1
|
Dawe HR, Di Meglio P. The Aryl Hydrocarbon Receptor (AHR): Peacekeeper of the Skin. Int J Mol Sci 2025; 26:1618. [PMID: 40004095 PMCID: PMC11855870 DOI: 10.3390/ijms26041618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
In the last decade, the aryl hydrocarbon receptor (AHR) has emerged as a critical peacekeeper for the maintenance of healthy skin. The evolutionary conservation of AHR implied physiological functions for this receptor, beyond the detoxification of man-made compounds, a notion further supported by the existence of physiological AHR ligands, notably derivates of tryptophan by the host and host microbiome. The UV light-derived ligand, 6-formylindolo[3,2-b]carbazole (FICZ), anticipated a role for AHR in skin, a UV light-exposed organ, where physiological AHR activation promotes a healthy skin barrier and constrains inflammation. The clinical development of tapinarof, the first topical AHR modulating drug for inflammatory skin disease, approved by the FDA for mild-to-moderate psoriasis and poised for approval in atopic dermatitis, supports the therapeutic targeting of the AHR pathway to harness its beneficial effect in skin inflammation. Here, we describe how a tightly controlled, physiological activation of the AHR pathway maintains skin homeostasis, and discuss how the pathway is dysregulated in psoriasis and atopic dermatitis, identifying areas offering opportunities for alternative therapeutic approaches, for further investigation.
Collapse
Affiliation(s)
- Hannah R. Dawe
- St John’s Institute of Dermatology, King’s College London, London SE1 9RT, UK;
- KHP Centre for Translational Medicine, London SE1 9RT, UK
| | - Paola Di Meglio
- St John’s Institute of Dermatology, King’s College London, London SE1 9RT, UK;
- KHP Centre for Translational Medicine, London SE1 9RT, UK
| |
Collapse
|
2
|
Freisenhausen JC, Luo L, Kelemen E, Elton J, Skoog V, Pivarcsi A, Sonkoly E. RNA Sequencing Reveals the Long Non-Coding RNA Signature in Psoriasis Keratinocytes and Identifies CYDAER as a Long Non-Coding RNA Regulating Epidermal Differentiation. Exp Dermatol 2025; 34:e70054. [PMID: 39953783 PMCID: PMC11829188 DOI: 10.1111/exd.70054] [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: 08/17/2024] [Revised: 12/19/2024] [Accepted: 01/26/2025] [Indexed: 02/17/2025]
Abstract
Psoriasis is a common chronic inflammatory skin disease determined by genetic and environmental factors, resulting in the activation of IL-23/IL-17-mediated immune response, epidermal hyperproliferation, and keratinocyte activation. Long non-coding RNAs (lncRNAs) are non-protein-coding transcripts > 500 nucleotides with diverse regulatory functions; their role in epidermal dysfunction in psoriasis is poorly understood. To identify epidermal transcripts with potential roles in psoriasis, including lncRNAs, we performed RNA sequencing on keratinocytes from psoriasis and healthy skin. We identified 889 differentially expressed lncRNAs, many of which with yet unknown functions. RP11-295G20.2 was identified as a lncRNA significantly induced in psoriasis keratinocytes, and this was verified by qRT-PCR and by single-molecule in situ hybridisation. Analysis of subcellular fractions of epidermis revealed a cytoplasmic localisation in line with results of single molecule in situ hybridisation. We report that RP11-295G20.2 has a skin-enriched expression, and within skin it is mainly expressed in suprabasal epidermal layers. Moreover, RP11-295G20.2 is induced by the key psoriasis cytokine IL-17A and shows a dynamic regulation during keratinocyte differentiation with upregulation during early differentiation and downregulation in the late stage. Knockdown of RP11-295G20.2 in keratinocytes promotes terminal differentiation. Based on our findings, we named RP11-295G20.2 Cytoplasmic Differentiation-Associated Epidermal RNA, CYDAER. In summary, our study provides a comprehensive characterisation of the non-coding RNA landscape of psoriasis keratinocytes and identifies CYDAER as a skin-enriched lncRNA regulating keratinocyte differentiation. Our data suggest that overexpression of CYDAER may contribute to altered differentiation in psoriatic epidermis.
Collapse
Affiliation(s)
- Jan Cedric Freisenhausen
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
- Division of Dermatology and Venereology, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| | - Longlong Luo
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
- Division of Dermatology and Venereology, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| | - Evelyn Kelemen
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
| | - Jonathan Elton
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
- Division of Dermatology and Venereology, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| | - Viktor Skoog
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
| | - Andor Pivarcsi
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
- Division of Dermatology and Venereology, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| | - Enikö Sonkoly
- Dermatology and Venereology, Department of Medical SciencesUppsala UniversityUppsalaSweden
- Department of Medical Biochemistry and MicrobiologyUppsala UniversityUppsalaSweden
- Division of Dermatology and Venereology, Department of Medicine SolnaKarolinska InstitutetStockholmSweden
| |
Collapse
|
3
|
Tsoi L, Dong Y, Patrick M, Sarkar M, Zhang H, Bogle R, Zhang Z, Dand N, Paulsen M, Ljungman M, Betz RC, Petukhova L, Christiano A, Simpson M, Modlin R, Khanna D, Barker J, Budunova I, Gharaee-Kermani M, Billi A, Elder J, Kahlenberg JM, Gudjonsson J. IL-1 signaling enrichment in inflammatory skin disease loci with higher-risk allele frequencies in African ancestry. RESEARCH SQUARE 2025:rs.3.rs-5724270. [PMID: 39975900 PMCID: PMC11838759 DOI: 10.21203/rs.3.rs-5724270/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Inflammatory skin diseases (ISDs) exhibit varying prevalence across different ancestry background and geographical regions. Genetic research for complex ISDs has predominantly centered on European Ancestry (EurA) populations and genetic effects on immune cell responses but generally failed to consider contributions from other cell types in skin. Here, we utilized 273 genetic signals from seven different ISDs: acne, alopecia areata (AA), atopic dermatitis (AD), psoriasis, systemic lupus erythematosus (SLE), systemic sclerosis (SSc), and vitiligo, to demonstrate enriched IL1 signaling in keratinocytes, particularly in signals with higher risk allele frequencies in the African ancestry. Using a combination of ATAC-seq, Bru-seq, and promoter capture Hi-C, we revealed potential regulatory mechanisms of the acne locus on chromosome 2q13. We further demonstrated differential responses in keratinocytes upon IL1β stimulation, including the pro-inflammatory mediators CCL5, IL36G, and CXCL8. Taken together, our findings highlight IL1 signaling in epidermal keratinocytes as a contributor to ancestry-related differences in ISDs.
Collapse
Affiliation(s)
| | | | | | | | | | - Rachael Bogle
- Department of Dermatology, INSERM 1098, Franche comté university, Besançon university hospital
| | | | | | | | | | | | | | | | | | - Robert Modlin
- University of California Los Angeles, David Geffen School of Medicine
| | | | | | | | | | | | - James Elder
- Department of Dermatology, University of Michigan, 1500 East Medical Center
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, 48109, USA
| | | |
Collapse
|
4
|
Mai J, Qian Q, Gao H, Fan Z, Zeng J, Xiao J. scTWAS Atlas: an integrative knowledgebase of single-cell transcriptome-wide association studies. Nucleic Acids Res 2025; 53:D1195-D1204. [PMID: 39420631 PMCID: PMC11701648 DOI: 10.1093/nar/gkae931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Single-cell transcriptome-wide association studies (scTWAS) is a new method for conducting TWAS analysis at the cellular level to identify gene-trait associations with higher precision. This approach helps overcome the challenge of interpreting cell-type heterogeneity in traditional TWAS results. As the field of scTWAS rapidly advances, there is a growing need for additional database platforms to integrate this wealth of data and knowledge effectively. To address this gap, we present scTWAS Atlas (https://ngdc.cncb.ac.cn/sctwas/), a comprehensive database of scTWAS information integrating literature curation and data analysis. The current version of scTWAS Atlas amasses 2,765,211 associations encompassing 34 traits, 30 cell types, 9 cell conditions and 16,470 genes. The database features visualization tools, including an interactive knowledge graph that integrates single-cell expression quantitative trait loci (sc-eQTL) and scTWAS associations to build a multi-omics level regulatory network at the cellular level. Additionally, scTWAS Atlas facilitates cross-cell-type analysis, highlighting cell-type-specific and shared TWAS genes. The database is designed with user-friendly interfaces and allows for easy browsing, searching, and downloading of relevant information. Overall, scTWAS Atlas is instrumental in exploring the genetic regulatory mechanisms at the cellular level and shedding light on the role of various cell types in biological processes, offering novel insights for human health research.
Collapse
Affiliation(s)
- Jialin Mai
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiheng Qian
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Gao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuojing Fan
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingyao Zeng
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingfa Xiao
- National Genomics Data Center, China National Center for Bioinformation, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Dave K, Patel D, Dave N, Jain M. Genomic strategies for drug repurposing. J Egypt Natl Canc Inst 2024; 36:35. [PMID: 39523244 DOI: 10.1186/s43046-024-00245-z] [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: 06/17/2024] [Accepted: 09/29/2024] [Indexed: 11/16/2024] Open
Abstract
Functional genomics, a multidisciplinary subject, investigates the functions of genes and their products in biological systems to better understand diseases and find new drugs. Drug repurposing is an economically efficient approach that entails discovering novel therapeutic applications for already-available medications. Genomics enables the identification of illness and therapeutic molecular characteristics and interactions, which in turn facilitates the process of drug repurposing. Techniques like gene expression profiling and Mendelian randomization are helpful in identifying possible medication candidates. Progress in computer science allows for the investigation and modeling of gene expression networks that involve large amounts of data. The amalgamation of data concerning DNA, RNA, and protein functions bears similarity to pharmacogenomics, a crucial aspect in crafting cancer therapeutics. Functional genomics in drug discovery, particularly for cancer, is still not thoroughly investigated, despite the existence of a significant amount of literature on the subject. Next-generation sequencing and proteomics present highly intriguing opportunities. Publicly available databases and mining techniques facilitate the development of cancer treatments based on functional genomics. Broadening the exploration and utilization of functional genomics holds significant potential for advancing drug discovery and repurposing, particularly within the realm of oncology.
Collapse
Affiliation(s)
- Kirtan Dave
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India.
- Bioinformatics Laboratory, Research & Development Cell, Parul University, Vadodara, Gujarat, India.
| | - Dhaval Patel
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Nischal Dave
- Bioinformatics Laboratory, Research & Development Cell, Parul University, Vadodara, Gujarat, India
| | - Mukul Jain
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara, Gujarat, India
- Cell & Developmental Biology Lab, Research and Development Cell, Parul University, Vadodara, Gujarat, India
| |
Collapse
|
6
|
Ramessur R, Saklatvala J, Budu-Aggrey A, Ostaszewski M, Möbus L, Greco D, Ndlovu M, Mahil SK, Barker JN, Brown S, Paternoster L, Dand N, Simpson MA, Smith CH. Exploring the Link Between Genetic Predictors of Cardiovascular Disease and Psoriasis. JAMA Cardiol 2024; 9:1009-1017. [PMID: 39292496 PMCID: PMC11411451 DOI: 10.1001/jamacardio.2024.2859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/13/2024] [Indexed: 09/19/2024]
Abstract
Importance The epidemiological link between immune-mediated diseases (IMIDs) and cardiovascular disease has often been attributed to systemic inflammation. However, the direction of causality and the biological mechanisms linking cardiovascular disease with IMIDs are incompletely understood. Given the robust epidemiological association and the growing body of supportive mechanistic evidence, psoriasis is an exemplary IMID model for exploring this relationship. Objective To assess the bidirectional relationships between genetic predictors of psoriasis and the 2 major forms of cardiovascular disease, coronary artery disease (CAD) and stroke, and to evaluate the association between genetic predictors of cardiovascular disease with 9 other IMIDs. Design, Setting, and Participants This was a genetic association study using mendelian randomization (MR), a powerful genetic tool to help distinguish causation from associations observed in epidemiological studies, to provide supportive evidence for causality between traits. The study conducted 2-sample MR analyses using summary-level data from large-scale genome-wide association meta-analysis studies (GWAS) for each trait. The analysis focused on individuals of European descent from GWAS meta-analyses, involving CAD, stroke, psoriasis, and 9 other IMIDs. Data were analyzed from January 2023 to May 2024. Exposures Genetic predictors of CAD, stroke, psoriasis, and 9 other IMIDs. Main Outcomes and Measures The primary outcomes were the associations of genetic predictors of CAD and stroke with the risk of psoriasis and 9 other IMIDs, determined using inverse-variance weighted (IVW) MR estimates. Results This study included 181 249 cases and 1 165 690 controls with CAD, 110 182 cases and 1 503 898 controls with stroke, 36 466 cases and 458 078 controls with psoriasis, for a total of approximately 3 400 000 individuals, and 9 other IMIDs. In contrast to previous assumptions, genetic predictors of psoriasis were found to have no association with CAD or stroke. In the reverse direction, genetic predictors of both CAD (MR estimate IVW odds ratio [OR], 1.07; 95% CI, 1.04-1.10; P = .003) and stroke (IVW OR, 1.22; 95% CI, 1.05-1.41; P = .01) were found to have risk-increasing associations with psoriasis. Adjusting for stroke rendered the associations of genetically predicted CAD with psoriasis risk nonsignificant (and vice versa), suggesting that a shared effect underlying genetic risk for CAD and stroke associates with increased psoriasis risk. No risk-increasing associations were observed for genetic predictors of cardiovascular disease with other common IMIDs, including rheumatoid arthritis and inflammatory bowel disease. Conclusions and Relevance Findings of this mendelian randomization study indicate that genetic predictors of cardiovascular disease were associated with increased psoriasis risk with no reciprocal effect or association with other IMIDs. Elucidating mechanisms underpinning this association could lead to novel therapeutic approaches in both diseases.
Collapse
Affiliation(s)
- Ravi Ramessur
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jake Saklatvala
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Ashley Budu-Aggrey
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lena Möbus
- Finnish Hub for Development and Validation of Integrated Approaches, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Matladi Ndlovu
- Department of Immunology Research, UCB, Brussels, Belgium
| | - Satveer K. Mahil
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jonathan N. Barker
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Sara Brown
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Department of Dermatology, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, United Kingdom
| | - Nick Dand
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Michael A. Simpson
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Catherine H. Smith
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
7
|
Rossi S, Richards EL, Orozco G, Eyre S. Functional Genomics in Psoriasis. Int J Mol Sci 2024; 25:7349. [PMID: 39000456 PMCID: PMC11242296 DOI: 10.3390/ijms25137349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Psoriasis is an autoimmune cutaneous condition that significantly impacts quality of life and represents a burden on society due to its prevalence. Genome-wide association studies (GWASs) have pinpointed several psoriasis-related risk loci, underlining the disease's complexity. Functional genomics is paramount to unveiling the role of such loci in psoriasis and disentangling its complex nature. In this review, we aim to elucidate the main findings in this field and integrate our discussion with gold-standard techniques in molecular biology-i.e., Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-and high-throughput technologies. These tools are vital to understanding how disease risk loci affect gene expression in psoriasis, which is crucial in identifying new targets for personalized treatments in advanced precision medicine.
Collapse
Affiliation(s)
| | | | | | - Stephen Eyre
- Centre for Genetics and Genomics versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK; (S.R.); (E.L.R.); (G.O.)
| |
Collapse
|
8
|
Wu J, Ma Y, Yang J, Tian Y. Exposure to Air Pollution, Genetic Susceptibility, and Psoriasis Risk in the UK. JAMA Netw Open 2024; 7:e2421665. [PMID: 39012635 PMCID: PMC11252902 DOI: 10.1001/jamanetworkopen.2024.21665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/17/2024] [Indexed: 07/17/2024] Open
Abstract
IMPORTANCE Psoriasis is a common autoinflammatory disease influenced by complex interactions between environmental and genetic factors. The influence of long-term air pollution exposure on psoriasis remains underexplored. OBJECTIVE To examine the association between long-term exposure to air pollution and psoriasis and the interaction between air pollution and genetic susceptibility for incident psoriasis. DESIGN, SETTING, AND PARTICIPANTS This prospective cohort study used data from the UK Biobank. The analysis sample included individuals who were psoriasis free at baseline and had available data on air pollution exposure. Genetic analyses were restricted to White participants. Data were analyzed between November 1 and December 10, 2023. EXPOSURES Exposure to nitrogen dioxide (NO2), nitrogen oxides (NOx), fine particulate matter with a diameter less than 2.5 µm (PM2.5), and particulate matter with a diameter less than 10 µm (PM10) and genetic susceptibility for psoriasis. MAIN OUTCOMES AND MEASURES To ascertain the association of long-term exposure to NO2, NOx, PM2.5, and PM10 with the risk of psoriasis, a Cox proportional hazards model with time-varying air pollution exposure was used. Cox models were also used to explore the potential interplay between air pollutant exposure and genetic susceptibility for the risk of psoriasis incidence. RESULTS A total of 474 055 individuals were included, with a mean (SD) age of 56.54 (8.09) years and 257 686 (54.36%) female participants. There were 9186 participants (1.94%) identified as Asian or Asian British, 7542 (1.59%) as Black or Black British, and 446 637 (94.22%) as White European. During a median (IQR) follow-up of 11.91 (11.21-12.59) years, 4031 incident psoriasis events were recorded. There was a positive association between the risk of psoriasis and air pollutant exposure. For every IQR increase in PM2.5, PM10, NO2, and NOx, the hazard ratios (HRs) were 1.41 (95% CI, 1.35-1.46), 1.47 (95% CI, 1.41-1.52), 1.28 (95% CI, 1.23-1.33), and 1.19 (95% CI, 1.14-1.24), respectively. When comparing individuals in the lowest exposure quartile (Q1) with those in the highest exposure quartile (Q4), the multivariate-adjusted HRs were 2.01 (95% CI, 1.83-2.20) for PM2.5, 2.21 (95% CI, 2.02-2.43) for PM10, 1.64 (95% CI, 1.49-1.80) for NO2, and 1.34 (95% CI, 1.22-1.47) for NOx. Moreover, significant interactions between air pollution and genetic predisposition for incident psoriasis were observed. In the subset of 446 637 White individuals, the findings indicated a substantial risk of psoriasis development in participants exposed to the highest quartile of air pollution levels concomitant with high genetic risk compared with those in the lowest quartile of air pollution levels with low genetic risk (PM2.5: HR, 4.11; 95% CI, 3.46-4.90; PM10: HR, 4.29; 95% CI, 3.61-5.08; NO2: HR, 2.95; 95% CI, 2.49-3.50; NOx: HR, 2.44; 95% CI, 2.08-2.87). CONCLUSIONS AND RELEVANCE In this prospective cohort study of the association between air pollution and psoriasis, long-term exposure to air pollution was associated with increased psoriasis risk. There was an interaction between air pollution and genetic susceptibility on psoriasis risk.
Collapse
Affiliation(s)
- Junhui Wu
- School of Nursing, Peking University, Beijing, China
| | - Yudiyang Ma
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People’s Hospital, Yichang, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, China
- Hubei Provincial Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, China
| | - Yaohua Tian
- Ministry of Education Key Laboratory of Environment and Health, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Shellard EM, Rane SS, Eyre S, Warren RB. Functional Genomics and Insights into the Pathogenesis and Treatment of Psoriasis. Biomolecules 2024; 14:548. [PMID: 38785955 PMCID: PMC11117854 DOI: 10.3390/biom14050548] [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: 03/11/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Psoriasis is a lifelong, systemic, immune mediated inflammatory skin condition, affecting 1-3% of the world's population, with an impact on quality of life similar to diseases like cancer or diabetes. Genetics are the single largest risk factor in psoriasis, with Genome-Wide Association (GWAS) studies showing that many psoriasis risk genes lie along the IL-23/Th17 axis. Potential psoriasis risk genes determined through GWAS can be annotated and characterised using functional genomics, allowing the identification of novel drug targets and the repurposing of existing drugs. This review is focused on the IL-23/Th17 axis, providing an insight into key cell types, cytokines, and intracellular signaling pathways involved. This includes examination of currently available biological treatments, time to relapse post drug withdrawal, and rates of primary/secondary drug failure, showing the need for greater understanding of the underlying genetic mechanisms of psoriasis and how they can impact treatment. This could allow for patient stratification towards the treatment most likely to reduce the burden of disease for the longest period possible.
Collapse
Affiliation(s)
- Elan May Shellard
- Faculty of Biology, Medicine and Health, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - Shraddha S. Rane
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, The University of Manchester, Manchester M13 9PT, UK; (S.S.R.); (S.E.)
| | - Stephen Eyre
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, NIHR Manchester Biomedical Research Centre, The University of Manchester, Manchester M13 9PT, UK; (S.S.R.); (S.E.)
| | - Richard B. Warren
- Dermatology Centre, Northern Care Alliance NHS Foundation Trust, Manchester M6 8HD, UK;
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M23 9LT, UK
| |
Collapse
|
10
|
Guo J, Luo Q, Li C, Liang H, Cao Q, Li Z, Chen G, Yu X. Evidence for the gut-skin axis: Common genetic structures in inflammatory bowel disease and psoriasis. Skin Res Technol 2024; 30:e13611. [PMID: 38348734 PMCID: PMC10862160 DOI: 10.1111/srt.13611] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/26/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) and psoriasis (Ps) are common immune-mediated diseases that exhibit clinical comorbidity, possibly due to a common genetic structure. However, the exact mechanism remains unknown. METHODS The study population consisted of IBD and Ps genome-wide association study (GWAS) data. Genetic correlations were first evaluated. Then, the overall evaluation employed LD score regression (LDSC), while the local assessment utilized heritability estimation from summary statistics (HESS). Causality assessment was conducted through two-sample Mendelian randomization (2SMR), and genetic overlap analysis utilized the conditional false discovery rate/conjunctional FDR (cond/conjFDR) method. Finally, LDSC applied to specifically expressed genes (LDSC-SEG) was performed at the tissue level. For IBD and Ps-specific expressed genes, genetic correlation, causality, shared genetics, and trait-specific associated tissues were methodically examined. RESULTS At the genomic level, both overall and local genetic correlations were found between IBD and Ps. MR analysis indicated a positive causal relationship between Ps and IBD. The conjFDR analysis with a threshold of < 0.01 identified 43 loci shared between IBD and Ps. Subsequent investigations into disease-associated tissues indicated a close association of IBD and Ps with whole blood, lung, spleen, and EBV-transformed lymphocytes. CONCLUSION The current research offers a novel perspective on the association between IBD and Ps. It contributes to an enhanced comprehension of the genetic structure and mechanisms of comorbidities in both diseases.
Collapse
Affiliation(s)
- Jinyan Guo
- Department of Anorectal SurgeryJiangmen Wuyi Hospital of Traditional Chinese MedicineJiangmenChina
| | - Qinghua Luo
- Clinical Medical CollegeJiangxi University of Chinese MedicineNanchangChina
| | - Chunsheng Li
- Department of Anorectal SurgeryJiangmen Wuyi Hospital of Traditional Chinese MedicineJiangmenChina
| | - Hong Liang
- Department of Anorectal SurgeryJiangmen Wuyi Hospital of Traditional Chinese MedicineJiangmenChina
| | - Qiurui Cao
- Department of Anorectal SurgeryJiangmen Wuyi Hospital of Traditional Chinese MedicineJiangmenChina
| | - Zihao Li
- Department of Anorectal SurgeryJiangmen Wuyi Hospital of Traditional Chinese MedicineJiangmenChina
| | - Guanghua Chen
- Department of Anorectal SurgeryAffiliated Hospital of Jiangxi University of Chinese MedicineNanchangChina
| | - Xuchao Yu
- Department of Anorectal SurgeryAffiliated Hospital of Jiangxi University of Chinese MedicineNanchangChina
| |
Collapse
|
11
|
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
|
12
|
Stacey VM, Kõks S. Genome-Wide Differential Transcription of Long Noncoding RNAs in Psoriatic Skin. Int J Mol Sci 2023; 24:16344. [PMID: 38003532 PMCID: PMC10671291 DOI: 10.3390/ijms242216344] [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: 10/19/2023] [Revised: 11/11/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) may contribute to the formation of psoriatic lesions. The present study's objective was to identify long lncRNA genes that are differentially expressed in patient samples of psoriasis through computational analysis techniques. By using previously published RNA sequencing data from psoriatic and healthy patients (n = 324), we analysed the differential expression of lncRNAs to determine transcripts of heightened expression. We computationally screened lncRNA transcripts as annotated by GENCODE across the human genome and compared transcription in psoriatic and healthy samples from two separate studies. We observed 54 differentially expressed genes as seen in two independent datasets collected from psoriasis and healthy patients. We also identified the differential expression of LINC01215 and LINC1206 associated with the cell cycle pathway and psoriasis pathogenesis. SH3PXD2A-AS1 was identified as a participant in the STAT3/SH3PXD2A-AS1/miR-125b/STAT3 positive feedback loop. Both the SH3PXD2A-AS1 and CERNA2 genes have already been recognised as part of the IFN-γ signalling pathway regulation. Additionally, EPHA1-AS1, CYP4Z2P and SNHG12 gene upregulation have all been previously linked to inflammatory skin diseases. Differential expression of various lncRNAs affects the pathogenesis of psoriasis. Further characterisation of lncRNAs and their functions are important for developing our understanding of psoriasis.
Collapse
Affiliation(s)
- Valerie M. Stacey
- Perron Institute for Neurological and Translational Science, 8 Verdun Street, Nedlands, WA 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, 8 Verdun Street, Nedlands, WA 6009, Australia;
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA 6150, Australia
| |
Collapse
|