1
|
Ebrahimi Samani S, Tatsukawa H, Hitomi K, Kaartinen MT. Transglutaminase 1: Emerging Functions beyond Skin. Int J Mol Sci 2024; 25:10306. [PMID: 39408635 PMCID: PMC11476513 DOI: 10.3390/ijms251910306] [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: 08/29/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
Transglutaminase enzymes catalyze Ca2+- and thiol-dependent posttranslational modifications of glutamine-residues that include esterification, hydrolysis and transamidation, which results in covalent protein-protein crosslinking. Among the eight transglutaminase family members in mammals, transglutaminase 1 (TG1) plays a crucial role in skin barrier formation via crosslinking and insolubilizing proteins in keratinocytes. Despite this established function in skin, novel functions have begun merging in normal tissue homeostasis as well as in pathologies. This review summarizes our current understanding of the structure, activation, expression and activity patterns of TG1 and discusses its putative novel role in other tissues, such as in vascular integrity, and in diseases, such as cancer and fibrosis.
Collapse
Affiliation(s)
- Sahar Ebrahimi Samani
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada;
| | - Hideki Tatsukawa
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; (H.T.); (K.H.)
| | - Kiyotaka Hitomi
- Department of Basic Medicinal Sciences, Graduate School of Pharmaceutical Sciences, Nagoya University, Nagoya 464-8601, Japan; (H.T.); (K.H.)
| | - Mari T. Kaartinen
- Division of Experimental Medicine, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3A 0C7, Canada;
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
2
|
Zakiudin DP, Rø ADB, Videm V, Øien T, Simpson MR. Systemic inflammatory proteins in offspring following maternal probiotic supplementation for atopic dermatitis prevention. Clin Mol Allergy 2023; 21:5. [PMID: 37516841 PMCID: PMC10386175 DOI: 10.1186/s12948-023-00186-3] [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: 03/05/2023] [Accepted: 07/13/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND Maternal probiotic supplementation has a promising effect on atopic dermatitis (AD) prevention in infancy. In the randomised controlled study, Probiotics in the Prevention of Allergy among Children in Trondheim (ProPACT), maternal probiotics reduced the cumulative incidence of AD in their offspring by 40% at 2 years of age. However, our understanding on how probiotics prevented AD is still limited, and the role of inflammatory proteins in infants following maternal probiotic supplementation is unclear. We hypothesised that maternal probiotics lowered pro-inflammatory proteins and increased anti-inflammatory proteins in their 2-year-old children as a mechanism of AD prevention. We aimed to explore this hypothesis and the association between these proteins and the presence of AD, severity of AD, and the degree of preventive effect of probiotics. METHODS Plasma samples were collected from 2-year-old children (n = 202) during the ProPACT study, a randomised placebo-controlled trial of maternal probiotic supplementation. These samples were analysed for 92 inflammatory proteins using a multiplex proximity extension assay. Associations between inflammatory proteins and the presence and severity of AD, and the degree of preventive effect, was estimated individually using regression analysis and then collectively using unsupervised cluster analysis. RESULTS Several proteins were observed to differ between the groups. The probiotic group had lower CCL11 and IL-17C, while children with AD had higher IL-17C, MCP-4, uPA, and CD6. Cytokine CCL20 and IL-18 had moderate correlation (r = 0.35 and r = 0.46) with the severity of AD. The cluster analysis revealed that children in the cluster of samples with the highest value of immune checkpoint receptors and inflammatory suppressor enzymes showed the greatest AD preventive effect from probiotics. CONCLUSIONS The proteins associated with both maternal probiotic supplementation and the presence and severity of AD warrant attention because of their potential biological relevance. Cluster analysis may provide a new insight when considering which subgroups benefit from probiotic supplementation. Larger studies are needed to confirm the results. TRIAL REGISTRATION NUMBER The study was retrospectively registered at ClinicalTrials.gov (NCT00159523) on 12nd September 2005.
Collapse
Affiliation(s)
- Dinastry Pramadita Zakiudin
- Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Håkon Jarls Gate 11, 7030, Trondheim, Norway.
- Clinic for Laboratory Medicine, St Olavs Hospital, Trondheim, Norway.
| | - Anne Dorthea Bjerkenes Rø
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Vibeke Videm
- Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Immunology and Transfusion Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Torbjørn Øien
- Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Håkon Jarls Gate 11, 7030, Trondheim, Norway
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Håkon Jarls Gate 11, 7030, Trondheim, Norway
| |
Collapse
|
3
|
Integrative analysis to explore the biological association between environmental skin diseases and ambient particulate matter. Sci Rep 2022; 12:9750. [PMID: 35697899 PMCID: PMC9192598 DOI: 10.1038/s41598-022-13001-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Although numerous experimental studies have suggested a significant association between ambient particulate matter (PM) and respiratory damage, the etiological relationship between ambient PM and environmental skin diseases is not clearly understood. Here, we aimed to explore the association between PM and skin diseases through biological big data analysis. Differential gene expression profiles associated with PM and environmental skin diseases were retrieved from public genome databases. The co-expression among them was analyzed using a text-mining-based network analysis software. Activation/inhibition patterns from RNA-sequencing data performed with PM2.5-treated normal human epidermal keratinocytes (NHEK) were overlapped to select key regulators of the analyzed pathways. We explored the adverse effects of PM on the skin and attempted to elucidate their relationships using public genome data. We found that changes in upstream regulators and inflammatory signaling networks mediated by MMP-1, MMP-9, PLAU, S100A9, IL-6, and S100A8 were predicted as the key pathways underlying PM-induced skin diseases. Our integrative approach using a literature-based co-expression analysis and experimental validation not only improves the reliability of prediction but also provides assistance to clarify underlying mechanisms of ambient PM-induced dermal toxicity that can be applied to screen the relationship between other chemicals and adverse effects.
Collapse
|
4
|
Jung N, Kong T, Yu Y, Park H, Lee E, Yoo S, Baek S, Lee S, Kang KS. Immunomodulatory Effect of Epidermal Growth Factor Secreted by Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells on Atopic Dermatitis. Int J Stem Cells 2022; 15:311-323. [PMID: 35220283 PMCID: PMC9396020 DOI: 10.15283/ijsc21173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives Human mesenchymal stem cells (MSCs) are emerging as a treatment for atopic dermatitis (AD), a chronic inflammatory skin disorder that affects a large number of people across the world. Treatment of AD using human umbilical cord blood-derived MSCs (hUCB-MSCs) has recently been studied. However, the mechanism underlying their effect needs to be studied continuously. Thus, the objective of this study was to investigate the immunomodulatory effect of epidermal growth factor (EGF) secreted by hUCB-MSCs on AD. Methods and Results To explore the mechanism involved in the therapeutic effect of MSCs for AD, a secretome array was performed using culture medium of hUCB-MSCs. Among the list of genes common for epithelium development and skin diseases, we focused on the function of EGF. To elucidate the effect of EGF secreted by hUCB-MSCs, EGF was downregulated in hUCB-MSCs using EGF-targeting small interfering RNA. These cells were then co-cultured with keratinocytes, Th2 cells, and mast cells. Depletion of EGF disrupted immunomodulatory effects of hUCB-MSCs on these AD-related inflammatory cells. In a Dermatophagoides farinae-induced AD mouse model, subcutaneous injection of hUCB-MSCs ameliorated gross scoring, histopathologic damage, and mast cell infiltration. It also significantly reduced levels of inflammatory cytokines including interleukin (IL)-4, tumor necrosis factor (TNF)-α, thymus and activation-regulated chemokine (TARC), and IL-22, as well as IgE levels. These therapeutic effects were significantly attenuated at all evaluation points in mice injected with EGF-depleted hUCB-MSCs. Conclusions EGF secreted by hUCB-MSCs can improve AD by regulating inflammatory responses of keratinocytes, Th2 cells, and mast cells.
Collapse
Affiliation(s)
- Namhee Jung
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - TaeHo Kong
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Yeonsil Yu
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Hwanhee Park
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Eunjoo Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - SaeMi Yoo
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - SongYi Baek
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
| | - Kyung-Sun Kang
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co., Ltd., Seoul, Korea
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
5
|
Ottman N, Barrientos‐Somarribas M, Fyhrquist N, Alexander H, Wisgrill L, Olah P, Tsoka S, Greco D, Levi‐Schaffer F, Soumelis V, Schröder JM, Kere J, Nestle FO, Barker J, Ranki A, Lauerma A, Homey B, Andersson B, Alenius H. Microbial and transcriptional differences elucidate atopic dermatitis heterogeneity across skin sites. Allergy 2021; 76:1173-1187. [PMID: 33001460 PMCID: PMC8246754 DOI: 10.1111/all.14606] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 12/29/2022]
Abstract
It is well established that different sites in healthy human skin are colonized by distinct microbial communities due to different physiological conditions. However, few studies have explored microbial heterogeneity between skin sites in diseased skin, such as atopic dermatitis (AD) lesions. To address this issue, we carried out deep analysis of the microbiome and transcriptome in the skin of a large cohort of AD patients and healthy volunteers, comparing two physiologically different sites: upper back and posterior thigh. Microbiome samples and biopsies were obtained from both lesional and nonlesional skin to identify changes related to the disease process. Transcriptome analysis revealed distinct disease-related gene expression profiles depending on anatomical location, with keratinization dominating the transcriptomic signatures in posterior thigh, and lipid metabolism in the upper back. Moreover, we show that relative abundance of Staphylococcus aureus is associated with disease severity in the posterior thigh, but not in the upper back. Our results suggest that AD may select for similar microbes in different anatomical locations-an "AD-like microbiome," but distinct microbial dynamics can still be observed when comparing posterior thigh to upper back. This study highlights the importance of considering the variability across skin sites when studying the development of skin inflammation.
Collapse
Affiliation(s)
- Noora Ottman
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
| | | | - Nanna Fyhrquist
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- Human Microbiome Research Program University of Helsinki Helsinki Finland
| | - Helen Alexander
- St John's Institute of Dermatology Guy's and St Thomas' NHS Foundation Trust and King's College London London UK
| | - Lukas Wisgrill
- Division of Neonatology Pediatric Intensive Care and Neuropediatrics Medical University of Vienna Vienna Austria
| | - Peter Olah
- Department of Dermatology University Hospital Duesseldorf Duesseldorf Germany
- Department of Dermatology, Venereology and Oncodermatology University of Pécs Pécs Hungary
| | - Sophia Tsoka
- Department of Informatics Faculty of Natural and Mathematical Sciences King’s College London London UK
| | - Dario Greco
- Faculty of Medicine and Life Sciences University of Tampere Tampere Finland
- Institute of Biomedical Technology University of Tampere Tampere Finland
- Institute of Biotechnology University of Helsinki Helsinki Finland
| | - Francesca Levi‐Schaffer
- Pharmacology Unit School of Pharmacy Faculty of Medicine The Institute for Drug Research The Hebrew University of Jerusalem Jerusalem Israel
| | | | - Jens M. Schröder
- Department of Dermatology University Hospital Schleswig‐Holstein Kiel Germany
| | - Juha Kere
- Department of Biosciences and Nutrition Karolinska Institutet Stockholm Sweden
- School of Basic and Medical Biosciences King’s College London London UK
| | - Frank O. Nestle
- Cutaneous Medicine Unit St. John’s Institute of Dermatology and Biomedical Research Centre Faculty of Life Sciences and Medicine King’s College London London UK
| | - Jonathan Barker
- St John’s Institute of Dermatology Division of Genetics and Molecular Medicine Faculty of Life Sciences and Medicine Kings College London London UK
| | - Annamari Ranki
- Department of Dermatology, Allergology and Venereology Inflammation Centre University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Antti Lauerma
- Department of Dermatology, Allergology and Venereology Inflammation Centre University of Helsinki and Helsinki University Hospital Helsinki Finland
| | - Bernhard Homey
- Department of Dermatology University Hospital Duesseldorf Duesseldorf Germany
| | - Björn Andersson
- Department of Cell and Molecular Biology Karolinska Institutet Stockholm Sweden
| | - Harri Alenius
- Institute of Environmental Medicine Karolinska Institutet Stockholm Sweden
- Human Microbiome Research Program University of Helsinki Helsinki Finland
| |
Collapse
|
6
|
Corzo-León DE, MacCallum DM, Munro CA. Host Responses in an Ex Vivo Human Skin Model Challenged With Malassezia sympodialis. Front Cell Infect Microbiol 2021; 10:561382. [PMID: 33552997 PMCID: PMC7859105 DOI: 10.3389/fcimb.2020.561382] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
Malassezia species are a major part of the normal mycobiota and colonize mainly sebum-rich skin regions of the body. This group of fungi cause a variety of infections such as pityriasis versicolor, folliculitis, and fungaemia. In particular, Malassezia sympodialis and its allergens have been associated with non-infective inflammatory diseases such as seborrheic dermatitis and atopic eczema. The aim of this study was to investigate the host response to M. sympodialis on oily skin (supplemented with oleic acid) and non-oily skin using an ex vivo human skin model. Host-pathogen interactions were analyzed by SEM, histology, gene expression, immunoassays and dual species proteomics. The skin response to M. sympodialis was characterized by increased expression of the genes encoding β-defensin 3 and RNase7, and by high levels of S100 proteins in tissue. Supplementation of oleic acid onto skin was associated with direct contact of yeasts with keratinocytes and epidermal damage. In oily conditions, there was increased expression of IL18 but no expression of antimicrobial peptide genes in the skin’s response to M. sympodialis. In supernatants from inoculated skin plus oleic acid, TNFα, IL-6, and IL1-β levels were decreased and IL-18 levels were significantly increased.
Collapse
Affiliation(s)
- Dora E Corzo-León
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Donna M MacCallum
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Carol A Munro
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
7
|
Abstract
Atopic dermatitis is a common inflammatory skin disorder characterised by recurrent eczematous lesions and intense itch. The disorder affects people of all ages and ethnicities, has a substantial psychosocial impact on patients and relatives, and is the leading cause of the global burden from skin disease. Atopic dermatitis is associated with increased risk of multiple comorbidities, including food allergy, asthma, allergic rhinitis, and mental health disorders. The pathophysiology is complex and involves a strong genetic predisposition, epidermal dysfunction, and T-cell driven inflammation. Although type-2 mechanisms are dominant, there is increasing evidence that the disorder involves multiple immune pathways. Currently, there is no cure, but increasing numbers of innovative and targeted therapies hold promise for achieving disease control, including in patients with recalcitrant disease. We summarise and discuss advances in our understanding of the disease and their implications for prevention, management, and future research.
Collapse
Affiliation(s)
- Sinéad M Langan
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK; St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK; Health Data Research UK, London, UK.
| | - Alan D Irvine
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland; Dermatology, Children's Health Ireland, Crumlin, Ireland; National Children's Research Centre, Dublin, Ireland
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
8
|
Zimmer SE, Kowalczyk AP. The desmosome as a model for lipid raft driven membrane domain organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183329. [PMID: 32376221 DOI: 10.1016/j.bbamem.2020.183329] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/20/2020] [Accepted: 04/23/2020] [Indexed: 01/14/2023]
Abstract
Desmosomes are cadherin-based adhesion structures that mechanically couple the intermediate filament cytoskeleton of adjacent cells to confer mechanical stress resistance to tissues. We have recently described desmosomes as mesoscale lipid raft membrane domains that depend on raft dynamics for assembly, function, and disassembly. Lipid raft microdomains are regions of the plasma membrane enriched in sphingolipids and cholesterol. These domains participate in membrane domain heterogeneity, signaling and membrane trafficking. Cellular structures known to be dependent on raft dynamics include the post-synaptic density in neurons, the immunological synapse, and intercellular junctions, including desmosomes. In this review, we discuss the current state of the desmosome field and put forward new hypotheses for the role of lipid rafts in desmosome adhesion, signaling and epidermal homeostasis. Furthermore, we propose that differential lipid raft affinity of intercellular junction proteins is a central driving force in the organization of the epithelial apical junctional complex.
Collapse
Affiliation(s)
- Stephanie E Zimmer
- Graduate Program in Biochemistry, Cell and Developmental Biology, Emory University, Atlanta, GA 30322, United States of America; Department of Cell Biology, Emory University, Atlanta, GA 30322, United States of America
| | - Andrew P Kowalczyk
- Department of Cell Biology, Emory University, Atlanta, GA 30322, United States of America; Department of Dermatology, Emory University, Atlanta, GA 30322, United States of America.
| |
Collapse
|
9
|
Igalan from Inula helenium (L.) suppresses the atopic dermatitis-like response in stimulated HaCaT keratinocytes via JAK/STAT3 signaling. Inflamm Res 2020; 69:309-319. [DOI: 10.1007/s00011-020-01322-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/20/2020] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
|
10
|
Review-Current Concepts in Inflammatory Skin Diseases Evolved by Transcriptome Analysis: In-Depth Analysis of Atopic Dermatitis and Psoriasis. Int J Mol Sci 2020; 21:ijms21030699. [PMID: 31973112 PMCID: PMC7037913 DOI: 10.3390/ijms21030699] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/14/2022] Open
Abstract
During the last decades, high-throughput assessment of gene expression in patient tissues using microarray technology or RNA-Seq took center stage in clinical research. Insights into the diversity and frequency of transcripts in healthy and diseased conditions provide valuable information on the cellular status in the respective tissues. Growing with the technique, the bioinformatic analysis toolkit reveals biologically relevant pathways which assist in understanding basic pathophysiological mechanisms. Conventional classification systems of inflammatory skin diseases rely on descriptive assessments by pathologists. In contrast to this, molecular profiling may uncover previously unknown disease classifying features. Thereby, treatments and prognostics of patients may be improved. Furthermore, disease models in basic research in comparison to the human disease can be directly validated. The aim of this article is not only to provide the reader with information on the opportunities of these techniques, but to outline potential pitfalls and technical limitations as well. Major published findings are briefly discussed to provide a broad overview on the current findings in transcriptomics in inflammatory skin diseases.
Collapse
|
11
|
Zhu J, Wang Z, Chen F. Association of Key Genes and Pathways with Atopic Dermatitis by Bioinformatics Analysis. Med Sci Monit 2019; 25:4353-4361. [PMID: 31184315 PMCID: PMC6582687 DOI: 10.12659/msm.916525] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Atopic dermatitis is a chronic inflammatory disease of the skin. It has a high prevalence worldwide and affected persons are prone to recurrent attacks, seriously affecting the physical and mental of patients. The exact etiology of the disease is still unclear. Material/Methods There are 7 datasets on atopic dermatitis in the Gene Expression Omnibus database, including 142 lesional and 134 non-lesional skin biopsy samples. Differential analysis was performed after datasets were integrated by robust multi-array average method. Functional modules of GSE99802 were explored by weighted gene co-expression network analysis. The 4 most important modules were enriched into the pathways by Metascape. Results Significantly differentially expressed genes included 41 upregulated and 10 downregulated genes. The following 5 of the most important upregulated genes had the strongest association with atopic dermatitis. SERPINB3&4 promote inflammation and impaired skin barrier function in the early stage of atopic dermatitis. S100A9 aggravates the inflammatory response by inducing the activation of toll-like receptor 4, neutrophil chemotaxis, neutrophilic inflammation, and the amplification of interleukin-8. MMP1 is the key protease of skin collagen degradation, keeping the extracellular matrix in dynamic balance. MMP12 induces the aggregation of various inflammatory cells into inflammatory tissue. The enriched pathways of each module mainly include Cellular responses to external stimuli, Metabolism of RNA and Translation, and Infectious disease. Conclusions The associated pathways and genes not only help us understand the molecular mechanism of the disease, but also provide research directions or targets for accurate diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Zheng Wang
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Fengzhe Chen
- Department of Infectious Diseases, Qilu Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
12
|
Ianiri G, Heitman J, Scheynius A. The Skin Commensal Yeast Malassezia globosa Thwarts Bacterial Biofilms to Benefit the Host. J Invest Dermatol 2019; 138:1026-1029. [PMID: 29681390 DOI: 10.1016/j.jid.2018.01.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 01/09/2018] [Indexed: 12/11/2022]
Abstract
Malassezia are abundant, lipid-dependent, commensal yeasts in the skin microbiome that also have a pathogenic lifestyle associated with several common skin disorders. Malassezia genomes encode myriad lipases and proteases thought to mediate lipid utilization and pathogenesis. Li et al. report the biochemical characterization of a unique secreted aspartyl protease produced by Malassezia globosa, MgSAP1, and demonstrate its active role in hindering biofilm formation of the bacterium Staphylococcus aureus. Because biofilms are an established virulence attribute of S. aureus, this study reveals a potential benefit to the host of the fungal aspartyl protease MgSAP1 and opens the door for the investigation of the roles of such molecules in microbial interactions and their possible effects on the host.
Collapse
Affiliation(s)
- Giuseppe Ianiri
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Joseph Heitman
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Annika Scheynius
- Science for Life Laboratory, Department of Clinical Science and Education, Karolinska Institutet, and Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden.
| |
Collapse
|
13
|
Ghosh D, Bernstein JA, Khurana Hershey GK, Rothenberg ME, Mersha TB. Leveraging Multilayered "Omics" Data for Atopic Dermatitis: A Road Map to Precision Medicine. Front Immunol 2018; 9:2727. [PMID: 30631320 PMCID: PMC6315155 DOI: 10.3389/fimmu.2018.02727] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022] Open
Abstract
Atopic dermatitis (AD) is a complex multifactorial inflammatory skin disease that affects ~280 million people worldwide. About 85% of AD cases begin in childhood, a significant portion of which can persist into adulthood. Moreover, a typical progression of children with AD to food allergy, asthma or allergic rhinitis has been reported (“allergic march” or “atopic march”). AD comprises highly heterogeneous sub-phenotypes/endotypes resulting from complex interplay between intrinsic and extrinsic factors, such as environmental stimuli, and genetic factors regulating cutaneous functions (impaired barrier function, epidermal lipid, and protease abnormalities), immune functions and the microbiome. Though the roles of high-throughput “omics” integrations in defining endotypes are recognized, current analyses are primarily based on individual omics data and using binary clinical outcomes. Although individual omics analysis, such as genome-wide association studies (GWAS), can effectively map variants correlated with AD, the majority of the heritability and the functional relevance of discovered variants are not explained or known by the identified variants. The limited success of singular approaches underscores the need for holistic and integrated approaches to investigate complex phenotypes using trans-omics data integration strategies. Integrating omics layers (e.g., genome, epigenome, transcriptome, proteome, metabolome, lipidome, exposome, microbiome), which often have complementary and synergistic effects, might provide the opportunity to capture the flow of information underlying AD disease manifestation. Overlapping genes/candidates derived from multiple omics types include FLG, SPINK5, S100A8, and SERPINB3 in AD pathogenesis. Overlapping pathways include macrophage, endothelial cell and fibroblast activation pathways, in addition to well-known Th1/Th2 and NFkB activation pathways. Interestingly, there was more multi-omics overlap at the pathway level than gene level. Further analysis of multi-omics overlap at the tissue level showed that among 30 tissue types from the GTEx database, skin and esophagus were significantly enriched, indicating the biological interconnection between AD and food allergy. The present work explores multi-omics integration and provides new biological insights to better define the biological basis of AD etiology and confirm previously reported AD genes/pathways. In this context, we also discuss opportunities and challenges introduced by “big omics data” and their integration.
Collapse
Affiliation(s)
- Debajyoti Ghosh
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Jonathan A Bernstein
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| | - Tesfaye B Mersha
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
14
|
Traks T, Keermann M, Karelson M, Rätsep R, Reimann E, Silm H, Vasar E, Kõks S, Kingo K. Polymorphisms in melanocortin system and MYG1 genes are associated with vitiligo. J Eur Acad Dermatol Venereol 2018; 33:e65-e67. [PMID: 30051642 DOI: 10.1111/jdv.15195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- T Traks
- Department of Dermatology and Venerology, University of Tartu, Tartu, Estonia
| | - M Keermann
- Department of Dermatology and Venerology, University of Tartu, Tartu, Estonia.,Clinic of Dermatology, Tartu University Hospital, Tartu, Estonia
| | - M Karelson
- Department of Dermatology and Venerology, University of Tartu, Tartu, Estonia.,Clinic of Dermatology, Tartu University Hospital, Tartu, Estonia
| | - R Rätsep
- Department of Physiology, University of Tartu, Tartu, Estonia
| | - E Reimann
- Department of Pathophysiology, University of Tartu, Tartu, Estonia.,Department of Reproductive Biology, Estonian University of Life Sciences, Tartu, Estonia
| | - H Silm
- Department of Dermatology and Venerology, University of Tartu, Tartu, Estonia.,Clinic of Dermatology, Tartu University Hospital, Tartu, Estonia
| | - E Vasar
- Department of Physiology, University of Tartu, Tartu, Estonia
| | - S Kõks
- Department of Pathophysiology, University of Tartu, Tartu, Estonia.,Department of Reproductive Biology, Estonian University of Life Sciences, Tartu, Estonia
| | - K Kingo
- Department of Dermatology and Venerology, University of Tartu, Tartu, Estonia.,Clinic of Dermatology, Tartu University Hospital, Tartu, Estonia
| |
Collapse
|
15
|
Czarnowicki T, Dohlman AB, Malik K, Antonini D, Bissonnette R, Chan TC, Zhou L, Wen HC, Estrada Y, Xu H, Bryson C, Shen J, Lala D, Ma'ayan A, McGeehan G, Gregg R, Guttman-Yassky E. Effect of short-term liver X receptor activation on epidermal barrier features in mild to moderate atopic dermatitis: A randomized controlled trial. Ann Allergy Asthma Immunol 2018; 120:631-640.e11. [PMID: 29567358 DOI: 10.1016/j.anai.2018.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/27/2018] [Accepted: 03/12/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Liver X receptors (LXRs) are involved in maintaining epidermal barrier and suppressing inflammatory responses in model systems. The LXR agonist VTP-38543 showed promising results in improving barrier function and inflammatory responses in model systems. OBJECTIVE To assess the safety, tolerability, cellular and molecular changes, and clinical efficacy of the topical VTP-38543 in adults with mild to moderate atopic dermatitis (AD). METHODS A total of 104 ambulatory patients with mild to moderate AD were enrolled in this randomized, double-blind, vehicle-controlled trial between December 2015 and September 2016. VTP-38543 cream in 3 concentrations (0.05%, 0.15%, and 1.0%) or placebo was applied twice daily for 28 days. Pretreatment and posttreatment skin biopsy specimens were obtained from a subset of 33 patients. Changes in SCORing of Atopic Dermatitis, Eczema Area and Severity Index, Investigator's Global Assessment, and tissue biomarkers (by real-time polymerase chain reaction and immunostaining) were evaluated. RESULTS Topical VTP-38543 was safe and well tolerated. VTP-38543 significantly increased messenger RNA (mRNA) expression of epidermal barrier differentiation (loricrin and filaggrin, P = .02) and lipid (adenosine triphosphate-binding cassette subfamily G member 1 and sterol regulatory element binding protein 1c, P < .01) measures and reduced epidermal hyperplasia markers (thickness, keratin 16 mRNA). VTP-38543 nonsignificantly suppressed cellular infiltrates and down-regulated mRNA expression of several TH17/TH22-related (phosphatidylinositol 3, S100 calcium-binding protein A12) and innate immunity (interleukin 6) markers. CONCLUSION Topical VTP-38543 is safe and well tolerated. Its application led to improvement in barrier differentiation and lipids. Longer-term studies are needed to clarify whether a barrier-based approach can induce meaningful suppression of immune abnormalities. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT02655679.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York
| | - Anders B Dohlman
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai School, New York, New York
| | - Kunal Malik
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; College of Medicine, SUNY Downstate, Brooklyn, New York
| | - Diane Antonini
- Vitae Pharmaceuticals Inc, an Allergan affiliate, Irvine, California
| | | | - Tom C Chan
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Lisa Zhou
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Huei-Chi Wen
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yeriel Estrada
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hui Xu
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Catherine Bryson
- Vitae Pharmaceuticals Inc, an Allergan affiliate, Irvine, California
| | - Jie Shen
- Allergan plc, Irvine, California
| | - Deepak Lala
- Vitae Pharmaceuticals Inc, an Allergan affiliate, Irvine, California
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai School, New York, New York
| | - Gerard McGeehan
- Vitae Pharmaceuticals Inc, an Allergan affiliate, Irvine, California
| | - Richard Gregg
- Vitae Pharmaceuticals Inc, an Allergan affiliate, Irvine, California
| | - Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York.
| |
Collapse
|
16
|
Hardman CS, Chen YL, Salimi M, Jarrett R, Johnson D, Järvinen VJ, Owens RJ, Repapi E, Cousins DJ, Barlow JL, McKenzie ANJ, Ogg G. CD1a presentation of endogenous antigens by group 2 innate lymphoid cells. Sci Immunol 2017; 2:eaan5918. [PMID: 29273672 PMCID: PMC5826589 DOI: 10.1126/sciimmunol.aan5918] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023]
Abstract
Group 2 innate lymphoid cells (ILC2) are effectors of barrier immunity, with roles in infection, wound healing, and allergy. A proportion of ILC2 express MHCII (major histocompatibility complex II) and are capable of presenting peptide antigens to T cells and amplifying the subsequent adaptive immune response. Recent studies have highlighted the importance of CD1a-reactive T cells in allergy and infection, activated by the presentation of endogenous neolipid antigens and bacterial components. Using a human skin challenge model, we unexpectedly show that human skin-derived ILC2 can express CD1a and are capable of presenting endogenous antigens to T cells. CD1a expression is up-regulated by TSLP (thymic stromal lymphopoietin) at levels observed in the skin of patients with atopic dermatitis, and the response is dependent on PLA2G4A. Furthermore, this pathway is used to sense Staphylococcus aureus by promoting Toll-like receptor-dependent CD1a-reactive T cell responses to endogenous ligands. These findings define a previously unrecognized role for ILC2 in lipid surveillance and identify shared pathways of CD1a- and PLA2G4A-dependent ILC2 inflammation amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Clare S Hardman
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Yi-Ling Chen
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Maryam Salimi
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rachael Jarrett
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Valtteri J Järvinen
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Raymond J Owens
- Oxford Protein Production Facility-UK, Harwell and Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Emmanouela Repapi
- Computational Biology Research Group, Weatherall Institute of Molecular Medicine, Oxford, UK
| | - David J Cousins
- Department of Infection, Immunity and Inflammation, NIHR Leicester Respiratory Biomedical Research Unit, University of Leicester, Leicester, UK
- MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, King's College London, London, UK
| | | | | | - Graham Ogg
- Medical Research Council (MRC) Human Immunology Unit, Weatherall Institute of Molecular Medicine, National Institute for Health Research (NIHR) Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
De Vuyst E, Salmon M, Evrard C, Lambert de Rouvroit C, Poumay Y. Atopic Dermatitis Studies through In Vitro Models. Front Med (Lausanne) 2017; 4:119. [PMID: 28791291 PMCID: PMC5523664 DOI: 10.3389/fmed.2017.00119] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/11/2017] [Indexed: 11/13/2022] Open
Abstract
Atopic dermatitis (AD) is a complex inflammatory skin condition that is not fully understood. Epidermal barrier defects and Th2 immune response dysregulations are thought to play crucial roles in the pathogenesis of the disease. A vicious circle takes place between these alterations, and it can further be complicated by additional genetic and environmental factors. Studies investigating in more depth the etiology of the disease are thus needed in order to develop functional treatments. In recent years, there have been significant advances regarding in vitro models reproducing important features of AD. However, since a lot of models have been developed, finding the appropriate experimental setting can be difficult. Therefore, herein, we review the different types of in vitro models mimicking features of AD. The simplest models are two-dimensional culture systems composed of immune cells or keratinocytes, whereas three-dimensional skin or epidermal equivalents reconstitute more complex stratified tissues exhibiting barrier properties. In those models, hallmarks of AD are obtained, either by challenging tissues with interleukin cocktails overexpressed in AD epidermis or by silencing expression of pivotal genes encoding epidermal barrier proteins. Tissue equivalents cocultured with lymphocytes or containing AD patient cells are also described. Furthermore, each model is placed in its study context with a brief summary of the main results obtained. In conclusion, the described in vitro models are useful tools to better understand AD pathogenesis, but also to screen new compounds in the field of AD, which probably will open the way to new preventive or therapeutic strategies.
Collapse
Affiliation(s)
- Evelyne De Vuyst
- Cell and Tissue Laboratory, URPhyM-NARILIS, University of Namur, Namur, Belgium
| | | | - Céline Evrard
- Cell and Tissue Laboratory, URPhyM-NARILIS, University of Namur, Namur, Belgium
| | | | - Yves Poumay
- Cell and Tissue Laboratory, URPhyM-NARILIS, University of Namur, Namur, Belgium
| |
Collapse
|
18
|
Kruglikov IL, Wollina U. Local effects of adipose tissue in psoriasis and psoriatic arthritis. PSORIASIS-TARGETS AND THERAPY 2017; 7:17-25. [PMID: 29387604 PMCID: PMC5774600 DOI: 10.2147/ptt.s122959] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The structure and physiological state of the local white adipose tissue (WAT) located underneath the lesional psoriatic skin and inside of the joints affected by psoriatic arthritis play an important role in the pathophysiology of these diseases. WAT pads associated with inflammatory sites in psoriasis and psoriatic arthritis are, correspondingly, dermal WAT and articular adipose tissue; these pads demonstrate inflammatory phenotypes in both diseases. Such local WAT inflammation could be the primary effect in the pathophysiology of psoriasis leading to the modification of the local expression of adipokines, a change in the structure of the basement membrane and the release of keratinocytes with consequent epidermal hyperproliferation during psoriasis. Similar articular adipose tissue inflammation can lead to the induction of structural modifications and synovial inflammation in the joints of patients with psoriatic arthritis.
Collapse
Affiliation(s)
| | - Uwe Wollina
- Department of Dermatology and Allergology, Hospital Dresden-Friedrichstadt Academic Teaching Hospital of the Technical University of Dresden, Dresden, Germany
| |
Collapse
|
19
|
Chen W, Wang T, Pino-Yanes M, Forno E, Liang L, Yan Q, Hu D, Weeks DE, Baccarelli A, Acosta-Perez E, Eng C, Han YY, Boutaoui N, Laprise C, Davies GA, Hopkin JM, Moffatt MF, Cookson WOCM, Canino G, Burchard EG, Celedón JC. An epigenome-wide association study of total serum IgE in Hispanic children. J Allergy Clin Immunol 2017; 140:571-577. [PMID: 28069425 DOI: 10.1016/j.jaci.2016.11.030] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/26/2016] [Accepted: 11/24/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Total IgE is a therapeutic target in patients with allergic diseases. DNA methylation in white blood cells (WBCs) was associated with total IgE levels in an epigenome-wide association study of white subjects. Whether DNA methylation of eosinophils explains these findings is insufficiently understood. METHODS We tested for association between genome-wide DNA methylation in WBCs and total IgE levels in 2 studies of Hispanic children: the Puerto Rico Genetics of Asthma and Lifestyle Study (PR-GOAL; n = 306) and the Genes-environments and Admixture in Latino Americans (GALA II) study (n = 573). Whole-genome methylation of DNA from WBCs was measured by using the Illumina Infinium HumanMethylation450 BeadChip. Total IgE levels were measured by using the UniCAP 100 system. In PR-GOAL WBC types (ie, neutrophils, eosinophils, basophils, lymphocytes, and monocytes) in peripheral blood were measured by using Coulter Counter techniques. In the GALA II study WBC types were imputed. Multivariable linear regression was used for the analysis of DNA methylation and total IgE levels, which was first conducted separately for each cohort, and then results from the 2 cohorts were combined in a meta-analysis. RESULTS CpG sites in multiple genes, including novel findings and results previously reported in white subjects, were significantly associated with total IgE levels. However, adjustment for WBC types resulted in markedly fewer significant sites. Top findings from this adjusted meta-analysis were in the genes ZFPM1 (P = 1.5 × 10-12), ACOT7 (P = 2.5 × 10-11), and MND1 (P = 1.4 × 10-9). CONCLUSIONS In an epigenome-wide association study adjusted for WBC types (including eosinophils), methylation changes in genes enriched in pathways relevant to asthma and immune responses were associated with total IgE levels among Hispanic children.
Collapse
Affiliation(s)
- Wei Chen
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Ting Wang
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Maria Pino-Yanes
- Instituto de Salud Carlos III, CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Erick Forno
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Liming Liang
- Departments of Epidemiology and Biostatistics, Harvard School of Public Health, Boston, Mass
| | - Qi Yan
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Donglei Hu
- Department of Therapeutic Sciences and Bioengineering, University of California at San Francisco, San Francisco, Calif
| | - Daniel E Weeks
- Departments of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pa
| | - Andrea Baccarelli
- Department of Environmental Medicine, Harvard School of Public Health, Boston, Mass
| | - Edna Acosta-Perez
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, Puerto Rico
| | - Celeste Eng
- Department of Therapeutic Sciences and Bioengineering, University of California at San Francisco, San Francisco, Calif
| | - Yueh-Ying Han
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | - Nadia Boutaoui
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa
| | | | - Gwyneth A Davies
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Julian M Hopkin
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Miriam F Moffatt
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | | | - Glorisa Canino
- Behavioral Sciences Research Institute and Department of Pediatrics, University of Puerto Rico, San Juan, Puerto Rico
| | - Esteban G Burchard
- Department of Therapeutic Sciences and Bioengineering, University of California at San Francisco, San Francisco, Calif
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh, Pittsburgh, Pa.
| |
Collapse
|
20
|
Global gene regulation during activation of immunoglobulin class switching in human B cells. Sci Rep 2016; 6:37988. [PMID: 27897229 PMCID: PMC5126563 DOI: 10.1038/srep37988] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 11/03/2016] [Indexed: 02/06/2023] Open
Abstract
Immunoglobulin class switch recombination (CSR) to IgE is a tightly regulated process central to atopic disease. To profile the B-cell transcriptional responses underlying the activation of the germinal centre activities leading to the generation of IgE, naïve human B-cells were stimulated with IL-4 and anti-CD40. Gene expression and alternative splicing were profiled over 12 days using the Affymetrix Human Exon 1.0 ST Array. A total of 1,399 genes, forming 13 temporal profiles were differentially expressed. CCL22 and CCL17 were dramatically induced but followed a temporal trajectory distinct from classical mediators of isotype switching. AICDA, NFIL3, IRF4, XBP1 and BATF3 shared a profile with several genes involved in innate immunity, but with no recognised role in CSR. A transcription factor BHLHE40 was identified at the core of this profile. B-cell activation was also accompanied by variation in exon retention affecting >200 genes including CCL17. The data indicate a circadian component and central roles for the Th2 chemokines CCL22 and CCL17 in the activation of CSR.
Collapse
|
21
|
Olivry T, Mayhew D, Paps JS, Linder KE, Peredo C, Rajpal D, Hofland H, Cote-Sierra J. Early Activation of Th2/Th22 Inflammatory and Pruritogenic Pathways in Acute Canine Atopic Dermatitis Skin Lesions. J Invest Dermatol 2016; 136:1961-1969. [PMID: 27342734 DOI: 10.1016/j.jid.2016.05.117] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/16/2022]
Abstract
Determining inflammation and itch pathway activation in patients with atopic dermatitis (AD) is fraught with the inability to precisely assess the age of skin lesions, thus affecting the analysis of time-dependent mediators. To characterize inflammatory events occurring during early experimental acute AD lesions, biopsy samples were collected 6, 24, and 48 hours after epicutaneous application of Dermatophagoides farinae house dust mites to sensitized atopic dogs. The skin transcriptome was assessed using a dog-specific microarray and quantitative PCR. Acute canine AD skin lesions had a significant up-regulation of genes encoding T helper (Th) 2 (e.g., IL4, IL5, IL13, IL31, and IL33), Th9 (IL9), and Th22 (IL22) cytokines as well as Th2-promoting chemokines such as CCL5 and CCL17. Proinflammatory (e.g., IL6, LTB, and IL18) cytokines were also up-regulated. Other known pruritogenic pathways were also activated: there was significant up-regulation of genes encoding proteases cathepsin S (CTSS), mast cell chymase (CMA1), tryptase (TPS1) and mastin, neuromedin-B (NMB), nerve growth factor (NGF), and leukotriene-synthesis enzymes (ALOX5, ALOX5AP, and LTA4H). Experimental acute canine house dust mite-induced AD lesions exhibit an activation of innate and adaptive immune responses and pruritogenic pathways similar to those seen in humans with acute AD, thereby validating this model to test innovative therapeutics modalities for this disease.
Collapse
Affiliation(s)
- Thierry Olivry
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - David Mayhew
- Computational Biology, Target Sciences, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Judy S Paps
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Keith E Linder
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA; Department of Population Health and Pathobiology, College of Veterinary Medicine, Research Triangle Park, North Carolina, USA
| | - Carlos Peredo
- Stiefel, GlaxoSmithKline, Research Triangle Park, North Carolina, USA.
| | - Deepak Rajpal
- Safety Assessment, Platform Technology and Science, GlaxoSmithKline, Research Triangle Park, NC, USA
| | - Hans Hofland
- Stiefel, GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | | |
Collapse
|
22
|
Jarrett R, Ogg G. Lipid-specific T cells and the skin. Br J Dermatol 2016; 175 Suppl 2:19-25. [DOI: 10.1111/bjd.14908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2016] [Indexed: 12/23/2022]
Affiliation(s)
- R. Jarrett
- MRC Human Immunology Unit; Weatherall Institute of Molecular Medicine; NIHR Biomedical Research Centre; Radcliffe Department of Medicine; University of Oxford; Oxford OX3 9DS U.K
| | - G. Ogg
- MRC Human Immunology Unit; Weatherall Institute of Molecular Medicine; NIHR Biomedical Research Centre; Radcliffe Department of Medicine; University of Oxford; Oxford OX3 9DS U.K
| |
Collapse
|
23
|
Sun Y, Sheshadri N, Zong WX. SERPINB3 and B4: From biochemistry to biology. Semin Cell Dev Biol 2016; 62:170-177. [PMID: 27637160 DOI: 10.1016/j.semcdb.2016.09.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/08/2016] [Accepted: 09/12/2016] [Indexed: 12/15/2022]
Abstract
Human SERPINB3 and SERPINB4 are evolutionary duplicated serine/cysteine protease inhibitors. Genomic analysis indicates that these paralogous genes were encoded from independent loci arising from tandem gene duplication. Although the two molecules share 92% identity of their amino acid sequences, they are distinct in the Reactive Center Loop (RCL) including a hinge region and catalytic sequences which accounts for altered substrate specificity. Elevated expression of the two molecules has been reported to contribute to numerous pathological conditions such as inflammatory diseases and cancer. In this review, we focus on summarizing the biochemical features of SERPINB3/B4 and discussing the mechanistic basis for their biological functions and implications in human diseases.
Collapse
Affiliation(s)
- Yu Sun
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Namratha Sheshadri
- National Cancer Institute, Center for Cancer Research, Frederick, MD 21702, United States
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, United States.
| |
Collapse
|
24
|
Jarrett R, Salio M, Lloyd-Lavery A, Subramaniam S, Bourgeois E, Archer C, Cheung KL, Hardman C, Chandler D, Salimi M, Gutowska-Owsiak D, de la Serna JB, Fallon PG, Jolin H, Mckenzie A, Dziembowski A, Podobas EI, Bal W, Johnson D, Moody DB, Cerundolo V, Ogg G. Filaggrin inhibits generation of CD1a neolipid antigens by house dust mite-derived phospholipase. Sci Transl Med 2016; 8:325ra18. [PMID: 26865566 PMCID: PMC4872823 DOI: 10.1126/scitranslmed.aad6833] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Atopic dermatitis is a common pruritic skin disease in which barrier dysfunction and cutaneous inflammation contribute to pathogenesis. Mechanisms underlying the associated inflammation are not fully understood, and although Langerhans cells expressing the nonclassical major histocompatibility complex (MHC) family member CD1a are known to be enriched within lesions, their role in clinical disease pathogenesis has not been studied. We observed that house dust mite (HDM) allergen generates neolipid antigens presented by CD1a to T cells in the blood and skin lesions of affected individuals. HDM-responsive CD1a-reactive T cells increased in frequency after birth in individuals with atopic dermatitis and showed rapid effector function, consistent with antigen-driven maturation. In HDM-challenged human skin, we observed phospholipase A2 (PLA2) activity in vivo. CD1a-reactive T cell activation was dependent on HDM-derived PLA2, and such cells infiltrated the skin after allergen challenge. Moreover, we observed that the skin barrier protein filaggrin, insufficiency of which is associated with atopic skin disease, inhibited PLA2 activity and decreased CD1a-reactive PLA2-generated neolipid-specific T cell activity from skin and blood. The most widely used classification schemes of hypersensitivity suggest that nonpeptide stimulants of T cells act as haptens that modify peptides or proteins; however, our results show that HDM proteins may also generate neolipid antigens that directly activate T cells. These data define PLA2 inhibition as a function of filaggrin, supporting PLA2 inhibition as a therapeutic approach.
Collapse
Affiliation(s)
- Rachael Jarrett
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Mariolina Salio
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Antonia Lloyd-Lavery
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Sumithra Subramaniam
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Elvire Bourgeois
- Division of Rheumatology, Immunology and Allergy, Department of Medicine Brigham and Women’s Hospital, Harvard Medical School, 1 Jimmy Fund Way, Boston, Massachusetts, 02114, USA
| | - Charles Archer
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Ka Lun Cheung
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Clare Hardman
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - David Chandler
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Maryam Salimi
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Danuta Gutowska-Owsiak
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Jorge Bernardino de la Serna
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Padraic G. Fallon
- Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- Institute of Molecular Medicine, St James’s Hospital, Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Childrens Hospital, Dublin, Ireland
| | - Helen Jolin
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | | | - Andrzej Dziembowski
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Ewa Izabela Podobas
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - David Johnson
- Department of Plastic and Reconstructive Surgery, John Radcliffe Hospital, Oxford University Hospitals NHS Trust, UK
| | - D Branch Moody
- Division of Rheumatology, Immunology and Allergy, Department of Medicine Brigham and Women’s Hospital, Harvard Medical School, 1 Jimmy Fund Way, Boston, Massachusetts, 02114, USA
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, University of Oxford, UK
| |
Collapse
|
25
|
Ghosh D, Ding L, Sivaprasad U, Geh E, Biagini Myers J, Bernstein JA, Khurana Hershey GK, Mersha TB. Multiple Transcriptome Data Analysis Reveals Biologically Relevant Atopic Dermatitis Signature Genes and Pathways. PLoS One 2015; 10:e0144316. [PMID: 26717000 PMCID: PMC4696650 DOI: 10.1371/journal.pone.0144316] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Several studies have identified genes that are differentially expressed in atopic dermatitis (AD) compared to normal skin. However, there is also considerable variation in the list of differentially expressed genes (DEGs) reported by different groups and the exact cause of AD is still not fully understood. Using a rank-based approach, we analyzed gene expression data from five different microarray studies, comprising a total of 127 samples and more than 250,000 transcripts. A total of 89 AD gene expression signatures '89ADGES', including FLG gene, were identified to show dysregulation consistently across these studies. Using a Support Vector Machine, we showed that the '89ADGES' discriminates AD from normal skin with 98% predictive accuracy. Functional annotation of these genes implicated their roles in immune responses (e.g., betadefensin, microseminoprotein), keratinocyte differentiation/epidermal development (e.g., FLG, CORIN, AQP, LOR, KRT16), inflammation (e.g., IL37, IL27RA, CCL18) and lipid metabolism (e.g., AKR1B10, FAD7, FAR2). Subsequently, we validated a subset of signature genes using quantitative PCR in a mouse model. Using a bioinformatic approach, we identified keratinocyte pathway over-represented (P = <0.0006) among the 89 signature genes. Keratinocytes are known to play a major role in barrier function due to their location in the epidermis. Our result suggests that besides immune- mediated pathway, skin barrier pathways such as the keratinocyte differentiation pathway play a key role in AD pathogenesis. A better understanding of the role of keratinocytes in AD will be important for developing novel "barrier therapy" for this disease.
Collapse
Affiliation(s)
- Debajyoti Ghosh
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, United States of America
| | - Lili Ding
- Division of Biostatistics and Epidemiology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, United States of America
| | - Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, United States of America
| | - Esmond Geh
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, United States of America
| | - Jocelyn Biagini Myers
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, United States of America
| | - Jonathan A. Bernstein
- Division of Immunology, Allergy & Rheumatology, Department of Internal Medicine, University of Cincinnati, Cincinnati, United States of America
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, United States of America
| | - Tesfaye B. Mersha
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, United States of America
- * E-mail:
| |
Collapse
|
26
|
Chae M, Jung JY, Bae IH, Kim HJ, Lee TR, Shin DW. Lipin-1 expression is critical for keratinocyte differentiation. J Lipid Res 2015; 57:563-73. [PMID: 26658689 DOI: 10.1194/jlr.m062588] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Indexed: 12/19/2022] Open
Abstract
Lipin-1 is an Mg(2+)-dependent phosphatidate phosphatase that facilitates the dephosphorylation of phosphatidic acid to generate diacylglycerol. Little is known about the expression and function of lipin-1 in normal human epidermal keratinocytes (NHEKs). Here, we demonstrate that lipin-1 is present in basal and spinous layers of the normal human epidermis, and lipin-1 expression is gradually downregulated during NHEK differentiation. Interestingly, lipin-1 knockdown (KD) inhibited keratinocyte differentiation and caused G1 arrest by upregulating p21 expression. Cell cycle arrest by p21 is required for commitment of keratinocytes to differentiation, but must be downregulated for the progress of keratinocyte differentiation. Therefore, reduced keratinocyte differentiation results from sustained upregulation of p21 by lipin-1 KD. Lipin-1 KD also decreased the phosphorylation/activation of protein kinase C (PKC)α, whereas lipin-1 overexpression increased PKCα phosphorylation. Treatment with PKCα inhibitors, like lipin-1 KD, stimulated p21 expression, while lipin-1 overexpression reduced p21 expression, implicating PKCα in lipin-1-induced regulation of p21 expression. Taken together, these results suggest that lipin-1-mediated downregulation of p21 is critical for the progress of keratinocyte differentiation after the initial commitment of keratinocytes to differentiation induced by p21, and that PKCα is involved in p21 expression regulation by lipin-1.
Collapse
Affiliation(s)
- Minjung Chae
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Ji-Yong Jung
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Il-Hong Bae
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Hyoung-June Kim
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Tae Ryong Lee
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Dong Wook Shin
- Bioscience Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
27
|
Ewald DA, Malajian D, Krueger JG, Workman CT, Wang T, Tian S, Litman T, Guttman-Yassky E, Suárez-Fariñas M. Meta-analysis derived atopic dermatitis (MADAD) transcriptome defines a robust AD signature highlighting the involvement of atherosclerosis and lipid metabolism pathways. BMC Med Genomics 2015; 8:60. [PMID: 26459294 PMCID: PMC4603338 DOI: 10.1186/s12920-015-0133-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 09/11/2015] [Indexed: 12/24/2022] Open
Abstract
Background Atopic dermatitis (AD) is a common inflammatory skin disease with limited treatment options. Several microarray experiments have been conducted on lesional/LS and non-lesional/NL AD skin to develop a genomic disease phenotype. Although these experiments have shed light on disease pathology, inter-study comparisons reveal large differences in resulting sets of differentially expressed genes (DEGs), limiting the utility of direct comparisons across studies. Methods We carried out a meta-analysis combining 4 published AD datasets to define a robust disease profile, termed meta-analysis derived AD (MADAD) transcriptome. Results This transcriptome enriches key AD pathways more than the individual studies, and associates AD with novel pathways, such as atherosclerosis signaling (IL-37, selectin E/SELE). We identified wide lipid abnormalities and, for the first time in vivo, correlated Th2 immune activation with downregulation of key epidermal lipids (FA2H, FAR2, ELOVL3), emphasizing the role of cytokines on the barrier disruption in AD. Key AD “classifier genes” discriminate lesional from nonlesional skin, and may evaluate therapeutic responses. Conclusions Our meta-analysis provides novel and powerful insights into AD disease pathology, and reinforces the concept of AD as a systemic disease. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0133-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David A Ewald
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA. .,Molecular Biomedicine, LEO Pharma AS, Ballerup, Denmark. .,Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Dana Malajian
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA. .,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Columbia University, College of Physicians and Surgeons, New York, NY, USA.
| | - James G Krueger
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA.
| | - Christopher T Workman
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | - Tianjiao Wang
- School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, Jilin, 130012, China.
| | - Suyan Tian
- School of Life Science, Jilin University, 2699 Qianjin Street, Changchun, Jilin, 130012, China.
| | - Thomas Litman
- Molecular Biomedicine, LEO Pharma AS, Ballerup, Denmark.
| | - Emma Guttman-Yassky
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA. .,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Mayte Suárez-Fariñas
- The Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA. .,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn Institute for Genomics and Multiscale Biology at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
28
|
Rouaud-Tinguely P, Boudier D, Marchand L, Barruche V, Bordes S, Coppin H, Roth MP, Closs B. From the morphological to the transcriptomic characterization of a compromised three-dimensional in vitro model mimicking atopic dermatitis. Br J Dermatol 2015; 173:1006-14. [PMID: 26147950 DOI: 10.1111/bjd.14012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disease in which skin barrier function is disrupted. In this AD environment, proinflammatory cytokines are upregulated, promoting a vicious circle of inflammation. Although several three-dimensional in vitro models mimicking AD have been published, no study has presented a fully characterized and controlled model of AD-related inflammation. OBJECTIVES To develop and characterize, from the morphological to the molecular level, a compromised reconstructed epidermis (RE) mimicking AD-related inflammation in vitro. METHODS Normal human keratinocytes were used to generate RE, treated or not with an inflammatory cocktail (polyinosinic-polycytidylic acid, tumour necrosis factor-α, interleukin-4 and interleukin-13). RESULTS The inflammatory cocktail induces some modifications observed in patients with AD: (i) it leads to spongiosis; (ii) it alters early and terminal differentiation proteins; (iii) it increases thymic stromal lymphopoietin and interleukin-8 secretion by keratinocytes and (iv) it results in a specific gene expression pattern. CONCLUSIONS The inflammatory context contributes to the morphological, functional and transcriptomic changes observed in AD skin. As a result, this compromised RE model shares some characteristics with those found in AD skin and thus can be used as a relevant tool for screening formulations and drugs for the treatment of AD.
Collapse
Affiliation(s)
| | - D Boudier
- R&D Department, SILAB, BP 213, 19108, Brive CEDEX, France
| | - L Marchand
- R&D Department, SILAB, BP 213, 19108, Brive CEDEX, France
| | - V Barruche
- R&D Department, SILAB, BP 213, 19108, Brive CEDEX, France
| | - S Bordes
- R&D Department, SILAB, BP 213, 19108, Brive CEDEX, France
| | - H Coppin
- Centre de Physiopathologie de Toulouse Purpan, Inserm U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - M P Roth
- Centre de Physiopathologie de Toulouse Purpan, Inserm U1043, CNRS U5282, Université de Toulouse, Toulouse, France
| | - B Closs
- R&D Department, SILAB, BP 213, 19108, Brive CEDEX, France
| |
Collapse
|
29
|
Sivaprasad U, Kinker KG, Ericksen MB, Lindsey M, Gibson AM, Bass SA, Hershey NS, Deng J, Medvedovic M, Khurana Hershey GK. SERPINB3/B4 contributes to early inflammation and barrier dysfunction in an experimental murine model of atopic dermatitis. J Invest Dermatol 2014; 135:160-169. [PMID: 25111616 PMCID: PMC4268075 DOI: 10.1038/jid.2014.353] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 07/08/2014] [Accepted: 07/31/2014] [Indexed: 01/21/2023]
Abstract
Serine proteases are critical for epidermal barrier homeostasis, and their aberrant expression and/or activity is associated with chronic skin diseases. Elevated levels of the serine protease inhibitors SERPINB3 and SERPINB4 are seen in patients with atopic dermatitis and psoriasis. However, their mechanistic role in the skin is unknown. To evaluate the contribution of Serpinb3a (mouse homolog of SERPINB3 and SERPINB4) in atopic dermatitis, we examined the effect of topical Aspergillus fumigatus extract exposure in wild-type and Serpinb3a-null mice on transepidermal water loss (TEWL), sensitization, and inflammation. Allergen exposure induced Serpinb3a expression in the skin, along with increased TEWL, epidermal thickness, and skin inflammation, all of which were attenuated in the absence of Serpinb3a. Attenuated TEWL correlated with decreased expression of the pro-inflammatory marker S100A8. Silencing of SERPINB3/B4 in human keratinocytes decreased S100A8 expression, supporting a role for SERPINB3/B4 in the initiation of the acute inflammatory response. RNA-seq analysis following allergen exposure identified a network of pro-inflammatory genes induced in wild-type mice that was absent in Serpinb3a-null mice. In conclusion, Serpinb3a deficiency attenuates barrier dysfunction and the early inflammatory response following cutaneous allergen exposure, supporting a role for Serpinb3a (mice) and SERPINB3/B4 (humans) early in atopic dermatitis.
Collapse
Affiliation(s)
- Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kayla G Kinker
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark B Ericksen
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mark Lindsey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Aaron M Gibson
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stacey A Bass
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nicolas S Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jingyuan Deng
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
30
|
Cole C, Kroboth K, Schurch NJ, Sandilands A, Sherstnev A, O'Regan GM, Watson RM, McLean WHI, Barton GJ, Irvine AD, Brown SJ. Filaggrin-stratified transcriptomic analysis of pediatric skin identifies mechanistic pathways in patients with atopic dermatitis. J Allergy Clin Immunol 2014; 134:82-91. [PMID: 24880632 PMCID: PMC4090750 DOI: 10.1016/j.jaci.2014.04.021] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/28/2014] [Accepted: 04/24/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Atopic dermatitis (AD; eczema) is characterized by a widespread abnormality in cutaneous barrier function and propensity to inflammation. Filaggrin is a multifunctional protein and plays a key role in skin barrier formation. Loss-of-function mutations in the gene encoding filaggrin (FLG) are a highly significant risk factor for atopic disease, but the molecular mechanisms leading to dermatitis remain unclear. OBJECTIVE We sought to interrogate tissue-specific variations in the expressed genome in the skin of children with AD and to investigate underlying pathomechanisms in atopic skin. METHODS We applied single-molecule direct RNA sequencing to analyze the whole transcriptome using minimal tissue samples. Uninvolved skin biopsy specimens from 26 pediatric patients with AD were compared with site-matched samples from 10 nonatopic teenage control subjects. Cases and control subjects were screened for FLG genotype to stratify the data set. RESULTS Two thousand four hundred thirty differentially expressed genes (false discovery rate, P < .05) were identified, of which 211 were significantly upregulated and 490 downregulated by greater than 2-fold. Gene ontology terms for "extracellular space" and "defense response" were enriched, whereas "lipid metabolic processes" were downregulated. The subset of FLG wild-type cases showed dysregulation of genes involved with lipid metabolism, whereas filaggrin haploinsufficiency affected global gene expression and was characterized by a type 1 interferon-mediated stress response. CONCLUSION These analyses demonstrate the importance of extracellular space and lipid metabolism in atopic skin pathology independent of FLG genotype, whereas an aberrant defense response is seen in subjects with FLG mutations. Genotype stratification of the large data set has facilitated functional interpretation and might guide future therapy development.
Collapse
Affiliation(s)
- Christian Cole
- Division of Computational Biology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Karin Kroboth
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, Colleges of Life Sciences and Medicine, Dentistry & Nursing, University of Dundee, Dundee, United Kingdom
| | - Nicholas J Schurch
- Division of Computational Biology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Aileen Sandilands
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, Colleges of Life Sciences and Medicine, Dentistry & Nursing, University of Dundee, Dundee, United Kingdom
| | - Alexander Sherstnev
- Division of Computational Biology, College of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Grainne M O'Regan
- Department of Dermatology, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - Rosemarie M Watson
- Department of Dermatology, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - W H Irwin McLean
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, Colleges of Life Sciences and Medicine, Dentistry & Nursing, University of Dundee, Dundee, United Kingdom
| | - Geoffrey J Barton
- Division of Computational Biology, College of Life Sciences, University of Dundee, Dundee, United Kingdom.
| | - Alan D Irvine
- Department of Dermatology, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland; Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
| | - Sara J Brown
- Centre for Dermatology and Genetic Medicine, Division of Molecular Medicine, Colleges of Life Sciences and Medicine, Dentistry & Nursing, University of Dundee, Dundee, United Kingdom; National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland.
| |
Collapse
|
31
|
What does global gene expression profiling tell us about the pathogenesis of systemic sclerosis? Curr Opin Rheumatol 2014; 25:686-91. [PMID: 24061076 DOI: 10.1097/01.bor.0000434672.77891.41] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The purpose of this study is to review recent hypothesis-driven studies that utilize global gene expression data for elucidating the molecular basis of systemic sclerosis (SSc) and its various clinical manifestations. RECENT FINDINGS The longitudinal skin gene expression studies indicate that the previously identified molecular subsets are stable over time and might identify inherent subgroups of SSc patients. Skin transcript follow-up studies indicate that the Wnt/β-catenin pathway plays an important role in promotion of fibrogenesis in fibroblasts and preadipocytes. Furthermore, the transcript profile of sclerodermatous graft-versus-host disease (sclGVHD) mice resembles the skin transcriptomes of a subgroup of SSc patientswith IL13/IL4-inducible skin signature wherein the profibrotic chemokine CCL2 plays a key role. The comparison of skin biopsies from SSc patients to skin lesions of patients with cutaneous lupus and dermatomyositis has provided valuable information about the interferon (IFN) signature in these autoimmune diseases. Furthermore, plasma IFN-inducible chemokines correlate with the IFN gene expression score in SSc patients, enabling researchers to examine this molecular signature in large SSc cohorts with serum or plasma collection. SUMMARY Global gene expression profiling in skin and peripheral blood can contribute to a better understanding of SSc pathogenesis and identify novel biomarkers and therapeutic targets.
Collapse
|
32
|
Chen F, Li Z, Chen YPP. Determining common insertion sites based on retroviral insertion distribution across tumors. Comput Biol Chem 2014; 51:83-92. [PMID: 24675070 DOI: 10.1016/j.compbiolchem.2014.03.001] [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: 12/31/2013] [Revised: 02/24/2014] [Accepted: 03/03/2014] [Indexed: 10/25/2022]
Abstract
A CIS (common insertion site) indicates a genome region that is hit more frequently by retroviral insertions than expected by chance. Such a region is strongly related to cancer gene loci, which leads to the detection of cancer genes. An algorithm for detecting CISs should satisfy the following: (1) it does not require any prior knowledge of underlying insertion distribution; (2) it can resolve the insertion biases caused by hotspots; (3) it can detect CISs of any biological width; (4) it can identify noises resulting from statistic mistakes and non-CIS insertions; and (5) it can identify the widths of CISs as accurately as possible. We develop a method to resolve these difficulties. We verify a region's significance from two perspectives: distribution width and distribution depth. The former indicates how many insertions in a region while the latter evaluates the insertion distribution across the tumors in a region. We compare our method with kernel density estimation and sliding window on the simulated data, showing that our method not only identifies cancer-related insertions effectively, but also filters noises correctly. The experiments on the real data show that taking insertion distribution into account can highlight significant CISs. We detect 53 novel CISs, some of which have been proven correct by the biological literature.
Collapse
Affiliation(s)
- Feng Chen
- College of Information Science and Engineering, Henan University of Technology, Zhengzhou City, Henan Province 450001, China; Faculty of Science, Technology and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Zhoufang Li
- College of Information Science and Engineering, Henan University of Technology, Zhengzhou City, Henan Province 450001, China
| | - Yi-Ping Phoebe Chen
- Faculty of Science, Technology and Engineering, La Trobe University, Melbourne, Victoria 3086, Australia.
| |
Collapse
|
33
|
Chen F, Zhang Y, Chen YPP. Subdividing globally important zones based on data distribution across multiple genome fragments. Comput Biol Med 2014; 48:109-18. [PMID: 24674715 DOI: 10.1016/j.compbiomed.2014.02.004] [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: 08/19/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 11/15/2022]
Abstract
In multiple genome fragments, a globally important mode is a zone represented by a significant change, where the change has a similar impact on every related fragment in the zone. This zone may represent the cancer related genes involved in diverse tumors. Globally important zones are characterized by two features: (1) there are more data points in globally important zones than in other areas of fragments; (2) the data points are distributed evenly on as many genome fragments as possible. Globally important zone mining needs to contain the following features: (1) independent of data distribution; (2) noise filtering; (3) pattern boundary identification; and (4) zone ranking. We have developed a hierarchical and density-based method, called GIZFinder (globally important zone finder), to detect and rank such zones based on two criteria: distribution width and distribution depth. The comparisons on the simulated data shows our method performs significantly better than the kernel framework and the sliding window. By experimenting on real cancer gene data, we identify 53 novel cancer genes, some of which have been proven correct.
Collapse
Affiliation(s)
- Feng Chen
- College of Information Science and Engineering, Henan University of Technology, Zhengzhou, China; Department of Computer Science and Computer Engineering, La Trobe University, Melbourne, Australia
| | - Yuhong Zhang
- College of Information Science and Engineering, Henan University of Technology, Zhengzhou, China
| | - Yi-Ping Phoebe Chen
- Department of Computer Science and Computer Engineering, La Trobe University, Melbourne, Australia.
| |
Collapse
|
34
|
Hoppe T, Winge MCG, Bradley M, Nordenskjöld M, Vahlquist A, Törmä H, Berne B. Moisturizing treatment of patients with atopic dermatitis and ichthyosis vulgaris improves dry skin, but has a modest effect on gene expression regardless of FLG genotype. J Eur Acad Dermatol Venereol 2013; 29:174-7. [PMID: 24330146 DOI: 10.1111/jdv.12333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 11/12/2013] [Indexed: 12/26/2022]
Abstract
BACKGROUND Loss-of-function mutations in FLG (encoding filaggrin) are a predisposing factor for atopic dermatitis (AD) and cause ichthyosis vulgaris (IV). Patients with AD and IV display impaired skin barrier and dry skin, and altered epidermal expression of genes in pro-inflammatory and lipid metabolic pathways are often evident. OBJECTIVES To evaluate the effect of three different moisturizers on skin barrier function and epidermal gene expression in patients with AD/IV in relation to FLG mutation status. METHODS Patients (n = 43) were classified according to their FLG status: AD with FLG+/+ (n = 14), AD with FLG+/- (n = 14), and AD/IV with FLG-/- (n = 15). Dryness score and transepidermal water loss (TEWL) were monitored on volar forearms, and punch biopsies were taken for analysis of gene expression. Measurements were repeated after 4 weeks of treatment with either of two moisturizers on each forearm. RESULTS Treatment with any of the three moisturizers significantly reduced dryness score and TEWL in the group as a whole. FLG-/- patients displayed the largest reduction in dryness score. Only minute changes occurred in the mRNA expression of 15 selected epidermal genes. CONCLUSIONS Moisturizing treatment improves dry skin and certain aspects of abnormal skin barrier function, especially in patients with AD/IV and dual FLG mutations, but does not normalize the epidermal gene expression profile.
Collapse
Affiliation(s)
- T Hoppe
- Department of Medical Sciences, Dermatology and Venereology, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
35
|
Mihály J, Gericke J, Törőcsik D, Gáspár K, Szegedi A, Rühl R. Reduced lipoxygenase and cyclooxygenase mediated signaling in PBMC of atopic dermatitis patients. Prostaglandins Other Lipid Mediat 2013; 107:35-42. [DOI: 10.1016/j.prostaglandins.2013.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 10/26/2022]
|
36
|
Gaitanis G, Velegraki A, Mayser P, Bassukas ID. Skin diseases associated with Malassezia yeasts: facts and controversies. Clin Dermatol 2013; 31:455-463. [PMID: 23806162 DOI: 10.1016/j.clindermatol.2013.01.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The implication of the yeast genus Malassezia in skin diseases has been characterized by controversy, since the first description of the fungal nature of pityriasis versicolor in 1846 by Eichstedt. This is underscored by the existence of Malassezia yeasts as commensal but also by their implication in diseases with distinct absence of inflammation despite the heavy fungal load (pityriasis versicolor) or with characteristic inflammation (eg, seborrheic dermatitis, atopic dermatitis, folliculitis, or psoriasis). The description of 14 Malassezia species and subsequent worldwide epidemiologic studies did not reveal pathogenic species but rather disease-associated subtypes within species. Emerging evidence demonstrates that the interaction of Malassezia yeasts with the skin is multifaceted and entails constituents of the fungal wall (melanin, lipid cover), enzymes (lipases, phospholipases), and metabolic products (indoles), as well as the cellular components of the epidermis (keratinocytes, dendritic cells, and melanocytes). Understanding the complexity of their interactions will highlight the controversies on the clinical presentation of Malassezia-associated diseases and unravel the complexity of skin homeostatic mechanisms.
Collapse
Affiliation(s)
- Georgios Gaitanis
- Department of Skin and Venereal Diseases, University of Ioannina Medical School, Ioannina, Greece.
| | - Aristea Velegraki
- Mycology Laboratory, Microbiology Department, Medical School, National and Kapodistrian University of Athens, Athens Greece
| | - Peter Mayser
- Department of Dermatology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Ioannis D Bassukas
- Department of Skin and Venereal Diseases, University of Ioannina Medical School, Ioannina, Greece
| |
Collapse
|
37
|
Chisaguano AM, Montes R, Pérez-Berezo T, Castellote AI, Guerendiain M, Bustamante M, Morales E, García-Esteban R, Sunyer J, Franch À, López-Sabater MC. Gene expression of desaturase (FADS1 and FADS2) and Elongase (ELOVL5) enzymes in peripheral blood: association with polyunsaturated fatty acid levels and atopic eczema in 4-year-old children. PLoS One 2013; 8:e78245. [PMID: 24167612 PMCID: PMC3805510 DOI: 10.1371/journal.pone.0078245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 09/10/2013] [Indexed: 12/16/2022] Open
Abstract
Background It is unknown if changes in the gene expression of the desaturase and elongase enzymes are associated with abnormal n-6 long chain polyunsaturated fatty acid (LC-PUFA) levels in children with atopic eczema (AE). We analyzed whether mRNA-expression of genes encoding key enzymes of LC-PUFA synthesis (FADS1, FADS2 and ELOVL5) is associated with circulating LC-PUFA levels and risk of AE in 4-year-old children. Methods AE (n=20) and non-AE (n=104) children participating in the Sabadell cohort within the INfancia y Medio Ambiente (INMA) Project were included in the present study. RT-PCR with TaqMan Low-Density Array cards was used to measure the mRNA-expression of FADS1, FADS2 and ELOVL5. LC-PUFA levels were measured by fast gas chromatography in plasma phospholipids. The relationship of gene expression with LC-PUFA levels and enzyme activities was evaluated by Pearson’s rank correlation coefficient, and logistic regression models were used to study its association with risk of developing AE. Results Children with AE had lower levels of several n-6 PUFA members, dihomo-γ-linolenic (DGLA) and arachidonic (AA) acids. mRNA-expression levels of FADS1 and 2 strongly correlated with DGLA levels and with D6D activity. FADS2 and ELOVL5 mRNA-expression levels were significantly lower in AE than in non-AE children (-40.30% and -20.36%; respectively), but no differences were found for FADS1. Conclusions and Significance Changes in the mRNA-expression levels of FADS1 and 2 directly affect blood DGLA levels and D6D activity. This study suggests that lower mRNA-expressions of FADS2 and ELOVL5 are associated with higher risk of atopic eczema in young children.
Collapse
Affiliation(s)
- Aida Maribel Chisaguano
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
| | - Rosa Montes
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
| | - Teresa Pérez-Berezo
- Departament of Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Ana Isabel Castellote
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
| | - Marcela Guerendiain
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Mariona Bustamante
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
- Center for Genomic Regulation (CRG), Barcelona, Spain
| | - Eva Morales
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
| | - Raquel García-Esteban
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
| | - Jordi Sunyer
- Center for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
- Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain
| | - Àngels Franch
- Departament of Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - M. Carmen López-Sabater
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Spanish Government, Madrid, Spain
- * E-mail:
| |
Collapse
|
38
|
Dwivedi M, Laddha NC, Begum R. Correlation of increased MYG1 expression and its promoter polymorphism with disease progression and higher susceptibility in vitiligo patients. J Dermatol Sci 2013; 71:195-202. [PMID: 23706493 DOI: 10.1016/j.jdermsci.2013.04.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 03/14/2013] [Accepted: 04/19/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND MYG1 (Melanocyte proliferating gene 1 or C12orf10) -119C/G promoter and Arg4Gln structural polymorphisms have a functional impact on its regulation. The promoter polymorphism was shown to be associated with vitiligo in Caucasian population. OBJECTIVE The present study explores MYG1 polymorphisms and correlates them with MYG1 mRNA expression, disease onset and progression in vitiligo patients. METHODS Polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique was used for genotyping of MYG1 -119C/G promoter (rs1465073) and 11-12AA/GC structural polymorphisms (rs1534284-rs1534283; Arg4Gln) in 846 vitiligo patients and 726 age-matched unaffected controls. MYG1 mRNA levels were assessed in whole blood of 166 patients and 175 controls by Real-time PCR. RESULTS The MYG1 -119C/G promoter polymorphism was found to be in significant association with vitiligo being 'G' allele prevalent in patients. However, 11-12AA/GC structural polymorphism was prevalently monogenic in patients and controls with only MYG1 GC (4Arg) allele being present. Significant increase in MYG1 mRNA expression was observed in vitiligo patients compared to controls. The MYG1 mRNA expression was increased in patients with active and generalized vitiligo as compared to stable and localized vitiligo. MYG1 mRNA expression was increased in patients with susceptible -119 GG genotype compared to controls. Also, patients with susceptible -119 GG genotype had early age of onset of vitiligo. Moreover, patients with age groups 1-20 years and 21-40 years showed increased expression of MYG1 mRNA compared to those of controls. Female patients showed significant increase in MYG1 mRNA and early age of onset of vitiligo compared to male patients. CONCLUSION The present study suggests that MYG1 -119C/G promoter polymorphism may be a genetic risk factor for susceptibility and progression of vitiligo. The up-regulation of MYG1 transcript in patients with susceptible -119GG genotype advocates the crucial role of MYG1 in autoimmune pathogenesis of vitiligo.
Collapse
Affiliation(s)
- Mitesh Dwivedi
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390002, India
| | | | | |
Collapse
|
39
|
Fujii M, Nakashima H, Tomozawa J, Shimazaki Y, Ohyanagi C, Kawaguchi N, Ohya S, Kohno S, Nabe T. Deficiency of n-6 polyunsaturated fatty acids is mainly responsible for atopic dermatitis-like pruritic skin inflammation in special diet-fed hairless mice. Exp Dermatol 2013; 22:272-7. [DOI: 10.1111/exd.12120] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2013] [Indexed: 12/24/2022]
Affiliation(s)
- Masanori Fujii
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Hiroyuki Nakashima
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Junko Tomozawa
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Yuki Shimazaki
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Chie Ohyanagi
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Naomi Kawaguchi
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Susumu Ohya
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Shigekatsu Kohno
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| | - Takeshi Nabe
- Department of Pharmacology; Division of Pathological Sciences; Kyoto Pharmaceutical University; Kyoto; Japan
| |
Collapse
|
40
|
Liedén A, Winge MCG, Sääf A, Kockum I, Ekelund E, Rodriguez E, Fölster-Holst R, Franke A, Illig T, Tengvall-Linder M, Baurecht H, Weidinger S, Wahlgren CF, Nordenskjöld M, Bradley M. Genetic variation in the epidermal transglutaminase genes is not associated with atopic dermatitis. PLoS One 2012. [PMID: 23189155 PMCID: PMC3506648 DOI: 10.1371/journal.pone.0049694] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Atopic dermatitis (AD) is a common chronic inflammatory skin disorder where epidermal barrier dysfunction is a major factor in the pathogenesis. The identification of AD susceptibility genes related to barrier dysfunction is therefore of importance. The epidermal transglutaminases (TGM1, TGM3 and TGM5) encodes essential cross-linking enzymes in the epidermis. Objective To determine whether genetic variability in the epidermal transglutaminases contributes to AD susceptibility. Methods Forty-seven single nucleotide polymorphisms (SNPs) in the TGM1, TGM3 and TGM5 gene region were tested for genetic association with AD, independently and in relation to FLG genotype, using a pedigree disequilibrium test (PDT) in a Swedish material consisting of 1753 individuals from 539 families. In addition, a German case-control material, consisting of 533 AD cases and 1996 controls, was used for in silico analysis of the epidermal TGM regions. Gene expression of the TGM1, TGM3 and TGM5 gene was investigated by relative quantification with Real Time PCR (qRT-PCR). Immunohistochemical (IHC) analysis was performed to detect TG1, TG3 and TG5 protein expression in the skin of patients and healthy controls. Results PDT analysis identified a significant association between the TGM1 SNP rs941505 and AD with allergen-specific IgE in the Swedish AD family material. However, the association was not replicated in the German case-control material. No significant association was detected for analyzed SNPs in relation to FLG genotype. TG1, TG3 and TG5 protein expression was detected in AD skin and a significantly increased TGM3 mRNA expression was observed in lesional skin by qRT-PCR. Conclusion Although TGM1 and TGM3 may be differentially expressed in AD skin, the results from the genetic analysis suggest that genetic variation in the epidermal transglutaminases is not an important factor in AD susceptibility.
Collapse
Affiliation(s)
- Agne Liedén
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Mårten C. G. Winge
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Dermatology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
- * E-mail:
| | - Annika Sääf
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Elisabeth Ekelund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Elke Rodriguez
- Department of Dermatology, University of Kiel, Kiel, Germany
| | | | - Andre Franke
- Institute of Clinical Molecular Biology, Christian- Albrechts-Universität zu Kiel, Kiel, Germany
| | - Thomas Illig
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Maria Tengvall-Linder
- Clinical Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Carl-Fredrik Wahlgren
- Dermatology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Nordenskjöld
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Maria Bradley
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Dermatology Unit, Department of Medicine Solna, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
41
|
Roth W, Kumar V, Beer HD, Richter M, Wohlenberg C, Reuter U, Thiering S, Staratschek-Jox A, Hofmann A, Kreusch F, Schultze JL, Vogl T, Roth J, Reichelt J, Hausser I, Magin TM. Keratin 1 maintains skin integrity and participates in an inflammatory network in skin through interleukin-18. J Cell Sci 2012; 125:5269-79. [PMID: 23132931 DOI: 10.1242/jcs.116574] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Keratin 1 (KRT1) and its heterodimer partner keratin 10 (KRT10) are major constituents of the intermediate filament cytoskeleton in suprabasal epidermis. KRT1 mutations cause epidermolytic ichthyosis in humans, characterized by loss of barrier integrity and recurrent erythema. In search of the largely unknown pathomechanisms and the role of keratins in barrier formation and inflammation control, we show here that Krt1 is crucial for maintenance of skin integrity and participates in an inflammatory network in murine keratinocytes. Absence of Krt1 caused a prenatal increase in interleukin-18 (IL-18) and the S100A8 and S100A9 proteins, accompanied by a barrier defect and perinatal lethality. Depletion of IL-18 partially rescued Krt1(-/-) mice. IL-18 release was keratinocyte-autonomous, KRT1 and caspase-1 dependent, supporting an upstream role of KRT1 in the pathology. Finally, transcriptome profiling revealed a Krt1-mediated gene expression signature similar to atopic eczema and psoriasis, but different from Krt5 deficiency and epidermolysis bullosa simplex. Our data suggest a functional link between KRT1 and human inflammatory skin diseases.
Collapse
Affiliation(s)
- Wera Roth
- Translational Centre for Regenerative Medicine and Institute of Biology, University of Leipzig, 04103 Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mills KJ, Hu P, Henry J, Tamura M, Tiesman JP, Xu J. Dandruff/seborrhoeic dermatitis is characterized by an inflammatory genomic signature and possible immune dysfunction: transcriptional analysis of the condition and treatment effects of zinc pyrithione. Br J Dermatol 2012; 166 Suppl 2:33-40. [PMID: 22670617 DOI: 10.1111/j.1365-2133.2012.10863.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Dandruff/seborrhoeic dermatitis is a common scalp condition that is characterized by flakes, pruritus and sometimes mild erythema. These symptoms reflect tissue level events that are poorly understood at the molecular level. OBJECTIVES The purpose of this work was: (i) to compare gene expression profiles in subjects with dandruff vs. those of subjects without dandruff to determine the key physiological disruptions manifest in the condition; and (ii) to determine the effect on this profile of treatment with a shampoo containing potentiated zinc pyrithione (ZPT). METHODS In study 1, scalp biopsies were taken from 16 normal subjects and from involved and uninvolved sites in 15 subjects with dandruff. In study 2, 30 subjects with dandruff were treated for 3 weeks with a commercial ZPT shampoo (n = 15) or a vehicle (n = 15), and scalp lesional biopsies were collected at baseline and end of study for transcriptomic analysis. RNA was extracted from all biopsies and Affymetrix gene chips were used to analyse transcriptomic profiles, followed by bioinformatic analysis. RESULTS Analysis of study 1 biopsies revealed more than 7000 individual probes differentially regulated in dandruff lesional skin relative to normal. Enriched Gene Ontology categories included: lipid metabolism, immune response, response to stimulus, apoptosis, cell proliferation, and epidermal development. The most striking feature of lesional skin relative to normal was the reciprocal expression of induced inflammatory genes and repressed lipid metabolism genes. Induced inflammatory genes were also enriched in dandruff uninvolved skin, suggesting the existence of predisposing factors associated with inflammation. Many genes increased in lesional skin were increased at the level of protein in stratum corneum samples (e.g. IL-1RA, S100A8, S100A9, S100A11, IL-8). Under conditions known to improve overall scalp condition, the ZPT shampoo treatment in study 2 produced a transcriptomic profile resembling that of normal scalp skin. CONCLUSIONS These data provide novel insights into the nature of dandruff and the therapeutic action of potentiated ZPT-containing shampoo, and provide a basis to explore many new mechanistic questions related to these topics.
Collapse
Affiliation(s)
- K J Mills
- The Procter and Gamble Company, Miami Valley Innovation Center, 11810 East Miami River Road, Cincinnati, OH 45252, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Characterization of EGFR and ErbB2 expression in atopic dermatitis patients. Arch Dermatol Res 2012; 304:773-80. [DOI: 10.1007/s00403-012-1242-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 04/04/2012] [Accepted: 04/18/2012] [Indexed: 11/26/2022]
|
44
|
Abstract
In the last 15 years, the genus Malassezia has been a topic of intense basic research on taxonomy, physiology, biochemistry, ecology, immunology, and metabolomics. Currently, the genus encompasses 14 species. The 1996 revision of the genus resulted in seven accepted taxa: M. furfur, M. pachydermatis, M. sympodialis, M. globosa, M. obtusa, M. restricta, and M. slooffiae. In the last decade, seven new taxa isolated from healthy and lesional human and animal skin have been accepted: M. dermatis, M. japonica, M. yamatoensis, M. nana, M. caprae, M. equina, and M. cuniculi. However, forthcoming multidisciplinary research is expected to show the etiopathological relationships between these new species and skin diseases. Hitherto, basic and clinical research has established etiological links between Malassezia yeasts, pityriasis versicolor, and sepsis of neonates and immunocompromised individuals. Their role in aggravating seborrheic dermatitis, dandruff, folliculitis, and onychomycosis, though often supported by histopathological evidence and favorable antifungal therapeutic outcomes, remains under investigation. A close association between skin and Malassezia IgE binding allergens in atopic eczema has been shown, while laboratory data support a role in psoriasis exacerbations. Finally, metabolomic research resulted in the proposal of a hypothesis on the contribution of Malassezia-synthesized aryl hydrocarbon receptor (AhR) ligands to basal cell carcinoma through UV radiation-induced carcinogenesis.
Collapse
|
45
|
Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, Yao Y, Fiorentino D. Interferon and biologic signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PLoS One 2012; 7:e29161. [PMID: 22235269 PMCID: PMC3250414 DOI: 10.1371/journal.pone.0029161] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 11/22/2011] [Indexed: 01/08/2023] Open
Abstract
Background Dermatomyositis (DM) is an autoimmune disease that mainly affects the skin, muscle, and lung. The pathogenesis of skin inflammation in DM is not well understood. Methodology and Findings We analyzed genome-wide expression data in DM skin and compared them to those from healthy controls. We observed a robust upregulation of interferon (IFN)-inducible genes in DM skin, as well as several other gene modules pertaining to inflammation, complement activation, and epidermal activation and differentiation. The interferon (IFN)-inducible genes within the DM signature were present not only in DM and lupus, but also cutaneous herpes simplex-2 infection and to a lesser degree, psoriasis. This IFN signature was absent or weakly present in atopic dermatitis, allergic contact dermatitis, acne vulgaris, systemic sclerosis, and localized scleroderma/morphea. We observed that the IFN signature in DM skin appears to be more closely related to type I than type II IFN based on in vitro IFN stimulation expression signatures. However, quantitation of IFN mRNAs in DM skin shows that the majority of known type I IFNs, as well as IFN g, are overexpressed in DM skin. In addition, both IFN-beta and IFN-gamma (but not other type I IFN) transcript levels were highly correlated with the degree of the in vivo IFN transcriptional response in DM skin. Conclusions and Significance As in the blood and muscle, DM skin is characterized by an overwhelming presence of an IFN signature, although it is difficult to conclusively define this response as type I or type II. Understanding the significance of the IFN signature in this wide array of inflammatory diseases will be furthered by identification of the nature of the cells that both produce and respond to IFN, as well as which IFN subtype is biologically active in each diseased tissue.
Collapse
Affiliation(s)
- David Wong
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Bory Kea
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Rob Pesich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Brandon W. Higgs
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - Wei Zhu
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - Patrick Brown
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yihong Yao
- MedImmune, Translational Sciences, One MedImmune Way, Gaithersburg, Maryland, United States of America
| | - David Fiorentino
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
46
|
Filaggrin genotype determines functional and molecular alterations in skin of patients with atopic dermatitis and ichthyosis vulgaris. PLoS One 2011; 6:e28254. [PMID: 22164253 PMCID: PMC3229525 DOI: 10.1371/journal.pone.0028254] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 11/04/2011] [Indexed: 12/16/2022] Open
Abstract
Background Several common genetic and environmental disease mechanisms are important for the pathophysiology behind atopic dermatitis (AD). Filaggrin (FLG) loss-of-function is of great significance for barrier impairment in AD and ichthyosis vulgaris (IV), which is commonly associated with AD. The molecular background is, however, complex and various clusters of genes are altered, including inflammatory and epidermal-differentiation genes. Objective The objective was to study whether the functional and molecular alterations in AD and IV skin depend directly on FLG loss-of-function, and whether FLG genotype determines the type of downstream molecular pathway affected. Methods and Findings Patients with AD/IV (n = 43) and controls (n = 15) were recruited from two Swedish outpatient clinics and a Swedish AD family material with known FLG genotype. They were clinically examined and their medical history recorded using a standardized questionnaire. Blood samples and punch biopsies were taken and trans-epidermal water loss (TEWL) and skin pH was assessed with standard techniques. In addition to FLG genotyping, the STS gene was analyzed to exclude X-linked recessive ichthyosis (XLI). Microarrays and quantitative real-time PCR were used to compare differences in gene expression depending on FLG genotype. Several different signalling pathways were altered depending on FLG genotype in patients suffering from AD or AD/IV. Disease severity, TEWL and pH follow FLG deficiency in the skin; and the number of altered genes and pathways are correlated to FLG mRNA expression. Conclusions We emphasize further the role of FLG in skin-barrier integrity and the complex compensatory activation of signalling pathways. This involves inflammation, epidermal differentiation, lipid metabolism, cell signalling and adhesion in response to FLG-dependent skin-barrier dysfunction.
Collapse
|
47
|
Coda AB, Sinha AA. Integration of genome-wide transcriptional and genetic profiles provides insights into disease development and clinical heterogeneity in Alopecia areata. Genomics 2011; 98:431-9. [DOI: 10.1016/j.ygeno.2011.08.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 08/22/2011] [Accepted: 08/31/2011] [Indexed: 12/22/2022]
|
48
|
Abstract
Filaggrin variants are well-established risk factors for atopic eczema (AE). Recent studies suggest additional epidermal differentiation complex (EDC) gene associations with AE. In this issue, Marenholz and colleagues confirm this prediction and show that a small proline-rich protein 3 (SPRR3) variant confers susceptibility to AE. This finding suggests that further genetic and functional characterization of SPRR3 should be performed in patients with AE.
Collapse
Affiliation(s)
- David P Kelsell
- Centre for Cutaneous Research, The Blizard Institute, Barts & The London School of Medicine and Dentistry, Queen Mary, University of London, London, UK.
| | | |
Collapse
|
49
|
Bchetnia M, Tremblay ML, Leclerc G, Dupérée A, Powell J, McCuaig C, Morin C, Legendre-Guillemin V, Laprise C. Expression signature of epidermolysis bullosa simplex. Hum Genet 2011; 131:393-406. [DOI: 10.1007/s00439-011-1077-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 07/29/2011] [Indexed: 01/24/2023]
|
50
|
Al-Shobaili HA. Update on the genetics characterization of vitiligo. Int J Health Sci (Qassim) 2011; 5:167-79. [PMID: 23267294 PMCID: PMC3521835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
Vitiligo is an autoimmune skin disorder in which autoimmune-mediated destruction of melanocytes caused depigmentation of skin patches. The complex genetics of vitiligo involves multiple susceptibility loci, genetic heterogeneity and incomplete penetrance with gene-gene and gene-environment interactions. In order to clarify the genetic factors, two different principal approaches have applied for the identification of genomic regions or candidate genes that mediate susceptibility to vitiligo. First approach is the genome-wide linkage analyses, which is conducted by scanning of entire human genome for genomic regions that are linked to the development of vitiligo. The other approach is functional candidate gene association (FCGA) analyses that detect specific candidate genes, which are expected to involve in disease on the basis of their priori biological functions. Genomic-wide scans have provided a strong support for vitiligo susceptibility genes on chromosomes 4q13-q21, 1p31, 7q22, 8p12 and 17p13, while loci of interest at 6p, 6q, 14q, 9q, 13q, 19p and 22q required further follow-up. Whereas, FCGA studies have identified some candidate genes which are associated with vitiligo, such as HLA, AIRE, VIT1, CAT, FOXD3, ESR1, COMT, PTPN22, NALP1, PDGFRA, MYG1, MITF, CD117, XBP1, FAS, COX2, EDN1 and ACE, but few of them reports now appear to be false-positive. This review will provides an update on genetics of vitiligo based on the identification of novel candidate genes that represent, in my opinion as optimal utility for future therapeutic targets in the pathogenesis of vitiligo.
Collapse
Affiliation(s)
- Hani A. Al-Shobaili
- Department of Dermatology, College of Medicine, Qassim University, Saudi Arabia
| |
Collapse
|