1
|
Zhao SS, Yiu ZZN. Genetically proxied IL-6 receptor inhibition is associated with increased risk of atopic dermatitis. J Allergy Clin Immunol 2024; 154:666-669. [PMID: 38801923 DOI: 10.1016/j.jaci.2024.05.016] [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: 12/15/2023] [Revised: 04/08/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Dermatitis has been reported after initiation of IL-6 receptor (IL-6R) inhibitors (IL-6Ri), while genetic association studies of atopic dermatitis (AD) have implicated IL-6R pathway signaling. However, causality remains unclear. As the indications for IL-6Ri expand, so do the clinical importance of determining whether there is mechanistic evidence linking it to AD. OBJECTIVE Our aim was to examine the association between IL-6Ri and risk of AD. METHODS To genetically mimic IL-6Ri, we selected single-nucleotide polymorphisms within or near the IL6R gene associated with C-reactive protein at genome-wide significance among 343,524 individuals. Genetic data were obtained from 10,788 individuals with AD and 30,047 controls of European ancestry. We used inverse variance-weighted and pleiotropy-robust methods and examined genetic confounding using colocalization. Analyses were replicated by using 13,473 Finnish and 2,385 East Asian individuals with AD. The results from 3 independent analyses were pooled by meta-analysis. RESULTS Genetically proxied IL-6Ri was associated with increased risk of AD (odds ratio [OR] = 1.78 per 4.4-mg/L reduction in C-reactive protein level [95% CI = 1.28-2.48] [P = 6.5 × 10-4]). The results were replicated using Finnish outcome data (OR = 2.07 [95% CI = 1.58-2.72] [P = 1.57 × 10-7]) and Eastern Asian data (OR = 1.68 [95% CI = 1.12-2.54] [P = .013]). Meta-analysis of 3 independent populations (OR = 1.89 [95% CI = 1.57-2.28] [P = 2.68 × 10-11]) showed no evidence of heterogeneity (P = .65). We found no statistical evidence for pleiotropy or genetic confounding. CONCLUSION This genetic investigation provides consistent evidence (across independent multiancestry populations) that IL-6R signaling is causally implicated in AD susceptibility. Clinicians should remain vigilant for adverse effects resembling AD when using IL-6R inhibitors for immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Sizheng Steven Zhao
- Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biological Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| | - Zenas Z N Yiu
- Dermatology Centre, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
2
|
Li CY, Chang WC, Chen MH, Tu PC, Chen TL, Chen CC, Chang YT, Chen YY, Bai YM. Correlation of Disease Severity, Proinflammatory Cytokines, and Reduced Brain Gray Matter Volumes in Patients with Atopic Dermatitis. Dermatitis 2024; 35:489-497. [PMID: 38634841 DOI: 10.1089/derm.2023.0340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Background: Atopic dermatitis (AD) is a chronic inflammatory skin disease. However, few studies have investigated brain changes associated with chronic inflammation. We hypothesized that chronic inflammation might be related to brain structural alterations in patients with AD. Objectives: To investigate the association between disease severity (Eczema Area and Severity Index [EASI]), proinflammatory cytokines, and differences in brain gray matter (GM) volume in patients with AD. Methods: Nineteen patients with AD and 19 age- and sex-matched healthy subjects were enrolled. All participants underwent clinical assessment and brain magnetic resonance imaging. Voxel-based morphometry was performed to analyze GM volume differences. Results: Patients with AD exhibited significantly decreased GM volume in many brain regions, such as bilateral precentral gyrus, right frontal pole, and right middle temporal gyrus (P < 0.001), compared with healthy subjects. Notably, in patients with AD, the GM volume in right middle temporal gyrus was negatively associated with both EASI score and proinflammatory cytokines (sIL-2R [soluble interleukin 2 receptor] and TNF-α receptor-1), whereas the GM volume in left precentral gyrus was negatively associated with both EASI score and proinflammatory cytokines (sIL-2R and CRP). Conclusion: Patients with AD demonstrated significant brain GM volume reduction in many brain regions, which is related to disease severity and proinflammatory cytokines.
Collapse
Affiliation(s)
- Cheng-Yuan Li
- From the Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dermatology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wan-Chen Chang
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mu-Hong Chen
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Pei-Chi Tu
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Philosophy of Mind and Cognition, National Yang-Ming University, Taipei, Taiwan
| | - Tai-Li Chen
- From the Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dermatology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Chiang Chen
- From the Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dermatology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yun-Ting Chang
- From the Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Dermatology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - You-Yin Chen
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ya-Mei Bai
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
3
|
Wu W, Su H, Chen Z, Wei S. Exploring novel drug targets for atopic dermatitis through plasma proteome with genome. Arch Dermatol Res 2024; 316:521. [PMID: 39136778 DOI: 10.1007/s00403-024-03262-z] [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: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 10/25/2024]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease with a complex and heterogeneous clinical presentation, leading to treatment limitations. Therefore, there is an urgent demand for new therapeutic drug targets. This study utilized Summary-data-based Mendelian randomization (SMR) to identify potential drug targets for AD. Summary statistics for 2,940 human plasma proteins were obtained from the UK Biobank, while AD statistics came from the Early Genetics and Epidemiology of Life Processes consortium and the FinnGen consortium. Furthermore, subsequent colocalization analyses confirmed the causal roles of candidate proteins. Moreover, Phenome-Wide Association Studies (PheWAS), protein-protein interaction (PPI), enrichment analysis, and single cell-type expression analysis provided additional insights. Additionally, drug prediction, druggability prediction, and molecular docking informed the discovery of novel drug targets. SMR analysis showed that eight plasma proteins were causally associated with AD: PVALB and TST were associated with a reduced risk of AD, while CA14, ECM1, IL22, IL6R, IL18R1, and MMP12 were associated with an increased risk of AD. Colocalization analysis confirmed significant associations for TST, IL22, and CA14. PheWAS further revealed that candidate drug targets were mainly linked to other allergic diseases. The corresponding protein-coding genes are predominantly expressed in melanocytes, T cells, and macrophages in skin tissue. Importantly, these proteins were identified to be involved in cytokine-cytokine receptor interaction, Th17 cell differentiation, and the JAK-STAT signaling pathway. All of these proteins are druggable, and six of them show great potential as drug targets. In conclusion, this study identified eight plasma proteins causally associated with AD and provided new insights into the etiology and potential drug targets for AD.
Collapse
Affiliation(s)
- Wenhua Wu
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hao Su
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zhengrui Chen
- The Second School of Clinical Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shanshan Wei
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Cao Z, Li Q, Li Y, Wu J. Identification of plasma protein markers of allergic disease risk: a mendelian randomization approach to proteomic analysis. BMC Genomics 2024; 25:503. [PMID: 38773393 PMCID: PMC11110418 DOI: 10.1186/s12864-024-10412-0] [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: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND While numerous allergy-related biomarkers and targeted treatment strategies have been developed and employed, there are still signifcant limitations and challenges in the early diagnosis and targeted treatment for allegic diseases. Our study aims to identify circulating proteins causally associated with allergic disease-related traits through Mendelian randomization (MR)-based analytical framework. METHODS Large-scale cis-MR was employed to estimate the effects of thousands of plasma proteins on five main allergic diseases. Additional analyses including MR Steiger analyzing and Bayesian colocalisation, were performed to test the robustness of the associations; These findings were further validated utilizing meta-analytical methods in the replication analysis. Both proteome- and transcriptome-wide association studies approach was applied, and then, a protein-protein interaction was conducted to examine the interplay between the identified proteins and the targets of existing medications. RESULTS Eleven plasma proteins were identified with links to atopic asthma (AA), atopic dermatitis (AD), and allergic rhinitis (AR). Subsequently, these proteins were classified into four distinct target groups, with a focus on tier 1 and 2 targets due to their higher potential to become drug targets. MR analysis and extra validation revealed STAT6 and TNFRSF6B to be Tier 1 and IL1RL2 and IL6R to be Tier 2 proteins with the potential for AA treatment. Two Tier 1 proteins, CRAT and TNFRSF6B, and five Tier 2 proteins, ERBB3, IL6R, MMP12, ICAM1, and IL1RL2, were linked to AD, and three Tier 2 proteins, MANF, STAT6, and TNFSF8, to AR. CONCLUSION Eleven Tier 1 and 2 protein targets that are promising drug target candidates were identified for AA, AD, and AR, which influence the development of allergic diseases and expose new diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Ziqin Cao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410000, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Qiangxiang Li
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China
| | - Yajia Li
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Jianhuang Wu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha, 410000, China.
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| |
Collapse
|
5
|
Dmytrzak A, Lewandowska K, Boroń A, Łoniewska B, Grzesch N, Brodkiewicz A, Clark JSC, Ciechanowicz A, Kostrzewa-Nowak D. No Association of Polymorphisms in the Genes Encoding Interleukin-6 and Interleukin-6 Receptor Subunit Alpha with the Risk of Keloids in Polish Patients. Int J Mol Sci 2024; 25:5284. [PMID: 38791322 PMCID: PMC11121548 DOI: 10.3390/ijms25105284] [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: 04/11/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
A keloid is a benign fibroproliferative hypertrophy of scar tissue that extends outside the original wound and invades adjacent healthy skin. Keloid formation is thought to be a complex process including overactivity of the interleukin-6 signaling pathway and genetic susceptibility. The aim of the study was to investigate possible associations between rs1800797, rs1800796, and rs1800795 polymorphisms in the promoter of the IL6 gene encoding interleukin-6 and the rs2228145 polymorphism in the IL6R gene encoding the interleukin-6 receptor subunit alpha with the predisposition to keloids in Polish patients. The genetic polymorphisms were identified either using Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) or sequencing of samples of genomic DNA extracted from blood leukocytes of 86 adult patients with keloids and 100 newborns comprising a control group. No significant differences in the distributions of IL6 or IL6R alleles or genotypes were found between keloid patients and newborn controls. There were also no significant differences between both groups in the distribution of IL6 haplotypes. The IL6 rs1800797, rs1800796 and rs1800795 and IL6R rs2228145 polymorphisms were not found to predispose individuals in the study group to keloids. IL6 promoter haplotypes were not found to be associated with a higher risk of keloids in the studied group.
Collapse
Affiliation(s)
| | - Klaudyna Lewandowska
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| | - Agnieszka Boroń
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| | - Beata Łoniewska
- Department of Neonatal Diseases, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Natalie Grzesch
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| | - Andrzej Brodkiewicz
- Department of Pediatrics, Child Nephrology, Dialysotherapy and Management of Acute Poisoning, Pomeranian Medical University, 70-780 Szczecin, Poland;
| | - Jeremy S. C. Clark
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| | - Andrzej Ciechanowicz
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| | - Dorota Kostrzewa-Nowak
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.L.); (A.B.); (N.G.); (J.S.C.C.); (A.C.)
| |
Collapse
|
6
|
Schuler CF, Tsoi LC, Billi AC, Harms PW, Weidinger S, Gudjonsson JE. Genetic and Immunological Pathogenesis of Atopic Dermatitis. J Invest Dermatol 2024; 144:954-968. [PMID: 38085213 PMCID: PMC11040454 DOI: 10.1016/j.jid.2023.10.019] [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: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024]
Abstract
Type 2 immune-mediated diseases give a clear answer to the issue of nature (genetics) versus nurture (environment). Both genetics and environment play vital complementary roles in the development of atopic dermatitis (AD). As a key component of the atopic march, AD demonstrates the interactive nature of genetic and environmental contributions to atopy. From sequence variants in the epithelial barrier gene encoding FLG to the hygiene hypothesis, AD combines a broad array of contributions into a single syndrome. This review will focus on the genetic contribution to AD and where genetics facilitates the elicitation or enhancement of AD pathogenesis.
Collapse
Affiliation(s)
- Charles F Schuler
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lam C Tsoi
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison C Billi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul W Harms
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephan Weidinger
- Department of Dermatology, Venereology, and Allergology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Johann E Gudjonsson
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
7
|
Saheb Kashaf S, Kong HH. Adding Fuel to the Fire? The Skin Microbiome in Atopic Dermatitis. J Invest Dermatol 2024; 144:969-977. [PMID: 38530677 PMCID: PMC11034722 DOI: 10.1016/j.jid.2024.01.011] [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: 12/08/2023] [Accepted: 01/07/2024] [Indexed: 03/28/2024]
Abstract
Atopic dermatitis (AD) is a multifactorial, heterogeneous disease characterized by epidermal barrier dysfunction, immune system dysregulation, and skin microbiome alterations. Skin microbiome studies in AD have demonstrated that disease flares are associated with microbial shifts, particularly Staphylococcus aureus predominance. AD-associated S. aureus strains differ from those in healthy individuals across various genomic loci, including virulence factors, adhesion proteins, and proinflammatory molecules-which may contribute to complex microbiome barrier-immune system interactions in AD. Different microbially based treatments for AD have been explored, and their future therapeutic successes will depend on a deeper understanding of the potential microbial contributions to the disease.
Collapse
Affiliation(s)
- Sara Saheb Kashaf
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA; Pritzker School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Heidi H Kong
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
8
|
Rusiñol L, Puig L. Multi-Omics Approach to Improved Diagnosis and Treatment of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2024; 25:1042. [PMID: 38256115 PMCID: PMC10815999 DOI: 10.3390/ijms25021042] [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: 12/16/2023] [Revised: 01/07/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Psoriasis and atopic dermatitis fall within the category of cutaneous immune-mediated inflammatory diseases (IMIDs). The prevalence of IMIDs is increasing in industrialized societies, influenced by both environmental changes and a genetic predisposition. However, the exact immune factors driving these chronic, progressive diseases are not fully understood. By using multi-omics techniques in cutaneous IMIDs, it is expected to advance the understanding of skin biology, uncover the underlying mechanisms of skin conditions, and potentially devise precise and personalized approaches to diagnosis and treatment. We provide a narrative review of the current knowledge in genomics, epigenomics, and proteomics of atopic dermatitis and psoriasis. A literature search was performed for articles published until 30 November 2023. Although there is still much to uncover, recent evidence has already provided valuable insights, such as proteomic profiles that permit differentiating psoriasis from mycosis fungoides and β-defensin 2 correlation to PASI and its drop due to secukinumab first injection, among others.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain
| |
Collapse
|
9
|
Budu-Aggrey A, Kilanowski A, Sobczyk MK, Shringarpure SS, Mitchell R, Reis K, Reigo A, Mägi R, Nelis M, Tanaka N, Brumpton BM, Thomas LF, Sole-Navais P, Flatley C, Espuela-Ortiz A, Herrera-Luis E, Lominchar JVT, Bork-Jensen J, Marenholz I, Arnau-Soler A, Jeong A, Fawcett KA, Baurecht H, Rodriguez E, Alves AC, Kumar A, Sleiman PM, Chang X, Medina-Gomez C, Hu C, Xu CJ, Qi C, El-Heis S, Titcombe P, Antoun E, Fadista J, Wang CA, Thiering E, Wu B, Kress S, Kothalawala DM, Kadalayil L, Duan J, Zhang H, Hadebe S, Hoffmann T, Jorgenson E, Choquet H, Risch N, Njølstad P, Andreassen OA, Johansson S, Almqvist C, Gong T, Ullemar V, Karlsson R, Magnusson PKE, Szwajda A, Burchard EG, Thyssen JP, Hansen T, Kårhus LL, Dantoft TM, Jeanrenaud ACSN, Ghauri A, Arnold A, Homuth G, Lau S, Nöthen MM, Hübner N, Imboden M, Visconti A, Falchi M, Bataille V, Hysi P, Ballardini N, Boomsma DI, Hottenga JJ, Müller-Nurasyid M, Ahluwalia TS, Stokholm J, Chawes B, Schoos AMM, Esplugues A, Bustamante M, Raby B, Arshad S, German C, Esko T, Milani LA, Metspalu A, Terao C, Abuabara K, Løset M, Hveem K, Jacobsson B, Pino-Yanes M, Strachan DP, Grarup N, Linneberg A, Lee YA, Probst-Hensch N, Weidinger S, Jarvelin MR, Melén E, Hakonarson H, Irvine AD, Jarvis D, Nijsten T, Duijts L, Vonk JM, Koppelmann GH, Godfrey KM, Barton SJ, Feenstra B, Pennell CE, Sly PD, Holt PG, Williams LK, Bisgaard H, Bønnelykke K, Curtin J, Simpson A, Murray C, Schikowski T, Bunyavanich S, Weiss ST, Holloway JW, Min JL, Brown SJ, Standl M, Paternoster L. European and multi-ancestry genome-wide association meta-analysis of atopic dermatitis highlights importance of systemic immune regulation. Nat Commun 2023; 14:6172. [PMID: 37794016 PMCID: PMC10550990 DOI: 10.1038/s41467-023-41180-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 08/24/2023] [Indexed: 10/06/2023] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory skin condition and prior genome-wide association studies (GWAS) have identified 71 associated loci. In the current study we conducted the largest AD GWAS to date (discovery N = 1,086,394, replication N = 3,604,027), combining previously reported cohorts with additional available data. We identified 81 loci (29 novel) in the European-only analysis (which all replicated in a separate European analysis) and 10 additional loci in the multi-ancestry analysis (3 novel). Eight variants from the multi-ancestry analysis replicated in at least one of the populations tested (European, Latino or African), while two may be specific to individuals of Japanese ancestry. AD loci showed enrichment for DNAse I hypersensitivity and eQTL associations in blood. At each locus we prioritised candidate genes by integrating multi-omic data. The implicated genes are predominantly in immune pathways of relevance to atopic inflammation and some offer drug repurposing opportunities.
Collapse
Affiliation(s)
- Ashley Budu-Aggrey
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, England
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Anna Kilanowski
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, Munich, Germany
- Pettenkofer School of Public Health, Ludwig-Maximilians University Munich, Munich, Germany
| | - Maria K Sobczyk
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, England
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | | | - Ruth Mitchell
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, England
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Kadri Reis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anu Reigo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mari Nelis
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Core Facility of Genomics, University of Tartu, Tartu, Estonia
| | - Nao Tanaka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, 7030, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, 7600, Norway
- Clinic of Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, 7030, Norway
| | - Laurent F Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, 7030, Norway
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Pol Sole-Navais
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christopher Flatley
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Espuela-Ortiz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Esther Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Jesus V T Lominchar
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Ingo Marenholz
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Aleix Arnau-Soler
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ayoung Jeong
- Swiss Tropical and Public Health Institute, CH-4123, Basel, Switzerland
- University of Basel, CH-4001, Basel, Switzerland
| | - Katherine A Fawcett
- Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Hansjorg Baurecht
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Elke Rodriguez
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Ashish Kumar
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Solna, Sweden
| | - Patrick M Sleiman
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Rhythm Pharmaceuticals, 222 Berkley Street, Boston, 02116, USA
| | - Xiao Chang
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Carolina Medina-Gomez
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Chen Hu
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Cheng-Jian Xu
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- Centre for Individualized Infection Medicine, CiiM, a joint venture between Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Cancan Qi
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Sarah El-Heis
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Philip Titcombe
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Elie Antoun
- Faculty of Medicine, University of Southampton, Southampton, UK
- Institute of Developmental Sciences, University of Southampton, Southampton, UK
| | - João Fadista
- Department of Bioinformatics & Data Mining, Måløv, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Carol A Wang
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Elisabeth Thiering
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, Munich, Germany
| | - Baojun Wu
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Medicine, Henry Ford Health, Detroit, MI, 48104, USA
| | - Sara Kress
- Environmental Epidemiology of Lung, Brain and Skin Aging, IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Dilini M Kothalawala
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Latha Kadalayil
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jiasong Duan
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas Hoffmann
- Institute for Human Genetics, UCSF, San Francisco, CA, 94143, USA
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, 94158, USA
| | | | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Neil Risch
- Institute for Human Genetics, UCSF, San Francisco, CA, 94143, USA
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, CA, 94158, USA
| | - Pål Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, NO-5021, Bergen, Norway
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, 0450, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, 0450, Oslo, Norway
| | - Stefan Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, NO-5020, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, NO-5021, Bergen, Norway
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Pediatric Lung and Allergy Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Tong Gong
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Vilhelmina Ullemar
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Agnieszka Szwajda
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Esteban G Burchard
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Jacob P Thyssen
- Department of Dermatology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Line L Kårhus
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Thomas M Dantoft
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
| | - Alexander C S N Jeanrenaud
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ahla Ghauri
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Arnold
- Clinic and Polyclinic of Dermatology, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Susanne Lau
- Department of Pediatric Respiratory Medicine, Immunology, and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Norbert Hübner
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
| | - Medea Imboden
- Swiss Tropical and Public Health Institute, CH-4123, Basel, Switzerland
- University of Basel, CH-4001, Basel, Switzerland
| | - Alessia Visconti
- Department of Twin Research & Genetics Epidemiology, Kings College London, London, UK
| | - Mario Falchi
- Department of Twin Research & Genetics Epidemiology, Kings College London, London, UK
| | - Veronique Bataille
- Department of Twin Research & Genetics Epidemiology, Kings College London, London, UK
- Dermatology Department, West Herts NHS Trust, Watford, UK
| | - Pirro Hysi
- Department of Twin Research & Genetics Epidemiology, Kings College London, London, UK
| | - Natalia Ballardini
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Solna, Sweden
| | - Dorret I Boomsma
- Dept Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
- Institute for Health and Care Research (EMGO), VU University, Amsterdam, the Netherlands
| | - Jouke J Hottenga
- Dept Biological Psychology, Netherlands Twin Register, VU University, Amsterdam, the Netherlands
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Tarunveer S Ahluwalia
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie M Schoos
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Ana Esplugues
- Nursing School, University of Valencia, FISABIO-University Jaume I-University of Valencia, Valencia, Spain
- Joint Research Unit of Epidemiology and Environmental Health, CIBERESP, Valencia, Spain
| | - Mariona Bustamante
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Benjamin Raby
- Channing Division of Network Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Syed Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- David Hide Asthma and Allergy Research Centre, Isle of Wight, UK
| | | | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lili A Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Katrina Abuabara
- Department of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Mari Løset
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, 7030, Norway
- Department of Dermatology, Clinic of Orthopaedy, Rheumatology and Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, 7030, Norway
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, Norway
| | - Bo Jacobsson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Genetics and Bioinformatics, Norwegian Institute of Public Health, Oslo, Norway
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, La Laguna, Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - David P Strachan
- Population Health Research Institute, St George's, University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, København, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Young-Ae Lee
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Clinic for Pediatric Allergy, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nicole Probst-Hensch
- Swiss Tropical and Public Health Institute, CH-4123, Basel, Switzerland
- University of Basel, CH-4001, Basel, Switzerland
| | - Stephan Weidinger
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health,Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Erik Melén
- Department of Clinical Science and Education Södersjukhuset, Karolinska Institutet, Solna, Sweden
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Divisions of Human Genetics and Pulmonary Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Faculty of Medicine, University of Iceland, 101, Reykjavík, Iceland
| | - Alan D Irvine
- Department of Clinical Medicine, Trinity College, Dublin, Ireland
| | - Deborah Jarvis
- Respiratory Epidemiology, Occupational Medicine and Public Health, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Medical Research Council and Public Health England Centre for Environment and Health, London, United Kingdom
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Judith M Vonk
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Gerard H Koppelmann
- University of Groningen, University Medical Center Groningen, Department of Pediatric Pulmonology and Pediatric Allergy, Beatrix Children's Hospital, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | - Craig E Pennell
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter D Sly
- Children's Health and Environment Program, Child Health Research Centre, The University of Queensland, South Brisbane, 4101, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, 4072, QLD, Australia
| | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research (CIGMA), Department of Medicine, Henry Ford Health, Detroit, MI, 48104, USA
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - John Curtin
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, England
| | - Angela Simpson
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, England
| | - Clare Murray
- Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, England
| | - Tamara Schikowski
- Environmental Epidemiology of Lung, Brain and Skin Aging, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Supinda Bunyavanich
- Division of Allergy and Immunology, Department of Pediatrics, and Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Josine L Min
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, England
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England
| | - Sara J Brown
- Centre for Genomics and Experimental Medicine, Institute for Genetics and Cancer, University of Edinburgh, Crewe Road, Edinburgh, UK EH4 2XU, Scotland
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Lung Research (DZL), Munich, Germany
| | - Lavinia Paternoster
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, England.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, England.
| |
Collapse
|
10
|
Pessôa R, Clissa PB, Sanabani SS. The Interaction between the Host Genome, Epigenome, and the Gut-Skin Axis Microbiome in Atopic Dermatitis. Int J Mol Sci 2023; 24:14322. [PMID: 37762624 PMCID: PMC10532357 DOI: 10.3390/ijms241814322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that occurs in genetically predisposed individuals. It involves complex interactions among the host immune system, environmental factors (such as skin barrier dysfunction), and microbial dysbiosis. Genome-wide association studies (GWAS) have identified AD risk alleles; however, the associated environmental factors remain largely unknown. Recent evidence suggests that altered microbiota composition (dysbiosis) in the skin and gut may contribute to the pathogenesis of AD. Examples of environmental factors that contribute to skin barrier dysfunction and microbial dysbiosis in AD include allergens, irritants, pollution, and microbial exposure. Studies have reported alterations in the gut microbiome structure in patients with AD compared to control subjects, characterized by increased abundance of Clostridium difficile and decreased abundance of short-chain fatty acid (SCFA)-producing bacteria such as Bifidobacterium. SCFAs play a critical role in maintaining host health, and reduced SCFA production may lead to intestinal inflammation in AD patients. The specific mechanisms through which dysbiotic bacteria and their metabolites interact with the host genome and epigenome to cause autoimmunity in AD are still unknown. By understanding the combination of environmental factors, such as gut microbiota, the genetic and epigenetic determinants that are associated with the development of autoantibodies may help unravel the pathophysiology of the disease. This review aims to elucidate the interactions between the immune system, susceptibility genes, epigenetic factors, and the gut microbiome in the development of AD.
Collapse
Affiliation(s)
- Rodrigo Pessôa
- Postgraduate Program in Translational Medicine, Department of Medicine, Federal University of Sao Paulo (UNIFESP), Sao Paulo 04039-002, Brazil;
| | | | - Sabri Saeed Sanabani
- Laboratory of Medical Investigation LIM-56, Division of Dermatology, Medical School, University of Sao Paulo, Sao Paulo 05508-220, Brazil
- Laboratory of Medical Investigation Unit 03, Clinics Hospital, Faculty of Medicine, University of Sao Paulo, Sao Paulo 05403-000, Brazil
- Laboratory of Dermatology and Immunodeficiency LIM56/03, Instituto de Medicina Tropical de Sao Paulo, Faculdade de Medicina, University of Sao Paulo, Av. Dr. Eneas de Carvalho Aguiar, 470 3º Andar, Sao Paulo 05403-000, Brazil
| |
Collapse
|
11
|
Ma CS. T-helper-2 cells and atopic disease: lessons learnt from inborn errors of immunity. Curr Opin Immunol 2023; 81:102298. [PMID: 36870225 DOI: 10.1016/j.coi.2023.102298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 03/06/2023]
Abstract
Inborn errors of immunity (IEI) are caused by monogenic variants that affect the host response to bacterial, viral, and fungal pathogens. As such, individuals with IEI often present with severe, recurrent, and life-threatening infections. However, the spectrum of disease due to IEI is very broad and extends to include autoimmunity, malignancy, and atopic diseases such as eczema, atopic dermatitis, and food and environmental allergies. Here, I review IEI that affect cytokine signaling pathways that dysregulate CD4+ T-cell differentiation, resulting in increased T-helper-2 (Th2) cell development, function, and pathogenicity. These are elegant examples of how rare IEI can provide unique insights into more common pathologies such as allergic disease that are impacting the general population at increased frequency.
Collapse
Affiliation(s)
- Cindy S Ma
- Garvan Institute of Medical Research, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Australia.
| |
Collapse
|
12
|
Fadadu RP, Abuabara K, Balmes JR, Hanifin JM, Wei ML. Air Pollution and Atopic Dermatitis, from Molecular Mechanisms to Population-Level Evidence: A Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2526. [PMID: 36767891 PMCID: PMC9916398 DOI: 10.3390/ijerph20032526] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
Atopic dermatitis (AD) has increased in prevalence to become the most common inflammatory skin condition globally, and geographic variation and migration studies suggest an important role for environmental triggers. Air pollution, especially due to industrialization and wildfires, may contribute to the development and exacerbation of AD. We provide a comprehensive, multidisciplinary review of existing molecular and epidemiologic studies on the associations of air pollutants and AD symptoms, prevalence, incidence, severity, and clinic visits. Cell and animal studies demonstrated that air pollutants contribute to AD symptoms and disease by activating the aryl hydrocarbon receptor pathway, promoting oxidative stress, initiating a proinflammatory response, and disrupting the skin barrier function. Epidemiologic studies overall report that air pollution is associated with AD among both children and adults, though the results are not consistent among cross-sectional studies. Studies on healthcare use for AD found positive correlations between medical visits for AD and air pollutants. As the air quality worsens in many areas globally, it is important to recognize how this can increase the risk for AD, to be aware of the increased demand for AD-related medical care, and to understand how to counsel patients regarding their skin health. Further research is needed to develop treatments that prevent or mitigate air pollution-related AD symptoms.
Collapse
Affiliation(s)
- Raj P. Fadadu
- Department of Dermatology, University of California, San Francisco, CA 94115, USA
- Dermatology Service, San Francisco VA Health Care System, San Francisco, CA 94121, USA
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Katrina Abuabara
- Department of Dermatology, University of California, San Francisco, CA 94115, USA
- School of Public Health, University of California, Berkeley, CA 94720, USA
| | - John R. Balmes
- School of Public Health, University of California, Berkeley, CA 94720, USA
- Division of Occupational and Environmental Medicine, University of California, San Francisco, CA 94143, USA
| | - Jon M. Hanifin
- Department of Dermatology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maria L. Wei
- Department of Dermatology, University of California, San Francisco, CA 94115, USA
- Dermatology Service, San Francisco VA Health Care System, San Francisco, CA 94121, USA
| |
Collapse
|
13
|
Makowska K, Nowaczyk J, Blicharz L, Waśkiel-Burnat A, Czuwara J, Olszewska M, Rudnicka L. Immunopathogenesis of Atopic Dermatitis: Focus on Interleukins as Disease Drivers and Therapeutic Targets for Novel Treatments. Int J Mol Sci 2023; 24:ijms24010781. [PMID: 36614224 PMCID: PMC9820829 DOI: 10.3390/ijms24010781] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Atopic dermatitis is a chronic, recurrent inflammatory skin disorder manifesting by eczematous lesions and intense pruritus. Atopic dermatitis develops primarily as a result of an epidermal barrier defect and immunological imbalance. Advances in understanding these pathogenetic hallmarks, and particularly the complex role of interleukins as atopic dermatitis drivers, resulted in achieving significant therapeutic breakthroughs. Novel medications involve monoclonal antibodies specifically blocking the function of selected interleukins and small molecules such as Janus kinase inhibitors limiting downstream signaling to reduce the expression of a wider array of proinflammatory factors. Nevertheless, a subset of patients remains refractory to those treatments, highlighting the complexity of atopic dermatitis immunopathogenesis in different populations. In this review, we address the immunological heterogeneity of atopic dermatitis endotypes and phenotypes and present novel interleukin-oriented therapies for this disease.
Collapse
|
14
|
Huang S, Wang H, Zheng H, Li W, Shi J, Shen C, Tao R. Association between IL-6 polymorphisms and Atopic Dermatitis in Chinese Han children. Front Pediatr 2023; 11:1156659. [PMID: 37025289 PMCID: PMC10070860 DOI: 10.3389/fped.2023.1156659] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction Atopic Dermatitis (AD) is a chronic inflammatory skin disease that affects almost 20% of children and 2 -10% of adults worldwide. Previous studies revealed that Interleukin-6 (IL-6) plays an essential role in autoimmune and chronic inflammatory diseases. This study aims to investigate the associations between IL-6 polymorphisms and AD. Methods Blood samples were collected from 132 AD patients and 100 controls, and single nucleotide polymorphisms (SNPs) in IL-6 (rs2069840 (C/G), rs2066992 (G/T), rs2069837 (A/G) and rs1800796 (G/C)) were analyzed using Multiplex PCR-Based Next Generation Sequencing (NGS). Results Results showed that the A/G genotype of IL-6/rs2069837 was significantly associated with a 1.933-fold increased risk of AD compared to those patients with A/A genotype (OR 1.933; 95%CI 1.086-3.438; p=0.024). The combined A/G-G/G genotype raised AD risk by 1.856 times compared to patients with the A/A genotype in dominant model (OR: 1.856; 95% CI: 1.056-3.261; p=0.030). No association was observed for 3 other SNPs and 4 haplotypes. Discussion These findings suggested that the A/G genotype of IL-6/rs2069837 was more susceptible to AD than A/A genotype in Chinese Han children, indicating the risk role of IL-6/rs2069837 in the occurrence of AD.
Collapse
Affiliation(s)
- Shuangshuang Huang
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Hao Wang
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Huiwen Zheng
- Department of Dermatology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Wei Li
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianrong Shi
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chen Shen
- Department of Data and Information, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Sino-Finland Joint AI Laboratory for Child Health of Zhejiang Province, Hangzhou, China
| | - Ran Tao
- Department of Clinical Laboratory, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Correspondence: Ran Tao
| |
Collapse
|
15
|
Chen Y, Chen W. Genome-Wide Integration of Genetic and Genomic Studies of Atopic Dermatitis: Insights into Genetic Architecture and Pathogenesis. J Invest Dermatol 2022; 142:2958-2967.e8. [PMID: 35577104 DOI: 10.1016/j.jid.2022.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/23/2022]
Abstract
Atopic dermatitis (AD) is a common heterogeneous, chronic, itching, and inflammatory skin disease. Genetic studies have identified multiple AD susceptibility genes. However, the genetic architecture of AD has not been elucidated. In this study, we conducted a large-scale meta-analysis of AD (35,647 cases and 1,013,885 controls) to characterize the genetic basis of AD. The heritability of AD in different datasets varied from 0.6 to 7.1%. We identified 31 previously unreported genes by integrating multiomics data. Among the 31 genes, MCL1 was identified as a potential treatment target for AD by mediating gene‒drug interactions. Tissue enrichment analyses and phenome-wide association study provided strong support for the role of the hemic and immune systems in AD. Across 1,207 complex traits and diseases, genetic correlations indicated that AD shared links with multiple respiratory phenotypes. The phenome-wide Mendelian randomization analysis (Mendelian randomization‒phenome-wide association study) revealed that the age of onset of diabetes exhibited a positive causal effect on AD (inverse-variance weighted β = 0.39, SEM = 0.09, P = 2.77 × 10-5). Overall, these results provide important insights into the genetic architecture of AD and will lead to a more thorough and complete understanding of the molecular mechanisms underlying AD.
Collapse
Affiliation(s)
- Yanxuan Chen
- Department of General Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wenyan Chen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
16
|
Nelson RW, Geha RS, McDonald DR. Inborn Errors of the Immune System Associated With Atopy. Front Immunol 2022; 13:860821. [PMID: 35572516 PMCID: PMC9094424 DOI: 10.3389/fimmu.2022.860821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Atopic disorders, including atopic dermatitis, food and environmental allergies, and asthma, are increasingly prevalent diseases. Atopic disorders are often associated with eosinophilia, driven by T helper type 2 (Th2) immune responses, and triggered by disrupted barrier function leading to abnormal immune priming in a susceptible host. Immune deficiencies, in contrast, occur with a significantly lower incidence, but are associated with greater morbidity and mortality. A subset of atopic disorders with eosinophilia and elevated IgE are associated with monogenic inborn errors of immunity (IEI). In this review, we discuss current knowledge of IEI that are associated with atopy and the lessons these immunologic disorders provide regarding the fundamental mechanisms that regulate type 2 immunity in humans. We also discuss further mechanistic insights provided by animal models.
Collapse
Affiliation(s)
- Ryan W Nelson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Douglas R McDonald
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Alsabbagh M, Ismaeel A. The role of cytokines in atopic dermatitis: a breakthrough in immunopathogenesis and treatment. ACTA DERMATOVENEROLOGICA ALPINA PANNONICA ET ADRIATICA 2022. [DOI: 10.15570/actaapa.2022.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
18
|
Chen Y, Lian P, Peng Z, Wazir J, Ma C, Wei L, Li L, Liu J, Zhao C, Pu W, Wang H, Su Z. Alpha-7 nicotinic acetylcholine receptor agonist alleviates psoriasis-like inflammation through inhibition of the STAT3 and NF-κB signaling pathway. Cell Death Dis 2022; 8:141. [PMID: 35351863 PMCID: PMC8964744 DOI: 10.1038/s41420-022-00943-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/17/2022] [Accepted: 03/10/2022] [Indexed: 12/03/2022]
Abstract
Psoriasis is a chronic inflammatory cutaneous disease; it has been discovered that stimulation of the nervous system increases susceptibility to psoriasis. Although the cholinergic anti-inflammatory pathway, which is mediated by the alpha-7 nicotinic acetylcholine receptor (α7nAChR), is critical for controlling multiple types of inflammation, its expression pattern and pathogenesis function in psoriatic lesioned skin tissue are unknown. We hereby analyzed the expression of α7nAchR in human and mouse psoriatic skin tissue. In vivo, PNU-282987 or Methyllycaconitine, a specific agonist or antagonist of α7nAchR, were administered to imiquimod (IMQ)-induced psoriatic mouse models. The macroscopic appearance and histopathological features of the psoriatic mice skin were evaluated. In addition, cell proliferation and differentiation markers were investigated. The level of pro-inflammatory cytokines released from the lesioned skin, as well as the activation of the relevant signaling pathways, were measured. Our findings indicated that psoriatic lesional skin expressed an increased level of α7nAChR, with its tissue distribution being primarily in skin keratinocytes and macrophages. In an IMQ-induced murine psoriasis model, α7nAChR agonist PNU-282987 treatment alleviated psoriasis-like inflammation by down-regulating the expression of multiple types of pro-inflammatory mediators and normalized keratinocyte proliferation and differentiation, whereas α7nAChR antagonist treatment exacerbated its effect. Mechanically, we observed that activation of the α7nAChR inhibited the activation of the STAT3 and NF-κB signaling pathways in in vitro cultured HaCaT cells induced by Th17-related cytokine IL-6/IL-22 or Th1-related cytokine TNF-α. Taken together, these findings demonstrate that attenuation of psoriatic inflammation via the cholinergic anti-inflammatory pathway is dependent on α7nAChR activation.
Collapse
Affiliation(s)
- Yiwen Chen
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China.,State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Panpan Lian
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Ziqi Peng
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Chujun Ma
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China
| | - Lulu Wei
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Jun Liu
- Department of Dermatology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210008, Nanjing, P.R. China
| | - Chen Zhao
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, 210093, P.R. China.
| | - Zhonglan Su
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, P.R. China.
| |
Collapse
|
19
|
Bosma AL, Ascott A, Iskandar R, Farquhar K, Matthewman J, Langendam MW, Mulick A, Abuabara K, Williams HC, Spuls PI, Langan SM, Middelkamp-Hup MA. Classifying atopic dermatitis: a systematic review of phenotypes and associated characteristics. J Eur Acad Dermatol Venereol 2022; 36:807-819. [PMID: 35170821 PMCID: PMC9307020 DOI: 10.1111/jdv.18008] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/10/2021] [Accepted: 01/18/2022] [Indexed: 11/28/2022]
Abstract
Atopic dermatitis is a heterogeneous disease, accompanied by a wide variation in disease presentation and the potential to identify many phenotypes that may be relevant for prognosis and treatment. We aimed to systematically review previously reported phenotypes of atopic dermatitis and any characteristics associated with them. Ovid EMBASE, Ovid MEDLINE and Web of Science were searched from inception till the 12th of February 2021 for studies attempting to classify atopic dermatitis. Primary outcomes are atopic dermatitis phenotypes and characteristics associated with them in subsequent analyses. A secondary outcome is the methodological approach used to derive them. In total, 8,511 records were found. By focusing only on certain clinical phenotypes, 186 studies were eligible for inclusion. The majority of studies were hospital-based (59%, 109/186) and cross-sectional (76%, 141/186). The number of included patients ranged from seven to 526,808. Data-driven approaches to identify phenotypes were only used in a minority of studies (7%, 13/186). Ninety-one studies (49%) investigated a phenotype based on disease severity. A phenotype based on disease trajectory, morphology and eczema herpeticum was investigated in 56 (30%), 22 (12%) and 11 (6%) studies, respectively. Thirty-six studies (19%) investigated morphological characteristics in other phenotypes. Investigated associated characteristics differed between studies. In conclusion, we present an overview of phenotype definitions used in literature for severity, trajectory, morphology and eczema herpeticum, including associated characteristics. There is a lack of uniform and consistent use of atopic dermatitis phenotypes across studies.
Collapse
Affiliation(s)
- A L Bosma
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| | - A Ascott
- Department of Dermatology, University Hospitals Sussex NHS Foundation Trust, Worthing, United Kingdom
| | - R Iskandar
- Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - J Matthewman
- Department of Non-communicable disease epidemiology, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - M W Langendam
- Department of Epidemiology and Data Science, UMC, location Amsterdam Medical Center, University of Amsterdam, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - A Mulick
- Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - K Abuabara
- Department of Dermatology, University of California San Francisco, United States
| | - H C Williams
- Centre of Evidence-Based Dermatology, University of Nottingham, United Kingdom
| | - P I Spuls
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| | - S M Langan
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands.,Faculty of Epidemiology and Population Health, School of Hygiene and Tropical Medicine, London, United Kingdom
| | - M A Middelkamp-Hup
- Department of Dermatology, UMC, location Academic Medical Center, University of Amsterdam, Amsterdam Public Health, Infection and Immunity, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Martínez-Pérez C, Kay C, Meehan J, Gray M, Dixon JM, Turnbull AK. The IL6-like Cytokine Family: Role and Biomarker Potential in Breast Cancer. J Pers Med 2021; 11:1073. [PMID: 34834425 PMCID: PMC8624266 DOI: 10.3390/jpm11111073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
IL6-like cytokines are a family of regulators with a complex, pleiotropic role in both the healthy organism, where they regulate immunity and homeostasis, and in different diseases, including cancer. Here we summarise how these cytokines exert their effect through the shared signal transducer IL6ST (gp130) and we review the extensive evidence on the role that different members of this family play in breast cancer. Additionally, we discuss how the different cytokines, their related receptors and downstream effectors, as well as specific polymorphisms in these molecules, can serve as predictive or prognostic biomarkers with the potential for clinical application in breast cancer. Lastly, we also discuss how our increasing understanding of this complex signalling axis presents promising opportunities for the development or repurposing of therapeutic strategies against cancer and, specifically, breast neoplasms.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Charlene Kay
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - James Meehan
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Mark Gray
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - J. Michael Dixon
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
| | - Arran K. Turnbull
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| |
Collapse
|
21
|
Adikusuma W, Irham LM, Chou WH, Wong HSC, Mugiyanto E, Ting J, Perwitasari DA, Chang WP, Chang WC. Drug Repurposing for Atopic Dermatitis by Integration of Gene Networking and Genomic Information. Front Immunol 2021; 12:724277. [PMID: 34721386 PMCID: PMC8548825 DOI: 10.3389/fimmu.2021.724277] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/15/2021] [Indexed: 12/02/2022] Open
Abstract
Atopic Dermatitis (AD) is a chronic and relapsing skin disease. The medications for treating AD are still limited, most of them are topical corticosteroid creams or antibiotics. The current study attempted to discover potential AD treatments by integrating a gene network and genomic analytic approaches. Herein, the Single Nucleotide Polymorphism (SNPs) associated with AD were extracted from the GWAS catalog. We identified 70 AD-associated loci, and then 94 AD risk genes were found by extending to proximal SNPs based on r2 > 0.8 in Asian populations using HaploReg v4.1. Next, we prioritized the AD risk genes using in silico pipelines of bioinformatic analysis based on six functional annotations to identify biological AD risk genes. Finally, we expanded them according to the molecular interactions using the STRING database to find the drug target genes. Our analysis showed 27 biological AD risk genes, and they were mapped to 76 drug target genes. According to DrugBank and Therapeutic Target Database, 25 drug target genes overlapping with 53 drugs were identified. Importantly, dupilumab, which is approved for AD, was successfully identified in this bioinformatic analysis. Furthermore, ten drugs were found to be potentially useful for AD with clinical or preclinical evidence. In particular, we identified filgotinub and fedratinib, targeting gene JAK1, as potential drugs for AD. Furthermore, four monoclonal antibody drugs (lebrikizumab, tralokinumab, tocilizumab, and canakinumab) were successfully identified as promising for AD repurposing. In sum, the results showed the feasibility of gene networking and genomic information as a potential drug discovery resource.
Collapse
Affiliation(s)
- Wirawan Adikusuma
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Faculty of Health Science, University of Muhammadiyah Mataram, Mataram, Indonesia
| | - Lalu Muhammad Irham
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Faculty of Pharmacy, University of Ahmad Dahlan, Yogyakarta, Indonesia
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Henry Sung-Ching Wong
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Eko Mugiyanto
- Ph. D. Program in the Clinical Drug Development of Herbal Medicines, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Faculty of Health Science, University of Muhammadiyah Pekajangan Pekalongan, Pekalongan, Indonesia
| | - Jafit Ting
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | | | - Wei-Pin Chang
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University (TMU) Research Center of Cancer Translational Medicine, Taipei, Taiwan
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
22
|
Kern C, Wan J, LeWinn KZ, Ramirez FD, Lee Y, McCulloch CE, Langan SM, Abuabara K. Association of Atopic Dermatitis and Mental Health Outcomes Across Childhood: A Longitudinal Cohort Study. JAMA Dermatol 2021; 157:1200-1208. [PMID: 34468686 PMCID: PMC8411354 DOI: 10.1001/jamadermatol.2021.2657] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Research has highlighted associations between atopic dermatitis (AD) and mental health conditions in adults. However, literature on the development of mental health comorbidities in children is limited despite the large burden of pediatric AD worldwide. OBJECTIVE To examine the association between AD and internalizing behaviors and symptoms of depression at multiple points across childhood and adolescence and to explore potential mediating factors, including asthma/rhinitis, sleep, and inflammation. DESIGN, SETTING, AND PARTICIPANTS This longitudinal, population-based birth cohort study included children followed up from birth for a mean (SD) duration of 10.0 (2.9) years from the UK Avon Longitudinal Study of Parents and Children. Data were collected from September 6, 1990, to December 31, 2009. Data were analyzed from August 30, 2019, to July 30, 2020. EXPOSURES Annual period prevalence of AD assessed at 11 points from 6 months to 18 years of age, measured by standardized questions about flexural dermatitis. MAIN OUTCOMES AND MEASURES Symptoms of depression, measured using child-reported responses to the Short Moods and Feelings Questionnaire at 5 points from 10 to 18 years of age and internalizing behaviors, measured by maternal report of the Emotional Symptoms subscale of the Strength and Difficulties Questionnaire at 7 points from 4 to 16 years of age. RESULTS Among the 11 181 children included in the analysis (5721 male [51.2%]), the period prevalence of symptoms of depression ranged from 6.0% to 21.6%; for internalizing behaviors, from 10.4% to 16.0%. Although mild to moderate AD was not associated with symptoms of depression, it was associated with internalizing behaviors as early as 4 years of age (mean increased odds of 29%-84% across childhood in adjusted models). Severe AD was associated with symptoms of depression (adjusted odds ratio, 2.38; 95% CI, 1.21-4.72) and internalizing symptoms (adjusted odds ratio, 1.90; 95% CI, 1.14-3.16). Sleep quality mediated some of this association, but it was not explained by differences in sleep duration, asthma/rhinitis, or levels of inflammatory markers (interleukin 6 and C-reactive protein). CONCLUSIONS AND RELEVANCE Within this population-based birth cohort study in the UK, severe AD was associated with symptoms of depression and internalizing behaviors throughout childhood and adolescence. Risk of internalizing symptoms was increased even for children with mild AD beginning early in childhood, highlighting the importance of behavioral and mental health awareness in this population.
Collapse
Affiliation(s)
- Chloe Kern
- Department of Dermatology, University of California, San Francisco
| | - Joy Wan
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco
| | | | - Yong Lee
- Department of Dermatology, University of California, San Francisco
| | - Charles E McCulloch
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Sinéad M Langan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Katrina Abuabara
- Department of Dermatology, University of California, San Francisco.,Division of Epidemiology, School of Public Health, University of California, Berkeley
| |
Collapse
|
23
|
Chen YH, Spencer S, Laurence A, Thaventhiran JE, Uhlig HH. Inborn errors of IL-6 family cytokine responses. Curr Opin Immunol 2021; 72:135-145. [PMID: 34044328 PMCID: PMC8591178 DOI: 10.1016/j.coi.2021.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 01/25/2023]
Abstract
The IL-6 family of cytokines mediates functions in host protective immunity, development of multiple organs, tissue regeneration and metabolism. Inborn errors in cytokines or cytokine receptor units highlight specific roles for IL-6, IL-11, LIF, OSM, and CLC signaling whereas incomplete loss-of-function variants in the common receptor chain GP130 encoded by IL6ST or the transcription factor STAT3, as well as genes that affect either GP130 glycosylation (PGM3) or STAT3 transcriptional control (ZNF341) lead to complex phenotypes including features of hyper-IgE syndrome. Gain-of-function variants in the GP130-STAT3 signaling pathway cause immune dysregulation disorders. Insights into IL-6 family cytokine signaling inform on therapeutic application in immune-mediated disorders and potential side effects such as infection susceptibility.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sarah Spencer
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge, UK
| | - Arian Laurence
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK; Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, UK
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK; Biomedical Research Center, University of Oxford, Oxford, UK; Department of Pediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
24
|
Xu H, Liu J, Niu M, Song S, Wei L, Chen G, Ding Y, Wang Y, Su Z, Wang H. Soluble IL-6R-mediated IL-6 trans-signaling activation contributes to the pathological development of psoriasis. J Mol Med (Berl) 2021; 99:1009-1020. [PMID: 33835216 DOI: 10.1007/s00109-021-02073-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 02/08/2023]
Abstract
IL-6 has been suggested to function as an autocrine mitogen in the psoriatic epidermis. The biological activity of IL-6 relies on interactions with its receptors, including the membrane-bound IL-6 receptor (mIL-6R) and soluble IL-6 receptor (sIL-6R). Our study presents data showing that the levels of plasma IL-6 and sIL-6R were elevated in psoriatic patients. Genotyping of two single-nucleotide polymorphisms (SNPs) in IL-6R (rs4845617 and rs2228145) demonstrated that the SNP IL-6R (rs4845617) rather than IL-6R (rs2228145) shows a significant association with psoriasis (P = 0.006). To verify the functions of sIL-6R, cultured keratinocytes and imiquimod (IMQ)-induced psoriatic model mice were treated with sIL-6R. We found that the presence of sIL-6R in the HaCaT cell culture medium enhanced the IL-6-induced Stat3 activation, which resulted in abnormal keratinocyte proliferation and aberrant differentiation. Furthermore, the application of sIL-6R in vivo accelerated the pathological development of the disease. Our results demonstrate for the first time that genetic polymorphisms in the IL-6R gene are associated with psoriasis disease phenotypes in a Chinese psoriatic patient population; sIL-6R-mediated trans-signaling pathway plays a pivotal role in keratinocyte proliferation and differentiation, suggesting potential therapeutics for psoriasis. KEY MESSAGES: Patients with psoriasis displayed higher levels of IL-6 and sIL-6R compared with healthy controls. Analysis of genotypes revealed that IL-6R rs4845617 GG genotype associated with the risk of psoriasis. Supplement of sIL-6R further enhanced IL-6-induced Stat3 activation in keratinocytes. In vivo administration of sIL-6R accelerated, whereas sgp130FC alleviated, the pathological development of psoriasis.
Collapse
Affiliation(s)
- Hui Xu
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Jun Liu
- Department of Dermatology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Mengyuan Niu
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Shiyu Song
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Lulu Wei
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Gang Chen
- Department of Esthetic Plastic Surgery, The First Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yibing Ding
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Yong Wang
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China
| | - Zhonglan Su
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| | - Hongwei Wang
- Center for Translational Medicine, State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, 210093, People's Republic of China.
| |
Collapse
|
25
|
Tanaka N, Koido M, Suzuki A, Otomo N, Suetsugu H, Kochi Y, Tomizuka K, Momozawa Y, Kamatani Y, Ikegawa S, Yamamoto K, Terao C. Eight novel susceptibility loci and putative causal variants in atopic dermatitis. J Allergy Clin Immunol 2021; 148:1293-1306. [PMID: 34116867 DOI: 10.1016/j.jaci.2021.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 03/03/2021] [Accepted: 04/08/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is the most common allergic disease in the world. While genetic components play critical roles in its pathophysiology, a large proportion of its genetic background is still unexplored. OBJECTIVES This study sought to illuminate the genetic associations with AD using genome-wide association study (GWAS) and its downstream analyses. METHODS This study conducted a GWAS for AD comprising 2,639 cases and 115,648 controls in the Japanese population, followed by a trans-ethnic meta-analysis with UK Biobank data and downstream analyses including partitioning heritability analysis by linkage disequilibrium score regression. RESULTS This study identified 17 significant susceptibility loci, among which 4 loci-AFF1, ITGB8, EHMT1, and EGR2-were novel in the Japanese GWAS. The trans-ethnic meta-analysis revealed 4 additional novel loci, namely-ZBTB38,LOC105755953/LOC101928272, TRAF3, andIQGAP1. This study found a missense variant (R243W) with a deleterious functional effect in NLRP10 and a variant altering expression of CCDC80 via enhancer expression as highly likely causal variants. These 2 regions were Asian-specific, and these population-specific associations could be explained by the frequency of causal variants. The gene-based test showed SMAD4 as an additional novel significant locus. Downstream analyses revealed substantial overlap of GWAS significant signals in enhancers of skin cells and immune cells, especially CD4 T cells. A highly shared polygenic architecture of AD between Europeans and Asians was also found. CONCLUSIONS This study identified Japanese-specific loci and novel significant loci shared by different populations. Two putative causal variants were illuminated in Japanese-specific loci. Trans-ethnic analyses revealed strong heritability enrichment in immune-related pathways, and relevant cell types shared among populations.
Collapse
Affiliation(s)
- Nao Tanaka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Rheumatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaru Koido
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nao Otomo
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan; Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Suetsugu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan; Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuta Kochi
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | -
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Medical Sciences, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan; Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan.
| |
Collapse
|
26
|
Abstract
IL-6 is involved both in immune responses and in inflammation, hematopoiesis, bone metabolism and embryonic development. IL-6 plays roles in chronic inflammation (closely related to chronic inflammatory diseases, autoimmune diseases and cancer) and even in the cytokine storm of corona virus disease 2019 (COVID-19). Acute inflammation during the immune response and wound healing is a well-controlled response, whereas chronic inflammation and the cytokine storm are uncontrolled inflammatory responses. Non-immune and immune cells, cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) and transcription factors nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) play central roles in inflammation. Synergistic interactions between NF-κB and STAT3 induce the hyper-activation of NF-κB followed by the production of various inflammatory cytokines. Because IL-6 is an NF-κB target, simultaneous activation of NF-κB and STAT3 in non-immune cells triggers a positive feedback loop of NF-κB activation by the IL-6-STAT3 axis. This positive feedback loop is called the IL-6 amplifier (IL-6 Amp) and is a key player in the local initiation model, which states that local initiators, such as senescence, obesity, stressors, infection, injury and smoking, trigger diseases by promoting interactions between non-immune cells and immune cells. This model counters dogma that holds that autoimmunity and oncogenesis are triggered by the breakdown of tissue-specific immune tolerance and oncogenic mutations, respectively. The IL-6 Amp is activated by a variety of local initiators, demonstrating that the IL-6-STAT3 axis is a critical target for treating diseases.
Collapse
Affiliation(s)
- Toshio Hirano
- National Institutes for Quantum and Radiological Science and Technology, Anagawa, Inage-ku, Chiba, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
27
|
Hübenthal M, Löscher BS, Erdmann J, Franke A, Gola D, König IR, Emmert H. Current Developments of Clinical Sequencing and the Clinical Utility of Polygenic Risk Scores in Inflammatory Diseases. Front Immunol 2021; 11:577677. [PMID: 33633722 PMCID: PMC7901950 DOI: 10.3389/fimmu.2020.577677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 12/03/2022] Open
Abstract
In this mini-review, we highlight selected research by the Deutsche Forschungsgemeinschaft (DFG) Cluster of Excellence “Precision Medicine in Chronic Inflammation” focusing on clinical sequencing and the clinical utility of polygenic risk scores as well as its implication on precision medicine in the field of the inflammatory diseases inflammatory bowel disease, atopic dermatitis and coronary artery disease. Additionally, we highlight current developments and discuss challenges to be faced in the future. Exemplary, we point to residual challenges in detecting disease-relevant variants resulting from difficulties in the interpretation of candidate variants and their potential interactions. While polygenic risk scores represent promising tools for the stratification of patient groups, currently, polygenic risk scores are not accurate enough for clinical setting. Precision medicine, incorporating additional data from genomics, transcriptomics and proteomics experiments, may enable the identification of distinct disease pathogeneses. In the future, data-intensive biomedical innovation will hopefully lead to improved patient stratification for personalized medicine.
Collapse
Affiliation(s)
- Matthias Hübenthal
- Department of Dermatology, Quincke Research Center, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Britt-Sabina Löscher
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Damian Gola
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Inke R König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Hila Emmert
- Department of Dermatology, Quincke Research Center, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
28
|
Sokolik R, Iwaszko M, Świerkot J, Wysoczańska B, Korman L, Wiland P, Bogunia-Kubik K. Relationship Between Interleukin-6 -174G/C Genetic Variant and Efficacy of Methotrexate Treatment in Psoriatic Arthritis Patients. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:157-166. [PMID: 33536774 PMCID: PMC7850562 DOI: 10.2147/pgpm.s264555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Introduction The purpose of the study was to investigate whether single-nucleotide polymorphisms (SNPs) IL-6 −174 G/C and IL-6R Asp358Ala are associated with susceptibility to psoriatic arthritis (PsA) or affect response to treatment with methotrexate (MTX). Patients and Methods Seventy-four patients diagnosed with PsA and qualified for MTX treatment were enrolled to the study. The control group consisted of 120 healthy individuals. Polymorphisms IL-6 −174 G/C and IL-6R Asp358Ala were genotyped using a polymerase chain reaction (PCR) amplification employing LightSNiP assays. Results A significant association between the IL-6 −174 CC genotype and an improved clinical outcome of MTX therapy was observed. A good response was more frequently observed among PsA patients bearing the IL-6 −174 CC genotype than patients with the GC or GG genotypes (P = 0.007). On the other hand, patients carrying the IL-6 −174 GC genotype less frequently responded to MTX treatment as compared to patients with other genotypes (P = 0.006). With respect to the IL-6R Asp358Ala SNP, there were no significant differences in genotype and allelic frequencies in relation to clinical outcome of MTX treatment. No association was found between the IL-6 −174 G/C or IL-6R Asp358Ala SNPs and PsA susceptibility. Conclusion Results from this study provide evidence that the IL-6 −174 G/C polymorphism might influence efficacy of MTX treatment.
Collapse
Affiliation(s)
- Renata Sokolik
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Wrocław, Poland
| | - Milena Iwaszko
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jerzy Świerkot
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Wrocław, Poland
| | - Barbara Wysoczańska
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Lucyna Korman
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Wrocław, Poland
| | - Piotr Wiland
- Department of Rheumatology and Internal Medicine, Wrocław Medical University, Wrocław, Poland
| | - Katarzyna Bogunia-Kubik
- Laboratory of Clinical Immunogenetics and Pharmacogenetics, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
29
|
Yasuda-Sekiguchi F, Shiohama A, Fukushima A, Obata S, Mochimaru N, Honda A, Kawasaki H, Kubo A, Ebihara T, Amagai M, Sasaki T. Single nucleotide variations in genes associated with innate immunity are enriched in Japanese adult cases of face and neck type atopic dermatitis. J Dermatol Sci 2020; 101:93-100. [PMID: 33279384 DOI: 10.1016/j.jdermsci.2020.11.005] [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] [Received: 07/29/2020] [Revised: 10/16/2020] [Accepted: 11/08/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is heterogenous in terms of phenotype as well as genetic and environmental factors, while its associated genetic factors and pathophysiology are not fully understood. OBJECTIVE We identify novel genetic factors enriched in a subgroup of AD patients with characteristic clinical features. METHODS We clinically subgrouped 18 AD patients who exhibited distinctive characteristic of persistent skin eruption areas on the face and neck from 92 Japanese adult AD patients and identified disease-associated genetic factors enriched within the subgroup. Targeted resequencing and subsequent genetic association analyses were used to identify novel enriched genetic variations in the subgroup compared with the other AD patients. RESULTS Targeted resequencing of 648 skin associated genes revealed an enrichment of 12 single nucleotide variations (SNVs) in patients with face and neck AD (n = 18) compared with the general Japanese population in the database. Subsequent allele frequency comparison between the face and neck AD and non - face and neck AD subgroups revealed enrichment of five SNVs. Multivariate analysis using genotype data revealed that three SNVs in theTLR1, TIRAP, and PSAPL1 genes, two of the three genes are involved in the Toll-like receptor pathway, were significantly enriched in patients with face and neck AD. CONCLUSION These findings revealed that the SNVs in genes associated with the innate immune pathway are enriched in a subgroup of AD. The combinational approach of clinical subgrouping and genotyping is valuable for detecting novel disease-associated genetic factors.
Collapse
Affiliation(s)
| | - Aiko Shiohama
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; KOSÉ Endowed Program for Skin Care and Allergy Prevention, Keio University School of Medicine, Tokyo, Japan
| | - Ayano Fukushima
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Shoko Obata
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Naoko Mochimaru
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Aki Honda
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Kawasaki
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Medical Sciences Innovation Hub Program, RIKEN, Kanagawa, Japan; Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan
| | - Akiharu Kubo
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Tamotsu Ebihara
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; KOSÉ Endowed Program for Skin Care and Allergy Prevention, Keio University School of Medicine, Tokyo, Japan; Center for Integrative Medical Sciences, RIKEN, Kanagawa, Japan
| | - Takashi Sasaki
- Center for Supercentenarian Medical Research, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
30
|
The Active Compounds and Therapeutic Mechanisms of Pentaherbs Formula for Oral and Topical Treatment of Atopic Dermatitis Based on Network Pharmacology. PLANTS 2020; 9:plants9091166. [PMID: 32916837 PMCID: PMC7569866 DOI: 10.3390/plants9091166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/07/2023]
Abstract
To examine the molecular targets and therapeutic mechanism of a clinically proven Chinese medicinal pentaherbs formula (PHF) in atopic dermatitis (AD), we analyzed the active compounds and core targets, performed network and molecular docking analysis, and investigated interacting pathways. Information on compounds in PHF was obtained from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, and target prediction was performed using the Drugbank database. AD-related genes were gathered using the GeneCards and Online Mendelian Inheritance in Man (OMIM) databases. Network analysis was performed by Cytoscape software and protein-protein interaction was analyzed by the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). The Database for Annotation, Visualization and Integrated Discovery (DAVID) Bioinformatics Resources were applied for the enrichment analysis of the potential biological process and pathways associated with the intersection targets between PHF and AD. Autodock software was used to perform protein compound docking analysis. We identified 43 active compounds in PHF associated with 117 targets, and 57 active compounds associated with 107 targets that form the main pathways linked to oral and topical treatment of AD, respectively. Among them, quercetin, luteolin, and kaempferol are key chemicals targeting the core genes involved in the oral use of PHF against AD, while apigenin, ursolic acid, and rosmarinic acid could be used in topical treatment of PHF against AD. The compound–target–disease network constructed in the current study reveals close interactions between multiple components and multiple targets. Enrichment analysis further supports the biological processes and signaling pathways identified, indicating the involvement of IL-17 and tumor necrosis factor signaling pathways in the action of PHF on AD. Our data demonstrated the main compounds and potential pharmacological mechanisms of oral and topical application of PHF in AD.
Collapse
|
31
|
APRIL expression is upregulated in atopic dermatitis skin lesions and at sites of antigen driven allergic skin inflammation in mice. Clin Immunol 2020; 219:108556. [PMID: 32777271 DOI: 10.1016/j.clim.2020.108556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 11/24/2022]
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease. It is characterized by a defective skin barrier and a Th2 dominated skin inflammation. The TNF family member a proliferation-inducing ligand (APRIL) and its receptors transmembrane activator and calcium modulator and cyclophilin ligand interactor (TACI) and B cell maturation antigen (BCMA) are expressed by immune cells and epithelial cells including keratinocytes. We demonstrate that APRIL expression is upregulated in the epidermis of skin lesions from patients with AD as well as in mouse skin undergoing allergic inflammation elicited by epicutaneous (EC) sensitization with the antigen ovalbumin. We show that APRIL from OVA sensitized mouse skin causes keratinocytes to upregulate the expression of IL-6, an inflammatory cytokine implicated in AD pathogenesis. These results suggest a role for APRIL in allergic skin inflammation and a potential role for APRIL blockade in treating AD.
Collapse
|
32
|
Nousbeck J, McAleer M, Hurault G, Kenny E, Harte K, Kezic S, Tanaka R, Irvine A. MicroRNA analysis of childhood atopic dermatitis reveals a role for miR‐451a*. Br J Dermatol 2020; 184:514-523. [DOI: 10.1111/bjd.19254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Affiliation(s)
- J. Nousbeck
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
| | - M.A. McAleer
- National Children’s Research Centre Crumlin, Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| | - G. Hurault
- Department of Bioengineering Imperial College London London UK
| | - E. Kenny
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - K. Harte
- Department of Psychiatry Trinity Translational Medicine Institute St James’s Hospital Dublin Ireland
| | - S. Kezic
- Coronel Institute of Occupational Health Academic Medical Center Amsterdam the Netherlands
| | - R.J. Tanaka
- Department of Bioengineering Imperial College London London UK
| | - A.D. Irvine
- National Children’s Research Centre Crumlin, Dublin Ireland
- Clinical Medicine Trinity College Dublin Dublin Ireland
- Paediatric Dermatology Our Lady’s Children’s Hospital Crumlin, Dublin Ireland
| |
Collapse
|
33
|
Bretherick AD, Canela-Xandri O, Joshi PK, Clark DW, Rawlik K, Boutin TS, Zeng Y, Amador C, Navarro P, Rudan I, Wright AF, Campbell H, Vitart V, Hayward C, Wilson JF, Tenesa A, Ponting CP, Baillie JK, Haley C. Linking protein to phenotype with Mendelian Randomization detects 38 proteins with causal roles in human diseases and traits. PLoS Genet 2020; 16:e1008785. [PMID: 32628676 PMCID: PMC7337286 DOI: 10.1371/journal.pgen.1008785] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 04/21/2020] [Indexed: 01/25/2023] Open
Abstract
To efficiently transform genetic associations into drug targets requires evidence that a particular gene, and its encoded protein, contribute causally to a disease. To achieve this, we employ a three-step proteome-by-phenome Mendelian Randomization (MR) approach. In step one, 154 protein quantitative trait loci (pQTLs) were identified and independently replicated. From these pQTLs, 64 replicated locally-acting variants were used as instrumental variables for proteome-by-phenome MR across 846 traits (step two). When its assumptions are met, proteome-by-phenome MR, is equivalent to simultaneously running many randomized controlled trials. Step 2 yielded 38 proteins that significantly predicted variation in traits and diseases in 509 instances. Step 3 revealed that amongst the 271 instances from GeneAtlas (UK Biobank), 77 showed little evidence of pleiotropy (HEIDI), and 92 evidence of colocalization (eCAVIAR). Results were wide ranging: including, for example, new evidence for a causal role of tyrosine-protein phosphatase non-receptor type substrate 1 (SHPS1; SIRPA) in schizophrenia, and a new finding that intestinal fatty acid binding protein (FABP2) abundance contributes to the pathogenesis of cardiovascular disease. We also demonstrated confirmatory evidence for the causal role of four further proteins (FGF5, IL6R, LPL, LTA) in cardiovascular disease risk.
Collapse
Affiliation(s)
- Andrew D. Bretherick
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Oriol Canela-Xandri
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, Scotland, United Kingdom
| | - Peter K. Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - David W. Clark
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Konrad Rawlik
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, Scotland, United Kingdom
| | - Thibaud S. Boutin
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Yanni Zeng
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Carmen Amador
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh, Scotland, United Kingdom
| | - Albert Tenesa
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, Scotland, United Kingdom
| | - Chris P. Ponting
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
| | - J. Kenneth Baillie
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, Scotland, United Kingdom
| | - Chris Haley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, Scotland, United Kingdom
- The Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
34
|
Abstract
Primary atopic disorders describes a series of monogenic diseases that have allergy- or atopic effector–related symptoms as a substantial feature. The underlying pathogenic genetic lesions help illustrate fundamental pathways in atopy, opening up diagnostic and therapeutic options for further study in those patients, but ultimately for common allergic diseases as well. Key pathways affected in these disorders include T cell receptor and B cell receptor signaling, cytokine signaling, skin barrier function, and mast cell function, as well as pathways that have not yet been elucidated. While comorbidities such as classically syndromic presentation or immune deficiency are often present, in some cases allergy alone is the presenting symptom, suggesting that commonly encountered allergic diseases exist on a spectrum of monogenic and complex genetic etiologies that are impacted by environmental risk factors.
Collapse
Affiliation(s)
- Joshua D. Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
35
|
Polymorphisms in IL-2 and IL-6R increase serum levels of the respective interleukins in differentiated thyroid cancer. Meta Gene 2020. [DOI: 10.1016/j.mgene.2019.100621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
36
|
Polányi L, Niessen CM, Vohlen C, Stinn J, Kretschmer T, Jentgen V, Hirani D, Koningsbruggen-Rietschel SV, Dötsch J, Alejandre Alcazar MA. Intrauterine growth restriction induces skin inflammation, increases TSLP and impairs epidermal barrier function. J Mol Med (Berl) 2020; 98:279-289. [PMID: 31912169 DOI: 10.1007/s00109-019-01867-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/21/2019] [Accepted: 12/06/2019] [Indexed: 12/29/2022]
Abstract
Intrauterine growth restriction (IUGR) and low birth weight are risk factors for childhood asthma. Atopic march describes the progression from early dermatitis to asthma during life. Since inflammatory signaling is linked to increased airway resistance and lung remodeling in rats after IUGR, we queried if these findings are related to skin inflammatory response. Firstly, we induced IUGR in Wistar rats by isocaloric protein restriction during gestation. IUGR rats showed lower body weight at postnatal day 1 (P1), catch-up growth at P21, and similar body weight like controls at P90. At P1 and P90, mRNA of inflammatory as well as fibrotic markers and number of skin immune cells (macrophages) were increased after IUGR. Skin thymic stromal lymphopoietin (TSLP) mRNA at P1 and serum TSLP at P1 and P21 were elevated in IUGR. Moreover, IUGR impaired transepidermal water loss at P21 and P90. IUGR induced higher. Secondly, the increase of TEWL after Oxazolone treatment as a model of atopic dermatitis (AD) was greater in IUGR than in Co. Our data demonstrate an early inflammatory skin response, which is linked to persistent macrophage infiltration in the skin and impaired epidermal barrier function after IUGR. These findings coupled with elevated TSLP could underlie atopic diseases in rats after IUGR. KEY MESSAGES: • The present study shows that IUGR increases macrophage infiltration and induces an inflammatory and fibrotic gene expression pattern in the skin of newborn rats. • Early postnatal inflammatory response in the skin after IUGR is followed by impaired epidermal barrier function later in life. • IUGR aggravates transepidermal water loss in an experimental atopic dermatitis model, possibly through elevated TSLP in skin and serum. • Early anti-inflammatory treatment and targeting TSLP signaling could offer novel avenues for early prevention of atopic disorders and late asthma in high-risk infants.
Collapse
Affiliation(s)
- Laura Polányi
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics, Experimental Pulmonology, University of Cologne, Kerpener Strasse 62, D-50937, Cologne, Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carien M Niessen
- Department of Dermatology, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics, Experimental Pulmonology, University of Cologne, Kerpener Strasse 62, D-50937, Cologne, Germany.,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julia Stinn
- Department of Dermatology, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Tobias Kretschmer
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Vanessa Jentgen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics, Experimental Pulmonology, University of Cologne, Kerpener Strasse 62, D-50937, Cologne, Germany
| | - Dharmesh Hirani
- Center of Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Jörg Dötsch
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, Translational Experimental Pediatrics, Experimental Pulmonology, University of Cologne, Kerpener Strasse 62, D-50937, Cologne, Germany. .,Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany. .,Center of Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
37
|
Mucha S, Baurecht H, Novak N, Rodríguez E, Bej S, Mayr G, Emmert H, Stölzl D, Gerdes S, Jung ES, Degenhardt F, Hübenthal M, Ellinghaus E, Kässens JC, Wienbrandt L, Lieb W, Müller-Nurasyid M, Hotze M, Dand N, Grosche S, Marenholz I, Arnold A, Homuth G, Schmidt CO, Wehkamp U, Nöthen MM, Hoffmann P, Paternoster L, Standl M, Bønnelykke K, Ahluwalia TS, Bisgaard H, Peters A, Gieger C, Waldenberger M, Schulz H, Strauch K, Werfel T, Lee YA, Wolfien M, Rosenstiel P, Wolkenhauer O, Schreiber S, Franke A, Weidinger S, Ellinghaus D. Protein-coding variants contribute to the risk of atopic dermatitis and skin-specific gene expression. J Allergy Clin Immunol 2019; 145:1208-1218. [PMID: 31707051 DOI: 10.1016/j.jaci.2019.10.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/09/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Fifteen percent of atopic dermatitis (AD) liability-scale heritability could be attributed to 31 susceptibility loci identified by using genome-wide association studies, with only 3 of them (IL13, IL-6 receptor [IL6R], and filaggrin [FLG]) resolved to protein-coding variants. OBJECTIVE We examined whether a significant portion of unexplained AD heritability is further explained by low-frequency and rare variants in the gene-coding sequence. METHODS We evaluated common, low-frequency, and rare protein-coding variants using exome chip and replication genotype data of 15,574 patients and 377,839 control subjects combined with whole-transcriptome data on lesional, nonlesional, and healthy skin samples of 27 patients and 38 control subjects. RESULTS An additional 12.56% (SE, 0.74%) of AD heritability is explained by rare protein-coding variation. We identified docking protein 2 (DOK2) and CD200 receptor 1 (CD200R1) as novel genome-wide significant susceptibility genes. Rare coding variants associated with AD are further enriched in 5 genes (IL-4 receptor [IL4R], IL13, Janus kinase 1 [JAK1], JAK2, and tyrosine kinase 2 [TYK2]) of the IL13 pathway, all of which are targets for novel systemic AD therapeutics. Multiomics-based network and RNA sequencing analysis revealed DOK2 as a central hub interacting with, among others, CD200R1, IL6R, and signal transducer and activator of transcription 3 (STAT3). Multitissue gene expression profile analysis for 53 tissue types from the Genotype-Tissue Expression project showed that disease-associated protein-coding variants exert their greatest effect in skin tissues. CONCLUSION Our discoveries highlight a major role of rare coding variants in AD acting independently of common variants. Further extensive functional studies are required to detect all potential causal variants and to specify the contribution of the novel susceptibility genes DOK2 and CD200R1 to overall disease susceptibility.
Collapse
Affiliation(s)
- Sören Mucha
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Hansjörg Baurecht
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany; Department for Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Natalija Novak
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Elke Rodríguez
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Saptarshi Bej
- Department of Systems Biology and Bioinformatics, University of Rostock, Germany
| | - Gabriele Mayr
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Hila Emmert
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Dora Stölzl
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sascha Gerdes
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Eun Suk Jung
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany; Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Matthias Hübenthal
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany; Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Jan Christian Kässens
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Lars Wienbrandt
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank PopGen, Christian Albrechts University of Kiel, Kiel, Germany
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Chair of Genetic Epidemiology, IBE, Faculty of Medicine, Ludwig-Maximilians-University Munich, Germany; Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Melanie Hotze
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Nick Dand
- School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Sarah Grosche
- Pediatric Allergology, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany; Max-Delbrück-Centrum (MDC) for Molecular Medicine, Berlin, Germany
| | - Ingo Marenholz
- Pediatric Allergology, Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany; Max-Delbrück-Centrum (MDC) for Molecular Medicine, Berlin, Germany
| | - Andreas Arnold
- Clinic and Polyclinic of Dermatology, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine and Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Carsten O Schmidt
- Institute for Community Medicine, Study of Health in Pomerania/KEF, University Medicine Greifswald, Greifswald, Germany
| | - Ulrike Wehkamp
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Lavinia Paternoster
- Medical Research Council (MRC) Integrative Epidemiology Unit, Bristol Medical School, and the School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, Gentofte, Denmark
| | - Tarunveer S Ahluwalia
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, Gentofte, Denmark; Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, Gentofte, Denmark
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology and Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology and Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany; Chair of Genetic Epidemiology, IBE, Faculty of Medicine, Ludwig-Maximilians-University Munich, Germany
| | - Thomas Werfel
- Steno Diabetes Center Copenhagen, Gentofte, Denmark; Department of Dermatology and Allergy, Division of Immunodermatology and Allergy Research, Hannover Medical School, Hannover, Germany
| | - Young-Ae Lee
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany; School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Markus Wolfien
- Department of Systems Biology and Bioinformatics, University of Rostock, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University of Rostock, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany; First Medical Department, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Stephan Weidinger
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany.
| |
Collapse
|
38
|
Irvine A, Mina‐Osorio P. Disease trajectories in childhood atopic dermatitis: an update and practitioner's guide. Br J Dermatol 2019; 181:895-906. [PMID: 30758843 PMCID: PMC6899789 DOI: 10.1111/bjd.17766] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a heterogeneous disease with a multifactorial aetiology and complex pathophysiology. This heterogeneity translates into different trajectories of disease progression with respect to severity, persistence and risk of development of atopic comorbidities. Determining which possible disease trajectories or comorbidities any individual child might develop is challenging in clinical practice. Tools that help identify paediatric patients at higher risk of disease progression would greatly aid clinicians. METHODS We reviewed recent cohort studies to synthesize and simplify the epidemiological data to try to identify shared clinically relevant characteristics that may help physicians estimate the risk of disease progression in paediatric patients with AD. RESULTS Despite the variability in data collection and methods of analysis and their limitations, there are common patterns of early-childhood AD that may aid in the estimation of risk for disease progression. Factors associated with risk of AD progression include younger age of onset, family history of atopy, greater AD severity, filaggrin mutations, urban environment and polysensitization and/or allergic multimorbidity. Based on these factors, we provide a practitioner's guide for identifying, counselling and/or referring infants and children with AD at potentially higher risk of developing persistent AD and atopic comorbidities. We also present clinical scenarios to illustrate how these data relate to real-life situations. CONCLUSIONS Useful insights are provided for physicians and patients to inform them better about the risk of AD progression and to help guide care pathways for the paediatric population with AD. What's already known about this topic? The complex pathophysiology of atopic dermatitis (AD) translates into a heterogeneous clinical presentation and trajectories of disease progression. Although the consensus is that most paediatric patients with AD will eventually 'outgrow' the disease or follow the longitudinal trajectory known as the 'atopic march', a significant proportion will develop persistent AD and/or other atopic conditions. No known factors conclusively predict the risk of progression or development of comorbidities. What does this study add? Recent analyses of data from large cohorts of paediatric patients with AD have suggested the existence of potentially discrete clusters of patients who present with relatively common AD phenotypes. These studies have shed some light onto the factors associated with risk of progression, which we review in this article. A practitioner's guide with clinical scenarios is provided to help identify patients at high risk of progression to determine whether a patient should be monitored and/or would require specialist referral.
Collapse
Affiliation(s)
- A.D. Irvine
- Paediatric DermatologyOur Lady's Children's HospitalCrumlin, Dublin12Ireland
- National Children's Research CentreOur Lady's Children's HospitalCrumlin, Dublin12Ireland
- Clinical MedicineTrinity College DublinDublinIreland
| | | |
Collapse
|
39
|
Thériault S, Dina C, Messika-Zeitoun D, Le Scouarnec S, Capoulade R, Gaudreault N, Rigade S, Li Z, Simonet F, Lamontagne M, Clavel MA, Arsenault BJ, Boureau AS, Lecointe S, Baron E, Bonnaud S, Karakachoff M, Charpentier E, Fellah I, Roussel JC, Philippe Verhoye J, Baufreton C, Probst V, Roussel R, Redon R, Dagenais F, Pibarot P, Mathieu P, Le Tourneau T, Bossé Y, Schott JJ. Genetic Association Analyses Highlight IL6, ALPL, and NAV1 As 3 New Susceptibility Genes Underlying Calcific Aortic Valve Stenosis. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 12:e002617. [PMID: 32141789 DOI: 10.1161/circgen.119.002617] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Calcific aortic valve stenosis (CAVS) is a frequent and life-threatening cardiovascular disease for which there is currently no medical treatment available. To date, only 2 genes, LPA and PALMD, have been identified as causal for CAVS. We aimed to identify additional susceptibility genes for CAVS. METHODS A GWAS (genome-wide association study) meta-analysis of 4 cohorts, totaling 5115 cases and 354 072 controls of European descent, was performed. A TWAS (transcriptome-wide association study) was completed to integrate transcriptomic data from 233 human aortic valves. A series of post-GWAS analyses were performed, including fine-mapping, colocalization, phenome-wide association studies, pathway, and tissue enrichment as well as genetic correlation with cardiovascular traits. RESULTS In the GWAS meta-analysis, 4 loci achieved genome-wide significance, including 2 new loci: IL6 (interleukin 6) on 7p15.3 and ALPL (alkaline phosphatase) on 1p36.12. A TWAS integrating gene expression from 233 human aortic valves identified NAV1 (neuron navigator 1) on 1q32.1 as a new candidate causal gene. The CAVS risk alleles were associated with higher mRNA expression of NAV1 in valve tissues. Fine-mapping identified rs1800795 as the most likely causal variant in the IL6 locus. The signal identified colocalizes with the expression of the IL6 RNA antisense in various tissues. Phenome-wide association analyses in the UK Biobank showed colocalized associations between the risk allele at the IL6 lead variant and higher eosinophil count, pulse pressure, systolic blood pressure, and carotid artery procedures, implicating modulation of the IL6 pathways. The risk allele at the NAV1 lead variant colocalized with higher pulse pressure and higher prevalence of carotid artery stenosis. Association results at the genome-wide scale indicated genetic correlation between CAVS, coronary artery disease, and cardiovascular risk factors. CONCLUSIONS Our study implicates 3 new genetic loci in CAVS pathogenesis, which constitute novel targets for the development of therapeutic agents.
Collapse
Affiliation(s)
- Sébastien Thériault
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,Department of Molecular Biology, Medical Biochemistry and Pathology (S.T.), Laval University, Quebec City, Canada
| | - Christian Dina
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.)
| | - David Messika-Zeitoun
- Cardiology Department, AP-HP, Bichat Hospital (D.M.-Z.), Univ Paris 7, France.,INSERM U698 (D.M.-Z.), Univ Paris 7, France.,Division of Cardiology, University of Ottawa Heart Institute, ON, Canada (D.M.-Z.)
| | - Solena Le Scouarnec
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.)
| | - Romain Capoulade
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Nathalie Gaudreault
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada
| | - Sidwell Rigade
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.)
| | - Zhonglin Li
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada
| | - Floriane Simonet
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Maxime Lamontagne
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada
| | - Marie-Annick Clavel
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,Department of Medicine, (M.-A.C., B.J.A., P.P.), Laval University, Quebec City, Canada
| | - Benoit J Arsenault
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,Department of Medicine, (M.-A.C., B.J.A., P.P.), Laval University, Quebec City, Canada
| | - Anne-Sophie Boureau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Simon Lecointe
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Estelle Baron
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.)
| | - Stéphanie Bonnaud
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Matilde Karakachoff
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Eric Charpentier
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Imen Fellah
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Jean-Christian Roussel
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes.,Service de chirurgie Thoracique et CardioVasculaire, CHU Nantes (J.-C.R.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Jean Philippe Verhoye
- Service de chirurgie cardio vasculaire (J.P.V.), Hopital Pontchaillou, Inserm 1099, Rennes
| | | | - Vincent Probst
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Ronan Roussel
- Inserm U1138, Centre de Recherche des Cordeliers (R. Roussel).,University Paris Diderot, Paris University (R. Roussel).,Diabetology, Endocrinology & Nutrition Department, DHU FIRE, Hopital Bichat, AP-HP, Paris (R. Roussel)
| | -
- IRSA, Institut inter-Regional pour la Santé, La Riche, France (the D.E.S.I.R. Study Group)
| | - Richard Redon
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | | | - Philippe Pibarot
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,Department of Medicine, (M.-A.C., B.J.A., P.P.), Laval University, Quebec City, Canada
| | - Patrick Mathieu
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada
| | - Thierry Le Tourneau
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec (S.T., R.C., N.G., Z.L., M.L., M.-A.C., B.J.A., P.P., P.M., Y.B.), Laval University, Quebec City, Canada.,Department of Molecular Medicine (Y.B.), Laval University, Quebec City, Canada
| | - Jean-Jacques Schott
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France (C.D., S.L.S., R.C., S.R., F.S., A.-S.B., S.L., E.B., S.B., M.K., E.C., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.).,l'institut du thorax, CHU Nantes, Nantes, France (R.C., F.S., A.-S.B., S.L., S.B., M.K., I.F., J.-C.R., V.P., R. Redon, T.L.T., J.-J.S.), Hopital Pontchaillou, Inserm 1099, Rennes
| |
Collapse
|
40
|
McGowan LM, Davey Smith G, Gaunt TR, Richardson TG. Integrating Mendelian randomization and multiple-trait colocalization to uncover cell-specific inflammatory drivers of autoimmune and atopic disease. Hum Mol Genet 2019; 28:3293-3300. [PMID: 31276585 PMCID: PMC6859431 DOI: 10.1093/hmg/ddz155] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/07/2019] [Accepted: 06/26/2019] [Indexed: 01/22/2023] Open
Abstract
Immune-mediated diseases (IMDs) arise when tolerance is lost and chronic inflammation is targeted towards healthy tissues. Despite their growing prevalence, therapies to treat IMDs are lacking. Cytokines and their receptors orchestrate inflammatory responses by regulating elaborate signalling networks across multiple cell types making it challenging to pinpoint therapeutically relevant drivers of IMDs. We developed an analytical framework that integrates Mendelian randomization (MR) and multiple-trait colocalization (moloc) analyses to highlight putative cell-specific drivers of IMDs. MR evaluated causal associations between the levels of 10 circulating cytokines and 9 IMDs within human populations. Subsequently, we undertook moloc analyses to assess whether IMD trait, cytokine protein and corresponding gene expression are driven by a shared causal variant. Moreover, we leveraged gene expression data from three separate cell types (monocytes, neutrophils and T cells) to discern whether associations may be attributed to cell type-specific drivers of disease. MR analyses supported a causal role for IL-18 in inflammatory bowel disease (IBD) (P = 1.17 × 10-4) and eczema/dermatitis (P = 2.81 × 10-3), as well as associations between IL-2rα and IL-6R with several other IMDs. Moloc strengthened evidence of a causal association for these results, as well as providing evidence of a monocyte and neutrophil-driven role for IL-18 in IBD pathogenesis. In contrast, IL-2rα and IL-6R associations were found to be T cell specific. Our analytical pipeline can help to elucidate putative molecular pathways in the pathogeneses of IMDs, which could be applied to other disease contexts.
Collapse
Affiliation(s)
- Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Population Health Sciences Institute, University of Bristol, Bristol, BS8 2BN, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Population Health Sciences Institute, University of Bristol, Bristol, BS8 2BN, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Population Health Sciences Institute, University of Bristol, Bristol, BS8 2BN, UK
| |
Collapse
|
41
|
Revez JA, Bain LM, Watson RM, Towers M, Collins T, Killian KJ, O'Byrne PM, Gauvreau GM, Upham JW, Ferreira MA. Effects of interleukin-6 receptor blockade on allergen-induced airway responses in mild asthmatics. Clin Transl Immunology 2019; 8:e1044. [PMID: 31223480 PMCID: PMC6566140 DOI: 10.1002/cti2.1044] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/03/2019] [Accepted: 03/04/2019] [Indexed: 01/12/2023] Open
Abstract
Background Interleukin (IL)-6 signalling has been implicated in allergic asthma by animal, genetic association and clinical studies. In this study, we tested the hypothesis that tocilizumab (TCZ), a human monoclonal antibody that blocks IL-6 signalling, can prevent the development of allergen-induced bronchoconstriction in humans. Methods We performed a randomised, double-blind, placebo-controlled study, with eligible participants completing two allergen inhalation challenge tests, conducted before and after treatment with a single dose of TCZ or placebo. The primary efficacy endpoint was the magnitude of the late asthmatic response recorded between 3 and 7 after allergen challenge. The secondary efficacy endpoint was the early asthmatic response, measured 20 min to 2 h after allergen challenge. Results A total of 66 patients enrolled between September 2014 and August 2017, when the trial was stopped for futility based on results from an interim analysis. Eleven patients fulfilled all eligibility criteria assessed at baseline and were subsequently randomised to the TCZ (n = 6) or placebo (n = 5) groups. Both the primary and secondary efficacy endpoints were not significantly different between the two groups. Five patients reported adverse events (AEs), three in the TCZ group (11 AEs) and two in the placebo group (four AEs). Only one AE was TCZ-related (mild neutropenia), and there were no serious AEs. Significant treatment effects were observed for serum levels of C-reactive protein, IL-6 and soluble IL-6R levels. Conclusion In a small proof-of-concept clinical trial, we found no evidence that a single dose of tocilizumab was able to prevent allergen-induced bronchoconstriction. (Trial registered in the Australian New Zealand Clinical Trials Registry, number ACTRN12614000123640).
Collapse
Affiliation(s)
- Joana A Revez
- QIMR Berghofer Medical Research Institute Brisbane QLD Australia
| | - Lisa M Bain
- QIMR Berghofer Medical Research Institute Brisbane QLD Australia
| | - Rick M Watson
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
| | - Michelle Towers
- Diamantina Institute University of Queensland Brisbane QLD Australia
| | - Tina Collins
- Diamantina Institute University of Queensland Brisbane QLD Australia
| | - Kieran J Killian
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
| | - Paul M O'Byrne
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
| | - Gail M Gauvreau
- Division of Respirology Department of Medicine McMaster University Hamilton ON Canada
| | - John W Upham
- Diamantina Institute University of Queensland Brisbane QLD Australia
| | | |
Collapse
|
42
|
Liu L, Luo Y, Zhou M, Lu Y, Xing M, Ru Y, Sun X, Chen X, Li S, Hong S, Wang S, Li F, Li B, Li X. Tripterygium agents for the treatment of atopic eczema: A Bayesian analysis of randomized controlled trials. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152914. [PMID: 30991183 DOI: 10.1016/j.phymed.2019.152914] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/24/2019] [Accepted: 04/02/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Atopic eczema is a common and recrudescent skin disorder. Tripterygium agents (TA), extracted from Tripterygium wilfordii hook F, a traditional Chinese medicine, have been used as a supplemental therapy for treating eczema empirically in recent years. PURPOSE To investigate the efficacy and safety of TA for treating atopic eczema. STUDY DESIGN Systematic review and Bayesian analysis. METHODS PubMed, Embase, Cochrane Central Register of Controlled Trials, CNKI, Chinese Scientific Journals Database, the Wan Fang Database, and Chinese Biomedicine databases were systematically searched from their respective inception dates to October 2, 2018. Randomized controlled trials (RCTs) related to TA used alone or in combination with other drugs were included. Meta-analysis was conducted by RevMan 5.3 software, and Bayesian analysis was performed in Stata 15.0 and R (V.3.4.0) package gemtc software. The Cochrane risk-of-bias tool and Jadad score were applied to assess the quality of all trials. RESULTS Thirteen trials involving 1385 patients were analyzed. Meta-analysis showed that, when treating atopic eczema patients, TA combined with other drugs were strongly synergistic (p < 0.00001). Among all combinations, the efficacy of TA combined with Diyin tablet (DYP) and topical glucocorticoids (TG) (RR: 0.06, 95%CI [0.01, 0.53]), as well as with compound glycyrrhizin (CG) (RR: 0.36, 95%CI [0.14,0.94]) was superior. Among the different combined medications, the best curative effect was achieved with TA combined with DYP and TG (98.2%), followed by TA combined with CG (85.3%), with TG (51.0%), or with Fuyang granule (FG) (49.9%). Reproductive system dysfunction was the main adverse events in patients treated with TA (RR: 6.23, 95%CI [1.12, 34.62]). Immunoglobulin E (IgE) levels were significantly decreased, after treatment with TA (p = 0.04). Subgroup analysis indicated no statistically significant difference in eczema-related cytokines (p = 0.44). Recurrence rates of using TA and other drugs were similar (p = 0.40). CONCLUSION TA appear to be effective in some therapies when treating patients with atopic eczema, but with apparent side effects. It cannot be concluded that TA can be generally used for eczema in the clinic, because of the small sample size. Further multi-center studies with large samples, and high-quality RCTs should be conducted to clarify the efficacy and safety of TA for treating eczema.
Collapse
Affiliation(s)
- Liu Liu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Mi Zhou
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yi Lu
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Meng Xing
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiaoying Sun
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xi Chen
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Su Li
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Seokgyeong Hong
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Shoumei Wang
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Fulun Li
- Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Bin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Li
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
43
|
Düsterhöft S, Lokau J, Garbers C. The metalloprotease ADAM17 in inflammation and cancer. Pathol Res Pract 2019; 215:152410. [PMID: 30992230 DOI: 10.1016/j.prp.2019.04.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/05/2019] [Accepted: 04/05/2019] [Indexed: 12/23/2022]
Abstract
Proteolytic cleavage of transmembrane proteins is an important post-translational modification that regulates the biological function of numerous transmembrane proteins. Among the 560 proteases encoded in the human genome, the metalloprotease A Disintegrin and Metalloprotease 17 (ADAM17) has gained much attention in recent years and has emerged as a central regulatory hub in inflammation, immunity and cancer development. In order to do so, ADAM17 cleaves a variety of substrates, among them the interleukin-6 receptor (IL-6R), the pro-inflammatory cytokine tumor necrosis factor α (TNFα) and most ligands of the epidermal growth factor receptor (EGFR). This review article provides an overview of the functions of ADAM17 with a special focus on its cellular regulation. It highlights the importance of ADAM17 to understand the biology of IL-6 and TNFα and their role in inflammatory diseases. Finally, the role of ADAM17 in the formation and progression of different tumor entities is discussed.
Collapse
Affiliation(s)
- Stefan Düsterhöft
- Institute for Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
44
|
Murakami M, Kamimura D, Hirano T. Pleiotropy and Specificity: Insights from the Interleukin 6 Family of Cytokines. Immunity 2019; 50:812-831. [DOI: 10.1016/j.immuni.2019.03.027] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
|
45
|
Cai XY, Cheng L, Yu CX, Wu YY, Fang L, Zheng XD, Zhou FS, Sheng YJ, Zhu J, Zheng J, Wu YY, Xiao FL. GWAS Follow-up Study Discovers a Novel Genetic Signal on 10q21.2 for Atopic Dermatitis in Chinese Han Population. Front Genet 2019; 10:174. [PMID: 30915103 PMCID: PMC6422937 DOI: 10.3389/fgene.2019.00174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/18/2019] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disease with high heritability. Two susceptibility loci have been confirmed in our previous AD genome-wide association study (GWAS). To look for additional genetic factors in Chinese Han ethnicity, we performed a large-scale GWAS follow-up study. Forty-nine top single nucleotide polymorphisms (SNPs) that had never been reported previously were genotyped using Sequenom Massarray system in an independent cohort, which consist of northern Chinese (1634 cases and 1263 controls) and southern Chinese (2985 cases and 9526 controls). Association analyses were performed using PLINK 2 software. Three SNPs in northern and ten SNPs in southern were found exhibiting association evidence with AD (P < 0.05). Finally, SNP rs224108 on 10q21.2 showed high significance for AD in joint analysis of GWAS and replication study (Pmeta = 4.55 × 10−9, OR = 1.21), and was confirmed as an independent genetic marker by Linkage disequilibrium calculation and conditional logistic regression analysis. Bioinformatics analysis strongly suggested that rs224108 may have the potential to alter the target gene expression through non-coding epigenetic regulation effects. Meanwhile, SNP rs11150780 on 17q25.3 was also found suggestive association with AD (Pmeta = 7.64 × 10−7, OR = 1.18). Our findings confirmed a novel susceptibility signal on 10q21.2 for AD in Chinese Han population and advanced the understanding of the genetic contribution to AD.
Collapse
Affiliation(s)
- Xin-Ying Cai
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Lu Cheng
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Chong-Xian Yu
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Yan-Yan Wu
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Ling Fang
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Xiao-Dong Zheng
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Fu-Sheng Zhou
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Yu-Jun Sheng
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Jun Zhu
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Jie Zheng
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Yuan-Yuan Wu
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| | - Feng-Li Xiao
- Institute of Dermatology and Department of Dermatology of First Affiliated Hospital, Anhui Medical University, Hefei, China.,Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China.,State Key Laboratory Incubation Base of Dermatology, Anhui Medical University, Hefei, China
| |
Collapse
|
46
|
Lau S, Matricardi PM, Wahn U, Lee YA, Keil T. Allergy and atopy from infancy to adulthood. Ann Allergy Asthma Immunol 2019; 122:25-32. [DOI: 10.1016/j.anai.2018.05.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/01/2018] [Accepted: 05/14/2018] [Indexed: 11/29/2022]
|
47
|
Ghosh D, Bernstein JA, Khurana Hershey GK, Rothenberg ME, Mersha TB. Leveraging Multilayered "Omics" Data for Atopic Dermatitis: A Road Map to Precision Medicine. Front Immunol 2018; 9:2727. [PMID: 30631320 PMCID: PMC6315155 DOI: 10.3389/fimmu.2018.02727] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a complex multifactorial inflammatory skin disease that affects ~280 million people worldwide. About 85% of AD cases begin in childhood, a significant portion of which can persist into adulthood. Moreover, a typical progression of children with AD to food allergy, asthma or allergic rhinitis has been reported (“allergic march” or “atopic march”). AD comprises highly heterogeneous sub-phenotypes/endotypes resulting from complex interplay between intrinsic and extrinsic factors, such as environmental stimuli, and genetic factors regulating cutaneous functions (impaired barrier function, epidermal lipid, and protease abnormalities), immune functions and the microbiome. Though the roles of high-throughput “omics” integrations in defining endotypes are recognized, current analyses are primarily based on individual omics data and using binary clinical outcomes. Although individual omics analysis, such as genome-wide association studies (GWAS), can effectively map variants correlated with AD, the majority of the heritability and the functional relevance of discovered variants are not explained or known by the identified variants. The limited success of singular approaches underscores the need for holistic and integrated approaches to investigate complex phenotypes using trans-omics data integration strategies. Integrating omics layers (e.g., genome, epigenome, transcriptome, proteome, metabolome, lipidome, exposome, microbiome), which often have complementary and synergistic effects, might provide the opportunity to capture the flow of information underlying AD disease manifestation. Overlapping genes/candidates derived from multiple omics types include FLG, SPINK5, S100A8, and SERPINB3 in AD pathogenesis. Overlapping pathways include macrophage, endothelial cell and fibroblast activation pathways, in addition to well-known Th1/Th2 and NFkB activation pathways. Interestingly, there was more multi-omics overlap at the pathway level than gene level. Further analysis of multi-omics overlap at the tissue level showed that among 30 tissue types from the GTEx database, skin and esophagus were significantly enriched, indicating the biological interconnection between AD and food allergy. The present work explores multi-omics integration and provides new biological insights to better define the biological basis of AD etiology and confirm previously reported AD genes/pathways. In this context, we also discuss opportunities and challenges introduced by “big omics data” and their integration.
Collapse
Affiliation(s)
- Debajyoti Ghosh
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Jonathan A Bernstein
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Tesfaye B Mersha
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
48
|
Zhu Z, Lee PH, Chaffin MD, Chung W, Loh PR, Lu Q, Christiani DC, Liang L. A genome-wide cross-trait analysis from UK Biobank highlights the shared genetic architecture of asthma and allergic diseases. Nat Genet 2018; 50:857-864. [PMID: 29785011 PMCID: PMC5980765 DOI: 10.1038/s41588-018-0121-0] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 03/27/2018] [Indexed: 01/10/2023]
Abstract
Clinical and epidemiological data suggest that asthma and allergic
diseases are associated and may share a common genetic etiology. We analyzed
genome-wide single-nucleotide polymorphism (SNP) data for asthma and allergic
diseases in 33,593 cases and 76,768 controls of European ancestry from the UK
Biobank. Two publicly available independent genome wide association studies
(GWAS) were used for replication. We have found a strong genome-wide genetic
correlation between asthma and allergic diseases (rg
= 0.75, P =
6.84×10−62). Cross trait analysis identified 38
genome-wide significant loci, including 7 novel shared loci. Computational
analysis showed that shared genetic loci are enriched in immune/inflammatory
systems and tissues with epithelium cells. Our work identifies common genetic
architectures shared between asthma and allergy and will help to advance our
understanding of the molecular mechanisms underlying co-morbid asthma and
allergic diseases.
Collapse
Affiliation(s)
- Zhaozhong Zhu
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Phil H Lee
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Mark D Chaffin
- Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Wonil Chung
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Po-Ru Loh
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Medical and Population Genetics Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Quan Lu
- Program in Molecular and Integrative Physiological Sciences, Departments of Environmental Health and Genetics & Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.,Pulmonary and Critical Care Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Liming Liang
- Program in Genetic Epidemiology and Statistical Genetics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA. .,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA.
| |
Collapse
|
49
|
Tamura Y, Phan C, Tu L, Le Hiress M, Thuillet R, Jutant EM, Fadel E, Savale L, Huertas A, Humbert M, Guignabert C. Ectopic upregulation of membrane-bound IL6R drives vascular remodeling in pulmonary arterial hypertension. J Clin Invest 2018; 128:1956-1970. [PMID: 29629897 DOI: 10.1172/jci96462] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by a progressive accumulation of pulmonary artery smooth muscle cells (PA-SMCs) in pulmonary arterioles leading to the narrowing of the lumen, right heart failure, and death. Although most studies have supported the notion of a role for IL-6/glycoprotein 130 (gp130) signaling in PAH, it remains unclear how this signaling pathway determines the progression of the disease. Here, we identify ectopic upregulation of membrane-bound IL-6 receptor (IL6R) on PA-SMCs in PAH patients and in rodent models of pulmonary hypertension (PH) and demonstrate its key role for PA-SMC accumulation in vitro and in vivo. Using Sm22a-Cre Il6rfl/fl, which lack Il6r in SM22A-expressing cells, we found that these animals are protected against chronic hypoxia-induced PH with reduced PA-SMC accumulation, revealing the potent pro-survival potential of membrane-bound IL6R. Moreover, we determine that treatment with IL6R-specific antagonist reverses experimental PH in two rat models. This therapeutic strategy holds promise for future clinical studies in PAH.
Collapse
|
50
|
Kim JH, Lee SY, Kang MJ, Yoon J, Jung S, Cho HJ, Kim HB, Hong SJ. Association of Genetic Polymorphisms with Atopic Dermatitis, Clinical Severity and Total IgE: A Replication and Extended Study. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:397-405. [PMID: 29949836 PMCID: PMC6021591 DOI: 10.4168/aair.2018.10.4.397] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 11/20/2022]
Abstract
Purpose Atopic dermatitis (AD) is a common and chronic inflammatory skin disease affecting up to 20% of children and 3% of adults worldwide. Although previous reports including genome-wide association study (GWAS) approaches have identified several risk factors that appear to be associated with AD development, replication studies are lacking. In our current study, we replicated the associations between candidate susceptibility loci and AD. Methods A total of 885 Korean subjects (425 AD patients and 460 unaffected controls) were genotyped for 17 single nucleotide polymorphisms (SNPs) from previous GWASs and meta-analyses of AD and from immune-related genes. Results Several SNPs showed significant associations with AD in the case-control analysis (minimum P=0.005 at rs17389644), suggesting that these polymorphisms may be related to this disease. In addition, several SNPs showed significant signals (minimum P=0.004 at rs6473227) in severe AD compared to unaffected controls. In additional linear regression analysis, a few genotypes appeared to have potential effects on the SCORing AD (SCORAD) values (minimum P=0.003 at rs13361382 on TMEM232) and immunoglobulin E (IgE) levels (minimum P<0.0001 at rs4713555 near HLA-DRB1 and HLA-DQA1) in AD patients. Conclusions Our replication and extended study provide additional supporting information on the genetic associations (especially, variants in TMEM232 and nearby to IL21 and HLA-DRB1/HLA-DQA1) related to AD, its clinical severity and IgE involvement.
Collapse
Affiliation(s)
- Jeong Hyun Kim
- Department of Medicine, University of Ulsan Collage of Medicine, Seoul, Korea
| | - So Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Environmental Health Center, Asan Medical Center, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Mi Jin Kang
- Asan Institute for Life Sciences, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Jisun Yoon
- Department of Pediatrics, Childhood Asthma Atopy Center, Environmental Health Center, Asan Medical Center, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Sungsu Jung
- Department of Pediatrics, Childhood Asthma Atopy Center, Environmental Health Center, Asan Medical Center, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Hyun Ju Cho
- Department of Pediatrics, Mediplex Sejong Hospital, Incheon, Korea
| | - Hyo Bin Kim
- Department of Pediatrics, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Soo Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Environmental Health Center, Asan Medical Center, University of Ulsan Collage of Medicine, Seoul, Korea.
| |
Collapse
|