kim
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Elizabeth J Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia.,Center for Drug Safety and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
21
|
Nasal DNA methylation at three CpG sites predicts childhood allergic disease. Nat Commun 2022; 13:7415. [PMID: 36456559 PMCID: PMC9715628 DOI: 10.1038/s41467-022-35088-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Childhood allergic diseases, including asthma, rhinitis and eczema, are prevalent conditions that share strong genetic and environmental components. Diagnosis relies on clinical history and measurements of allergen-specific IgE. We hypothesize that a multi-omics model could accurately diagnose childhood allergic disease. We show that nasal DNA methylation has the strongest predictive power to diagnose childhood allergy, surpassing blood DNA methylation, genetic risk scores, and environmental factors. DNA methylation at only three nasal CpG sites classifies allergic disease in Dutch children aged 16 years well, with an area under the curve (AUC) of 0.86. This is replicated in Puerto Rican children aged 9-20 years (AUC 0.82). DNA methylation at these CpGs additionally detects allergic multimorbidity and symptomatic IgE sensitization. Using nasal single-cell RNA-sequencing data, these three CpGs associate with influx of T cells and macrophages that contribute to allergic inflammation. Our study suggests the potential of methylation-based allergy diagnosis.
Collapse
|
22
|
Prasad V, Abdullah M, Nordin F, Subha ST. Prevalence, causes and treatments of allergic rhinitis in Malaysia: a literature review. THE EGYPTIAN JOURNAL OF OTOLARYNGOLOGY 2022. [DOI: 10.1186/s43163-022-00361-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractRecently, allergic rhinitis (AR) cases have been surging rapidly in many developing countries. However, the prevalence, risk factors and treatment modalities for AR within the Malaysian population have not been thoroughly evaluated. The present study aimed to provide a complete review of literature on allergic rhinitis in Malaysia. Available data indicated that the prevalence of AR varied across different age groups and regions in Malaysia, but there is an increasing trend among the younger population. The key contributing factor is exposure and sensitization towards several airborne allergens, mainly house dust mites, cat fur and fungi, with possible genetic predispositions. In addition, variations in the symptom severity may be associated with racial orientations. For management of the disease, standard prescriptions of conventional drugs (antihistamines, intranasal corticosteroids and nasal decongestants) remain as the treatment of choice. A small proportion of the native residents tend to resort to alternative approaches of self-healing through aromatherapy and natural food consumption such as tiger’s milk mushroom and Tualang honey. In conclusion, the increase in the cases of AR in Malaysia is due to environmental and genetic factors which requires proper medical intervention as treatment strategies. The utilization of holistic approaches requires further studies and clear understanding prior to their integration into the standard of care. There are still many gaps in the knowledge and management of allergic rhinitis which demands further attention from the research community.
Collapse
|
23
|
Kelleher MM, Phillips R, Brown SJ, Cro S, Cornelius V, Carlsen KCL, Skjerven HO, Rehbinder EM, Lowe AJ, Dissanayake E, Shimojo N, Yonezawa K, Ohya Y, Yamamoto-Hanada K, Morita K, Axon E, Cork M, Cooke A, Van Vogt E, Schmitt J, Weidinger S, McClanahan D, Simpson E, Duley L, Askie LM, Williams HC, Boyle RJ. Skin care interventions in infants for preventing eczema and food allergy. Cochrane Database Syst Rev 2022; 11:CD013534. [PMID: 36373988 PMCID: PMC9661877 DOI: 10.1002/14651858.cd013534.pub3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Eczema and food allergy are common health conditions that usually begin in early childhood and often occur in the same people. They can be associated with an impaired skin barrier in early infancy. It is unclear whether trying to prevent or reverse an impaired skin barrier soon after birth is effective for preventing eczema or food allergy. OBJECTIVES Primary objective To assess the effects of skin care interventions such as emollients for primary prevention of eczema and food allergy in infants. Secondary objective To identify features of study populations such as age, hereditary risk, and adherence to interventions that are associated with the greatest treatment benefit or harm for both eczema and food allergy. SEARCH METHODS We performed an updated search of the Cochrane Skin Specialised Register, CENTRAL, MEDLINE, and Embase in September 2021. We searched two trials registers in July 2021. We checked the reference lists of included studies and relevant systematic reviews, and scanned conference proceedings to identify further references to relevant randomised controlled trials (RCTs). SELECTION CRITERIA: We included RCTs of skin care interventions that could potentially enhance skin barrier function, reduce dryness, or reduce subclinical inflammation in healthy term (> 37 weeks) infants (≤ 12 months) without pre-existing eczema, food allergy, or other skin condition. Eligible comparisons were standard care in the locality or no treatment. Types of skin care interventions could include moisturisers/emollients; bathing products; advice regarding reducing soap exposure and bathing frequency; and use of water softeners. No minimum follow-up was required. DATA COLLECTION AND ANALYSIS This is a prospective individual participant data (IPD) meta-analysis. We used standard Cochrane methodological procedures, and primary analyses used the IPD dataset. Primary outcomes were cumulative incidence of eczema and cumulative incidence of immunoglobulin (Ig)E-mediated food allergy by one to three years, both measured at the closest available time point to two years. Secondary outcomes included adverse events during the intervention period; eczema severity (clinician-assessed); parent report of eczema severity; time to onset of eczema; parent report of immediate food allergy; and allergic sensitisation to food or inhalant allergen. MAIN RESULTS We identified 33 RCTs comprising 25,827 participants. Of these, 17 studies randomising 5823 participants reported information on one or more outcomes specified in this review. We included 11 studies, randomising 5217 participants, in one or more meta-analyses (range 2 to 9 studies per individual meta-analysis), with 10 of these studies providing IPD; the remaining 6 studies were included in the narrative results only. Most studies were conducted at children's hospitals. Twenty-five studies, including all those contributing data to meta-analyses, randomised newborns up to age three weeks to receive a skin care intervention or standard infant skin care. Eight of the 11 studies contributing to meta-analyses recruited infants at high risk of developing eczema or food allergy, although the definition of high risk varied between studies. Durations of intervention and follow-up ranged from 24 hours to three years. All interventions were compared against no skin care intervention or local standard care. Of the 17 studies that reported information on our prespecified outcomes, 13 assessed emollients. We assessed most of the evidence in the review as low certainty and had some concerns about risk of bias. A rating of some concerns was most often due to lack of blinding of outcome assessors or significant missing data, which could have impacted outcome measurement but was judged unlikely to have done so. We assessed the evidence for the primary food allergy outcome as high risk of bias due to the inclusion of only one trial, where findings varied based on different assumptions about missing data. Skin care interventions during infancy probably do not change the risk of eczema by one to three years of age (risk ratio (RR) 1.03, 95% confidence interval (CI) 0.81 to 1.31; risk difference 5 more cases per 1000 infants, 95% CI 28 less to 47 more; moderate-certainty evidence; 3075 participants, 7 trials) or time to onset of eczema (hazard ratio 0.86, 95% CI 0.65 to 1.14; moderate-certainty evidence; 3349 participants, 9 trials). Skin care interventions during infancy may increase the risk of IgE-mediated food allergy by one to three years of age (RR 2.53, 95% CI 0.99 to 6.49; low-certainty evidence; 976 participants, 1 trial) but may not change risk of allergic sensitisation to a food allergen by age one to three years (RR 1.05, 95% CI 0.64 to 1.71; low-certainty evidence; 1794 participants, 3 trials). Skin care interventions during infancy may slightly increase risk of parent report of immediate reaction to a common food allergen at two years (RR 1.27, 95% CI 1.00 to 1.61; low-certainty evidence; 1171 participants, 1 trial); however, this was only seen for cow's milk, and may be unreliable due to over-reporting of milk allergy in infants. Skin care interventions during infancy probably increase risk of skin infection over the intervention period (RR 1.33, 95% CI 1.01 to 1.75; risk difference 17 more cases per 1000 infants, 95% CI one more to 38 more; moderate-certainty evidence; 2728 participants, 6 trials) and may increase the risk of infant slippage over the intervention period (RR 1.42, 95% CI 0.67 to 2.99; low-certainty evidence; 2538 participants, 4 trials) and stinging/allergic reactions to moisturisers (RR 2.24, 95% 0.67 to 7.43; low-certainty evidence; 343 participants, 4 trials), although CIs for slippages and stinging/allergic reactions were wide and include the possibility of no effect or reduced risk. Preplanned subgroup analyses showed that the effects of interventions were not influenced by age, duration of intervention, hereditary risk, filaggrin (FLG) mutation, chromosome 11 intergenic variant rs2212434, or classification of intervention type for risk of developing eczema. We could not evaluate these effects on risk of food allergy. Evidence was insufficient to show whether adherence to interventions influenced the relationship between skin care interventions and eczema or food allergy development. AUTHORS' CONCLUSIONS Based on low- to moderate-certainty evidence, skin care interventions such as emollients during the first year of life in healthy infants are probably not effective for preventing eczema; may increase risk of food allergy; and probably increase risk of skin infection. Further study is needed to understand whether different approaches to infant skin care might prevent eczema or food allergy.
Collapse
Affiliation(s)
- Maeve M Kelleher
- National Heart & Lung Institute, Section of Inflammation and Repair, Imperial College London, London, UK
| | - Rachel Phillips
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Sara J Brown
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Suzie Cro
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | | | - Karin C Lødrup Carlsen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Håvard O Skjerven
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Eva M Rehbinder
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
| | - Adrian J Lowe
- Allergy and Lung Health Unit, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Eishika Dissanayake
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Naoki Shimojo
- Center for Preventive Medical Sciences, Chiba University, Chiba, Japan
| | - Kaori Yonezawa
- Department of Midwifery and Women's Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | | | - Kumiko Morita
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Emma Axon
- Centre of Evidence Based Dermatology, University of Nottingham, Nottingham, UK
| | - Michael Cork
- Sheffield Dermatology Research, Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield, Sheffield, UK
| | - Alison Cooke
- Division of Nursing, Midwifery and Social Work, School of Health Sciences, The University of Manchester, Manchester, UK
| | - Eleanor Van Vogt
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Jochen Schmitt
- Center for Evidence-Based Healthcare, Faculty of Medicine Carl Gustav Carus, Technischen Universität (TU) Dresden, Dresden, Germany
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Scheswig-Holstein, Kiel, Germany
| | - Danielle McClanahan
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA
| | - Eric Simpson
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA
| | - Lelia Duley
- Nottingham Clinical Trials Unit, University of Nottingham, Nottingham, UK
| | - Lisa M Askie
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, Australia
| | - Hywel C Williams
- Centre of Evidence Based Dermatology, University of Nottingham, Nottingham, UK
| | - Robert J Boyle
- National Heart & Lung Institute, Section of Inflammation and Repair, Imperial College London, London, UK
- Cochrane Skin, Centre of Evidence Based Dermatology, University of Nottingham, Nottingham, UK
| |
Collapse
|
24
|
Jia G, Zhong X, Im HK, Schoettler N, Pividori M, Hogarth DK, Sperling AI, White SR, Naureckas ET, Lyttle CS, Terao C, Kamatani Y, Akiyama M, Matsuda K, Kubo M, Cox NJ, Ober C, Rzhetsky A, Solway J. Discerning asthma endotypes through comorbidity mapping. Nat Commun 2022; 13:6712. [PMID: 36344522 PMCID: PMC9640644 DOI: 10.1038/s41467-022-33628-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Asthma is a heterogeneous, complex syndrome, and identifying asthma endotypes has been challenging. We hypothesize that distinct endotypes of asthma arise in disparate genetic variation and life-time environmental exposure backgrounds, and that disease comorbidity patterns serve as a surrogate for such genetic and exposure variations. Here, we computationally discover 22 distinct comorbid disease patterns among individuals with asthma (asthma comorbidity subgroups) using diagnosis records for >151 M US residents, and re-identify 11 of the 22 subgroups in the much smaller UK Biobank. GWASs to discern asthma risk loci for individuals within each subgroup and in all subgroups combined reveal 109 independent risk loci, of which 52 are replicated in multi-ancestry meta-analysis across different ethnicity subsamples in UK Biobank, US BioVU, and BioBank Japan. Fourteen loci confer asthma risk in multiple subgroups and in all subgroups combined. Importantly, another six loci confer asthma risk in only one subgroup. The strength of association between asthma and each of 44 health-related phenotypes also varies dramatically across subgroups. This work reveals subpopulations of asthma patients distinguished by comorbidity patterns, asthma risk loci, gene expression, and health-related phenotypes, and so reveals different asthma endotypes.
Collapse
Affiliation(s)
- Gengjie Jia
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Institute of Genomics and Systems Biology, University of Chicago, Chicago, IL, 60637, USA
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong, 518120, China
| | - Xue Zhong
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Hae Kyung Im
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Nathan Schoettler
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Milton Pividori
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - D Kyle Hogarth
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Anne I Sperling
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Steven R White
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | | | | | - Chikashi Terao
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, 420-8527, Japan
- Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Yoichiro Kamatani
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masato Akiyama
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Nancy J Cox
- Department of Medicine and Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Andrey Rzhetsky
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA.
- Institute of Genomics and Systems Biology, University of Chicago, Chicago, IL, 60637, USA.
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
- Committee on Genomics, Genetics, and Systems Biology, University of Chicago, Chicago, IL, 60637, USA.
| | - Julian Solway
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
25
|
Wang H, Han H, Niu Y, Li X, Du X, Wang Q. LPP polymorphisms are risk factors for allergic rhinitis in the Chinese Han population. Cytokine 2022; 159:156027. [PMID: 36084606 DOI: 10.1016/j.cyto.2022.156027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/17/2022] [Accepted: 08/26/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Lipoma preferred partner (LPP) polymorphisms are related to immune diseases, but the role of LPP gene in the pathogenesis of allergic rhinitis (AR) is unclear. The current study aimed to explore the contribution of LPP variants to AR susceptibility in the Chinese Han population. METHODS A total of 992 healthy controls and 992 patients with AR were recruited. Agena MassARRAY system was applied for genotyping. Odds ratios (OR) and 95% confidence intervals (CI) adjusted by age, sex, and body mass index (BMI) were calculated to conduct the risk assessment of LPP variants in people with a predisposition to AR. Additionally, multifactor dimensionality reduction (MDR) was applied to identify high-order interaction models for AR risk. RESULTS We found that rs2030519-G (p = 0.027, OR: 1.15, 95% CI: 1.02-1.31), rs6780858-G (p = 0.019, OR: 1.16, 95% CI: 1.03-1.32), and rs60946162-T (p = 0.014, OR: 1.18, 95% CI: 1.03-1.34) were associated with increased susceptibility to AR. Subgroup analyses indicated the interaction of LPP polymorphisms in terms of age, gender, and BMI with AR susceptibility (p < 0.05, OR > 1). MDR analysis revealed that rs60946162 had the information gain (0.40%) of individual attribute regarding AR. CONCLUSION Our results first determined that rs2030519, rs6780858, and rs60946162 were correlated with increased susceptibility to AR in the Chinese Han population, which add to our understanding of the impact of LPP gene variants on AR development.
Collapse
Affiliation(s)
- Haiying Wang
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China
| | - Hui Han
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China
| | - Yongliang Niu
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China
| | - Xiaobo Li
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China
| | - Xintao Du
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China
| | - Qiang Wang
- Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu 719300, China.
| |
Collapse
|
26
|
Lu C, Liu Z, Liao H, Yang W, Li Q, Liu Q. Effects of early life exposure to home environmental factors on childhood allergic rhinitis: Modifications by outdoor air pollution and temperature. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 244:114076. [PMID: 36113271 DOI: 10.1016/j.ecoenv.2022.114076] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND There is growing evidence that allergic rhinitis (AR) is associated with indoor environmental factors, but their role in childhood AR during early life remains unclear. OBJECTIVE To investigate the association of preconceptional, prenatal, early postnatal, and current exposure to home environmental factors with childhood AR, and to further explore whether this association can be interacted by outdoor air pollution and temperature. METHODS A retrospective cohort study of 8689 preschool children was conducted during 2019-2020 in Changsha, China. A standard questionnaire was used to collect data on each family's health outcomes and home environments. We considered home environmental exposures during one year before conception, pregnancy, first year of life, and past year. Associations of indoor air pollution and allergens with AR were assessed by multiple logistic regression models. RESULTS Pre-birth exposure to indoor air pollution emitted by new furniture or redecoration and dampness related allergen derived from mold/damp stains and mold/damp clothes or bedding during 1 year before conception and pregnancy was significantly associated with increased AR, with adjusted ORs (95% CI) ranging from 1.35 (1.05-1.75) to 1.87 (1.55-2.27). Childhood AR was also significantly related with post-birth exposure to dampness related indoor allergen including mold/damp stains and mold/damp clothes or bedding in first year and past year and pollen allergen including total and nonflowing plants in past year, with a range of ORs (95% CI) from 1.20 (1.01-1.42) to 1.79 (1.42-2.27). We identified that pre-birth, particularly in utero exposure to both indoor air pollution from renovation and dampness related allergens, played a key role in AR development compared to post-birth exposures, and accumulative effect was observed with the highest risk of AR. High exposure to traffic-related air pollution (TRAP) including outdoor PM2.5, NO2, CO, and O3, as well as living near traffic road not only significantly increased adverse effect of home environmental factors but also decreased protective effect of household dogs on childhood AR. Early life exposure to low temperature in pregnancy and high temperature in first year significantly increased AR risk of home environmental exposure. Sensitivity analysis indicated that some sub-groups were more susceptible to AR risk of home environmental exposure. CONCLUSION Our study suggests that pre-birth exposure to home environmental factors played an important role in AR development and this effect can be interacted by TRAP and temperature, which supports a hypothesis of "(pre)fetal origin of childhood AR".
Collapse
Affiliation(s)
- Chan Lu
- XiangYa School of Public Health, Central South University, Changsha, China.
| | - Zijing Liu
- XiangYa School of Public Health, Central South University, Changsha, China.
| | - Hongsen Liao
- XiangYa School of Public Health, Central South University, Changsha, China.
| | - Wenhui Yang
- XiangYa School of Public Health, Central South University, Changsha, China.
| | - Qin Li
- XiangYa School of Public Health, Central South University, Changsha, China.
| | - Qin Liu
- XiangYa School of Public Health, Central South University, Changsha, China.
| |
Collapse
|
27
|
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
|
28
|
Do-Umehara HC, Chen C, Zhang Q, Schleimer RP, Budinger GRS, Liu J. Suppression of Allergic Asthma by Loss of Function of Miz1-Mediated Th1 Skewing. Am J Respir Cell Mol Biol 2022; 67:346-359. [PMID: 35833903 DOI: 10.1165/rcmb.2022-0135oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Asthma is the most prevalent chronic respiratory disease worldwide. There is currently no cure, and it remains an important cause of morbidity and mortality. Here we report that lung-specific loss of function of the transcription factor c-Myc-interacting zinc finger protein-1 (Miz1) upregulates the pro-T helper 1 (Th1) cytokine interleukin 12 (IL-12). Upregulation of IL-12 in turn stimulates a Th1 response, thereby counteracting T helper 2 (Th2) response and preventing the allergic response in mouse models of house dust mite (HDM)- and ovalbumin (OVA)-induced asthma. Using transgenic mice expressing Cre under a cell-specific promoter, we demonstrate that Miz1 acts in lung epithelial cells and dendritic cells in asthma. Chromatin immunoprecipitation (ChIP) coupled with high-throughput DNA sequencing (ChIP-seq) or quantitative PCR (ChIP-qPCR) reveals the binding of Miz1 on the Il12 promoter indicating direct repression of IL-12 by Miz1. Additionally, histone deacetylase 1 (HDAC1) is recruited to the Il12 promoter in a Miz1-depdenent manner, suggesting epigenetic repression of Il12 by Miz1. Furthermore, Miz1 is upregulated in human asthmatic samples as well as in asthmatic mice. Our data together suggest that Miz1 is upregulated during asthma, which in turn promotes asthma pathogenesis by preventing Th1 skewing through the transcriptional repression of IL-12.
Collapse
Affiliation(s)
| | - Cong Chen
- Northwestern University, Chicago, Illinois, United States
| | - Qiao Zhang
- Northwestern University - Chicago, 205058, Chicago, Illinois, United States
| | - Robert P Schleimer
- Feinberg School of Medicine, Northwestern University, Division of Allergy-Immunology, Chicago, Illinois, United States
| | - G R Scott Budinger
- Northwestern University, Pulmonary and Critical Care Medicine, Chicago, Illinois, United States
| | - Jing Liu
- University of Illinois at Chicago College of Medicine, 12247, Chicago, Illinois, United States;
| |
Collapse
|
29
|
RORA polymorphisms are risk factors for allergic rhinitis susceptibility in the Shaanxi Han population. Int Immunopharmacol 2022; 108:108874. [DOI: 10.1016/j.intimp.2022.108874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/13/2022] [Accepted: 05/15/2022] [Indexed: 11/19/2022]
|
30
|
Johnson RK, Brunetti T, Quinn K, Doenges K, Campbell M, Arehart C, Taub MA, Mathias RA, Reisdorph N, Barnes KC, Daya M. Discovering metabolite quantitative trait loci in asthma using an isolated population. J Allergy Clin Immunol 2022; 149:1807-1811.e16. [PMID: 34780848 PMCID: PMC9081120 DOI: 10.1016/j.jaci.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Integration of metabolomics with genetics may advance understanding of disease pathogenesis but has been underused in asthma genetic studies. OBJECTIVE We sought to discover new genetic effects in asthma and to characterize the molecular consequences of asthma genetic risk through integration with the metabolome in a homogeneous population. METHODS From fasting serum samples collected on 348 Tangier Island residents, we quantified 2612 compounds using untargeted metabolomics. Genotyping was performed using Illumina's MEGA array imputed to the TOPMed reference panel. To prioritize metabolites for genome-wide association analysis, we performed a metabolome-wide association study with asthma, selecting asthma-associated metabolites with heritability q value less than 0.01 for genome-wide association analysis. We also tested the association between all metabolites and 8451 candidate asthma single nucleotide polymorphisms previously associated with asthma in the UK Biobank. We followed up significant associations by characterizing shared genetic signal for metabolites and asthma using colocalization analysis. For detailed Methods, please see this article's Online Repository at www.jacionline.org. RESULTS A total of 60 metabolites were associated with asthma (P < .01), including 40 heritable metabolites tested in genome-wide association analysis. We observed a strong association peak for the endocannabinoid linoleoyl ethanolamide on chromosome 6 in VNN1 (P < 2.7 × 10-9). We found strong evidence (colocalization posterior probability >75%) for a shared causal variant between 3 metabolites and asthma, including the polyamine acisoga and variants in LPP, and derivative leukotriene B4 and intergenic variants in chr10p14. CONCLUSIONS We identified novel metabolite quantitative trait loci with asthma associations. Identification and characterization of these genetically driven metabolites may provide insight into the functional consequences of genetic risk factors for asthma.
Collapse
Affiliation(s)
- Randi K Johnson
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo.
| | - Tonya Brunetti
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Kevin Quinn
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Katrina Doenges
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Monica Campbell
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Christopher Arehart
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Md
| | - Rasika A Mathias
- Division of Allergy & Clinical Immunology, Department of Medicine, Johns Hopkins University, Baltimore, Md
| | - Nichole Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Kathleen C Barnes
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Michelle Daya
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| |
Collapse
|
31
|
Genome-Wide Association Study of Fluorescent Oxidation Products Accounting for Tobacco Smoking Status in Adults from the French EGEA Study. Antioxidants (Basel) 2022; 11:antiox11050802. [PMID: 35624665 PMCID: PMC9137810 DOI: 10.3390/antiox11050802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/13/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress (OS) is the main pathophysiological mechanism involved in several chronic diseases, including asthma. Fluorescent oxidation products (FlOPs), a global biomarker of damage due to OS, is of growing interest in epidemiological studies. We conducted a genome-wide association study (GWAS) of the FlOPs level in 1216 adults from the case-control and family-based EGEA study (mean age 43 years old, 51% women, and 23% current smokers) to identify genetic variants associated with FlOPs. The GWAS was first conducted in the whole sample and then stratified according to smoking status, the main exogenous source of reactive oxygen species. Among the top genetic variants identified by the three GWAS, those located in BMP6 (p = 3 × 10−6), near BMPER (p = 9 × 10−6), in GABRG3 (p = 4 × 10−7), and near ATG5 (p = 2 × 10−9) are the most relevant because of both their link to biological pathways related to OS and their association with several chronic diseases for which the role of OS in their pathophysiology has been pointed out. BMP6 and BMPER are of particular interest due to their involvement in the same biological pathways related to OS and their functional interaction. To conclude, this study, which is the first GWAS of FlOPs, provides new insights into the pathophysiology of chronic OS-related diseases.
Collapse
|
32
|
Genetic predisposition of alopecia areata in jordanians: A case-control study. Heliyon 2022; 8:e09184. [PMID: 35392398 PMCID: PMC8980757 DOI: 10.1016/j.heliyon.2022.e09184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/30/2021] [Accepted: 03/17/2022] [Indexed: 11/29/2022] Open
Abstract
Alopecia areata (AA) is a common non-scarring hair loss disease of defined patterns with varied patches size and body sites. The etiology of AA has a complex basis of autoimmunity, environment, and genetic variations. The latter factor is found to play a crucial role in AA risk. Thus, this study aimed to investigate the potential impact of specific immune-related gene polymorphisms among a cohort of Jordanian patients, which was previously reported in other populations. Blood samples of AA patients and control subjects were collected for genomic DNA (gDNA) extraction. Targeted single nucleotide polymorphisms (SNPs) of MASP2, TLR1, CTLA4, and C11orf30 were genotyped in duplicate using the Sequenom MassARRAY® system (iPLEX GOLD). Genotype and allele analysis reveals statistical differences in TLR1 rs4833095 (allele C, P = 0.044), MASP2 rs2273346 (genotype AA, P = 0.0026), and C11orf30 rs2155219 (genotype GG, P = 0.0069) distribution. These findings present the significant contribution of genetic variations in AA susceptibility in the Jordanian population, which is infrequently studied.
Collapse
|
33
|
Exploring polygenic contributors to subgroups of comorbid conditions in autism spectrum disorder. Sci Rep 2022; 12:3416. [PMID: 35233033 PMCID: PMC8888546 DOI: 10.1038/s41598-022-07399-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/10/2022] [Indexed: 11/12/2022] Open
Abstract
Individuals with autism spectrum disorder (ASD) have heterogeneous comorbid conditions. This study examined whether comorbid conditions in ASD are associated with polygenic risk scores (PRS) of ASD or PRS of comorbid conditions in non-ASD specific populations. Genome-wide single nucleotide polymorphism (SNP) data were obtained from 1386 patients with ASD from the Autism Genetic Resource Exchange (AGRE) study. After excluding individuals with missing clinical information concerning comorbid conditions, a total of 707 patients were included in the study. A total of 18 subgroups of comorbid conditions (‘topics’) were identified using a machine learning algorithm, topic modeling. PRS for ASD were computed using a genome-wide association meta-analysis of 18,381 cases and 27,969 controls. From these 18 topics, Topic 6 (over-represented by allergies) (p = 1.72 × 10−3) and Topic 17 (over-represented by sensory processing issues such as low pain tolerance) (p = 0.037) were associated with PRS of ASD. The associations between these two topics and the multi-locus contributors to their corresponding comorbid conditions based on non-ASD specific populations were further explored. The results suggest that these two topics were not associated with the PRS of allergies and chronic pain disorder, respectively. Note that characteristics of the present AGRE sample and those samples used in the original GWAS for ASD, allergies, and chronic pain disorder, may differ due to significant clinical heterogeneity that exists in the ASD population. Additionally, the AGRE sample may be underpowered and therefore insensitive to weak PRS associations due to a relatively small sample size. Findings imply that susceptibility genes of ASD may contribute more to the occurrence of allergies and sensory processing issues in individuals with ASD, compared with the susceptibility genes for their corresponding phenotypes in non-ASD individuals. Since these comorbid conditions (i.e., allergies and pain sensory issues) may not be attributable to the corresponding comorbidity-specific biological factors in non-ASD individuals, clinical management for these comorbid conditions may still depend on treatments for core symptoms of ASD.
Collapse
|
34
|
Racca F, Pellegatta G, Cataldo G, Vespa E, Carlani E, Pelaia C, Paoletti G, Messina MR, Nappi E, Canonica GW, Repici A, Heffler E. Type 2 Inflammation in Eosinophilic Esophagitis: From Pathophysiology to Therapeutic Targets. Front Physiol 2022; 12:815842. [PMID: 35095572 PMCID: PMC8790151 DOI: 10.3389/fphys.2021.815842] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated disease of the esophagus characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation, whose incidence is rising. It significantly affects patients’ quality of life and, if left untreated, results in fibrotic complications. Although broad consensus has been achieved on first-line therapy, a subset of patients remains non-responder to standard therapy. The pathogenesis of EoE is multifactorial and results from the complex, still mostly undefined, interaction between genetics and intrinsic factors, environment, and antigenic stimuli. A deep understanding of the pathophysiology of this disease is pivotal for the development of new therapies. This review provides a comprehensive description of the pathophysiology of EoE, starting from major pathogenic mechanisms (genetics, type 2 inflammation, epithelial barrier dysfunction, gastroesophageal reflux, allergens, infections and microbiota) and subsequently focusing on the single protagonists of type 2 inflammation (involved cells, cytokines, soluble effectors, surface proteins and transcription factors) that could represent present and future therapeutic targets, while summarizing previous therapeutic approaches in literature.
Collapse
Affiliation(s)
- Francesca Racca
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- *Correspondence: Francesca Racca,
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giuseppe Cataldo
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Edoardo Vespa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Carlani
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Maria Rita Messina
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
35
|
Radakovics K, Battin C, Leitner J, Geiselhart S, Paster W, Stöckl J, Hoffmann-Sommergruber K, Steinberger P. A Highly Sensitive Cell-Based TLR Reporter Platform for the Specific Detection of Bacterial TLR Ligands. Front Immunol 2022; 12:817604. [PMID: 35087538 PMCID: PMC8786796 DOI: 10.3389/fimmu.2021.817604] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/17/2021] [Indexed: 12/04/2022] Open
Abstract
Toll-like receptors (TLRs) are primary pattern recognition receptors (PRRs), which recognize conserved microbial components. They play important roles in innate immunity but also in the initiation of adaptive immune responses. Impurities containing TLR ligands are a frequent problem in research but also for the production of therapeutics since TLR ligands can exert strong immunomodulatory properties even in minute amounts. Consequently, there is a need for sensitive tools to detect TLR ligands with high sensitivity and specificity. Here we describe the development of a platform based on a highly sensitive NF-κB::eGFP reporter Jurkat JE6-1 T cell line for the detection of TLR ligands. Ectopic expression of TLRs and their coreceptors and CRISPR/Cas9-mediated deletion of endogenously expressed TLRs was deployed to generate reporter cell lines selectively expressing functional human TLR2/1, TLR2/6, TLR4 or TLR5 complexes. Using well-defined agonists for the respective TLR complexes we could demonstrate high specificity and sensitivity of the individual reporter lines. The limit of detection for LPS was below 1 pg/mL and ligands for TLR2/1 (Pam3CSK4), TLR2/6 (Fsl-1) and TLR5 (flagellin) were detected at concentrations as low as 1.0 ng/mL, 0.2 ng/mL and 10 pg/mL, respectively. We showed that the JE6-1 TLR reporter cells have the utility to characterize different commercially available TLR ligands as well as more complex samples like bacterially expressed proteins or allergen extracts. Impurities in preparations of microbial compounds as well as the lack of specificity of detection systems can lead to erroneous results and currently there is no consensus regarding the involvement of TLRs in the recognition of several molecules with proposed immunostimulatory functions. This reporter system represents a highly suitable tool for the definition of structural requirements for agonists of distinct TLR complexes.
Collapse
Affiliation(s)
- Katharina Radakovics
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Claire Battin
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Judith Leitner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabine Geiselhart
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Paster
- Clinical Cell Biology and FACS Core Unit, St. Anna Children´s Cancer Research Institute (CCRI), Vienna, Austria
| | - Johannes Stöckl
- Division Regulation of the Immune System, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Karin Hoffmann-Sommergruber
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
36
|
Ranganathan Ganakammal S, Huang K, Walkiewicz M, Xirasagar S. Genomics technologies and bioinformatics in allergy and immunology. ALLERGIC AND IMMUNOLOGIC DISEASES 2022:221-260. [DOI: 10.1016/b978-0-323-95061-9.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
37
|
Choi BY, Han M, Kwak JW, Kim TH. Genetics and Epigenetics in Allergic Rhinitis. Genes (Basel) 2021; 12:2004. [PMID: 34946955 PMCID: PMC8700872 DOI: 10.3390/genes12122004] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of allergic rhinitis is associated with genetic, environmental, and epigenetic factors. Genotyping of single nucleotide polymorphisms (SNPs) is an advanced technique in the field of molecular genetics that is closely correlated with genome-wide association studies (GWASs) in large population groups with allergic diseases. Many recent studies have paid attention to the role of epigenetics, including alteration of DNA methylation, histone acetylation, and miRNA levels in the pathogenesis of allergic rhinitis. In this review article, genetics and epigenetics of allergic rhinitis, including information regarding functions and significance of previously known and newly-discovered genes, are summarized. Directions for future genetic and epigenetic studies of allergic rhinitis are also proposed.
Collapse
Affiliation(s)
| | | | | | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea; (B.Y.C.); (M.H.); (J.W.K.)
| |
Collapse
|
38
|
Drenos F. Systems epidemiology of metabolomics measures reveals new relationships between lipoproteins and other small molecules. Metabolomics 2021; 18:1. [PMID: 34919182 PMCID: PMC8683390 DOI: 10.1007/s11306-021-01856-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 11/20/2021] [Indexed: 11/15/2022]
Abstract
INTRODUCTION The study of lipoprotein metabolism at the population level can provide valuable information for the organisation of lipoprotein related processes in the body. To use this information towards interventional hypotheses generation and testing, we need to be able to identify the mechanistic connections among the large number of observed correlations between the measured components of the system. OBJECTIVES To use population level metabolomics information to gain insight on their biochemical networks and metabolism. METHODS Genetic and metabolomics information for 230 metabolic measures, predominately lipoprotein related, from a targeted nuclear magnetic resonance approach, in two samples of an established European cohort, totalling more than 9400 individuals analysed using phenotypic and genetic correlations, as well as Mendelian Randomisation. RESULTS More than 20,500 phenotypic correlations were identified in the data, with almost 2000 also showing evidence of strong genetic correlation. Mendelian randomisation, provided evidence for a causal effect between 9496 pairs of metabolic measures, mainly between lipoprotein traits. The results provide insights on the organisation of lipoproteins in three distinct classes, the heterogeneity between HDL particles, and the association, or lack of, between CLA, glycolysis markers, such as glucose and citrate, and glycoproteins with lipids subfractions. Two examples for the use of the approach in systems biology of lipoproteins are presented. CONCLUSIONS Genetic variation can be used to infer the underlying mechanisms for the associations between lipoproteins for hypothesis generation and confirmation, and, together with biological information, to map complex biological processes.
Collapse
Affiliation(s)
- Fotios Drenos
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK.
- Institute of Cardiovascular Sciences, UCL, London, WC1E 6JF, UK.
| |
Collapse
|
39
|
The Vista of Application of Specific Anaphylaxis Accurate Diagnosis Based on DNA Single-Nucleotide Methylation Sites. CONTRAST MEDIA & MOLECULAR IMAGING 2021; 2021:8202068. [PMID: 34908915 PMCID: PMC8635942 DOI: 10.1155/2021/8202068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/21/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022]
Abstract
Anaphylaxis has rapidly spread around the world in the last several decades. Environmental factors seem to play a major role, and epigenetic marks, especially DNA methylation, get more attention. We discussed several GEO opening data classifications with TOP 100 specific methylation region values (normalized M-values on line) by machine learning, which are remarkable to classify specific anaphylaxis after monoallergen exposure. Then, we sequenced the whole-genome DNA methylation of six people (3 wormwood monoallergen atopic rhinitis patients and 3 normal-immune people) during the pollen season and analyzed the difference of the single nucleotide and DNA region. The results' divergences were obvious (the differential single nucleotides were mostly distributed in nongene regions but the differential DNA regions of GWAS, on the other hand), which may have caused most single nucleotides to be concealed in the regions' sequences. Therefore, we suggest that we should conduct more “pragmatic” and directly find special single-nucleotide changes after exposure to atopic allergens instead of complex correlativity. It is possible to try to use DNA methylation marks to accurately diagnose anaphylaxis and form a machine learning classification based on the single methylated CpGs.
Collapse
|
40
|
Kim SY, Kim EG, Kim M, Hong JY, Kim GE, Jung JH, Park M, Kim MJ, Kim YH, Sohn MH, Kim KW. Genome-wide association study identifies BTNL2 associated with atopic asthma in children. Medicine (Baltimore) 2021; 100:e27626. [PMID: 34871226 PMCID: PMC8568460 DOI: 10.1097/md.0000000000027626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 10/06/2021] [Indexed: 01/05/2023] Open
Abstract
Asthma is a heterogeneous disease characterized by chronic airway inflammation with a genetic predisposition. Butyrophilin-like 2 (BTNL2) is a member of the immunoglobulin superfamily that plays an important role in regulating T cell activation and immune homeostasis. Here, we aimed to investigate the association of the genetic variants of BTNL2 with childhood asthma and asthma-related traits by utilizing extreme asthma phenotypes and employing a genome-wide association study. Our study included 243 children with well-defined moderate to severe atopic asthma and 134 healthy children with no history of allergic diseases and allergic sensitization. DNA from these subjects was genotyped using AxiomTM Genome-Wide Array Plates. Although no single nucleotide polymorphisms (SNPs) reached a genome-wide threshold of significance, 3 SNPs, rs3817971, rs41355746, and rs41441651, at BTNL2 were significantly associated with moderate to severe atopic asthma after performing Bonferroni correction. These SNPs were also associated with the risk of allergic sensitization toward house dust mites and the presence and degree of bronchial hyperresponsiveness. Thus, we identified that BTNL2 was associated with atopic moderate to severe persistent asthma in Korean children, and this may play an important role in disease development and susceptibility.
Collapse
Affiliation(s)
- Soo Yeon Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Gyul Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mina Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jung Yeon Hong
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Cardiovascular Disease Research, Department for Chronic Disease Convergence Research, Korea National Institute of Health, Cheongju, Republic of Korea
| | - Ga Eun Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Jae Hwa Jung
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mireu Park
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Jung Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Hee Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myung Hyun Sohn
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung Won Kim
- Department of Pediatrics, Severance Hospital, Institute of Allergy, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
41
|
Childs CE, Munblit D, Ulfman L, Gómez-Gallego C, Lehtoranta L, Recker T, Salminen S, Tiemessen M, Collado MC. Potential Biomarkers, Risk Factors and their Associations with IgE-mediated Food Allergy in Early Life: A Narrative Review. Adv Nutr 2021; 13:S2161-8313(22)00081-3. [PMID: 34596662 PMCID: PMC8970818 DOI: 10.1093/advances/nmab122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. In the past few decades, the prevalence of allergic disease has been on the rise worldwide. Identified risk factors for food allergy include family history, mode of delivery, variations in infant feeding practices, prior diagnosis of other atopic diseases such as eczema, and social economic status. Identifying reliable biomarkers which predict the risk of developing food allergy in early life would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. There is also the potential to identify new therapeutic targets. This narrative review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy and synthesizes the currently available data indicating potential biomarkers. While there is a large body of research evidence available within each field of potential risk factors, there are very limited number of studies which span multiple methodological fields, for example including immunology, microbiome, genetic/epigenetic factors and dietary assessment. We recommend that further collaborative research with detailed cohort phenotyping is required to identify biomarkers, and whether these vary between at-risk populations and the wider population. The low incidence of oral food challenge confirmed food allergy in the general population, and the complexities of designing nutritional intervention studies will provide challenges for researchers to address in generating high quality, reliable and reproducible research findings. STATEMENT OF SIGNIFICANCE Food allergy affects the quality of life of millions of people worldwide and presents a significant psychological and financial burden for both national and international public health. Identifying reliable biomarkers which predict the risk of developing food allergy would be valuable in both preventing morbidity and mortality and by making current interventions available at the earliest opportunity. This review provides details on the genetic, epigenetic, dietary and microbiome influences upon the development of food allergy. This helps in identifying reliable biomarkers to predict the risk of developing food allergy, which could be valuable in both preventing morbidity and mortality and by making interventions available at the earliest opportunity.
Collapse
Affiliation(s)
- Caroline E Childs
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Daniel Munblit
- Imperial College London, London, United Kingdom,Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child’s Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia,Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | - Carlos Gómez-Gallego
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
42
|
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
|
43
|
Ding X, Zhao Y, Yuan H, Zhang Y, Gao Y. Role of PVT1 polymorphisms in the glioma susceptibility and prognosis. Eur J Cancer Prev 2021; 30:400-408. [PMID: 33443959 DOI: 10.1097/cej.0000000000000636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Genetic factors play a crucial role in the glioma risk and prognosis of glioma patients. To explore the role of plasmacytoma variant translocation 1 (PVT1) polymorphism in the susceptibility and survival of glioma in the Chinese Han population, we conducted a case-control study. METHODS The three single-nucleotide polymorphisms (SNPs) in PVT1 were genotyped using Agena MassARRAY from 575 patients with glioma and 500 healthy controls. We used the χ2 test to analyze the differences in distribution of allele and genotype between the cases and controls. Odds ratio and 95% confidence interval (CI) were calculated by logistic regression analysis to evaluate the association SNPs with glioma risk. The effects of polymorphisms and clinical features on survival of glioma patients were evaluated using the log-rank test, Kaplan-Meier and Cox regression analysis. RESULTS We found that rs13255292 was associated with a decreased risk of glioma in the recessive model in overall or male; and rs4410871 was significantly associated with an increased the risk of glioma in age ≤40 years old or female. Moreover, the extent of resection and chemotherapy were found to be key prognostic factors in survival of glioma patients. However, the gender, age, tumor grade, radiotherapy and PVT1 polymorphisms have no effect on prognosis of glioma patients. CONCLUSIONS Our results indicated that PVT1 polymorphisms (rs13255292 and rs4410871) were associated with glioma susceptibility, but have no effect on prognosis of glioma patients. Further studies with large samples are required to confirm the results.
Collapse
Affiliation(s)
| | | | | | | | - Ya Gao
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
44
|
Komlósi ZI, van de Veen W, Kovács N, Szűcs G, Sokolowska M, O'Mahony L, Akdis M, Akdis CA. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med 2021; 85:100995. [PMID: 34364680 DOI: 10.1016/j.mam.2021.100995] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic disease of the airways, which affects more than 350 million people worldwide. It is the most common chronic disease in children, affecting at least 30 million children and young adults in Europe. Asthma is a complex, partially heritable disease with a marked heterogeneity. Its development is influenced both by genetic and environmental factors. The most common, as well as the most well characterized subtype of asthma is allergic eosinophilic asthma, which is characterized by a type 2 airway inflammation. The prevalence of asthma has substantially increased in industrialized countries during the last 60 years. The mechanisms underpinning this phenomenon are incompletely understood, however increased exposure to various environmental pollutants probably plays a role. Disease inception is thought to be enabled by a disadvantageous shift in the balance between protective and harmful lifestyle and environmental factors, including exposure to protective commensal microbes versus infection with pathogens, collectively leading to airway epithelial cell damage and disrupted barrier integrity. Epithelial cell-derived cytokines are one of the main drivers of the type 2 immune response against innocuous allergens, ultimately leading to infiltration of lung tissue with type 2 T helper (TH2) cells, type 2 innate lymphoid cells (ILC2s), M2 macrophages and eosinophils. This review outlines the mechanisms responsible for the orchestration of type 2 inflammation and summarizes the novel findings, including but not limited to dysregulated epithelial barrier integrity, alarmin release and innate lymphoid cell stimulation.
Collapse
Affiliation(s)
- Zsolt I Komlósi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary.
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Nóra Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Lung Health Hospital, Munkácsy Mihály Str. 70, 2045, Törökbálint, Hungary
| | - Gergő Szűcs
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Tömő Str. 25-29, 1083, Budapest, Hungary
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Ireland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
45
|
Jamialahmadi O, Bianco C, Pelusi S, Romeo S, Valenti L. Reply to: "Polygenic risk score: A promising predictor for hepatocellular carcinoma in the population with non-alcoholic fatty liver disease". J Hepatol 2021; 74:1494-1496. [PMID: 33676949 DOI: 10.1016/j.jhep.2021.02.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Oveis Jamialahmadi
- Department of Clinical and Molecular Medicine, University of Gothenburg, Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cristiana Bianco
- Precision Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F Sforza 35, 20122, Milan, Italy
| | - Serena Pelusi
- Precision Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F Sforza 35, 20122, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Romeo
- Department of Clinical and Molecular Medicine, University of Gothenburg, Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Science, University Magna Graecia, Catanzaro, Italy
| | - Luca Valenti
- Precision Medicine - Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via F Sforza 35, 20122, Milan, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
46
|
Karunas AS, Fedorova YY, Gimalova GF, Etkina EI, Khusnutdinova EK. Association of Gasdermin B Gene GSDMB Polymorphisms with Risk of Allergic Diseases. Biochem Genet 2021; 59:1527-1543. [PMID: 33963941 DOI: 10.1007/s10528-021-10073-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 04/22/2021] [Indexed: 01/05/2023]
Abstract
The GSDMB gene encodes gasdermin B from the family of gasdermin domain-containing proteins involved in various cellular processes related to tumor development and progression, such as differentiation, cell cycle control and apoptosis. Previously, we conducted GWAS on asthma in the Volga-Ural region of Russia and found SNPs associated with asthma with genome-wide significance (rs9303277, rs8067378, rs2290400, rs7216389, rs4795405) and located in the chromosomal region 17q12-q21, which contains IKZF3 (IKAROS family zinc finger 3), ZPBP2 (zona pellucida binding protein-like), GSDMB (gasdermin B), ORMDL3 (orosomucoid 1-like 3) and LRRC3C (leucine-rich repeat-containing 3C) genes. In the present study, we investigated the association of SNPs of the GSDMB gene with the development of various allergic diseases and their combined manifestations in individuals of Russian, Tatar and Bashkir ethnic origin. Our results revealed that polymorphic variants rs7216389, rs2290400 and rs2305480 are associated with the development of allergic diseases as well as with asthma and asthma combined with allergic rhinitis. We did not reveal the association of rs7216389 and rs2290400 with the development of allergic rhinitis and atopic dermatitis in the groups of patients without asthma symptoms. This may reflect a more important role of these SNPs in the development of asthma.
Collapse
Affiliation(s)
- Alexandra S Karunas
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia. .,Saint Petersburg State University, Saint Petersburg, Russia. .,Bashkir State Medical University of the Ministry of Healthcare of the Russian Federation, Ufa, Russia.
| | - Yuliya Yu Fedorova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia. .,Saint Petersburg State University, Saint Petersburg, Russia.
| | - Galiya F Gimalova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia.,Saint Petersburg State University, Saint Petersburg, Russia
| | - Esfir I Etkina
- Bashkir State Medical University of the Ministry of Healthcare of the Russian Federation, Ufa, Russia
| | - Elza K Khusnutdinova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa, Russia.,Saint Petersburg State University, Saint Petersburg, Russia.,Bashkir State University, Ufa, Russia
| |
Collapse
|
47
|
Zhang Y, Tan M, Qian X, Li C, Yue L, Liu Y, Shi S. Interaction between early-life pet exposure and methylation pattern of ADAM33 on allergic rhinitis among children aged 3-6 years in China. Allergy Asthma Clin Immunol 2021; 17:44. [PMID: 33933154 PMCID: PMC8088023 DOI: 10.1186/s13223-021-00526-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/11/2021] [Indexed: 12/01/2022] Open
Abstract
Background Recent research has pointed out the important roles of epigenetic modifications in the development and persistence of allergic rhinitis (AR), especially in relation to DNA methylation of disease-associated genes. We investigated whether AR susceptibility genes were epigenetically regulated, and whether methylation modulation of these genes in response to early-life environment could be a molecular mechanism underlying the risk for AR onset in a cohort of children aged 3–6 years in China. Methods Peripheral blood mononuclear cell (PBMC) samples were collected from 130 children patients, aged 3–6 years and diagnosed with AR; and 154 matched controls to detect promoter methylation in 25 AR susceptibility genes with the MethylTarget approach. Methylation levels were compared for each CpG site, each amplified region, and each gene. In addition, the relationship among DNA methylation, early-life environmental risk factors and AR onset were assessed. Results Maternal allergic history (P = 0.0390) and pet exposure (P = 0.0339) were significantly associated with increased AR risk. Differential methylation analyses were successfully performed for 507 CpG sites, 34 amplified regions and 17 genes and significant hypomethylation was observed in the promoter region of ADAM33 in AR patients [multiple test-corrected (FDR) P-value < 0.05]. Spearman correlation analysis revealed that the hypomethylation of ADAM33 was significantly associated with higher eosinophil counts (Spearman’s ρ: − 0.187, P-value = 0.037). According to the results of the multiple regression analysis, after adjusting for cofounders, the interaction of early-life pet exposure with methylation level of ADAM33 increased the risk for AR onset 1.423 times more in children (95% CI = 0.0290–4.109, P-value = 0.005). Conclusion This study provides evidence that early-life pet exposure and low methylation level of ADAM33 increase AR risk in children, and the interaction between pet exposure and methylation level of ADAM33 may play an important role in the development of AR.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Meiyu Tan
- Department of Laboratory Diagnosis, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xiaoqiong Qian
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Cong Li
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Lei Yue
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yuehong Liu
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Song Shi
- Department of Otorhinolaryngology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
48
|
Ma C, Storer CE, Chandran U, LaFramboise WA, Petrosko P, Frank M, Hartman DJ, Pantanowitz L, Haritunians T, Head RD, Liu TC. Crohn's disease-associated ATG16L1 T300A genotype is associated with improved survival in gastric cancer. EBioMedicine 2021; 67:103347. [PMID: 33906066 PMCID: PMC8099593 DOI: 10.1016/j.ebiom.2021.103347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND A non-synonymous single nucleotide polymorphism of the ATG16L1 gene, T300A, is a major Crohn's disease (CD) susceptibility allele, and is known to be associated with increased apoptosis induction in the small intestinal crypt base in CD subjects and mouse models. We hypothesized that ATG16L1 T300A genotype also correlates with increased tumor apoptosis and therefore could lead to superior clinical outcome in cancer subjects. METHODS T300A genotyping by Taqman assay was performed for gastric carcinoma subjects who underwent resection from two academic medical centers. Transcriptomic analysis was performed by RNA-seq on formalin-fixed paraffin-embedded cancerous tissue. Tumor apoptosis and autophagy were determined by cleaved caspase-3 and p62 immunohistochemistry, respectively. The subjects' genotypes were correlated with demographics, various histopathologic features, transcriptome, and clinical outcome. FINDINGS Of the 220 genotyped subjects, 163 (74%) subjects carried the T300A allele(s), including 55 (25%) homozygous and 108 (49%) heterozygous subjects. The T300A/T300A subjects had superior overall survival than the other groups. Their tumors were associated with increased CD-like lymphoid aggregates and increased tumor apoptosis without concurrent increase in tumor mitosis or defective autophagy. Transcriptomic analysis showed upregulation of WNT/β-catenin signaling and downregulation of PPAR, EGFR, and inflammatory chemokine pathways in tumors of T300A/T300A subjects. INTERPRETATION Gastric carcinoma of subjects with the T300A/T300A genotype is associated with repressed EGFR and PPAR pathways, increased tumor apoptosis, and improved overall survival. Genotyping gastric cancer subjects may provide additional insight for clinical stratification.
Collapse
Affiliation(s)
- Changqing Ma
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-610, Pittsburgh, PA 15213, United States.
| | - Chad E Storer
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Uma Chandran
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - William A LaFramboise
- UPMC Hillman Cancer Center, Cancer Genomics Facility, Pittsburgh, PA 15232, United States
| | - Patricia Petrosko
- UPMC Hillman Cancer Center, Cancer Genomics Facility, Pittsburgh, PA 15232, United States
| | - Madison Frank
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-610, Pittsburgh, PA 15213, United States
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-610, Pittsburgh, PA 15213, United States
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh School of Medicine, 200 Lothrop Street, A-610, Pittsburgh, PA 15213, United States
| | - Talin Haritunians
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Ta-Chiang Liu
- Departments of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8118, Saint Louis, MO 63110, United States.
| |
Collapse
|
49
|
The intersect of genetics, environment, and microbiota in asthma-perspectives and challenges. J Allergy Clin Immunol 2021; 147:781-793. [PMID: 33678251 DOI: 10.1016/j.jaci.2020.08.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/07/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023]
Abstract
In asthma, a significant portion of the interaction between genetics and environment occurs through microbiota. The proposed mechanisms behind this interaction are complex and at times contradictory. This review covers recent developments in our understanding of this interaction: the "microbial hypothesis" and the "farm effect"; the role of endotoxin and genetic variation in pattern recognition systems; the interaction with allergen exposure; the additional involvement of host gut and airway microbiota; the role of viral respiratory infections in interaction with the 17q21 and CDHR3 genetic loci; and the importance of in utero and early-life timing of exposures. We propose a unified framework for understanding how all these phenomena interact to drive asthma pathogenesis. Finally, we point out some future challenges for continued research in this field, in particular the need for multiomic integration, as well as the potential utility of asthma endotyping.
Collapse
|
50
|
AllerScreener – A Server for Allergenicity and Cross-Reactivity Prediction. CYBERNETICS AND INFORMATION TECHNOLOGIES 2020. [DOI: 10.2478/cait-2020-0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Abstract
Allergenicity of proteins is a subtle property encoded in their structures. The prediction of allergenicity of novel proteins saves time and resources for subsequent experimental work. In the host antigen-presenting cells, the allergens are processed as antigens by the means of Human Leukocyte Antigens (HLA) class II proteins. Sometimes, people allergic to a given protein show allergic reaction to a different protein, even when the two proteins have different routes of exposure. This phenomenon is termed cross-reactivity. Here, we describe a server for allergenicity and cross-reactivity prediction based on the abilities of allergenic proteins to generate binders to HLA class II proteins. The generated peptides are compared to HLA binders originating from known allergens. As a result, the server returns a list of common binders, origin proteins, and species. Different species generate common HLA binders and this determines their cross-reactivity. The server is named AllerScreener and is freely accessible at:
http://www.ddg-pharmfac.net/AllerScreener
.
Collapse
|