1
|
Kim J, Kim BE, Ahn K, Leung DYM. Skin Predictive Biomarkers for the Development of Atopic Dermatitis and Food Allergy in Infants. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:323-337. [PMID: 39155734 PMCID: PMC11331187 DOI: 10.4168/aair.2024.16.4.323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The pathogenesis of atopic dermatitis (AD) is multifactorial, involving a dynamic interplay between genetic susceptibility, skin-barrier dysfunction, microbiome alterations, and immune dysregulation, whereas food allergy (FA) arises from the interplay of transcutaneous sensitization to food allergens and failure in the induction of oral tolerance. Skin epicutaneous sensitization is commonly involved in the development of AD and FA. Although clinical trials have been conducted to prevent AD or FA by applications of emollients on the skin after birth, the results are not consistent. For more effective preventive strategies, reliable biomarkers are required to identify high-risk individuals. Skin tape stripping (STS) is a non-invasive technique for identifying these biomarkers in the skin. By analyzing the stratum corneum collected via STS, researchers can gain molecular or cellular insights into the early pathogenesis and potential progression of AD and FA. This review aims to elucidate the critical aspects of AD and FA, underlying their pathogenesis, early manifestations, and STS's potential as a tool for identifying predictive non-invasive biomarkers in infants prior to onset of clinical disease.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea
| | - Byung Eui Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology, Seoul, Korea.
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
2
|
Huang Y, Zhou W, Liu S, Zeng D, Zhou W. Association between polymorphisms and atopic dermatitis susceptibility: A systematic review and meta-analysis. Gene 2024; 913:148397. [PMID: 38513928 DOI: 10.1016/j.gene.2024.148397] [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: 12/10/2023] [Revised: 03/13/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
AIM Atopic dermatitis (AD) is a chronic pruritic inflammatory skin disease that is closely linked to genetic factors. Previous studies have revealed numerous single nucleotide polymorphisms (SNPs) that been related to susceptibility to AD; however, the results are conflicting. Therefore, a meta-analysis was conducted to assess the associations of these polymorphisms and AD risk. MATERIAL AND METHODS PubMed, Web of Science, Embase, Cochrane Library, and China National Knowledge Infrastructure databases were retrieved to identify eligible studies, with selected polymorphisms being reported in a minimum of three separate studies. The Newcastle-Ottawa Scale (NOS) was used to evaluate study quality. Review Manager 5.3 and STATA 14.0 were used to perform the meta-analysis. RESULTS After screening, 64 studies involving 13 genes (24 SNPs) were selected for inclusion in the meta-analysis. Nine SNPs were positively correlated with AD susceptibility [filaggrin (FLG) R501X, FLG 2282del4, chromosome 11q13.5 rs7927894, interleukin (IL)-17A rs2275913, IL-18 -137 G/C, Toll-like receptor 2 (TLR2) rs5743708, TLR2 A-16934 T, serine protease inhibitor Kazal type-5 (SPINK5) Asn368Ser, interferon-γ (IFN-γ) T874A] and one was negatively associated with AD susceptibility (IL-4 -1098 T/G). The 14 remaining SNPs were not significantly associated with AD susceptibility. CONCLUSIONS Nine SNPs that may be risk factors and one SNP that may be a protective factor for AD were identified, providing a reference for AD prediction, prevention, and therapy.
Collapse
Affiliation(s)
- Yunxia Huang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Department of Allergy, Chongqing General Hospital, Chongqing 400014, China
| | - Wei Zhou
- Department of Allergy, Chongqing General Hospital, Chongqing 400014, China
| | - Shunan Liu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Department of Allergy, Chongqing General Hospital, Chongqing 400014, China
| | - Dan Zeng
- Department of Allergy, Chongqing General Hospital, Chongqing 400014, China
| | - Weikang Zhou
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Department of Allergy, Chongqing General Hospital, Chongqing 400014, China.
| |
Collapse
|
3
|
Shen S, Sobczyk MK, Paternoster L, Brown SJ. From GWASs toward Mechanistic Understanding with Case Studies in Dermatogenetics. J Invest Dermatol 2024; 144:1189-1199.e8. [PMID: 38782533 DOI: 10.1016/j.jid.2024.03.013] [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: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 05/25/2024]
Abstract
Many human skin diseases result from the complex interplay of genetic and environmental mechanisms that are largely unknown. GWASs have yielded insight into the genetic aspect of complex disease by highlighting regions of the genome or specific genetic variants associated with disease. Leveraging this information to identify causal genes and cell types will provide insight into fundamental biology, inform diagnostics, and aid drug discovery. However, the etiological mechanisms from genetic variant to disease are still unestablished in most cases. There now exists an unprecedented wealth of data and computational methods for variant interpretation in a functional context. It can be challenging to decide where to start owing to a lack of consensus on the best way to identify causal genetic mechanisms. This article highlights 3 key aspects of genetic variant interpretation: prioritizing causal genes, cell types, and pathways. We provide a practical overview of the main methods and datasets, giving examples from recent atopic dermatitis studies to provide a blueprint for variant interpretation. A collection of resources, including brief description and links to the packages and web tools, is provided for researchers looking to start in silico follow-up genetic analysis of associated genetic variants.
Collapse
Affiliation(s)
- Silvia Shen
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom; Institute for Evolution and Ecology, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom.
| | - Maria K Sobczyk
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sara J Brown
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom; Department of Dermatology, NHS Lothian, Edinburgh, United Kingdom
| |
Collapse
|
4
|
Liu D, Liu G, Liu S. Promising Application, Efficient Production, and Genetic Basis of Mannosylerythritol Lipids. Biomolecules 2024; 14:557. [PMID: 38785964 PMCID: PMC11117751 DOI: 10.3390/biom14050557] [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: 04/11/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Mannosylerythritol lipids (MELs) are a class of glycolipids that have been receiving increasing attention in recent years due to their diverse biological activities. MELs are produced by certain fungi and display a range of bioactivities, making them attractive candidates for various applications in medicine, agriculture, and biotechnology. Despite their remarkable qualities, industrial-scale production of MELs remains a challenge for fungal strains. Excellent fungal strains and fermentation processes are essential for the efficient production of MELs, so efforts have been made to improve the fermentation yield by screening high-yielding strains, optimizing fermentation conditions, and improving product purification processes. The availability of the genome sequence is pivotal for elucidating the genetic basis of fungal MEL biosynthesis. This review aims to shed light on the applications of MELs and provide insights into the genetic basis for efficient MEL production. Additionally, this review offers new perspectives on optimizing MEL production, contributing to the advancement of sustainable biosurfactant technologies.
Collapse
Affiliation(s)
- Dun Liu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, China;
| | - Guanglei Liu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, China;
| | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Beibei, Chongqing 400716, China
| |
Collapse
|
5
|
Holden C, Soares P, Fidler K, Tavendale R, Felton J, Mukhopadhyay S. Children with asthma and eczema carrying filaggrin loss-of-function mutations have increased antibiotic use through to adulthood. Clin Exp Allergy 2024; 54:291-293. [PMID: 38100258 DOI: 10.1111/cea.14440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Affiliation(s)
- Ciara Holden
- Brighton & Sussex Medical School, Sussex, UK
- Academic Department of Paediatrics, Royal Alexandra Children's Hospital, Brighton, UK
| | - Patrícia Soares
- NOVA National School of Public Health, Public Health Research Centre, Comprehensive Health Research Center, NOVA University Lisbon, Lisbon, Portugal
| | - Katy Fidler
- Brighton & Sussex Medical School, Sussex, UK
- Academic Department of Paediatrics, Royal Alexandra Children's Hospital, Brighton, UK
| | - Roger Tavendale
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | | | - Somnath Mukhopadhyay
- Brighton & Sussex Medical School, Sussex, UK
- Academic Department of Paediatrics, Royal Alexandra Children's Hospital, Brighton, UK
| |
Collapse
|
6
|
Kobiela A, Hewelt-Belka W, Frąckowiak JE, Kordulewska N, Hovhannisyan L, Bogucka A, Etherington R, Piróg A, Dapic I, Gabrielsson S, Brown SJ, Ogg GS, Gutowska-Owsiak D. Keratinocyte-derived small extracellular vesicles supply antigens for CD1a-resticted T cells and promote their type 2 bias in the context of filaggrin insufficiency. Front Immunol 2024; 15:1369238. [PMID: 38585273 PMCID: PMC10995404 DOI: 10.3389/fimmu.2024.1369238] [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: 01/11/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
Introduction Exosome-enriched small extracellular vesicles (sEVs) are nanosized organelles known to participate in long distance communication between cells, including in the skin. Atopic dermatitis (AD) is a chronic inflammatory skin disease for which filaggrin (FLG) gene mutations are the strongest genetic risk factor. Filaggrin insufficiency affects multiple cellular function, but it is unclear if sEV-mediated cellular communication originating from the affected keratinocytes is also altered, and if this influences peptide and lipid antigen presentation to T cells in the skin. Methods Available mRNA and protein expression datasets from filaggrin-insufficient keratinocytes (shFLG), organotypic models and AD skin were used for gene ontology analysis with FunRich tool. sEVs secreted by shFLG and control shC cells were isolated from conditioned media by differential centrifugation. Mass spectrometry was carried out for lipidomic and proteomic profiling of the cells and sEVs. T cell responses to protein, peptide, CD1a lipid antigens, as well as phospholipase A2-digested or intact sEVs were measured by ELISpot and ELISA. Results Data analysis revealed extensive remodeling of the sEV compartment in filaggrin insufficient keratinocytes, 3D models and the AD skin. Lipidomic profiles of shFLGsEV showed a reduction in the long chain (LCFAs) and polyunsaturated fatty acids (PUFAs; permissive CD1a ligands) and increased content of the bulky headgroup sphingolipids (non-permissive ligands). This resulted in a reduction of CD1a-mediated interferon-γ T cell responses to the lipids liberated from shFLG-generated sEVs in comparison to those induced by sEVs from control cells, and an increase in interleukin 13 secretion. The altered sEV lipidome reflected a generalized alteration in the cellular lipidome in filaggrin-insufficient cells and the skin of AD patients, resulting from a downregulation of key enzymes implicated in fatty acid elongation and desaturation, i.e., enzymes of the ACSL, ELOVL and FADS family. Discussion We determined that sEVs constitute a source of antigens suitable for CD1a-mediated presentation to T cells. Lipids enclosed within the sEVs secreted on the background of filaggrin insufficiency contribute to allergic inflammation by reducing type 1 responses and inducing a type 2 bias from CD1a-restricted T cells, thus likely perpetuating allergic inflammation in the skin.
Collapse
Affiliation(s)
- Adrian Kobiela
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Weronika Hewelt-Belka
- Department of Analytical Chemistry, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Joanna E. Frąckowiak
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Kordulewska
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Lilit Hovhannisyan
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Bogucka
- The Mass Spectrometry Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Rachel Etherington
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Artur Piróg
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Irena Dapic
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Susanne Gabrielsson
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sara J. Brown
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Graham S. Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Danuta Gutowska-Owsiak
- Laboratory of Experimental and Translational Immunology, Intercollegiate Faculty of Biotechnology of the University of Gdańsk and the Medical University of Gdańsk, Gdańsk, Poland
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Rothenberg-Lausell C, Bar J, Del Duca E, Guttman-Yassky E. Diversity of atopic dermatitis and selection of immune targets. Ann Allergy Asthma Immunol 2024; 132:177-186. [PMID: 38008215 DOI: 10.1016/j.anai.2023.11.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
Atopic dermatitis (AD) is a heterogeneous immune-mediated skin disorder affecting people of all ages and ethnicities. Despite the development of targeted therapeutics such as biologics and Janus kinase inhibitors, attaining complete clinical efficacy remains difficult. This therapeutic challenge may be attributed to the complex pathogenesis of AD. Although the TH2 axis has been extensively studied, recent advancements have started to reveal the involvement of additional immune pathways including TH1, TH17, and TH22. Understanding the interplay of these immune axes may contribute to a more personalized therapeutic approach based on patients' molecular profile, with the prospect of improving clinical outcome. This review will discuss studies exploring the molecular profile of AD in both skin and blood across age, ethnicity/race, disease chronicity, IgE levels, filaggrin mutation status, and AD association with other atopic conditions. Moreover, it will explore the potential of personalized treatment strategies based on a patient's distinct immune signature.
Collapse
Affiliation(s)
- Camille Rothenberg-Lausell
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York; University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Jonathan Bar
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ester Del Duca
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York; Department of Dermatology, University of Rome La Sapienza, Rome, Italy
| | | |
Collapse
|
8
|
Lunjani N, Kerbelker T, Mdletshe FB, Hlela C, O’Mahony L. Phenotypes, endotypes and genotypes of atopic dermatitis and allergy in populations of African ancestry on the continent and diaspora. FRONTIERS IN ALLERGY 2024; 4:1203304. [PMID: 38327736 PMCID: PMC10847302 DOI: 10.3389/falgy.2023.1203304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 12/11/2023] [Indexed: 02/09/2024] Open
Abstract
Atopic dermatitis is a complex inflammatory condition characterized by synergist interactions between epidermal and immune related genotypes, skin barrier defects and immune dysregulation as well as microbial dysbiosis. Ethnicity-specific variations in clinical presentation, immune endotypes and genetic susceptibility have been described in diverse populations. We summarize available data with specific consideration of AD in populations of African ancestry. Some highlights include the observation of AD lesions on extensor surfaces, lichen planus-like AD, prurigo type AD and follicular AD in African populations. In addition, a consistent absence of dominant filaggrin gene defects has been reported. The detection of normal filaggrin protein content in AD skin implicates the contribution of alternative mechanisms in the pathogenesis of AD in African patients. Markedly high IgE has been described in paediatric and adult African AD. While Th2, Th22 and Th17 activation in African AD skin shares the same direction as with other populations, it has been noted that the magnitude of activation is dissimilar. Reduced Th17 cytokines have been observed in the circulation of moderate to severe paediatric AD.
Collapse
Affiliation(s)
- N. Lunjani
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Division of Dermatology, University of Cape Town, Cape Town, South Africa
| | - T. Kerbelker
- Department of Peadiatrics, University of Cape Town, Cape Town, South Africa
| | - F. B. Mdletshe
- Division of Otorhinolaryngology, University of Witwatersrand, Johannesburg, South Africa
| | - C. Hlela
- Division of Dermatology, University of Cape Town, Cape Town, South Africa
| | - L. O’Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
9
|
Traidl S, Heinrich L, Siegels D, Rösner L, Haufe E, Harder I, Abraham S, Ertner K, Kleinheinz A, Schäkel K, Wollenberg A, Effendy I, Quist S, Asmussen A, Wildberger J, Weisshaar E, Wiemers F, Brücher JJ, Weidinger S, Schmitt J, Werfel T. High recurrence rate of eczema herpeticum in moderate/severe atopic dermatitis -TREATgermany registry analysis. J Dtsch Dermatol Ges 2023; 21:1490-1498. [PMID: 37814394 DOI: 10.1111/ddg.15205] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 07/09/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Eczema herpeticum (EH) is a disseminated skin infection caused by herpes simplex virus in atopic dermatitis (AD) patients. The frequency of EH and the clinical features of EH patients have not yet been investigated in a larger cohort. METHODS We sought to investigate the TREATgermany cohort, a multicenter, non-interventional clinical registry of moderately to severely affected AD patients in Germany. Baseline characteristics of patients included between December 2017 and April 2021 were compared between patients without, single, and multiple EH. RESULTS Of the 893 patients, 195 (21.8%) had at least one EH. Of the 195 patients with EH, 107 had multiple EH (54.9%), representing 12.0% of the total study population. While there were no differences in demographic characteristics, previous treatment, and disease scores at enrollment (itch, IGA, oSCORAD, EASI), patients with EH had more frequent atopic comorbidities and sensitizations to house dust mite, food, and mold. DISCUSSION TREATgermany registry data suggest a high prevalence and recurrence rate of EH, while there appears to be no specific clinical phenotype, besides an increase in allergies, to identify EH patients in the daily routine.
Collapse
Affiliation(s)
- Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Luise Heinrich
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Doreen Siegels
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lennart Rösner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Eva Haufe
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Inken Harder
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Abraham
- Department of Dermatology, University Allergy Center, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | | | | | - Knut Schäkel
- Department of Dermatology, University Hospital, Heidelberg, Germany
| | - Andreas Wollenberg
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Ludwig Maximilian University, Munich, Germany
| | - Isaak Effendy
- Department of Dermatology, OWL University Hospital of Bielefeld University, Campus Clinic Bielefeld, Bielefeld, Germany
| | - Sven Quist
- Dermatology Clinic, Helix Medical Ecellence Center Mainz, Mainz, Germany
| | - Andrea Asmussen
- Practice Dr. med. Andrea Asmussen, Dermatology at Lesum, Bremen, Germany
| | - Julia Wildberger
- Practice Dr. med. Julia Wildberger Hautmedizin, Bad Soden, Germany
| | - Elke Weisshaar
- Division of Occupational Dermatology, Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | | | - Jens-Joachim Brücher
- Practice Dr. med. Jens-Joachim Brücher, Hautambulatorium Magdeburg, Magdeburg, Germany
| | - Stephan Weidinger
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jochen Schmitt
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| |
Collapse
|
10
|
Traidl S, Heinrich L, Siegels D, Rösner L, Haufe E, Harder I, Abraham S, Ertner K, Kleinheinz A, Schäkel K, Wollenberg A, Effendy I, Quist S, Asmussen A, Wildberger J, Weisshaar E, Wiemers F, Brücher JJ, Weidinger S, Schmitt J, Werfel T. Hohe Rezidivrate des Eczema herpeticatum bei mittelschwerer bis schwerer atopischer Dermatitis - eine TREATgermany Registeranalyse. J Dtsch Dermatol Ges 2023; 21:1490-1499. [PMID: 38082531 DOI: 10.1111/ddg.15205_g] [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: 10/11/2022] [Accepted: 07/09/2023] [Indexed: 12/18/2023]
Abstract
ZusammenfassungHintergrundDas Eczema herpeticatum (EH) ist eine disseminierte Hautinfektion, die durch Herpes‐simplex‐Viren bei Patienten mit atopischer Dermatitis (AD) verursacht wird. Die Häufigkeit des EH und die klinischen Charakteristika von EH Patienten wurden bisher noch nicht in einer größeren Kohorte untersucht.Methodik87 Patienten des TREATgermany Registers, einem multizentrischen, nichtinterventionellen klinischen Register mit moderat bis schwer betroffenen AD‐Patienten in Deutschland, wurden in dieser Analyse betrachtet. Patienten, die zwischen Dezember 2017 und April 2021 in das Register eingeschlossen wurden, wurden unterteilt in die Gruppen ohne, mit einem und mit mehreren EH und basierend auf den klinischen Charakteristika verglichen.ErgebnisseVon 893 Patienten berichteten 195 (21,8%) über mindestens eine EH. 107 der 195 Patienten mit EH hatten sogar mehrere EH in der Anamnese (54,9%), was 12,0% der gesamten Studienpopulation entspricht. Während hinsichtlich demographischer Merkmale, Vorbehandlungen und Krankheitsscores (Juckreiz, IGA, oSCORAD, EASI) keine Unterschiede festgestellt wurden, litten Patienten mit EH häufiger an atopischen Begleiterkrankungen und Sensibilisierungen gegen Hausstaubmilben, Nahrungsmittel und Schimmelpilze.SchlussfolgerungenDie Daten des TREATgermany‐Registers deuten auf eine hohe Prävalenz und Rezidivrate des EH hin, während es neben einer Häufung von Allergien keinen spezifischen klinischen Phänotyp zu geben scheint, um EH‐Patienten in der täglichen Routine zu identifizieren.
Collapse
Affiliation(s)
- Stephan Traidl
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| | - Luise Heinrich
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Doreen Siegels
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Lennart Rösner
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| | - Eva Haufe
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Inken Harder
- Zentrum für entzündliche Hauterkrankungen, Klinik für Dermatologie und Allergologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - Susanne Abraham
- Klinik für Dermatologie, Universitäts-Allergie-Centrum, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | | | | | | | - Andreas Wollenberg
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Ludwig Maximilian University, Munich, Germany
| | - Isaak Effendy
- Hautklinik, Universitätsklinikum OWL der Universität Bielefeld, Campus Klinikum Bielefeld
| | - Sven Quist
- Hautklinik, Helix Medical Ecellence Center Mainz
| | - Andrea Asmussen
- Praxis Dr. med. Andrea Asmussen, Hautarztpraxis an der Lesum, Bremen
| | | | - Elke Weisshaar
- Abteilung für Berufsdermatologie, Hautklinik, Ruprecht-Karls-Universität Heidelberg
| | | | | | - Stephan Weidinger
- Zentrum für entzündliche Hauterkrankungen, Klinik für Dermatologie und Allergologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - Jochen Schmitt
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Thomas Werfel
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| |
Collapse
|
11
|
Oh J, Oh HJ, Han KD, Gee HY, Lee JH. Increased Risk of Renal Malignancy in Patients with Moderate to Severe Atopic Dermatitis. Cancers (Basel) 2023; 15:5007. [PMID: 37894374 PMCID: PMC10605056 DOI: 10.3390/cancers15205007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/28/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Evidence for an association between atopic dermatitis (AD) and cancer is still insufficient. In particular, the association between the risk of renal malignancy and the severity of AD has not been thoroughly investigated. OBJECTIVE To investigate the risk of renal malignancy and determine the association between AD severity and cancer risk using data from the Korean National Health Insurance Service (KNHIS) database. METHODS We performed a population-based cohort study using the National Health Claims database from the NHIS in Korea. RESULTS We found a statistically significant association between AD and overall malignancy (for mild AD, hazard ratio (HR): 1.061, 95% confidence interval (CI): 1.006-1.118; for moderate to severe AD, HR: 1.061, 95% CI: 1.014-1.11) compared with the no AD group. The moderate to severe AD group showed a significantly increased risk for renal malignancy (adjusted HR: 1.533, 95% CI: 1.209-1.944) compared with the no AD group. LIMITATIONS Patient inclusion is solely based on diagnostic codes. We had no data about drug use, genetic factors, or other medical history that could affect the cancer risk. CONCLUSION In our large population-based cohort study, moderate to severe AD was associated with increased risk of renal malignancy. Regular check-ups for renal malignancy are recommended in this population.
Collapse
Affiliation(s)
- Jongwook Oh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Medicine, Physician-Scientist Program, Yonsei University Graduate School of Medicine, Seoul 03722, Republic of Korea
| | - Hyun Ju Oh
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kyung-Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ji Hyun Lee
- Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
12
|
Moran MC, Brewer MG, Schlievert PM, Beck LA. S. aureus virulence factors decrease epithelial barrier function and increase susceptibility to viral infection. Microbiol Spectr 2023; 11:e0168423. [PMID: 37737609 PMCID: PMC10581065 DOI: 10.1128/spectrum.01684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/03/2023] [Indexed: 09/23/2023] Open
Abstract
Individuals with atopic dermatitis (AD) are highly colonized by Staphylococcus aureus and are more susceptible to severe viral complications. We hypothesized that S. aureus secreted virulence factors may alter keratinocyte biology to enhance viral susceptibility through disruption of the skin barrier, impaired keratinocyte differentiation, and/or inflammation. To address this hypothesis, human keratinocytes were exposed to conditioned media from multiple S. aureus strains that vary in virulence factor production (USA300, HG003, and RN4220) or select purified virulence factors. We have identified the S. aureus enterotoxin-like superantigen SElQ, as a virulence factor of interest, since it is highly produced by USA300 and was detected on the skin of 53% of AD subjects (n = 72) in a study conducted by our group. Treatment with USA300 conditioned media or purified SElQ resulted in a significant increase in keratinocyte susceptibility to infection with vaccinia virus, and also significantly decreased barrier function. Importantly, we have previously demonstrated that keratinocyte differentiation influences susceptibility to viral infection, and our qPCR observations indicated that USA300 S. aureus and SElQ alter differentiation in keratinocytes. CRISPR/Cas9 was used to knock out CD40, a potential enterotoxin receptor on epithelial cells. We found that CD40 expression on keratinocytes was not completely necessary for SElQ-mediated responses, as measured by proinflammatory cytokine expression and barrier function. Together, these findings support that select S. aureus virulence factors, particularly SElQ, enhance the susceptibility of epidermal cells to viral infection, which may contribute to the increased cutaneous infections observed in individuals with AD. IMPORTANCE Staphylococcus aureus skin colonization and infection are frequently observed in individuals with atopic dermatitis. Many S. aureus strains belong to the clonal group USA300, and these strains produce superantigens including the staphylococcal enterotoxin-like Q (SElQ). Our studies highlight that SElQ may play a key role by altering keratinocyte differentiation and reducing barrier function; collectively, this may explain the AD-specific enhanced infection risk to cutaneous viruses. It is unclear what receptor mediates SElQ's effects on keratinocytes. We have shown that one putative surface receptor, CD40, was not critical for its effects on proinflammatory cytokine production or barrier function.
Collapse
Affiliation(s)
- Mary C. Moran
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
13
|
Hung YH, Liu HY, Chang R, Huang JY, Wu CD, Yen MS, Hung YM, Wei JCC, Wang PYP. Association between parental autoimmune disease and childhood atopic dermatitis varied by sex: a nationwide case-control study. Arch Dermatol Res 2023; 315:2011-2021. [PMID: 36892596 DOI: 10.1007/s00403-023-02582-w] [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: 11/07/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
Atopic dermatitis (AD) is a common inflammatory skin disorder induced by dysfunction of immune suppression sharing similar pathogenesis to autoimmune diseases. To explore the association between autoimmune diseases and AD in children, we linked the birth data from National Birth Registry with National Health Insurance Research Database. There were 1,174,941 children obtained from 2006 to 2012 birth cohort. A total of 312,329 children diagnosed with AD before 5 years old were compared to 862,612 children without AD in the control group. Conditional logistic regression was utilized to calculate adjusted odds ratio (OR) and Bonferroni-corrected confidence interval (CI) for overall significance level of 0.05. In 2006-2012 birth cohort, the prevalence rate of AD was 26.6% (95% CI 26.5, 26.7) before 5 years of age. Having parental autoimmune disease (including rheumatoid arthritis, systemic lupus erythematosus, Sjogren's syndrome, ankylosing spondylitis, and psoriasis) was associated with a significant higher risk of children AD development. The other associated factors were maternal obstetric complications (including gestational diabetes mellitus and cervical incompetence), parental systemic diseases (including anemia, hypertension, diabetes mellitus, chronic obstructive pulmonary disease, hyperthyroidism, and obstructive sleep apnea), and parental allergic disease (including asthma and AD). The subgroup analysis showed similar results between children's sexes. Moreover, maternal autoimmune disease had higher impact on the risk of developing AD in the child compared with paternal autoimmune disease. In conclusion, parental autoimmune diseases were found to be related to their children's AD before 5 years old.
Collapse
Affiliation(s)
- Yu-Hsuan Hung
- School of Medicine, Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsin-Yu Liu
- School of Medicine, Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Renin Chang
- Department of Emergency Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Jing-Yang Huang
- Department of Medical Research, Chung Shan Medical University, Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung City, 40201, Taiwan
| | - Cheng-Dong Wu
- School of Medicine, Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Man-Syuan Yen
- School of Medicine, Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yao-Min Hung
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital Taitung Branch, No.1000, Gengsheng Rd, Taitung City, 95050, Taiwan.
- College of Science and Engineering, National Taitung University, Taitung, Taiwan.
- College of Health and Nursing, Meiho University, Pingtung, Taiwan.
| | - James Cheng-Chung Wei
- Institute of Medicine, Chung Shan Medical University, No. 110, Sec. 1, Jianguo N. Rd., South District, Taichung City, 40201, Taiwan.
- Divison of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan.
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| | - Paul Yung-Pou Wang
- Division of Nephrology, Kaiser Permanente Baldwin Park Medical Center, Baldwin Park, CA, 91706, USA
| |
Collapse
|
14
|
Chawla HS, Kosta S, Namdeo C, Kataria R, Bhatia K, Sahu R, Joshi P. Genotype Study of Filaggrin Gene Loss-of-Function Mutations in Central India Population with Atopic Dermatitis and Ichthyosis Vulgaris. Indian Dermatol Online J 2023; 14:611-615. [PMID: 37727564 PMCID: PMC10506825 DOI: 10.4103/idoj.idoj_636_22] [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: 11/24/2022] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 09/21/2023] Open
Abstract
Background A genotype study of filaggrin gene loss-of-function mutations in central India can provide valuable insights into the prevalence and association of these mutations with atopic dermatitis (AD) and ichthyosis vulgaris (IV) in the region. The FLG R501X and 2282del4 are both genetic variants in the human gene called filaggrin gene (FLG), which encodes a protein that plays an important role in the formation and maintenance of the skin barrier. In this study, we determined the FLG R501X and 2282del4 variants association with both AD and IV in Central Indian populations. Materials and Methods This case-control study was conducted in the Departments of Dermatology and Molecular and Virology Research and Diagnostic Laboratory at Sri Aurobindo Medical College and Post Graduate Institute, Indore (Madhya Pradesh). The study was approved by the Clinical Research and Ethics Committee. A total of 180 patients aged between 3 months - 60 years who attended the skin outpatient department between March-2021 to June-2022 were recruited in this study. Among them, 60 patients were in AD-group, 60 patients in IV-group, and 60 patients were in the healthy control group. Polymerase chain reaction followed by restriction fragment length polymorphism (PCR-RFLP) was used in genotyping for FLG mutations (R501X and 2282del4). Results The most common FLG mutations were R501X (31.6% and 23.3%) and 2282del4 (18.3% and 13.3%) in AD and IV patients with heterozygous (AT) genotype, respectively. The combined mutation (FLG R501X and 2282del4) association was 10% and 5% in the AD and IV groups with heterozygous (AT) genotype, respectively, and in all the patients of control group with wild genotype (AA). There were no significant (P = 0.09) associations found with 2282del14 genotype. Conclusion The R501X mutation in the gene encoding filaggrin is one of the robust genetic associations of AD and IV. The 2282del4 polymorphism was marginally less as compared to R501X.
Collapse
Affiliation(s)
- Harsimran S. Chawla
- Department of Dermatology, Venerology and Leprosy, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Susmit Kosta
- Department of Molecular and Virology Research and Diagnostic Laboratory, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Chaitanya Namdeo
- Department of Dermatology, Venerology and Leprosy, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Rajesh Kataria
- Department of Dermatology, Venerology and Leprosy, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Kailash Bhatia
- Department of Dermatology, Venerology and Leprosy, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Roshni Sahu
- Department of Molecular and Virology Research and Diagnostic Laboratory, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| | - Pallavi Joshi
- Department of Molecular and Virology Research and Diagnostic Laboratory, Sri Aurobindo Medical College and Post Graduate Institute, Sri Aurobindo University, Indore, Madhya Pradesh, India
| |
Collapse
|
15
|
Pontikas A, Antonatos C, Evangelou E, Vasilopoulos Y. Candidate Gene Association Studies in Atopic Dermatitis in Participants of European and Asian Ancestry: A Systematic Review and Meta-Analysis. Genes (Basel) 2023; 14:1456. [PMID: 37510360 PMCID: PMC10379179 DOI: 10.3390/genes14071456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Atopic dermatitis (AD) has been extensively investigated for genetic associations utilizing both candidate gene approaches and genome-wide scans. Here, we comprehensively evaluated the available literature to determine the association of candidate genes in AD to gain additional insight into the etiopathogenesis of the disease. We systematically screened all studies that explored the association between polymorphisms and AD risks in cases of European and Asian ancestry and synthesized the available evidence through a random-effects meta-analysis. We identified 99 studies that met our inclusion/exclusion criteria that examined 17 candidate loci in Europeans and 14 candidate genes in Asians. We confirmed the significant associations between FLG variants in both European and Asian populations and AD risk, while synthesis of the available data revealed novel loci mapped to IL18 and TGFB1 genes in Europeans and IL12RB1 and MIF in Asians that have not yet been identified by genome-wide association studies. Our findings provide comprehensive evidence for AD risk loci in cases of both European and Asian ancestries, validating previous associations as well as revealing novel loci that could imply previously unexplored biological pathways.
Collapse
Affiliation(s)
- Alexandros Pontikas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, 45110 Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas, 45110 Ioannina, Greece
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| |
Collapse
|
16
|
Perälä M, Kaustio M, Salava A, Jakkula E, Pelkonen AS, Saarela J, Remitz A, Mäkelä MJ. RELEVANCE OF CODING VARIATION IN FILAGGRIN AND DOCK8 IN FINNISH PEDIATRIC PATIENTS WITH EARLY-ONSET MODERATE-TO-SEVERE ATOPIC DERMATITIS. JID INNOVATIONS 2023. [PMID: 37533579 PMCID: PMC10392095 DOI: 10.1016/j.xjidi.2023.100203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
Early-onset, persistent atopic dermatitis (AD) is proposed as a distinct subgroup that may have specific genotypic features. FLG gene loss-of-function variants are the best known genetic factors contributing to epidermal barrier impairment and eczema severity. In a cohort of 140 Finnish children with early-onset moderate-to-severe AD, we investigated the effect of coding variation in FLG and 13 other genes with epidermal barrier or immune function through the use of targeted amplicon sequencing and genotyping. A FLG loss-of-function variant (Arg501Ter, Ser761fs, Arg2447Ter, or Ser3247Ter) was identified in 20 of 140 patients showing higher transepidermal water loss values than patients without these variants. Total FLG loss-of-function variant frequency (7.14%) was significantly higher than in the general Finnish population (2.34%). When tested separately, only Arg2447Ter showed a significant association with AD (P = 0.003104). In addition, a modest association with moderate-to-severe pediatric AD was seen for rs12730241 and rs6587667 (FLG2:Gly137Glu). Loss-of-function variants, previously reported pathogenic variants, or statistically significant enrichment of nonsynonymous coding region variants were not found in the 13 candidate genes studied by amplicon sequencing. However, higher IgE and eosinophil counts were found in carriers of potentially pathogenic DOCK8 missense variants, suggesting that the role of DOCK8 variation in AD should be further investigated in larger cohorts.
Collapse
|
17
|
Jackson ND, Dyjack N, Goleva E, Bin L, Montgomery MT, Rios C, Everman JL, Taylor P, Bronchick C, Richers BN, Leung DY, Seibold MA. Atopic dermatitis complicated by recurrent eczema herpeticum is characterized by multiple, concurrent epidermal inflammatory endotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530316. [PMID: 36909594 PMCID: PMC10002633 DOI: 10.1101/2023.02.27.530316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
BACKGROUND A subgroup of atopic dermatitis (AD) patients suffer from recurrent, disseminated herpes simplex virus (HSV) skin infections, termed eczema herpeticum (EH), which can be life-threatening and contribute to AD morbidity. The pathobiology underlying ADEH is unknown. OBJECTIVE To determine transcriptional mechanisms of skin and immune system pathobiology that underlie ADEH disease. METHODS We performed whole transcriptome RNA-sequencing of non-lesional skin samples (epidermis, dermis) of AD patients with (ADEH + , n=15) and without (ADEH - , n=13) recurrent EH history, and healthy controls (HC, n=15). We also performed RNA-sequencing on plasmacytoid dendritic cells (pDCs) collected from these participants and infected in vitro with HSV-1. Differential expression, gene set enrichment, and endotyping analyses were performed. RESULTS ADEH + disease was characterized by dysregulation in skin gene expression, which was limited in dermis (differentially expressed genes [DEGs]=14) and widespread in epidermis (DEGs=129). ADEH + -upregulated epidermal DEGs were enriched in type 2 cytokine (T2) ( IL4R, CCL22, CRLF2, IL7R ), interferon ( CXCL10, ICAM1, IFI44 , and IRF7) , and IL-36γ ( IL36G ) inflammatory pathway genes. At a person-level, all ADEH + participants exhibited T2 and interferon endotypes and 87% were IL36G-high. In contrast, these endotypes were more variably expressed among ADEH - participants. ADEH + patient skin also exhibited dysregulation in epidermal differentiation complex (EDC) genes within the LCE, S100 , and SPRR families, which are involved in skin barrier function, inflammation, and antimicrobial activities. pDC transcriptional responses to HSV-1 infection were not altered by ADEH status. CONCLUSIONS ADEH + pathobiology is characterized by a unique, multi-faceted epidermal inflammation that accompanies dysregulation in the expression of EDC genes. Key Messages AD patients with a history of recurrent EH exhibit molecular skin pathobiology that is similar in form, but more severe in degree, than in AD patients without this complication. Non-lesional skin of ADEH + patients concurrently exhibits excessive type 2 cytokine, interferon, and IL-36γ-driven epidermal inflammation. Expression of these inflammatory skin endotypes among ADEH + patients is associated with dysregulation in expression of epidermal differentiation complex genes involved in barrier function, inflammation, and antimicrobial activity. Capsule Summary AD patients with a history of recurrent disseminated HSV-1 skin infections form a unique molecular skin endotype group that concurrently exhibits type 2 cytokine, interferon, and IL-36γ-driven skin inflammation, accompanied by dysregulation in expression of epidermal differentiation complex genes involved in barrier function, inflammation, and antimicrobial activity.
Collapse
|
18
|
Inborn Errors of Immunity Predisposing to Herpes Simplex Virus Infections of the Central Nervous System. Pathogens 2023; 12:pathogens12020310. [PMID: 36839582 PMCID: PMC9961685 DOI: 10.3390/pathogens12020310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023] Open
Abstract
Herpesvirus infections can lead to a number of severe clinical manifestations, particularly when involving the central nervous system (CNS), causing encephalitis and meningitis. However, understanding of the host factors conferring increased susceptibility to these diseases and their complications remains incomplete. Previous studies have uncovered defects in the innate Toll-like receptor 3 pathway and production of type I interferon (IFN-I) in children and adults that predispose them to herpes simplex encephalitis. More recently, there is accumulating evidence for an important role of IFN-independent cell-autonomous intrinsic mechanisms, including small nucleolar RNAs, RNA lariat metabolism, and autophagy, in restricting herpesvirus replication and conferring protection against CNS infection. The present review first describes clinical manifestations of HSV infection with a focus on neurological complications and then summarizes the host-pathogen interactions and innate immune pathways responsible for sensing herpesviruses and triggering antiviral responses and immunity. Next, we review the current landscape of inborn errors of immunity and the underlying genetic defects and disturbances of cellular immune pathways that confer increased susceptibility to HSV infection in CNS. Ultimately, we discuss some of the present outstanding unanswered questions relating to inborn errors of immunity and HSV CNS infection together with some perspectives and future directions for research in the pathogenesis of these severe diseases in humans.
Collapse
|
19
|
Incorporating genetics in identifying peanut allergy risk and tailoring allergen immunotherapy: A perspective on the genetic findings from the LEAP trial. J Allergy Clin Immunol 2023; 151:841-847. [PMID: 36732171 DOI: 10.1016/j.jaci.2022.12.819] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 02/04/2023]
Abstract
Examining the genetics of peanut allergy (PA) in the context of clinical trial interventions and outcomes provides an opportunity to not only understand gene-environment interactions for PA risk but to also understand the benefit of allergen immunotherapy. A consistent theme in the genetics of food allergy is that in keeping with the dual allergen exposure hypothesis, barrier- and immune-related genes are most commonly implicated in food allergy and tolerance. With a focus on PA, we review how genetic risk factors across 3 genes (FLG, MALT1, and HLA-DQA1) have helped delineate distinct allergic characteristics and outcomes in the context of environmental interventions in the Learning Early about Peanut Allergy (LEAP) study and other clinical trials. We specifically consider and present a framework for genetic risk prediction for the development of PA and discuss how genetics, age, and oral consumption intertwine to predict PA outcome. Although there is some promise in this proposed framework, a better understanding of the mechanistic pathways by which PA develops and persists is needed to develop targeted therapeutics for established disease. Only by understanding the mechanisms by which PA develops, persists, and resolves can we identify adjuvants to oral immunotherapy to make older children and adults immunologically similar to their younger, more malleable counterparts and thus more likely to achieve long-term tolerance.
Collapse
|
20
|
Chennareddy S, Brunner PM. Sphingolipids in viral skin superinfection: Friend or foe? J Allergy Clin Immunol 2023; 151:108-109. [PMID: 36241047 DOI: 10.1016/j.jaci.2022.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/16/2022] [Accepted: 09/28/2022] [Indexed: 02/04/2023]
Affiliation(s)
- Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York.
| |
Collapse
|
21
|
Broderick C, Ziehfreund S, van Bart K, Arents B, Eyerich K, Weidinger S, Rastrick J, Zink A, Flohr C. Biomarkers associated with the development of comorbidities in patients with atopic dermatitis: A systematic review. Allergy 2023; 78:84-120. [PMID: 36366871 PMCID: PMC10107168 DOI: 10.1111/all.15578] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/06/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Biomarkers associated with the development of comorbidities in atopic dermatitis (AD) patients have been reported, but have not yet been systematically reviewed. Seven electronic databases were searched, from database inception to September 2021. English language randomized controlled trials, prospective and retrospective cohort, and case-control studies that investigated the association between a biomarker and the development of comorbidities in AD patients were included. Two authors independently screened the records for eligibility, one extracted all data, and critically appraised the quality of studies and risk of bias. Fifty six articles met the inclusion criteria, evaluating 146 candidate biomarkers. The most frequently reported biomarkers were filaggrin mutations and allergen specific-IgE. Promising biomarkers include specific-IgE and/or skin prick tests predicting the development of asthma, and genetic polymorphisms predicting the occurrence of eczema herpeticum. The identified studies and biomarkers were highly heterogeneous, and associated with predominately moderate-to-high risk of bias across multiple domains. Overall, findings were inconsistent. High-quality studies assessing biomarkers associated with the development of comorbidities in people with AD are lacking. Harmonized datasets and independent validation studies are urgently needed.
Collapse
Affiliation(s)
- Conor Broderick
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | - Stefanie Ziehfreund
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Karin van Bart
- Royal College of Physicians, National Guideline Centre, London, UK
| | - Bernd Arents
- Dutch Association for People with Atopic Dermatitis, Nijkerk, The Netherlands
| | - Kilian Eyerich
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Flohr
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | | |
Collapse
|
22
|
de Lima LC, Cruz ÁA, Costa RDS, Silva HDS, Coelho RS, Teixeira HM, Oliveira PR, Barnes KC, Figueiredo CA, Carneiro VL. TSLP and IL25 variants are related to asthma and atopy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
23
|
Herpes Simplex Virus 1 Can Bypass Impaired Epidermal Barriers upon Ex Vivo Infection of Skin from Atopic Dermatitis Patients. J Virol 2022; 96:e0086422. [PMID: 35969080 PMCID: PMC9472615 DOI: 10.1128/jvi.00864-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To infect its human host, herpes simplex virus 1 (HSV-1) must overcome the protective barriers of skin and mucosa. Here, we addressed whether pathological skin conditions can facilitate viral entry via the skin surface and used ex vivo infection studies to explore viral invasion in atopic dermatitis (AD) skin characterized by disturbed barrier functions. Our focus was on the visualization of the onset of infection in single cells to determine the primary entry portals in the epidermis. After ex vivo infection of lesional AD skin, we observed infected cells in suprabasal layers indicating successful invasion in the epidermis via the skin surface which was never detected in control skin where only sample edges allowed viral access. The redistribution of filaggrin, loricrin, and tight-junction components in the lesional skin samples suggested multiple defective mechanical barriers. To dissect the parameters that contribute to HSV-1 invasion, we induced an AD-like phenotype by adding the Th2 cytokines interleukin 4 (IL-4) and IL-13 to healthy human skin samples. Strikingly, we detected infected cells in the epidermis, implying that the IL-4/IL-13-driven inflammation is sufficient to induce modifications allowing HSV-1 to penetrate the skin surface. In summary, not only did lesional AD skin facilitate HSV-1 penetration but IL-4/IL-13 responses alone allowed virus invasion. Our results suggest that the defective epidermal barriers of AD skin and the inflammation-induced altered barriers in healthy skin can make receptors accessible for HSV-1. IMPORTANCE Herpes simplex virus 1 (HSV-1) can target skin to establish primary infection in the epithelium. While the human skin provides effective barriers against viral invasion under healthy conditions, a prominent example of successful invasion is the disseminated HSV-1 infection in the skin of atopic dermatitis (AD) patients. AD is characterized by impaired epidermal barrier functions, chronic inflammation, and dysbiosis of skin microbiota. We addressed the initial invasion process of HSV-1 in atopic dermatitis skin to understand whether the physical barrier functions are sufficiently disturbed to allow the virus to invade skin and reach its receptors on skin cells. Our results demonstrate that HSV-1 can indeed penetrate and initiate infection in atopic dermatitis skin. Since treatment of skin with IL-4 and IL-13 already resulted in successful invasion, we assume that inflammation-induced barrier defects play an important role for the facilitated access of HSV-1 to its target cells.
Collapse
|
24
|
Gao JF, Tang L, Luo F, Zhang YY, Chen L, Ding H, Meng ZD. Nicotinamide mononucleotide ameliorates DNFB-induced atopic dermatitis-like symptoms in mice by blocking activation of ROS-mediated JAK2/STAT5 signaling pathway. Int Immunopharmacol 2022; 109:108812. [DOI: 10.1016/j.intimp.2022.108812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/28/2022]
|
25
|
Numata T, Harada K, Nakae S. Roles of Mast Cells in Cutaneous Diseases. Front Immunol 2022; 13:923495. [PMID: 35874756 PMCID: PMC9298983 DOI: 10.3389/fimmu.2022.923495] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/16/2022] [Indexed: 01/05/2023] Open
Abstract
Mast cells are present in all vascularized tissues of the body. They are especially abundant in tissues that are in frequent contact with the surrounding environment and act as potential sources of inflammatory and/or regulatory mediators during development of various infections and diseases. Mature mast cells’ cytoplasm contains numerous granules that store a variety of chemical mediators, cytokines, proteoglycans, and proteases. Mast cells are activated via various cell surface receptors, including FcϵRI, toll-like receptors (TLR), Mas-related G-protein-coupled receptor X2 (MRGPRX2), and cytokine receptors. IgE-mediated mast cell activation results in release of histamine and other contents of their granules into the extracellular environment, contributing to host defense against pathogens. TLRs, play a crucial role in host defense against various types of pathogens by recognizing pathogen-associated molecular patterns. On the other hand, excessive/inappropriate mast cell activation can cause various disorders. Here, we review the published literature regarding the known and potential inflammatory and regulatory roles of mast cells in cutaneous inflammation, including atopic dermatitis, psoriasis, and contact dermatitis GVHD, as well as in host defense against pathogens.
Collapse
Affiliation(s)
- Takafumi Numata
- Department of Dermatology, Tokyo Medical University, Tokyo, Japan
| | - Kazutoshi Harada
- Department of Dermatology, Tokyo Medical University, Tokyo, Japan
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan.,Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Saitama, Japan
| |
Collapse
|
26
|
Jing C, Guo J, Li Z, Xu X, Wang J, Zhai L, Liu J, Sun G, Wang F, Xu Y, Li Z, Zhao D, Jiang R, Sun L. Screening and Research on Skin Barrier Damage Protective Efficacy of Different Mannosylerythritol Lipids. Molecules 2022; 27:molecules27144648. [PMID: 35889520 PMCID: PMC9320248 DOI: 10.3390/molecules27144648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
Mannosylerythritol lipids (MELs) may prevent skin barrier damage, although their protective mechanisms and active monomeric constituents remain unclear. Here, three MELs were extracted from Candida antarctica cultures containing fermented olive oil then purified using silica gel-based column chromatography and semipreparative HPLC. All three compounds (MEL-A, MEL-B, MEL-C) were well separated and stable, and reliable materials were used for NMR and HRESIMS chemical structure determinations and for assessing MELs’ protective effects against skin damage. Notably, MEL-B and MEL-C effectively protected HaCaT cells from UVB-induced damage by upregulating the contents of filaggrin (FLG) and transglutaminase-1 (TGM1), as determined via ELISA. Moreover, MEL-B treatment (20 μg/mL) of UVB-irradiated HaCaT cells led to the upregulation of both the expression of mRNA genes and the key proteins FLG, LOR, and TGM1, which are known to be decreased in damaged skin cells. Additionally, histopathological analysis results revealed a markedly reduced intracellular vacuolation and cell damage, reflecting improved skin function after MEL-B treatment. Furthermore, immunofluorescence results revealed that MEL-B protected EpiKutis® three-dimensional cultured human skin cells from sodium dodecyl sulfate-induced damage by up-regulating FLG, LOR, and TGM1 expression. Accordingly, MELs’ protection against skin barrier damage depended on MEL-B monomeric constituent activities, thus highlighting their promise as beneficial ingredients for use in skin-care products.
Collapse
Affiliation(s)
- Chenxu Jing
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Jiling Guo
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Zhenzhuo Li
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Jing Wang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Lu Zhai
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Jianzeng Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Guang Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Fei Wang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Yangfen Xu
- Modern Hanfang Technology Company Limited, Guangzhou 510550, China; (Y.X.); (Z.L.)
| | - Zhaolian Li
- Modern Hanfang Technology Company Limited, Guangzhou 510550, China; (Y.X.); (Z.L.)
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Rui Jiang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Correspondence: (R.J.); (L.S.)
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Correspondence: (R.J.); (L.S.)
| |
Collapse
|
27
|
Moosbrugger-Martinz V, Leprince C, Méchin MC, Simon M, Blunder S, Gruber R, Dubrac S. Revisiting the Roles of Filaggrin in Atopic Dermatitis. Int J Mol Sci 2022; 23:5318. [PMID: 35628125 PMCID: PMC9140947 DOI: 10.3390/ijms23105318] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 12/31/2022] Open
Abstract
The discovery in 2006 that loss-of-function mutations in the filaggrin gene (FLG) cause ichthyosis vulgaris and can predispose to atopic dermatitis (AD) galvanized the dermatology research community and shed new light on a skin protein that was first identified in 1981. However, although outstanding work has uncovered several key functions of filaggrin in epidermal homeostasis, a comprehensive understanding of how filaggrin deficiency contributes to AD is still incomplete, including details of the upstream factors that lead to the reduced amounts of filaggrin, regardless of genotype. In this review, we re-evaluate data focusing on the roles of filaggrin in the epidermis, as well as in AD. Filaggrin is important for alignment of keratin intermediate filaments, control of keratinocyte shape, and maintenance of epidermal texture via production of water-retaining molecules. Moreover, filaggrin deficiency leads to cellular abnormalities in keratinocytes and induces subtle epidermal barrier impairment that is sufficient enough to facilitate the ingress of certain exogenous molecules into the epidermis. However, although FLG null mutations regulate skin moisture in non-lesional AD skin, filaggrin deficiency per se does not lead to the neutralization of skin surface pH or to excessive transepidermal water loss in atopic skin. Separating facts from chaff regarding the functions of filaggrin in the epidermis is necessary for the design efficacious therapies to treat dry and atopic skin.
Collapse
Affiliation(s)
- Verena Moosbrugger-Martinz
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (V.M.-M.); (S.B.); (R.G.)
| | - Corinne Leprince
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse University, CNRS UMR5051, Inserm UMR1291, UPS, 31059 Toulouse, France; (C.L.); (M.-C.M.); (M.S.)
| | - Marie-Claire Méchin
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse University, CNRS UMR5051, Inserm UMR1291, UPS, 31059 Toulouse, France; (C.L.); (M.-C.M.); (M.S.)
| | - Michel Simon
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse University, CNRS UMR5051, Inserm UMR1291, UPS, 31059 Toulouse, France; (C.L.); (M.-C.M.); (M.S.)
| | - Stefan Blunder
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (V.M.-M.); (S.B.); (R.G.)
| | - Robert Gruber
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (V.M.-M.); (S.B.); (R.G.)
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria; (V.M.-M.); (S.B.); (R.G.)
| |
Collapse
|
28
|
Abstract
Atopic dermatitis (AD) is a common disease with a broad spectrum of clinical manifestations. AD can manifest differently in adults than children. Core AD features are similar between children and adults overall, including lesions affecting flexural areas, presence of atopy, and xerosis. Adults have more signs of chronic disease, higher prevalence and different patterns of hand eczema, and a stronger relationship of disease activity with emotional factors, whereas children with AD have more exudative lesions, perifollicular accentuation, pityriasis alba, Dennie-Morgan folds, and seborrheic dermatitis-like presentation. These differences may be due in part to pathophysiologic differences in AD in children compared with adults. Atopic diseases commonly co-occur with AD, although most do not temporally have the "atopic march." Further research is warranted to better understand the differential roles of immune dysregulation, epidermal-barrier disruption, and dysbiosis between children and adults and determine whether such differences translate into differences in therapeutic efficacy.
Collapse
Affiliation(s)
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
29
|
Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, Rossi AB. Type 2 Inflammation Contributes to Skin Barrier Dysfunction in Atopic Dermatitis. JID INNOVATIONS 2022; 2:100131. [PMID: 36059592 PMCID: PMC9428921 DOI: 10.1016/j.xjidi.2022.100131] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
Skin barrier dysfunction, a defining feature of atopic dermatitis (AD), arises from multiple interacting systems. In AD, skin inflammation is caused by host-environment interactions involving keratinocytes as well as tissue-resident immune cells such as type 2 innate lymphoid cells, basophils, mast cells, and T helper type 2 cells, which produce type 2 cytokines, including IL-4, IL-5, IL-13, and IL-31. Type 2 inflammation broadly impacts the expression of genes relevant for barrier function, such as intracellular structural proteins, extracellular lipids, and junctional proteins, and enhances Staphylococcus aureus skin colonization. Systemic anti‒type 2 inflammation therapies may improve dysfunctional skin barrier in AD.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- CLDN, claudin
- FFA, free fatty acid
- ILC2, type 2 innate lymphoid cell
- Jaki, Jak inhibitor
- K, keratin
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NMF, natural moisturizing factor
- PAR, protease-activated receptor
- PDE-4, phosphodiesterase-4
- SC, stratum corneum
- SG, stratum granulosum
- TCI, topical calcineurin inhibitor
- TCS, topical corticosteroid
- TEWL, transepidermal water loss
- TJ, tight junction
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor alpha
- TYK, tyrosine kinase
- Th, T helper
- ZO, zona occludens
- hBD, human β-defensin
Collapse
Affiliation(s)
- Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA,Correspondence: Lisa A. Beck, Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, New York 14642, USA.
| | - Michael J. Cork
- Sheffield Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease (IICD), The University of Sheffield, The Medical School, Sheffield, United Kingdom
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
30
|
Jung K, Pawluk MA, Lane M, Nabai L, Granville DJ. Granzyme B in Epithelial Barrier Dysfunction and Related Skin Diseases. Am J Physiol Cell Physiol 2022; 323:C170-C189. [PMID: 35442832 DOI: 10.1152/ajpcell.00052.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The predominant function of the skin is to serve as a barrier - to protect against external insults and to prevent water loss. Junctional and structural proteins in the stratum corneum, the outermost layer of the epidermis, are critical to the integrity of the epidermal barrier as it balances ongoing outward migration, differentiation, and desquamation of keratinocytes in the epidermis. As such, epidermal barrier function is highly susceptible to upsurges of proteolytic activity in the stratum corneum and epidermis. Granzyme B is a serine protease scarce in healthy tissues but present at high levels in tissues encumbered by chronic inflammation. Discovered in the 1980s, Granzyme B is currently recognized for its intracellular roles in immune cell-mediated targeted apoptosis as well as extracellular roles in inflammation, chronic injuries, tissue remodeling, and processing of cytokines, matrix proteins, and autoantigens. Increasing evidence has emerged in recent years supporting a role for Granzyme B in promoting barrier dysfunction in the epidermis by direct cleavage of barrier proteins and eliciting immunoreactivity. Likewise, Granzyme B contributes to impaired epithelial function of the airways, retina, gut and vessels. In the present review, the role of Granzyme B in cutaneous epithelial dysfunction is discussed in the context of specific conditions with an overview of underlying mechanisms as well as utility of current experimental and therapeutic inhibitors.
Collapse
Affiliation(s)
- Karen Jung
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Professional Firefighters' Wound Healing Laboratory, VCHRI, Vancouver, British Columbia, Canada
| | - Megan A Pawluk
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Professional Firefighters' Wound Healing Laboratory, VCHRI, Vancouver, British Columbia, Canada
| | - Michael Lane
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Professional Firefighters' Wound Healing Laboratory, VCHRI, Vancouver, British Columbia, Canada
| | - Layla Nabai
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Professional Firefighters' Wound Healing Laboratory, VCHRI, Vancouver, British Columbia, Canada
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.,British Columbia Professional Firefighters' Wound Healing Laboratory, VCHRI, Vancouver, British Columbia, Canada
| |
Collapse
|
31
|
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
|
32
|
Luo X, Chen J, Yang H, Hu X, Alphonse MP, Shen Y, Kawakami Y, Zhou X, Tu W, Kawakami T, Wan M, Archer NK, Wang H, Gao P. Dendritic cell immunoreceptor drives atopic dermatitis by modulating oxidized CaMKII-involved mast cell activation. JCI Insight 2022; 7:152559. [PMID: 35113811 PMCID: PMC8983143 DOI: 10.1172/jci.insight.152559] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Allergens have been identified as potential triggers in patients with atopic dermatitis (AD). AD patients are highly sensitive to cockroach allergen. The underlying mechanism, however, remains undetermined. Here, we established a cockroach allergen-induced AD-like mouse model and demonstrated that repeated exposure to cockroach allergen led to aggravated mouse skin inflammation, characterized by increased type 2 immunity, type 2 innate lymphoid cells (ILC2s), and mast cells. Increased skin mast cells were also observed in AD patients. AD mice with mast cell-deficient mice (kitW-sh/W-sh) showed diminished skin inflammation, suggesting that mast cells are required in allergen-induced skin inflammation. Furthermore, dendritic cell immuno-receptor (DCIR) is up-regulated in skin mast cells of AD patients and mediates allergen binding and uptake. DCIR-/- mice or reconstituted kitW-sh/W-sh mice with DCIR-/- mast cells showed a significant reduction in AD-like inflammation. Both in vitro and in vivo analyses demonstrated that DCIR-/- mast cells had reduced IgE-mediated mast cell activation and passive cutaneous anaphylaxis. Mechanistically, DCIR regulates allergen-induced IgE-mediated mast cell ROS generation and oxidation of calmodulin kinase II (ox-CaMKII). ROS-resistant CaMKII (MM-VVδ) prevents allergen-induced mast cell activation and inflammatory mediator release. Our study reveals a previously unrecognized DCIR-ROS-CaMKII axis that controls allergen-induced mast cell activation and AD-like inflammation.
Collapse
Affiliation(s)
- Xiaoyan Luo
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Jingsi Chen
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Huan Yang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yuko Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Xiaoying Zhou
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Mei Wan
- Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Hua Wang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| |
Collapse
|
33
|
Atopic eczema: How genetic studies can contribute to understanding this complex trait. J Invest Dermatol 2022; 142:1015-1019. [PMID: 35007558 DOI: 10.1016/j.jid.2021.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 11/24/2022]
Abstract
Atopic eczema is an itchy inflammatory skin disease. This complex trait results from multiple genetic and environmental factors, but atopic eczema also shows great complexity in its heterogenous presentation, clinical signs and longitudinal trajectory, with or without co-morbid conditions. The past 50 years have produced substantial improvements in the management of atopic eczema, but many patients still suffer a burden of disease affecting personal, social and family life. Genetic research refocused interest on skin barrier function, but effective targeting of this central pathomechanism remains elusive. This Perspective highlights progress in understanding molecular mechanisms and translational opportunities for the future.
Collapse
|
34
|
Arehart CH, Daya M, Campbell M, Boorgula MP, Rafaels N, Chavan S, David G, Hanifin J, Slifka MK, Gallo RL, Hata T, Schneider LC, Paller AS, Ong PY, Spergel JM, Guttman-Yassky E, Leung DYM, Beck LA, Gignoux CR, Mathias RA, Barnes KC. Polygenic prediction of atopic dermatitis improves with atopic training and filaggrin factors. J Allergy Clin Immunol 2022; 149:145-155. [PMID: 34111454 PMCID: PMC8973457 DOI: 10.1016/j.jaci.2021.05.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 04/26/2021] [Accepted: 05/20/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND While numerous genetic loci associated with atopic dermatitis (AD) have been discovered, to date, work leveraging the combined burden of AD risk variants across the genome to predict disease risk has been limited. OBJECTIVES This study aims to determine whether polygenic risk scores (PRSs) relying on genetic determinants for AD provide useful predictions for disease occurrence and severity. It also explicitly tests the value of including genome-wide association studies of related allergic phenotypes and known FLG loss-of-function (LOF) variants. METHODS AD PRSs were constructed for 1619 European American individuals from the Atopic Dermatitis Research Network using an AD training dataset and an atopic training dataset including AD, childhood onset asthma, and general allergy. Additionally, whole genome sequencing data were used to explore genetic scoring specific to FLG LOF mutations. RESULTS Genetic scores derived from the AD-only genome-wide association studies were predictive of AD cases (PRSAD: odds ratio [OR], 1.70; 95% CI, 1.49-1.93). Accuracy was first improved when PRSs were built off the larger atopy genome-wide association studies (PRSAD+: OR, 2.16; 95% CI, 1.89-2.47) and further improved when including FLG LOF mutations (PRSAD++: OR, 3.23; 95% CI, 2.57-4.07). Importantly, while all 3 PRSs correlated with AD severity, the best prediction was from PRSAD++, which distinguished individuals with severe AD from control subjects with OR of 3.86 (95% CI, 2.77-5.36). CONCLUSIONS This study demonstrates how PRSs for AD that include genetic determinants across atopic phenotypes and FLG LOF variants may be a promising tool for identifying individuals at high risk for developing disease and specifically severe disease.
Collapse
Affiliation(s)
- Christopher H Arehart
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Michelle Daya
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Monica Campbell
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | | | - Nicholas Rafaels
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Sameer Chavan
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | | | - Jon Hanifin
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | - Mark K Slifka
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | - Richard L Gallo
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | - Tissa Hata
- Department of Dermatology, University of California San Diego, San Diego, Calif
| | | | - Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill; Department of Pediatrics (Dermatology), Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Peck Y Ong
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Los Angeles, Calif; Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Jonathan M Spergel
- Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | | | - Donald Y M Leung
- Division of Allergy and Immunology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Lisa A Beck
- Department of Dermatology, Medicine and Pathology, University of Rochester Medical Center, Rochester, NY
| | - Christopher R Gignoux
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University Department of Medicine, Baltimore, Md
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo.
| |
Collapse
|
35
|
Gallegos-Alcalá P, Jiménez M, Cervantes-García D, Salinas E. The Keratinocyte as a Crucial Cell in the Predisposition, Onset, Progression, Therapy and Study of the Atopic Dermatitis. Int J Mol Sci 2021; 22:ijms221910661. [PMID: 34639001 PMCID: PMC8509070 DOI: 10.3390/ijms221910661] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/27/2021] [Accepted: 09/27/2021] [Indexed: 12/24/2022] Open
Abstract
The keratinocyte (KC) is the main functional and structural component of the epidermis, the most external layer of the skin that is highly specialized in defense against external agents, prevention of leakage of body fluids and retention of internal water within the cells. Altered epidermal barrier and aberrant KC differentiation are involved in the pathophysiology of several skin diseases, such as atopic dermatitis (AD). AD is a chronic inflammatory disease characterized by cutaneous and systemic immune dysregulation and skin microbiota dysbiosis. Nevertheless, the pathological mechanisms of this complex disease remain largely unknown. In this review, we summarize current knowledge about the participation of the KC in different aspects of the AD. We provide an overview of the genetic predisposing and environmental factors, inflammatory molecules and signaling pathways of the KC that participate in the physiopathology of the AD. We also analyze the link among the KC, the microbiota and the inflammatory response underlying acute and chronic skin AD lesions.
Collapse
Affiliation(s)
- Pamela Gallegos-Alcalá
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Mariela Jiménez
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
| | - Daniel Cervantes-García
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- National Council of Science and Technology, Ciudad de México 03940, Mexico
| | - Eva Salinas
- Department of Microbiology, Center of Basic Science, Autonomous University of Aguascalientes, Aguascalientes 20100, Mexico; (P.G.-A.); (M.J.); (D.C.-G.)
- Correspondence: ; Tel.: +52-449-9108424
| |
Collapse
|
36
|
Traidl S, Roesner L, Zeitvogel J, Werfel T. Eczema herpeticum in atopic dermatitis. Allergy 2021; 76:3017-3027. [PMID: 33844308 DOI: 10.1111/all.14853] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases leading to pruritic skin lesions. A subset of AD patients exhibits a disseminated severe HSV infection called eczema herpeticum (EH) that can cause life-threatening complications. This review gives an overview of the clinical picture, and characteristics of the patients as well as the diagnosis and therapy of EH. A special focus lies on the pathophysiological hallmarks identified so far that predispose for EH. This aspect covers genetic aberrations, immunological changes, and environmental influences displaying a complex multifactorial situation, which is not completely understood. Type 2 skewing of virus-specific T cells in ADEH+ patients has been implicated in immune profile abnormalities, along with impaired functions of dendritic cells and natural killer cells. Furthermore, aberrations in interferon pathway-related genes such as IFNG and IFNGR1 have been identified to increase the risk of EH. IL-4, IL-25, and thymic stromal lymphopoietin (TSLP) are overexpressed in EH, whereas antimicrobial peptides like human β-defensins and LL-37 are reduced. Concerning the epidermal barrier, single nucleotide polymorphisms (SNPs) in skin barrier proteins such as filaggrin were identified in ADEH+ patients. A dysbalance of the skin microbiome also contributes to EH due to an increase of Staphylococcus aureus, which provides a supporting role to the viral infection via secreted toxins such as α-toxin. The risk of EH is reduced in AD patients treated with dupilumab. Further research is needed to identify and specifically target risk factors for EH in AD patients.
Collapse
Affiliation(s)
- Stephan Traidl
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Lennart Roesner
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Jana Zeitvogel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| |
Collapse
|
37
|
Bin L, Malley C, Taylor P, Preethi Boorgula M, Chavan S, Daya M, Mathias M, Shankar G, Rafaels N, Vergara C, Potee J, Campbell M, Hanifin JM, Simpson E, Schneider LC, Gallo RL, Hata T, Paller AS, De Benedetto A, Beck LA, Ong PY, Guttman‐Yassky E, Richers B, Baraghoshi D, Ruczinski I, Barnes KC, Leung DYM, Mathias RA. Whole genome sequencing identifies novel genetic mutations in patients with eczema herpeticum. Allergy 2021; 76:2510-2523. [PMID: 33548076 DOI: 10.1111/all.14762] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Eczema herpeticum (EH) is a rare complication of atopic dermatitis (AD) caused by disseminated herpes simplex virus (HSV) infection. The role of rare and/or deleterious genetic variants in disease etiology is largely unknown. This study aimed to identify genes that harbor damaging genetic variants associated with HSV infection in AD with a history of recurrent eczema herpeticum (ADEH+). METHODS Whole genome sequencing (WGS) was performed on 49 recurrent ADEH+ (≥3 EH episodes), 491 AD without a history of eczema herpeticum (ADEH-) and 237 non-atopic control (NA) subjects. Variants were annotated, and a gene-based approach (SKAT-O) was used to identify genes harboring damaging genetic variants associated with ADEH+. Genes identified through WGS were studied for effects on HSV responses and keratinocyte differentiation. RESULTS Eight genes were identified in the comparison of recurrent ADEH+to ADEH-and NA subjects: SIDT2, CLEC7A, GSTZ1, TPSG1, SP110, RBBP8NL, TRIM15, and FRMD3. Silencing SIDT2 and RBBP8NL in normal human primary keratinocytes (NHPKs) led to significantly increased HSV-1 replication. SIDT2-silenced NHPKs had decreased gene expression of IFNk and IL1b in response to HSV-1 infection. RBBP8NL-silenced NHPKs had decreased gene expression of IFNk, but increased IL1b. Additionally, silencing SIDT2 and RBBP8NL also inhibited gene expression of keratinocyte differentiation markers keratin 10 (KRT10) and loricrin (LOR). CONCLUSION SIDT2 and RBBP8NL participate in keratinocyte's response to HSV-1 infection. SIDT2 and RBBP8NL also regulate expression of keratinocyte differentiation genes of KRT10 and LOR.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics National Jewish Health Denver CO USA
| | - Claire Malley
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Patricia Taylor
- Department of Pediatrics National Jewish Health Denver CO USA
| | | | - Sameer Chavan
- Department of Medicine University of Colorado Aurora CO USA
| | - Michelle Daya
- Department of Medicine University of Colorado Aurora CO USA
| | - Malaika Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Gautam Shankar
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | | | | | | | | | | | - Eric Simpson
- Oregon Health & Science University Portland OR USA
| | | | - Richard L. Gallo
- Department of Dermatology University of California San Diego CA USA
| | - Tissa Hata
- Department of Dermatology University of California San Diego CA USA
| | - Amy S. Paller
- Northwestern University Feinberg School of Medicine Chicago IL USA
| | | | - Lisa A. Beck
- University of Rochester Medical Center Rochester NY USA
| | - Peck Y. Ong
- Children’s Hospital Los Angeles University of Southern California Los Angeles CA USA
| | | | | | | | - Ingo Ruczinski
- Department of Biostatistics Bloomberg School of Public Health Johns Hopkins University Baltimore MD USA
| | | | | | - Rasika A. Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| |
Collapse
|
38
|
Damour A, Garcia M, Seneschal J, Lévêque N, Bodet C. Eczema Herpeticum: Clinical and Pathophysiological Aspects. Clin Rev Allergy Immunol 2021; 59:1-18. [PMID: 31836943 DOI: 10.1007/s12016-019-08768-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in the world. AD is a complex pathology mainly characterized by an impaired skin barrier, immune response dysfunction, and unbalanced skin microbiota. Moreover, AD patients exhibit an increased risk of developing bacterial and viral infections. One of the most current, and potentially life-threatening, viral infection is caused by herpes simplex virus (HSV), which occurs in about 3% of AD patients under the name of eczema herpeticum (EH). Following a first part dedicated to the clinical features, virological diagnosis, and current treatments of EH, this review will focus on the description of the pathophysiology and, more particularly, the presently known predisposing factors to herpetic complications in AD patients. These factors include those related to impairment of the skin barrier such as deficit in filaggrin and anomalies in tight and adherens junctions. In addition, low production of the antimicrobial peptides cathelicidin LL-37 and human β-defensins; overexpression of cytokines such as interleukin (IL)-4, IL-13, IL-25, IL-33, and thymic stromal lymphopoietin (TSLP); or downregulation of type I to III interferons as well as defect in functions of immune cells such as dendritic, natural killer, and regulatory T cells have been involved. Otherwise, genetic polymorphisms and AD topical calcineurin inhibitor treatments have been associated with an increased risk of EH. Finally, dysbiosis of skin microbiota characterized in AD patients by Staphylococcus aureus colonization and toxin secretion, such as α-toxin, has been described as promoting HSV replication and could therefore contribute to EH.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Julien Seneschal
- INSERM U1035, BMGIC, Immuno-dermatologie ATIP-AVENIR, Bordeaux, France.,Département de Dermatologie and Dermatologie Pédiatrique, Centre national de référence pour les maladies rares de la peau, Hôpital Saint-André, Bordeaux, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.
| |
Collapse
|
39
|
Role of Antimicrobial Peptides in Skin Barrier Repair in Individuals with Atopic Dermatitis. Int J Mol Sci 2020; 21:ijms21207607. [PMID: 33066696 PMCID: PMC7589391 DOI: 10.3390/ijms21207607] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease that exhibits a complex interplay of skin barrier disruption and immune dysregulation. Patients with AD are susceptible to cutaneous infections that may progress to complications, including staphylococcal septicemia. Although most studies have focused on filaggrin mutations, the physical barrier and antimicrobial barrier also play critical roles in the pathogenesis of AD. Within the physical barrier, the stratum corneum and tight junctions play the most important roles. The tight junction barrier is involved in the pathogenesis of AD, as structural and functional defects in tight junctions not only disrupt the physical barrier but also contribute to immunological impairments. Furthermore, antimicrobial peptides, such as LL-37, human b-defensins, and S100A7, improve tight junction barrier function. Recent studies elucidating the pathogenesis of AD have led to the development of barrier repair therapy for skin barrier defects in patients with this disease. This review analyzes the association between skin barrier disruption in patients with AD and antimicrobial peptides to determine the effect of these peptides on skin barrier repair and to consider employing antimicrobial peptides in barrier repair strategies as an additional approach for AD management.
Collapse
|
40
|
Kobayashi T, Nagao K. Host-microbial dialogues in atopic dermatitis. Int Immunol 2020; 31:449-456. [PMID: 30877745 DOI: 10.1093/intimm/dxz026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/15/2019] [Indexed: 12/11/2022] Open
Abstract
Recent advances in sequencing technologies have revealed the diversity of microbes that reside on the skin surface which has enhanced our understanding on skin as an ecosystem, wherein the epidermis, immune cells and the microbiota engage in active dialogues that maintain barrier integrity and functional immunity. This mutual dialogue is altered in atopic dermatitis (AD), in which an impaired epidermal barrier, the skin microbial flora and aberrant immunity can form a vicious cycle that leads to clinical manifestations as eczematous dermatitis. Microbiome studies have revealed an altered microbial landscape in AD and genetic studies have identified genes that underlie barrier impairment and immune dysregulation. Shifting from the long-standing notion that AD was mediated by conventional allergic responses, emerging data suggest that it is a disorder of an altered host-microbial relationship with sophisticated pathophysiology. In this review, we will discuss recent advancements that suggest the roles of the skin microbiota in AD pathophysiology, genetic factors that mediate barrier impairment, dysbiosis and inflammation. Studies in mice, classic AD and monogenic disorders that manifest as AD collectively facilitate our understanding of AD pathophysiology and provide a foundation for novel therapeutic strategies.
Collapse
Affiliation(s)
- Tetsuro Kobayashi
- Cutaneous Leukocyte Biology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Keisuke Nagao
- Cutaneous Leukocyte Biology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
41
|
Smieszek SP, Welsh S, Xiao C, Wang J, Polymeropoulos C, Birznieks G, Polymeropoulos MH. Correlation of age-of-onset of Atopic Dermatitis with Filaggrin loss-of-function variant status. Sci Rep 2020; 10:2721. [PMID: 32066784 PMCID: PMC7026049 DOI: 10.1038/s41598-020-59627-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/30/2020] [Indexed: 12/31/2022] Open
Abstract
The genetic background of Atopic Dermatitis (AD) with chronic pruritus is complex. Filaggrin (FLG) is an essential gene in the epidermal barrier formation s. Loss-of-function (LOF) variants in FLG associated with skin barrier dysfunction constitute the most well-known genetic risk factor for AD. In this study, we focused on the frequency and effect of FLG loss-of-function variants in association with self-reported age-of-onset of AD. The dataset consisted of 386 whole-genome sequencing (WGS) samples. We observe a significant association between FLG LOF status and age-of-onset, with earlier age of onset of AD observed in the FLG LOF carrier group (p-value 0.0003, Wilcoxon two-sample test). We first tested this on the two most prevalent FLG variants. Interestingly, the effect is even stronger when considering all detected FLG LOF variants. Having two or more FLG LOF variants associates with the onset of AD at 2 years of age. In this study, we have shown enrichment of rare variants in the EDC region in cases compared with controls. Age-of-onset analysis shows not only the effect of the FLG and likely EDC variants in terms of the heightened risk of AD, but foremost enables to predict early-onset, lending further credence to the penetrance and causative effect of the identified variants. Understanding the genetic background and risk of early-onset is suggestive of skin barrier dysfunction etiology of AD with chronic pruritus
Collapse
Affiliation(s)
- S P Smieszek
- Vanda Pharmaceuticals Inc., Washington, DC, USA.
| | - S Welsh
- Vanda Pharmaceuticals Inc., Washington, DC, USA
| | - C Xiao
- Vanda Pharmaceuticals Inc., Washington, DC, USA
| | - J Wang
- Vanda Pharmaceuticals Inc., Washington, DC, USA
| | | | - G Birznieks
- Vanda Pharmaceuticals Inc., Washington, DC, USA
| | | |
Collapse
|
42
|
Hertz A, Azulay-Abulafia L, Nascimento APD, Ohara CY, Kuschnir FC, Porto LC. Analysis of filaggrin 2 gene polymorphisms in patients with atopic dermatitis. An Bras Dermatol 2020; 95:173-179. [PMID: 32151410 PMCID: PMC7175100 DOI: 10.1016/j.abd.2019.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 07/26/2019] [Indexed: 11/12/2022] Open
Abstract
Background Polymorphisms of the filaggrin 2 gene (rs 12568784 and rs 16899374) are associated with persistent atopic dermatitis in African American patients. Filaggrin 2 is a protein with a function similar to filaggrin and also encoded in the epidermal differentiation complex on chromosome 1q21. Objective To evaluate the polymorphisms in the filaggrin 2 gene (rs 12568784 and rs 16899374) in children and adults with atopic dermatitis and to verify the association of these with the severity of the clinical picture, presence of other allergic diseases, and socio-demographic factors. Method The study was carried out with patients and control group. Questionnaires were used to evaluate ethnicity, sex, age, family history, scoring, atopic dermatitis (SCORAD), among other parameters. Genotyping of the filaggrin 2 gene was performed by real-time polymerase chain reaction. Results Forty-eight patients and 83 controls were evaluated. No correlation was found between the variables studied in patients with atopic dermatitis and polymorphisms, no significant difference between the prevalence of polymorphisms in the patients and in the control group p > 0.05. Study limits The exclusive use of self-reported ethnicity information and the sample size. Results The results of this work can be an incentive for the study of the polymorphisms in atopic dermaititis, considering the characteristic of the Brazilian multi ethnic population. Conclusion This is an unpublished work in Brazil and the first study in the world to have a control group to evaluate alterations in the gene of filaggrin 2.
Collapse
Affiliation(s)
- Amanda Hertz
- Pediatric Dermatology Clinic, Instituto de Dermatologia Prof. Rubem David Azulay, Rio de Janeiro, RJ, Brazil.
| | - Luna Azulay-Abulafia
- Department of Dermatology, Instituto de Dermatologia Prof. Rubem David Azulay, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Adriana Paulino do Nascimento
- Histocompatibility and Cryopreservation Laboratory, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Cintya Yumi Ohara
- Department of Allergy and Immunopathology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fabio Chigres Kuschnir
- Department of Pediatrics, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Department of Allergy, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luís Cristovão Porto
- Histocompatibility and Cryopreservation Laboratory, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
43
|
Abstract
For the first time, we show that secreted staphylococcal virulence factors can be quantified at the protein level directly from skin swabs obtained from the skin of atopic dermatitis patients. This technique eliminates the need to culture Staphylococcus aureus and then test the strain’s potential to produce secreted virulence factors. Our methodology shows that secreted virulence factors are present on the skin of atopic patients and provides a more accurate means of evaluating the physiological impact of S. aureus in inflammatory diseases such as atopic dermatitis. Staphylococcus aureus is the leading cause of skin and soft tissue infections, bacteremia, infective endocarditis, osteoarticular, pleuropulmonary, and device-related infections. Virulence factors secreted by S. aureus, including superantigens and cytotoxins, play significant roles in driving disease. The ability to identify virulence factors present at the site of infection will be an important tool in better identifying and understanding how specific virulence factors contribute to disease. Previously, virulence factor production has been determined by culturing S. aureus isolates and detecting the mRNA of specific virulence factors. We demonstrated for the first time that virulence factors can be directly detected at the protein level from human samples, removing the need to first culture isolated bacteria. Superantigens and cytotoxins were detected and quantified with a Western dot blot assay by using reconstituted skin swabs obtained from patients with atopic dermatitis. This methodology will significantly enhance our ability to investigate the complex host-microbe environment and the effects various therapies have on virulence factor production. Overall, the ability to directly quantify virulence factors present at the site of infection or colonization will enhance our understanding of S. aureus-related diseases and help identify optimal treatments. IMPORTANCE For the first time, we show that secreted staphylococcal virulence factors can be quantified at the protein level directly from skin swabs obtained from the skin of atopic dermatitis patients. This technique eliminates the need to culture Staphylococcus aureus and then test the strain’s potential to produce secreted virulence factors. Our methodology shows that secreted virulence factors are present on the skin of atopic patients and provides a more accurate means of evaluating the physiological impact of S. aureus in inflammatory diseases such as atopic dermatitis.
Collapse
|
44
|
Alexander H, Paller AS, Traidl-Hoffmann C, Beck LA, De Benedetto A, Dhar S, Girolomoni G, Irvine AD, Spuls P, Su J, Thyssen JP, Vestergaard C, Werfel T, Wollenberg A, Deleuran M, Flohr C. The role of bacterial skin infections in atopic dermatitis: expert statement and review from the International Eczema Council Skin Infection Group. Br J Dermatol 2019; 182:1331-1342. [PMID: 31677162 PMCID: PMC7317931 DOI: 10.1111/bjd.18643] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/27/2022]
Abstract
Patients with atopic dermatitis (AD) have an increased risk of bacterial skin infections, which cause significant morbidity and, if untreated, may become systemic. Staphylococcus aureus colonizes the skin of most patients with AD and is the most common organism to cause infections. Overt bacterial infection is easily recognized by the appearance of weeping lesions, honey‐coloured crusts and pustules. However, the wide variability in clinical presentation of bacterial infection in AD and the inherent features of AD – cutaneous erythema and warmth, oozing associated with oedema, and regional lymphadenopathy – overlap with those of infection, making clinical diagnosis challenging. Furthermore, some features may be masked because of anatomical site‐ and skin‐type‐specific features, and the high frequency of S. aureus colonization in AD makes positive skin swab culture of suspected infection unreliable as a diagnostic tool. The host mechanisms and microbial virulence factors that underlie S. aureus colonization and infection in AD are incompletely understood. The aim of this article is to present the latest evidence from animal and human studies, including recent microbiome research, to define the clinical features of bacterial infections in AD, and to summarize our current understanding of the host and bacterial factors that influence microbial colonization and virulence.
Collapse
Affiliation(s)
- H Alexander
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, SE1 7EH, U.K
| | - A S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, U.S.A
| | - C Traidl-Hoffmann
- Chair and Institute of Environmental Medicine, UNIKA-T, Technical University of Munich and Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE, Christine Kühne Center for Allergy Research and Education, Davos, Switzerland
| | - L A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, U.S.A
| | - A De Benedetto
- Department of Dermatology, College of Medicine, University of Florida, Gainesville, FL, U.S.A
| | - S Dhar
- Department of Pediatric Dermatology, Institute of Child Health, Kolkata, India
| | - G Girolomoni
- Department of Medicine, Section of Dermatology and Venereology, University of Verona, Verona, Italy
| | - A D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland.,Dermatology, Children's Health Ireland, Dublin, Ireland.,National Children's Research Centre, Dublin, Ireland
| | - P Spuls
- Department of Dermatology, Amsterdam Public Health, Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - J Su
- Departments of Dermatology and Paediatrics, Murdoch Children's Research Institute, University of Melbourne and Monash University, Eastern Health, Melbourne, VIC, Australia
| | - J P Thyssen
- Department of Dermatology and Allergy, Herlev-Gentofte Hospital, Hellerup, Denmark
| | - C Vestergaard
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - T Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - A Wollenberg
- Department of Dermatology and Allergology, Ludwig Maximilian University, Munich, Germany
| | - M Deleuran
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - C Flohr
- Unit for Population-Based Dermatology Research, St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, SE1 7EH, U.K
| |
Collapse
|
45
|
Elias MS, Wright SC, Nicholson WV, Morrison KD, Prescott AR, Ten Have S, Whitfield PD, Lamond AI, Brown SJ. Functional and proteomic analysis of a full thickness filaggrin-deficient skin organoid model. Wellcome Open Res 2019; 4:134. [PMID: 31641698 PMCID: PMC6790913 DOI: 10.12688/wellcomeopenres.15405.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Atopic eczema is an itchy inflammatory disorder characterised by skin barrier dysfunction. Loss-of-function mutations in the gene encoding filaggrin (
FLG) are a major risk factor, but the mechanisms by which filaggrin haploinsufficiency leads to atopic inflammation remain incompletely understood. Skin as an organ that can be modelled using primary cells
in vitro provides the opportunity for selected genetic effects to be investigated in detail. Methods: Primary human keratinocytes and donor-matched primary fibroblasts from healthy individuals were used to create skin organoid models with and without siRNA-mediated knockdown of
FLG. Biological replicate sets of organoids were assessed using histological, functional and biochemical measurements. Results:FLG knockdown leads to subtle changes in histology and ultrastructure including a reduction in thickness of the stratum corneum and smaller, less numerous keratohyalin granules. Immature organoids showed some limited evidence of barrier impairment with
FLG knockdown, but the mature organoids showed no difference in transepidermal water loss, water content or dye penetration. There was no difference in epidermal ceramide content. Mass spectrometry proteomic analysis detected >8000 proteins per sample. Gene ontology and pathway analyses identified an increase in transcriptional and translational activity but a reduction in proteins contributing to terminal differentiation, including caspase 14, dermokine, AKT1 and TGF-beta-1. Aspects of innate and adaptive immunity were represented in both the up-regulated and down-regulated protein groups, as was the term ‘axon guidance’. Conclusions: This work provides further evidence for keratinocyte-specific mechanisms contributing to immune and neurological, as well as structural, aspects of skin barrier dysfunction. Individuals with filaggrin deficiency may derive benefit from future therapies targeting keratinocyte-immune crosstalk and neurogenic pruritus.
Collapse
Affiliation(s)
- Martina S Elias
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, DD1 9SY, UK
| | - Sheila C Wright
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, DD1 9SY, UK
| | - William V Nicholson
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, DD1 9SY, UK
| | - Kimberley D Morrison
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, DD1 9SY, UK
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Sara Ten Have
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Phillip D Whitfield
- Lipidomics Research Facility, Division of Biomedical Sciences, University of the Highlands and Islands, Inverness, Scotland, IV2 3JH, UK
| | - Angus I Lamond
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, Scotland, DD1 5EH, UK
| | - Sara J Brown
- Skin Research Group, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, Scotland, DD1 9SY, UK.,Department of Dermatology, Ninewells Hospital, Dundee, Scotland, DD1 9SY, UK
| |
Collapse
|
46
|
Nakajima S, Nomura T, Common J, Kabashima K. Insights into atopic dermatitis gained from genetically defined mouse models. J Allergy Clin Immunol 2019; 143:13-25. [PMID: 30612664 DOI: 10.1016/j.jaci.2018.11.014] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 01/01/2023]
Abstract
Atopic dermatitis (AD) is characterized by severe pruritus and recurrent eczema with a chronic disease course. Impaired skin barrier function, hyperactivated TH2 cell-type inflammation, and pruritus-induced scratching contribute to the disease pathogenesis of AD. Skin microbial alterations complicate the pathogenesis of AD further. Mouse models are a powerful tool to analyze such intricate pathophysiology of AD, with a caution that anatomy and immunology of the skin differ between human subjects and mice. Here we review recent understanding of AD etiology obtained using mouse models, which address the epidermal barrier, skin microbiome, TH2 immune response, and pruritus.
Collapse
Affiliation(s)
- Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - John Common
- Skin Research Institute of Singapore (SRIS), Singapore.
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Skin Research Institute of Singapore (SRIS), Singapore; Singapore Immunology Network, A*STAR, Singapore.
| |
Collapse
|
47
|
Prieux R, Eeman M, Rothen-Rutishauser B, Valacchi G. Mimicking cigarette smoke exposure to assess cutaneous toxicity. Toxicol In Vitro 2019; 62:104664. [PMID: 31669394 DOI: 10.1016/j.tiv.2019.104664] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 12/24/2022]
Abstract
Cigarette smoke stands among the most toxic environmental pollutants and is composed of thousands of chemicals including polycyclic aromatic hydrocarbons (PAHs). Despite restrict cigarette smoking ban in indoor or some outdoor locations, the risk of non-smokers to be exposed to environmental cigarette smoke is not yet eliminated. Beside the well-known effects of cigarette smoke to the respiratory and cardiovascular systems, a growing literature has shown during the last 3 decades its noxious effects also on cutaneous tissues. Being the largest organ as well as the interface between the outer environment and the body, human skin acts as a natural shield which is continuously exposed to harmful exogenous agents. Thus, a prolonged and/or repetitive exposure to significant levels of toxic smoke pollutants may have detrimental effects on the cutaneous tissue by disrupting the epidermal barrier function and by exacerbating inflammatory skin disorders (i.e. psoriasis, atopic dermatitis). With the development of very complex skin tissue models and sophisticated cigarette smoke exposure systems it has become important to better understand the toxicity pathways induced by smoke pollutants in more realistic laboratory conditions to find solutions for counteracting their effects. This review provides an update on the skin models currently available to study cigarette smoke exposure and the known pathways involved in cutaneous toxicity. In addition, the article will briefly cover the inflammatory skin pathologies potentially induced and/or exacerbated by cigarette smoke exposure.
Collapse
Affiliation(s)
- Roxane Prieux
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Marc Eeman
- Home & Personal Care, Dow Silicones Belgium, Seneffe, Belgium
| | | | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy; Plants for Human Health Institute, North Carolina State University, Kannapolis, United States; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
48
|
Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease. Ann Allergy Asthma Immunol 2019; 124:36-43. [PMID: 31622670 DOI: 10.1016/j.anai.2019.10.008] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/07/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To provide an overview of filaggrin biology and the role of filaggrin variants in atopic dermatitis (AD) and allergic disease. DATA SOURCES We performed a PubMed literature review consisting mainly of studies relating to filaggrin in the last 5 years. STUDY SELECTIONS We selected articles that were found in PubMed using the search terms filaggrin, atopic dermatitis, skin barrier, and atopy. RESULTS Filaggrin plays an important role in the development of AD and allergic disease. Novel methods in measuring filaggrin expression and identifying filaggrin mutations aid in stratifying this patient cohort. We review new insights into understanding the role of filaggrin in AD and allergic disease. CONCLUSION Filaggrin remains a very important player in the pathogenesis of atopic dermatitis and allergic disease. This review looks at recent studies that aid our understanding of this crucial epidermal protein.
Collapse
Affiliation(s)
| | - Alan D Irvine
- Department of Paediatric Dermatology, Our Lady's Children's Hospital Crumlin, Dublin, National Children's Research Centre, Crumlin and Clinical Medicine, Trinity College Dublin, Ireland.
| |
Collapse
|
49
|
Elias MS, Wright SC, Nicholson WV, Morrison KD, Prescott AR, Ten Have S, Whitfield PD, Lamond AI, Brown SJ. Proteomic analysis of a filaggrin-deficient skin organoid model shows evidence of increased transcriptional-translational activity, keratinocyte-immune crosstalk and disordered axon guidance. Wellcome Open Res 2019; 4:134. [DOI: 10.12688/wellcomeopenres.15405.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2019] [Indexed: 11/20/2022] Open
Abstract
Background:Atopic eczema is an itchy inflammatory disorder characterised by skin barrier dysfunction. Loss-of-function mutations in the gene encoding filaggrin (FLG) are a major risk factor, but the mechanisms by which filaggrin haploinsufficiency leads to atopic inflammation remain incompletely understood. Skin as an organ that can be modelled using primary cellsin vitroprovides the opportunity for selected genetic effects to be investigated in detail.Methods:Primary human keratinocytes and donor-matched primary fibroblasts from healthy individuals were used to create skin organoid models with and without siRNA-mediated knockdown ofFLG. Biological replicate sets of organoids were assessed using histological, functional and biochemical measurements.Results:FLGknockdown leads to subtle changes in histology and ultrastructure including a reduction in thickness of the stratum corneum and smaller, less numerous keratohyalin granules. Immature organoids showed evidence of barrier impairment withFLGknockdown, but the mature organoids showed no difference in transepidermal water loss, water content or dye penetration. There was no difference in epidermal ceramide content. Mass spectrometry proteomic analysis detected >8000 proteins per sample. Gene ontology and pathway analyses identified an increase in transcriptional and translational activity but a reduction in proteins contributing to terminal differentiation, including caspase 14, dermokine, AKT1 and TGF-beta-1. Aspects of innate and adaptive immunity were represented in both the up-regulated and down-regulated protein groups, as was the term ‘axon guidance’. Conclusions:This work provides further evidence for keratinocyte-specific mechanisms contributing to immune and neurological, as well as structural, aspects of skin barrier dysfunction. Individuals with filaggrin deficiency may derive benefit from future therapies targeting keratinocyte-immune crosstalk and neurogenic pruritus.
Collapse
|
50
|
de Carvalho-Siqueira GQ, Ananina G, de Souza BB, Borges MG, Ito MT, da Silva-Costa SM, de Farias Domingos I, Falcão DA, Lopes-Cendes I, Bezerra MAC, da Silva Araújo A, Lucena-Araújo AR, de Souza Gonçalves M, Saad STO, Costa FF, de Melo MB. Whole-exome sequencing indicates FLG2 variant associated with leg ulcers in Brazilian sickle cell anemia patients. Exp Biol Med (Maywood) 2019; 244:932-939. [PMID: 31079484 DOI: 10.1177/1535370219849592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Although sickle cell anemia results from homozygosity for a single mutation at position 7 of the β-globin chain, the clinical aspects of this condition are very heterogeneous. Complications include leg ulcers, which have a negative impact on patients’ quality of life and are related to the severity of the disease. Nevertheless, the complex pathogenesis of this complication has yet to be elucidated. To identify novel genes associated with leg ulcers in sickle cell anemia, we performed whole-exome sequencing of extreme phenotypes in a sample of Brazilian sickle cell anemia patients and validated our findings in another sample. Our discovery cohort consisted of 40 unrelated sickle cell anemia patients selected based on extreme phenotypes: 20 patients without leg ulcers, aged from 40 to 61 years, and 20 with chronic leg ulcers. DNA was extracted from peripheral blood leukocytes and used for whole-exome sequencing. After the bioinformatics analysis, eight variants were selected for validation by Sanger sequencing and TaqMan® genotyping in 293 sickle cell anemia patients (153 without leg ulcers) from two different locations in Brazil. After the validation, Fisher’s exact test revealed a statistically significant difference in a stop codon variant (rs12568784 G/T) in the FLG2 gene between the GT and GG genotypes ( P = 0.035). We highlight the importance of rs12568784 in leg ulcer development as this variant of the FLG2 gene results in impairment of the skin barrier, predisposing the individual to inflammation and infection. Additionally, we suggest that the remaining seven variants and the genes in which they occur could be strong candidates for leg ulcers in sickle cell anemia. Impact statement To our knowledge, the present study is the first to use whole-exome sequencing based on extreme phenotypes to identify new candidate genes associated with leg ulcers in sickle cell anemia patients. There are few studies about this complication; the pathogenesis remains complex and has yet to be fully elucidated. We identified interesting associations in genes never related with this complication to our knowledge, especially the variant in the FLG2 gene. The knowledge of variants related with leg ulcer in sickle cell anemia may lead to a better comprehension of the disease’s etiology, allowing prevention and early treatment options in risk genotypes while improving quality of life for these patients.
Collapse
Affiliation(s)
| | - Galina Ananina
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Bruno Batista de Souza
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Murilo Guimarães Borges
- 2 Department of Medical Genetics and Genome Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | - Mirta Tomie Ito
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Sueli Matilde da Silva-Costa
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| | - Igor de Farias Domingos
- 3 Genetics Postgraduate Program, Federal University of Pernambuco, Recife, PE 50670-901, Brazil
| | - Diego Arruda Falcão
- 3 Genetics Postgraduate Program, Federal University of Pernambuco, Recife, PE 50670-901, Brazil
| | - Iscia Lopes-Cendes
- 2 Department of Medical Genetics and Genome Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, SP 13083-887, Brazil
| | | | | | | | | | | | | | - Mônica Barbosa de Melo
- 1 Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Campinas, SP 13083-875, Brazil
| |
Collapse
|