1
|
Wang L, Zhu Y, Zhang N, Xian Y, Tang Y, Ye J, Reza F, He G, Wen X, Jiang X. The multiple roles of interferon regulatory factor family in health and disease. Signal Transduct Target Ther 2024; 9:282. [PMID: 39384770 PMCID: PMC11486635 DOI: 10.1038/s41392-024-01980-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024] Open
Abstract
Interferon Regulatory Factors (IRFs), a family of transcription factors, profoundly influence the immune system, impacting both physiological and pathological processes. This review explores the diverse functions of nine mammalian IRF members, each featuring conserved domains essential for interactions with other transcription factors and cofactors. These interactions allow IRFs to modulate a broad spectrum of physiological processes, encompassing host defense, immune response, and cell development. Conversely, their pivotal role in immune regulation implicates them in the pathophysiology of various diseases, such as infectious diseases, autoimmune disorders, metabolic diseases, and cancers. In this context, IRFs display a dichotomous nature, functioning as both tumor suppressors and promoters, contingent upon the specific disease milieu. Post-translational modifications of IRFs, including phosphorylation and ubiquitination, play a crucial role in modulating their function, stability, and activation. As prospective biomarkers and therapeutic targets, IRFs present promising opportunities for disease intervention. Further research is needed to elucidate the precise mechanisms governing IRF regulation, potentially pioneering innovative therapeutic strategies, particularly in cancer treatment, where the equilibrium of IRF activities is of paramount importance.
Collapse
Affiliation(s)
- Lian Wang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanghui Zhu
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yali Xian
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Tang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Ye
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fekrazad Reza
- Radiation Sciences Research Center, Laser Research Center in Medical Sciences, AJA University of Medical Sciences, Tehran, Iran
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang Wen
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
James AE, Abdalgani M, Khoury P, Freeman AF, Milner JD. T H2-driven manifestations of inborn errors of immunity. J Allergy Clin Immunol 2024; 154:245-254. [PMID: 38761995 DOI: 10.1016/j.jaci.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024]
Abstract
Monogenic lesions in pathways critical for effector functions responsible for immune surveillance, protection against autoinflammation, and appropriate responses to allergens and microorganisms underlie the pathophysiology of inborn errors of immunity (IEI). Variants in cytokine production, cytokine signaling, epithelial barrier function, antigen presentation, receptor signaling, and cellular processes and metabolism can drive autoimmunity, immunodeficiency, and/or allergic inflammation. Identification of these variants has improved our understanding of the role that many of these proteins play in skewing toward TH2-related allergic inflammation. Early-onset or atypical atopic disease, often in conjunction with immunodeficiency and/or autoimmunity, should raise suspicion for an IEI. This becomes a diagnostic dilemma if the initial clinical presentation is solely allergic inflammation, especially when the prevalence of allergic diseases is becoming more common. Genetic sequencing is necessary for IEI diagnosis and is helpful for early recognition and implementation of targeted treatment, if available. Although genetic evaluation is not feasible for all patients with atopy, identifying atopic patients with molecular immune abnormalities may be helpful for diagnostic, therapeutic, and prognostic purposes. In this review, we focus on IEI associated with TH2-driven allergic manifestations and classify them on the basis of the affected molecular pathways and predominant clinical manifestations.
Collapse
Affiliation(s)
- Alyssa E James
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Manar Abdalgani
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| | - Paneez Khoury
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Joshua D Milner
- Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, NY
| |
Collapse
|
3
|
Chaudhary F, Agrawal DK. Ethnic and Racial Disparities in Clinical Manifestations of Atopic Dermatitis. ARCHIVES OF INTERNAL MEDICINE RESEARCH 2024; 7:114-133. [PMID: 38938453 PMCID: PMC11210272 DOI: 10.26502/aimr.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Atopic dermatitis is a heterogenous inflammatory skin illness that may last for long time and affect people of different racial and ethnic backgrounds. The condition primarily appears in infants and young children. There are people living with atopic dermatitis in every country and every ethnic group, although the frequency of the disease varies greatly. Due to the varied clinical presentations that atopic dermatitis can have, it can be challenging to characterize and diagnose the disease, particularly in adults. Nevertheless, there exists a dearth of information pertaining to the various presentations of atopic dermatitis among individuals from diverse racial and cultural groups. This critical review article offers a succinct and comprehensive overview of the current findings on the epidemiology of atopic dermatitis with regards to ethnic and racial disparities. The findings hold potential significance in advancing the development of targeted treatments for personalized medicine approaches and enhancing the quality of life for patients with atopy.
Collapse
Affiliation(s)
- Fihr Chaudhary
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766
| |
Collapse
|
4
|
Wang Y, Chen X, Xu X, Yang J, Liu X, Sun G, Li Z. Weighted Gene Co-Expression Network Analysis Based on Stimulation by Lipopolysaccharides and Polyinosinic:polycytidylic Acid Provides a Core Set of Genes for Understanding Hemolymph Immune Response Mechanisms of Amphioctopus fangsiao. Animals (Basel) 2023; 14:80. [PMID: 38200810 PMCID: PMC10778463 DOI: 10.3390/ani14010080] [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: 07/10/2023] [Revised: 12/05/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The primary influencer of aquaculture quality in Amphioctopus fangsiao is pathogen infection. Both lipopolysaccharides (LPS) and polyinosinic:polycytidylic acid (Poly I:C) are recognized by the pattern recognition receptor (PRR) within immune cells, a system that frequently serves to emulate pathogen invasion. Hemolymph, which functions as a transport mechanism for immune cells, offers vital transcriptome information when A. fangsiao is exposed to pathogens, thereby contributing to our comprehension of the species' immune biological mechanisms. In this study, we conducted analyses of transcript profiles under the influence of LPS and Poly I:C within a 24 h period. Concurrently, we developed a Weighted Gene Co-expression Network Analysis (WGCNA) to identify key modules and genes. Further, we carried out Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses to investigate the primary modular functions. Co-expression network analyses unveiled a series of immune response processes following pathogen stress, identifying several key modules and hub genes, including PKMYT1 and NAMPT. The invaluable genetic resources provided by our results aid our understanding of the immune response in A. fangsiao hemolymph and will further our exploration of the molecular mechanisms of pathogen infection in mollusks.
Collapse
Affiliation(s)
- Yongjie Wang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xipan Chen
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xiaohui Xu
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Jianmin Yang
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai 264005, China;
| | - Guohua Sun
- School of Agriculture, Ludong University, Yantai 264025, China
| | - Zan Li
- School of Agriculture, Ludong University, Yantai 264025, China
| |
Collapse
|
5
|
Traidl S, Heinrich L, Siegels D, Rösner L, Haufe E, Harder I, Abraham S, Ertner K, Kleinheinz A, Schäkel K, Wollenberg A, Effendy I, Quist S, Asmussen A, Wildberger J, Weisshaar E, Wiemers F, Brücher JJ, Weidinger S, Schmitt J, Werfel T. High recurrence rate of eczema herpeticum in moderate/severe atopic dermatitis -TREATgermany registry analysis. J Dtsch Dermatol Ges 2023; 21:1490-1498. [PMID: 37814394 DOI: 10.1111/ddg.15205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 07/09/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Eczema herpeticum (EH) is a disseminated skin infection caused by herpes simplex virus in atopic dermatitis (AD) patients. The frequency of EH and the clinical features of EH patients have not yet been investigated in a larger cohort. METHODS We sought to investigate the TREATgermany cohort, a multicenter, non-interventional clinical registry of moderately to severely affected AD patients in Germany. Baseline characteristics of patients included between December 2017 and April 2021 were compared between patients without, single, and multiple EH. RESULTS Of the 893 patients, 195 (21.8%) had at least one EH. Of the 195 patients with EH, 107 had multiple EH (54.9%), representing 12.0% of the total study population. While there were no differences in demographic characteristics, previous treatment, and disease scores at enrollment (itch, IGA, oSCORAD, EASI), patients with EH had more frequent atopic comorbidities and sensitizations to house dust mite, food, and mold. DISCUSSION TREATgermany registry data suggest a high prevalence and recurrence rate of EH, while there appears to be no specific clinical phenotype, besides an increase in allergies, to identify EH patients in the daily routine.
Collapse
Affiliation(s)
- Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Luise Heinrich
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Doreen Siegels
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lennart Rösner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| | - Eva Haufe
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Inken Harder
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Susanne Abraham
- Department of Dermatology, University Allergy Center, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | | | | | - Knut Schäkel
- Department of Dermatology, University Hospital, Heidelberg, Germany
| | - Andreas Wollenberg
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Ludwig Maximilian University, Munich, Germany
| | - Isaak Effendy
- Department of Dermatology, OWL University Hospital of Bielefeld University, Campus Clinic Bielefeld, Bielefeld, Germany
| | - Sven Quist
- Dermatology Clinic, Helix Medical Ecellence Center Mainz, Mainz, Germany
| | - Andrea Asmussen
- Practice Dr. med. Andrea Asmussen, Dermatology at Lesum, Bremen, Germany
| | - Julia Wildberger
- Practice Dr. med. Julia Wildberger Hautmedizin, Bad Soden, Germany
| | - Elke Weisshaar
- Division of Occupational Dermatology, Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | | | - Jens-Joachim Brücher
- Practice Dr. med. Jens-Joachim Brücher, Hautambulatorium Magdeburg, Magdeburg, Germany
| | - Stephan Weidinger
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Jochen Schmitt
- Center for Evidence-Based Healthcare, University Hospital Carl Gustav Carus and Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
- Hannover Medical School, Cluster of Excellence RESIST (EXC 2155), Hannover, Germany
| |
Collapse
|
6
|
Traidl S, Heinrich L, Siegels D, Rösner L, Haufe E, Harder I, Abraham S, Ertner K, Kleinheinz A, Schäkel K, Wollenberg A, Effendy I, Quist S, Asmussen A, Wildberger J, Weisshaar E, Wiemers F, Brücher JJ, Weidinger S, Schmitt J, Werfel T. Hohe Rezidivrate des Eczema herpeticatum bei mittelschwerer bis schwerer atopischer Dermatitis - eine TREATgermany Registeranalyse. J Dtsch Dermatol Ges 2023; 21:1490-1499. [PMID: 38082531 DOI: 10.1111/ddg.15205_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 07/09/2023] [Indexed: 12/18/2023]
Abstract
ZusammenfassungHintergrundDas Eczema herpeticatum (EH) ist eine disseminierte Hautinfektion, die durch Herpes‐simplex‐Viren bei Patienten mit atopischer Dermatitis (AD) verursacht wird. Die Häufigkeit des EH und die klinischen Charakteristika von EH Patienten wurden bisher noch nicht in einer größeren Kohorte untersucht.Methodik87 Patienten des TREATgermany Registers, einem multizentrischen, nichtinterventionellen klinischen Register mit moderat bis schwer betroffenen AD‐Patienten in Deutschland, wurden in dieser Analyse betrachtet. Patienten, die zwischen Dezember 2017 und April 2021 in das Register eingeschlossen wurden, wurden unterteilt in die Gruppen ohne, mit einem und mit mehreren EH und basierend auf den klinischen Charakteristika verglichen.ErgebnisseVon 893 Patienten berichteten 195 (21,8%) über mindestens eine EH. 107 der 195 Patienten mit EH hatten sogar mehrere EH in der Anamnese (54,9%), was 12,0% der gesamten Studienpopulation entspricht. Während hinsichtlich demographischer Merkmale, Vorbehandlungen und Krankheitsscores (Juckreiz, IGA, oSCORAD, EASI) keine Unterschiede festgestellt wurden, litten Patienten mit EH häufiger an atopischen Begleiterkrankungen und Sensibilisierungen gegen Hausstaubmilben, Nahrungsmittel und Schimmelpilze.SchlussfolgerungenDie Daten des TREATgermany‐Registers deuten auf eine hohe Prävalenz und Rezidivrate des EH hin, während es neben einer Häufung von Allergien keinen spezifischen klinischen Phänotyp zu geben scheint, um EH‐Patienten in der täglichen Routine zu identifizieren.
Collapse
Affiliation(s)
- Stephan Traidl
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| | - Luise Heinrich
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Doreen Siegels
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Lennart Rösner
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| | - Eva Haufe
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Inken Harder
- Zentrum für entzündliche Hauterkrankungen, Klinik für Dermatologie und Allergologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - Susanne Abraham
- Klinik für Dermatologie, Universitäts-Allergie-Centrum, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | | | | | | | - Andreas Wollenberg
- Department of Dermatology and Allergy, University Hospital Augsburg, Augsburg, Ludwig Maximilian University, Munich, Germany
| | - Isaak Effendy
- Hautklinik, Universitätsklinikum OWL der Universität Bielefeld, Campus Klinikum Bielefeld
| | - Sven Quist
- Hautklinik, Helix Medical Ecellence Center Mainz
| | - Andrea Asmussen
- Praxis Dr. med. Andrea Asmussen, Hautarztpraxis an der Lesum, Bremen
| | | | - Elke Weisshaar
- Abteilung für Berufsdermatologie, Hautklinik, Ruprecht-Karls-Universität Heidelberg
| | | | | | - Stephan Weidinger
- Zentrum für entzündliche Hauterkrankungen, Klinik für Dermatologie und Allergologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | - Jochen Schmitt
- Zentrum für Evidenzbasierte Medizin, Universitätsklinikum Carl Gustav Carus und Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden
| | - Thomas Werfel
- Klinik für Dermatologie und Allergologie, Medizinische Hochschule Hannover, Hannover, Germany
- Medizinische Hochschule Hannover, Exzellenzcluster RESIST (EXC 2155), Hannover
| |
Collapse
|
7
|
Traidl S, Harries L, Kienlin P, Begemann G, Roesner LM, Werfel T. Dupilumab strengthens herpes simplex virus type 1-specific immune responses in atopic dermatitis. J Allergy Clin Immunol 2023; 152:1460-1469.e5. [PMID: 37660986 DOI: 10.1016/j.jaci.2023.08.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/22/2023] [Accepted: 08/03/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND Impaired virus clearance in a subgroup of atopic dermatitis (AD) patients can lead to severe herpes simplex virus (HSV) infections called eczema herpeticum (EH). We recently identified a type 2 skewed viral immune response in EH patients. Clinical data suggest a reduced incidence of EH in AD patients treated with dupilumab, although immunologic investigations of this phenomenon are still lacking. OBJECTIVE We examined the impact of dupilumab on the HSV type 1 (HSV-1) specific immune response in AD, focusing on patients with (ADEH+) and without (ADEH-) a history of EH. METHODS Sera and peripheral blood mononuclear cells were collected from ADEH+ and ADEH- patients, a subgroup of whom was receiving dupilumab treatment, and healthy controls. Serum samples were tested for IgE against HSV-1 glycoprotein D (n = 85). Peripheral blood mononuclear cells were stimulated with HSV peptides, and activated CD4+ and CD8+ cells were characterized by flow cytometry after magnetic enrichment via CD154 or CD137 (n = 60). Cytokine production of HSV-1-reactive T-cell lines (n = 33) and MHC-I tetramer+ (HSV-1-UL25) CD8+ T cells was investigated by bead assay and intracellular cytokine staining (n = 21). RESULTS We confirmed that HSV-1-specific IgE is elevated in ADEH+ patients. During dupilumab treatment, the IgE levels were significantly decreased, reaching levels of healthy controls. HSV-1-specific TC1 frequencies were elevated in ADEH- patients treated with dupilumab compared to dupilumab-negative patients. There were no changes in the frequencies of HSV-1-specific TH cells while receiving dupilumab therapy. AD patients receiving dupilumab exhibited elevated IFN-γ and reduced IL-4 production in HSV-1-UL25-epitope-specific T cells compared to dupilumab-negative patients. CONCLUSION Dupilumab may improve the HSV-1-specific immune response in AD as a result of an increased type I immune response and a reduction of HSV-1-specific IgE.
Collapse
Affiliation(s)
- Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| | - Leonard Harries
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Petra Kienlin
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Gabriele Begemann
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Lennart M Roesner
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Bratu D, Boda D, Caruntu C. Genomic, Epigenomic, Transcriptomic, Proteomic and Metabolomic Approaches in Atopic Dermatitis. Curr Issues Mol Biol 2023; 45:5215-5231. [PMID: 37367080 PMCID: PMC10297041 DOI: 10.3390/cimb45060331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with a high prevalence in the developed countries. It is associated with atopic and non-atopic diseases, and its close correlation with atopic comorbidities has been genetically demonstrated. One of the main roles of genetic studies is to comprehend the defects of the cutaneous barrier due to filaggrin deficit and epidermal spongiosis. Recently, epigenetic studies started to analyze the influence of the environmental factors on gene expression. The epigenome is considered to be a superior second code that controls the genome, which includes alterations of the chromatin. The epigenetic changes do not alter the genetic code, however, changes in the chromatin structure could activate or inhibit the transcription process of certain genes and consequently, the translation process of the new mRNA into a polypeptide chain. In-depth analysis of the transcriptomic, metabolomic and proteomic studies allow to unravel detailed mechanisms that cause AD. The extracellular space and lipid metabolism are associated with AD that is independent of the filaggrin expression. On the other hand, around 45 proteins are considered as the principal components in the atopic skin. Moreover, genetic studies based on the disrupted cutaneous barrier can lead to the development of new treatments targeting the cutaneous barrier or cutaneous inflammation. Unfortunately, at present, there are no target therapies that focus on the epigenetic process of AD. However, in the future, miR-143 could be an important objective for new therapies, as it targets the miR-335:SOX axis, thereby restoring the miR-335 expression, and repairing the cutaneous barrier defects.
Collapse
Affiliation(s)
- Dalia Bratu
- Department of Dermatology, ‘Colentina’ Clinical Hospital, 020125 Bucharest, Romania;
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Daniel Boda
- Department of Dermatology, ‘Carol Davila’ University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Dermatology, ‘Ponderas’ Academic Hospital, 014142 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Constantin Caruntu
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
9
|
Chiricozzi A, Maurelli M, Calabrese L, Peris K, Girolomoni G. Overview of Atopic Dermatitis in Different Ethnic Groups. J Clin Med 2023; 12:2701. [PMID: 37048783 PMCID: PMC10095524 DOI: 10.3390/jcm12072701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/16/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease with a high prevalence worldwide, including countries from Asia, Africa, and Latin America, and in different ethnic groups. In recent years, more attention has been placed on the heterogeneity of AD associated with multiple factors, including a patient's ethnic background, resulting in an increasing body of clinical, genetic, epidemiologic, and immune-phenotypic evidence that delineates differences in AD among racial groups. Filaggrin (FLG) mutations, the strongest genetic risk factor for the development of AD, are detected in up to 50% of European and 27% of Asian AD patients, but very rarely in Africans. Th2 hyperactivation is a common attribute of all ethnic groups, though the Asian endotype of AD is also characterized by an increased Th17-mediated signal, whereas African Americans show a strong Th2/Th22 signature and an absence of Th1/Th17 skewing. In addition, the ethnic heterogeneity of AD may hold important therapeutic implications as a patient's genetic predisposition may affect treatment response and, thereby, a tailored strategy that better targets the dominant immunologic pathways in each ethnic subgroup may be envisaged. Nevertheless, white patients with AD represent the largest ethnicity enrolled and tested in clinical trials and the most treated in a real-world setting, limiting investigations about safety and efficacy across different ethnicities. The purpose of this review is to describe the heterogeneity in the pathophysiology of AD across ethnicities and its potential therapeutic implications.
Collapse
Affiliation(s)
- Andrea Chiricozzi
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Martina Maurelli
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, 37126 Verona, Italy
| | - Laura Calabrese
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Ketty Peris
- UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
- Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giampiero Girolomoni
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, 37126 Verona, Italy
| |
Collapse
|
10
|
Genetic/Protein Association of Atopic Dermatitis and Tooth Agenesis. Int J Mol Sci 2023; 24:ijms24065754. [PMID: 36982827 PMCID: PMC10055628 DOI: 10.3390/ijms24065754] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/07/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Atopic dermatitis and abnormalities in tooth development (including hypomineralization, hypodontia and microdontia) have been observed to co-occur in some patients. A common pathogenesis pathway that involves genes and protein interactions has been hypothesized. This review aims to first provide a description of the key gene mutations and signaling pathways associated with atopic dermatitis and tooth agenesis (i.e., the absence of teeth due to developmental failure) and identify the possible association between the two diseases. Second, utilizing a list of genes most commonly associated with the two diseases, we conducted a protein–protein network interaction analysis using the STRING database and identified a novel association between the Wnt/β-catenin signaling pathway (major pathway responsible for TA) and desmosomal proteins (component of skin barrier that affect the pathogenesis of AD). Further investigation into the mechanisms that may drive their co-occurrence and underlie the development of the two diseases is warranted.
Collapse
|
11
|
Broderick C, Ziehfreund S, van Bart K, Arents B, Eyerich K, Weidinger S, Rastrick J, Zink A, Flohr C. Biomarkers associated with the development of comorbidities in patients with atopic dermatitis: A systematic review. Allergy 2023; 78:84-120. [PMID: 36366871 PMCID: PMC10107168 DOI: 10.1111/all.15578] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/06/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Biomarkers associated with the development of comorbidities in atopic dermatitis (AD) patients have been reported, but have not yet been systematically reviewed. Seven electronic databases were searched, from database inception to September 2021. English language randomized controlled trials, prospective and retrospective cohort, and case-control studies that investigated the association between a biomarker and the development of comorbidities in AD patients were included. Two authors independently screened the records for eligibility, one extracted all data, and critically appraised the quality of studies and risk of bias. Fifty six articles met the inclusion criteria, evaluating 146 candidate biomarkers. The most frequently reported biomarkers were filaggrin mutations and allergen specific-IgE. Promising biomarkers include specific-IgE and/or skin prick tests predicting the development of asthma, and genetic polymorphisms predicting the occurrence of eczema herpeticum. The identified studies and biomarkers were highly heterogeneous, and associated with predominately moderate-to-high risk of bias across multiple domains. Overall, findings were inconsistent. High-quality studies assessing biomarkers associated with the development of comorbidities in people with AD are lacking. Harmonized datasets and independent validation studies are urgently needed.
Collapse
Affiliation(s)
- Conor Broderick
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | - Stefanie Ziehfreund
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Karin van Bart
- Royal College of Physicians, National Guideline Centre, London, UK
| | - Bernd Arents
- Dutch Association for People with Atopic Dermatitis, Nijkerk, The Netherlands
| | - Kilian Eyerich
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine, Technical University of Munich, Munich, Germany.,Division of Dermatology and Venerology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Flohr
- Unit for Population-Based Dermatology Research, School of Basic and Medical Biosciences, St John's Institute of Dermatology, King's College London, London, UK
| | | |
Collapse
|
12
|
Lukhele S, Rabbo DA, Guo M, Shen J, Elsaesser HJ, Quevedo R, Carew M, Gadalla R, Snell LM, Mahesh L, Ciudad MT, Snow BE, You-Ten A, Haight J, Wakeham A, Ohashi PS, Mak TW, Cui W, McGaha TL, Brooks DG. The transcription factor IRF2 drives interferon-mediated CD8 + T cell exhaustion to restrict anti-tumor immunity. Immunity 2022; 55:2369-2385.e10. [PMID: 36370712 PMCID: PMC9809269 DOI: 10.1016/j.immuni.2022.10.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 08/10/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
Type I and II interferons (IFNs) stimulate pro-inflammatory programs that are critical for immune activation, but also induce immune-suppressive feedback circuits that impede control of cancer growth. Here, we sought to determine how these opposing programs are differentially induced. We demonstrated that the transcription factor interferon regulatory factor 2 (IRF2) was expressed by many immune cells in the tumor in response to sustained IFN signaling. CD8+ T cell-specific deletion of IRF2 prevented acquisition of the T cell exhaustion program within the tumor and instead enabled sustained effector functions that promoted long-term tumor control and increased responsiveness to immune checkpoint and adoptive cell therapies. The long-term tumor control by IRF2-deficient CD8+ T cells required continuous integration of both IFN-I and IFN-II signals. Thus, IRF2 is a foundational feedback molecule that redirects IFN signals to suppress T cell responses and represents a potential target to enhance cancer control.
Collapse
Affiliation(s)
- Sabelo Lukhele
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada.
| | - Diala Abd Rabbo
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Mengdi Guo
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - Jian Shen
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53226, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Heidi J Elsaesser
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Rene Quevedo
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Madeleine Carew
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Ramy Gadalla
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Laura M Snell
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lawanya Mahesh
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - M Teresa Ciudad
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Bryan E Snow
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Annick You-Ten
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Jillian Haight
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Andrew Wakeham
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada
| | - Pamela S Ohashi
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - Tak W Mak
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - Weiguo Cui
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53226, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tracy L McGaha
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8 Canada
| | - David G Brooks
- Princess Margaret Cancer Center, University Health Network, Toronto, ON, M5G 2M9 Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8 Canada.
| |
Collapse
|
13
|
Chong AC, Visitsunthorn K, Ong PY. Genetic/Environmental Contributions and Immune Dysregulation in Children with Atopic Dermatitis. J Asthma Allergy 2022; 15:1681-1700. [PMID: 36447957 PMCID: PMC9701514 DOI: 10.2147/jaa.s293900] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/11/2022] [Indexed: 08/01/2023] Open
Abstract
Atopic dermatitis (AD) is one of the most common skin conditions in humans. AD affects up to 20% of children worldwide and results in morbidity for both patients and their caregivers. The basis of AD is an interplay between genetics and the environment characterized by immune dysregulation. A myriad of mutations that compromise the skin barrier and/or immune function have been linked to AD. Of these, filaggrin gene (FLG) mutations are the most evidenced. Many other mutations have been implicated in isolated studies that are often unreplicated, creating an archive of genes with potential but unconfirmed relevance to AD. Harnessing big data, polygenic risk scores (PRSs) and genome-wide association studies (GWAS) may provide a more practical strategy for identifying the genetic signatures of AD. Epigenetics may also play a role. Staphylococcus aureus is the most evidenced microbial contributor to AD. Cutaneous dysbiosis may result in over-colonization by pathogenic strains and aberrant skin immunity and inflammation. Aeroallergens, air pollution, and climate are other key environmental contributors to AD. The right climate and/or commensals may improve AD for some patients.
Collapse
Affiliation(s)
- Albert C Chong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Peck Y Ong
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Division of Clinical Immunology and Allergy, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
14
|
The Genetics of Eczema Herpeticum. Clin Rev Allergy Immunol 2022; 63:390-397. [DOI: 10.1007/s12016-022-08953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 11/03/2022]
|
15
|
Jamerson TA, Li Q, Sreeskandarajan S, Budunova IV, He Z, Kang J, Gudjonsson JE, Patrick MT, Tsoi LC. Roles Played by Stress-Induced Pathways in Driving Ethnic Heterogeneity for Inflammatory Skin Diseases. Front Immunol 2022; 13:845655. [PMID: 35572606 PMCID: PMC9095822 DOI: 10.3389/fimmu.2022.845655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/23/2022] [Indexed: 12/25/2022] Open
Abstract
Immune-mediated skin conditions (IMSCs) are a diverse group of autoimmune diseases associated with significant disease burden. Atopic dermatitis and psoriasis are among the most common IMSCs in the United States and have disproportionate impact on racial and ethnic minorities. African American patients are more likely to develop atopic dermatitis compared to their European American counterparts; and despite lower prevalence of psoriasis among this group, African American patients can suffer from more extensive disease involvement, significant post-inflammatory changes, and a decreased quality of life. While recent studies have been focused on understanding the heterogeneity underlying disease mechanisms and genetic factors at play, little emphasis has been put on the effect of psychosocial or psychological stress on immune pathways, and how these factors contribute to differences in clinical severity, prevalence, and treatment response across ethnic groups. In this review, we explore the heterogeneity of atopic dermatitis and psoriasis between African American and European American patients by summarizing epidemiological studies, addressing potential molecular and environmental factors, with a focus on the intersection between stress and inflammatory pathways.
Collapse
Affiliation(s)
- Taylor A. Jamerson
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Qinmengge Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | | | - Irina V. Budunova
- Department of Dermatology, Northwestern Medicine, Northwestern University, Chicago, IL, United States,Department of Urology, Northwestern Medicine, Northwestern University, Chicago, IL, United States
| | - Zhi He
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Jian Kang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States
| | - Johann E. Gudjonsson
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Matthew T. Patrick
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Lam C. Tsoi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States,Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, United States,Department of Computational Medicine and Bioinformatics, Michigan Medicine, University of Michigan, Ann Arbor, MI, United States,*Correspondence: Lam C. Tsoi,
| |
Collapse
|
16
|
Traidl S, Roesner L, Zeitvogel J, Werfel T. Eczema herpeticum in atopic dermatitis. Allergy 2021; 76:3017-3027. [PMID: 33844308 DOI: 10.1111/all.14853] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 03/29/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases leading to pruritic skin lesions. A subset of AD patients exhibits a disseminated severe HSV infection called eczema herpeticum (EH) that can cause life-threatening complications. This review gives an overview of the clinical picture, and characteristics of the patients as well as the diagnosis and therapy of EH. A special focus lies on the pathophysiological hallmarks identified so far that predispose for EH. This aspect covers genetic aberrations, immunological changes, and environmental influences displaying a complex multifactorial situation, which is not completely understood. Type 2 skewing of virus-specific T cells in ADEH+ patients has been implicated in immune profile abnormalities, along with impaired functions of dendritic cells and natural killer cells. Furthermore, aberrations in interferon pathway-related genes such as IFNG and IFNGR1 have been identified to increase the risk of EH. IL-4, IL-25, and thymic stromal lymphopoietin (TSLP) are overexpressed in EH, whereas antimicrobial peptides like human β-defensins and LL-37 are reduced. Concerning the epidermal barrier, single nucleotide polymorphisms (SNPs) in skin barrier proteins such as filaggrin were identified in ADEH+ patients. A dysbalance of the skin microbiome also contributes to EH due to an increase of Staphylococcus aureus, which provides a supporting role to the viral infection via secreted toxins such as α-toxin. The risk of EH is reduced in AD patients treated with dupilumab. Further research is needed to identify and specifically target risk factors for EH in AD patients.
Collapse
Affiliation(s)
- Stephan Traidl
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Lennart Roesner
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Jana Zeitvogel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research Department of Dermatology and Allergy Hannover Medical School Hannover Germany
- Cluster of Excellence RESIST (EXC 2155) Hannover Medical School Hannover Germany
| |
Collapse
|
17
|
Bin L, Malley C, Taylor P, Preethi Boorgula M, Chavan S, Daya M, Mathias M, Shankar G, Rafaels N, Vergara C, Potee J, Campbell M, Hanifin JM, Simpson E, Schneider LC, Gallo RL, Hata T, Paller AS, De Benedetto A, Beck LA, Ong PY, Guttman‐Yassky E, Richers B, Baraghoshi D, Ruczinski I, Barnes KC, Leung DYM, Mathias RA. Whole genome sequencing identifies novel genetic mutations in patients with eczema herpeticum. Allergy 2021; 76:2510-2523. [PMID: 33548076 DOI: 10.1111/all.14762] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Eczema herpeticum (EH) is a rare complication of atopic dermatitis (AD) caused by disseminated herpes simplex virus (HSV) infection. The role of rare and/or deleterious genetic variants in disease etiology is largely unknown. This study aimed to identify genes that harbor damaging genetic variants associated with HSV infection in AD with a history of recurrent eczema herpeticum (ADEH+). METHODS Whole genome sequencing (WGS) was performed on 49 recurrent ADEH+ (≥3 EH episodes), 491 AD without a history of eczema herpeticum (ADEH-) and 237 non-atopic control (NA) subjects. Variants were annotated, and a gene-based approach (SKAT-O) was used to identify genes harboring damaging genetic variants associated with ADEH+. Genes identified through WGS were studied for effects on HSV responses and keratinocyte differentiation. RESULTS Eight genes were identified in the comparison of recurrent ADEH+to ADEH-and NA subjects: SIDT2, CLEC7A, GSTZ1, TPSG1, SP110, RBBP8NL, TRIM15, and FRMD3. Silencing SIDT2 and RBBP8NL in normal human primary keratinocytes (NHPKs) led to significantly increased HSV-1 replication. SIDT2-silenced NHPKs had decreased gene expression of IFNk and IL1b in response to HSV-1 infection. RBBP8NL-silenced NHPKs had decreased gene expression of IFNk, but increased IL1b. Additionally, silencing SIDT2 and RBBP8NL also inhibited gene expression of keratinocyte differentiation markers keratin 10 (KRT10) and loricrin (LOR). CONCLUSION SIDT2 and RBBP8NL participate in keratinocyte's response to HSV-1 infection. SIDT2 and RBBP8NL also regulate expression of keratinocyte differentiation genes of KRT10 and LOR.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics National Jewish Health Denver CO USA
| | - Claire Malley
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Patricia Taylor
- Department of Pediatrics National Jewish Health Denver CO USA
| | | | - Sameer Chavan
- Department of Medicine University of Colorado Aurora CO USA
| | - Michelle Daya
- Department of Medicine University of Colorado Aurora CO USA
| | - Malaika Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | - Gautam Shankar
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| | | | | | | | | | | | - Eric Simpson
- Oregon Health & Science University Portland OR USA
| | | | - Richard L. Gallo
- Department of Dermatology University of California San Diego CA USA
| | - Tissa Hata
- Department of Dermatology University of California San Diego CA USA
| | - Amy S. Paller
- Northwestern University Feinberg School of Medicine Chicago IL USA
| | | | - Lisa A. Beck
- University of Rochester Medical Center Rochester NY USA
| | - Peck Y. Ong
- Children’s Hospital Los Angeles University of Southern California Los Angeles CA USA
| | | | | | | | - Ingo Ruczinski
- Department of Biostatistics Bloomberg School of Public Health Johns Hopkins University Baltimore MD USA
| | | | | | - Rasika A. Mathias
- Division of Allergy and Clinical Immunology Johns Hopkins University Baltimore MD USA
| |
Collapse
|
18
|
Damour A, Garcia M, Seneschal J, Lévêque N, Bodet C. Eczema Herpeticum: Clinical and Pathophysiological Aspects. Clin Rev Allergy Immunol 2021; 59:1-18. [PMID: 31836943 DOI: 10.1007/s12016-019-08768-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in the world. AD is a complex pathology mainly characterized by an impaired skin barrier, immune response dysfunction, and unbalanced skin microbiota. Moreover, AD patients exhibit an increased risk of developing bacterial and viral infections. One of the most current, and potentially life-threatening, viral infection is caused by herpes simplex virus (HSV), which occurs in about 3% of AD patients under the name of eczema herpeticum (EH). Following a first part dedicated to the clinical features, virological diagnosis, and current treatments of EH, this review will focus on the description of the pathophysiology and, more particularly, the presently known predisposing factors to herpetic complications in AD patients. These factors include those related to impairment of the skin barrier such as deficit in filaggrin and anomalies in tight and adherens junctions. In addition, low production of the antimicrobial peptides cathelicidin LL-37 and human β-defensins; overexpression of cytokines such as interleukin (IL)-4, IL-13, IL-25, IL-33, and thymic stromal lymphopoietin (TSLP); or downregulation of type I to III interferons as well as defect in functions of immune cells such as dendritic, natural killer, and regulatory T cells have been involved. Otherwise, genetic polymorphisms and AD topical calcineurin inhibitor treatments have been associated with an increased risk of EH. Finally, dysbiosis of skin microbiota characterized in AD patients by Staphylococcus aureus colonization and toxin secretion, such as α-toxin, has been described as promoting HSV replication and could therefore contribute to EH.
Collapse
Affiliation(s)
- Alexia Damour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France
| | - Magali Garcia
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Julien Seneschal
- INSERM U1035, BMGIC, Immuno-dermatologie ATIP-AVENIR, Bordeaux, France.,Département de Dermatologie and Dermatologie Pédiatrique, Centre national de référence pour les maladies rares de la peau, Hôpital Saint-André, Bordeaux, France
| | - Nicolas Lévêque
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.,Laboratoire de Virologie et Mycobactériologie, CHU de Poitiers, Poitiers, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines EA 4331, Université de Poitiers, Poitiers, France.
| |
Collapse
|
19
|
Bai H, He Y, Ding Y, Chu Q, Lian L, Heifetz EM, Yang N, Cheng HH, Zhang H, Chen J, Song J. Genome-wide characterization of copy number variations in the host genome in genetic resistance to Marek's disease using next generation sequencing. BMC Genet 2020; 21:77. [PMID: 32677890 PMCID: PMC7364486 DOI: 10.1186/s12863-020-00884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 07/05/2020] [Indexed: 11/13/2022] Open
Abstract
Background Marek’s disease (MD) is a highly neoplastic disease primarily affecting chickens, and remains as a chronic infectious disease that threatens the poultry industry. Copy number variation (CNV) has been examined in many species and is recognized as a major source of genetic variation that directly contributes to phenotypic variation such as resistance to infectious diseases. Two highly inbred chicken lines, 63 (MD-resistant) and 72 (MD-susceptible), as well as their F1 generation and six recombinant congenic strains (RCSs) with varied susceptibility to MD, are considered as ideal models to identify the complex mechanisms of genetic and molecular resistance to MD. Results In the present study, to unravel the potential genetic mechanisms underlying resistance to MD, we performed a genome-wide CNV detection using next generation sequencing on the inbred chicken lines with the assistance of CNVnator. As a result, a total of 1649 CNV regions (CNVRs) were successfully identified after merging all the nine datasets, of which 90 CNVRs were overlapped across all the chicken lines. Within these shared regions, 1360 harbored genes were identified. In addition, 55 and 44 CNVRs with 62 and 57 harbored genes were specifically identified in line 63 and 72, respectively. Bioinformatics analysis showed that the nearby genes were significantly enriched in 36 GO terms and 6 KEGG pathways including JAK/STAT signaling pathway. Ten CNVRs (nine deletions and one duplication) involved in 10 disease-related genes were selected for validation by using quantitative real-time PCR (qPCR), all of which were successfully confirmed. Finally, qPCR was also used to validate two deletion events in line 72 that were definitely normal in line 63. One high-confidence gene, IRF2 was identified as the most promising candidate gene underlying resistance and susceptibility to MD in view of its function and overlaps with data from previous study. Conclusions Our findings provide valuable insights for understanding the genetic mechanism of resistance to MD and the identified gene and pathway could be considered as the subject of further functional characterization.
Collapse
Affiliation(s)
- Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, 225009, China.,Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA.,Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yanghua He
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA.,Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Yi Ding
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Qin Chu
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA.,Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Ling Lian
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Eliyahu M Heifetz
- Faculty of Health Sciences, Jerusalem College of Technology, 9116001, Jerusalem, Israel
| | - Ning Yang
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hans H Cheng
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA
| | - Huanmin Zhang
- USDA, Agricultural Research Service, Avian Disease and Oncology Laboratory, East Lansing, MI, 48823, USA
| | - Jilan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jiuzhou Song
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
20
|
Nomura T, Wu J, Kabashima K, Guttman-Yassky E. Endophenotypic Variations of Atopic Dermatitis by Age, Race, and Ethnicity. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 8:1840-1852. [DOI: 10.1016/j.jaip.2020.02.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022]
|
21
|
Replicated methylation changes associated with eczema herpeticum and allergic response. Clin Epigenetics 2019; 11:122. [PMID: 31443688 PMCID: PMC6706929 DOI: 10.1186/s13148-019-0714-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/22/2019] [Indexed: 01/06/2023] Open
Abstract
Background Although epigenetic mechanisms are important risk factors for allergic disease, few studies have evaluated DNA methylation differences associated with atopic dermatitis (AD), and none has focused on AD with eczema herpeticum (ADEH+). We will determine how methylation varies in AD individuals with/without EH and associated traits. We modeled differences in genome-wide DNA methylation in whole blood cells from 90 ADEH+, 83 ADEH−, and 84 non-atopic, healthy control subjects, replicating in 36 ADEH+, 53 ADEH−, and 55 non-atopic healthy control subjects. We adjusted for cell-type composition in our models and used genome-wide and candidate-gene approaches. Results We replicated one CpG which was significantly differentially methylated by severity, with suggestive replication at four others showing differential methylation by phenotype or severity. Not adjusting for eosinophil content, we identified 490 significantly differentially methylated CpGs (ADEH+ vs healthy controls, genome-wide). Many of these associated with severity measures, especially eosinophil count (431/490 sites). Conclusions We identified a CpG in IL4 associated with serum tIgE levels, supporting a role for Th2 immune mediating mechanisms in AD. Changes in eosinophil level, a measure of disease severity, are associated with methylation changes, providing a potential mechanism for phenotypic changes in immune response-related traits. Electronic supplementary material The online version of this article (10.1186/s13148-019-0714-1) contains supplementary material, which is available to authorized users.
Collapse
|
22
|
Wang S, Cao F, Gu X, Chen J, Xu R, Huang Y, Ying L. LncRNA XIST, as a ceRNA of miR-204, aggravates lipopolysaccharide-induced acute respiratory distress syndrome in mice by upregulating IRF2. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:2425-2434. [PMID: 31934069 PMCID: PMC6949564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a common clinical syndrome with high a mortality rate, which is associated with diffuse alveolar injury and capillary endothelial damage. In recent years, numerous studies have been performed to explore the roles of long non-coding RNAs (lncRNAs) in various diseases in which lncRNA serves as a microRNA (miRNA) sponge to regulate targeted gene expression. However, whether lncRNAs participate in ARDS progression remains unclear. MATERIALS/METHODS The dual-luciferase reporter assay was employed to identify the interaction between lncRNA XIST and miR-204, as well as the correlation between miR-204 and interferon regulatory factor 2 (IRF2). Then, PaO2/FiO2 was determined in lipopolysaccharide (LPS)-induced ARDS. In addition, the concentrations of cytokines, including IFN-γ, IL-6, IL-17, TNF-α, IL-1β, and IL-6R were analyzed by ELISA. lncRNA XIST, miR-204, and IRF2 levels were determined by qRT-PCR assay, and the IRF2 expression was evaluated by western blot. Furthermore, levels of inflammation and conditions of alveoli were evaluated by hematoxylin (H&E)-staining in LPS-induced ARDS. RESULTS Our findings indicated that lncRNA XIST served as a sponge for miR-204. miR-204 directly regulated IRF2, andlncRNA XIST upregulated IRF2 expression by targeting miR-204. LncRNA XIST and miR-204 inhibitors significantly decreased the PaO2/FiO2 ratio, whereas miR-204 and silencing of IRF2 significantly increased the PaO2/FiO2 ratio in LPS-induced ARDS. In addition, lncRNA XIST and miR-204 inhibitors significantly increased levels of IFN-γ, IL-6, IL-17, TNF-α, IL-1β, and IL-6R, whereas miR-204 and silencing of IRF2 dramatically decreased related cytokines in LPS-induced ARDS. Furthermore, we demonstrated that lncRNA XIST and miR-204 inhibitors aggravated inflammatory cell infiltration, alveolitis, and the degree of fibrosis, whereas miR-204 and silencing of IRF2 alleviated inflammation and conditions of the alveoli. CONCLUSION In this study, we verified that lncRNA XIST serves as a sponge for miR-204 to aggravate LPS-induced ARDS in mice by upregulating IRF2.
Collapse
Affiliation(s)
- Shuguang Wang
- Department of Emergency Intensive Care Unit, Ningbo No. 6 HospitalNingbo 315040, China
| | - Feng Cao
- Department of Emergency, Ningbo No. 6 HospitalNingbo 315040, China
| | - Xingsheng Gu
- Department of Emergency Intensive Care Unit, Ningbo No. 6 HospitalNingbo 315040, China
| | - Jianan Chen
- Department of Emergency Intensive Care Unit, Ningbo No. 6 HospitalNingbo 315040, China
| | - Ranxing Xu
- Department of Clinical Laboratory, Ningbo No. 6 HospitalNingbo 315040, China
| | - Yangneng Huang
- Department of Emergency, Ningbo No. 6 HospitalNingbo 315040, China
| | - Lina Ying
- Department of Clinical Laboratory, Ningbo No. 6 HospitalNingbo 315040, China
| |
Collapse
|
23
|
Brunner PM, Guttman-Yassky E. Racial differences in atopic dermatitis. Ann Allergy Asthma Immunol 2019; 122:449-455. [PMID: 30465859 DOI: 10.1016/j.anai.2018.11.015] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/04/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To summarize studies investigating ethnical and racial differences in atopic dermatitis (AD) epidemiology, clinical features, and skin and blood phenotypes. DATA SOURCES PubMed literature review (years 2000-2018). STUDY SELECTIONS Articles discussing primarily human disease. RESULTS Higher overall rates of AD were found in Africa and Oceania as opposed to India and Northern and Eastern Europe. In the United States, AD prevalence was found to be higher in African American (19.3%) compared with European American (16.1%) children. Although several studies have consistently found FLG loss-of-function mutations in up to 50% of European and 27% of Asian patients with AD, FLG mutations were 6 times less common in African American than in European American patients, even in patients with severe AD. Thus, FLG mutations seem to play less a pathogenic role in patients of African origin than in individuals of European or Asian ancestry. The immune phenotype of all ethnic groups was characterized by strong TH2 activation, but important differences in immune polarization exist among the different ethnicities. Asian patients with AD had stronger TH17/TH22 activation than African American and European American patients with AD, whereas African American patients had the highest serum IgE levels among all groups, while largely lacking TH1 and TH17 activation. CONCLUSION AD is a heterogeneous disease that has differences among various ethnic and racial groups, which might be important for the development of future, targeted treatments and for personalized medicine approaches.
Collapse
Affiliation(s)
- Patrick M Brunner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
24
|
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease. Genetic predisposition, epidermal barrier disruption, and dysregulation of the immune system are some of the critical components of AD. An impaired skin barrier may be the initial step in the development of the atopic march as well as AD, which leads to further skin inflammation and allergic sensitization. Type 2 cytokines as well as interleukin 17 and interleukin 22 contribute to skin barrier dysfunction and the development of AD. New insights into the pathophysiology of AD have focused on epidermal lipid profiles, neuroimmune interactions, and microbial dysbiosis. Newer therapeutic strategies focus on improving skin barrier function and targeting polarized immune pathways found in AD. Further understanding of AD pathophysiology will allow us to achieve a more precision medicine approach to the prevention and the treatment of AD.
Collapse
Affiliation(s)
- Jihyun Kim
- From the Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung Eui Kim
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado
| |
Collapse
|
25
|
Serra-Baldrich E, de Frutos J, Jáuregui I, Armario-Hita J, Silvestre J, Herraez L, Martín-Santiago A, Valero A, Sastre J. Changing perspectives in atopic dermatitis. Allergol Immunopathol (Madr) 2018; 46:397-412. [PMID: 29031890 DOI: 10.1016/j.aller.2017.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 07/29/2017] [Indexed: 01/25/2023]
Abstract
Atopic dermatitis (AD) is a multifaceted disease that involves a complex interplay between the skin and the immune system. The course of the disease depends strongly on the genetic background of the patient and on yet poorly-defined environmental factors. Changes in lifestyle could be behind the dramatic rise in the prevalence of AD across continents; including hygienic conditions, food, social habits, skin microbiome or exposure to a number of allergens. Although AD typically develops in childhood and disappears after a few years, in a relatively large number of patients it continues into adulthood. Adult AD can also appear de novo but it is often underdiagnosed and its treatment can be challenging. New, highly effective drugs are being developed to manage moderate and severe forms of the disease in adults. In this review, we highlight the most recent developments in diagnostic tools, current insights into the mechanistic basis of this disease, and therapeutic innovations.
Collapse
|
26
|
Kaufman BP, Guttman-Yassky E, Alexis AF. Atopic dermatitis in diverse racial and ethnic groups-Variations in epidemiology, genetics, clinical presentation and treatment. Exp Dermatol 2018; 27:340-357. [DOI: 10.1111/exd.13514] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Bridget P. Kaufman
- Department of Dermatology; Mount Sinai St. Luke's and Mount Sinai West; New York NY USA
| | - Emma Guttman-Yassky
- Department of Dermatology and the Laboratory for Inflammatory Skin Diseases; Icahn School of Medicine at Mount Sinai; New York NY USA
| | - Andrew F. Alexis
- Department of Dermatology; Mount Sinai St. Luke's and Mount Sinai West; New York NY USA
| |
Collapse
|
27
|
Simpson EL, Villarreal M, Jepson B, Rafaels N, David G, Hanifin J, Taylor P, Boguniewicz M, Yoshida T, De Benedetto A, Barnes KC, Leung DYM, Beck LA. Patients with Atopic Dermatitis Colonized with Staphylococcus aureus Have a Distinct Phenotype and Endotype. J Invest Dermatol 2018; 138:2224-2233. [PMID: 29604251 DOI: 10.1016/j.jid.2018.03.1517] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/31/2022]
Abstract
Patients with atopic dermatitis (AD) are commonly colonized with Staphylococcus aureus (AD S. aureus+), but what differentiates this group from noncolonized AD patients (AD S. aureus-) has not been well studied. To evaluate whether these two groups have unique phenotypic or endotypic features, we performed a multicenter, cross-sectional study enrolling AD S. aureus+ (n = 51) and AD S. aureus- (n = 45) participants defined by the presence or absence of S. aureus by routine culture techniques and nonatopic, noncolonized control individuals (NA S. aureus-) (n = 46). Filaggrin (FLG) genotypes were determined, and disease severity (Eczema Area and Severity Index, Rajka-Langeland Severity Score, Investigator's Global Assessment score, Numerical Rating Scale, and Dermatology Life Quality Index) was captured. Skin physiology was assessed (transepidermal water loss [TEWL], stratum corneum integrity, hydration, and pH), and serum biomarkers were also measured. We found that AD S. aureus+ patients had more severe disease based on all scoring systems except itch (Numerical Rating Scale), and they had higher levels of type 2 biomarkers (eosinophil count, tIgE, CCL17, and periostin). Additionally, AD S. aureus+ patients had significantly greater allergen sensitization (Phadiatop and tIgE), barrier dysfunction (TEWL and stratum corneum integrity), and serum lactate dehydrogenase (LDH) than both the AD S. aureus- and NA S. aureus- groups. FLG mutations did not associate with S. aureus+ colonization. In conclusion, adult patients with AD who are colonized on their skin with S. aureus have more severe disease, greater type 2 immune deviation, allergen sensitization, barrier disruption, and LDH level elevation than noncolonized patients with AD.
Collapse
Affiliation(s)
- Eric L Simpson
- Oregon Health & Science University, Portland, Oregon, USA
| | | | | | | | | | - Jon Hanifin
- Oregon Health & Science University, Portland, Oregon, USA
| | | | | | - Takeshi Yoshida
- University of Rochester Medical Center, Rochester, New York, USA
| | | | | | | | - Lisa A Beck
- University of Rochester Medical Center, Rochester, New York, USA.
| |
Collapse
|
28
|
Kawaguchi M, Oka T, Sugaya M, Suga H, Kimura T, Morimura S, Fujita H, Sato S. IRF-2 haploinsufficiency causes enhanced imiquimod-induced psoriasis-like skin inflammation. J Dermatol Sci 2018; 90:35-45. [PMID: 29305258 DOI: 10.1016/j.jdermsci.2017.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/24/2017] [Accepted: 12/20/2017] [Indexed: 01/05/2023]
Abstract
BACKGROUNDS IFN regulatory factor (IRF)-2 is one of the potential susceptibility genes for psoriasis, but how this gene influences psoriasis pathogenesis is unclear. Topical application of imiquimod (IMQ), a TLR7 ligand, induces psoriasis-like skin lesions in mice. OBJECTIVE The aim of this study was to investigate whether IRF-2 gene status would influence severity of skin disease in IMQ-treated mice. METHODS Imiquimod-induced psoriasis-like skin inflammation was assessed by clinical findings, histology, and cytokine expression. The effects of imiquimod or IFN on peritoneal macrophages were analyzed in vitro. RESULTS IMQ-induced skin inflammation assessed by clinical findings and histology was more severe in IRF-2+/- mice than in wild-type mice. In inflamed skin, mRNA expression levels of tumor necrosis factor (TNF)-α, IL-12/23p40, IL-17A, and IL-22 were significantly elevated in IRF-2+/- mice compared to wild-type mice. Stimulation of peritoneal macrophages by IMQ significantly increased mRNA levels of TNF-α, IL-12/23p40, IL-23p19, IL-12p35, and IL-36. Interestingly, macrophages from IRF-2+/- mice expressed higher levels of TNF-α, IL-12/23p40, and IL-36 compared to those from wild-type mice 24 h after stimulation, while they expressed similar levels of IL-12p35 and IL-23p19. Moreover, elevated mRNA expression of inducible nitric oxide synthase was observed only in IMQ-stimulated macrophages derived from IRF-2+/- mice, which correlated with angiogenesis in IMQ-treated ears of IRF-2+/- mice. CONCLUSIONS These results suggest that IRF-2 haploinsufficiency creates heightened biologic responses to IFN-α that phenotypically lead to enhanced angiogenesis and psoriasis-like inflammation within skin.
Collapse
Affiliation(s)
- Makiko Kawaguchi
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tomonori Oka
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Makoto Sugaya
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Hiraku Suga
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takayuki Kimura
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Sohshi Morimura
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Hideki Fujita
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Saintive S, Abad E, C Ferreira DD, Stambovsky M, Cavalcante FS, Gonçalves LS, Vidal F, dos Santos KRN. What is the role of Staphylococcus aureus and herpes virus infections in the pathogenesis of atopic dermatitis? Future Microbiol 2017; 12:1327-1334. [DOI: 10.2217/fmb-2017-0081] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic, relapsing disease. Genetic, environmental and immunological factors are involved in its pathophysiology. Individuals with AD have an increased predisposition to colonization and/or infection of the skin by various pathogens, especially Staphylococcus aureus and herpes simplex virus. The composition of their skin microbiome is also different, and changes during flares. The disease severity can be related to the degree of colonization by S. aureus. In addition, the presence of this bacterial species can predispose the host to more severe and disseminated viral infections. This article reviews the role of S. aureus and herpes virus infections and the skin microbiome in the pathogenesis of AD and their importance in the treatment and prevention strategies of this dermatosis.
Collapse
Affiliation(s)
- Simone Saintive
- Pediatric Dermatology Service, IPPMG–Martagão Gesteira Pediatric Institute–Federal University of Rio de Janeiro–UFRJ, Rio de Janeiro, Brazil
| | - Eliane Abad
- Pediatric Dermatology Service, IPPMG–Martagão Gesteira Pediatric Institute–Federal University of Rio de Janeiro–UFRJ, Rio de Janeiro, Brazil
| | - Dennis de C Ferreira
- Dentistry Graduate Program, School of Dentistry, Veiga de Almeida University, Rio de Janeiro, Brazil
- Post graduation Program in Dentistry, School of Dentistry, Estácio de Sá University, Rio de Janeiro, Brazil
| | - Mayra Stambovsky
- Dentistry Graduate Program, School of Dentistry, Veiga de Almeida University, Rio de Janeiro, Brazil
| | - Fernanda S Cavalcante
- Microbiology Department–Federal University of Rio de Janeiro–UFRJ, Campus Macaé, Rio de Janeiro, Brazil
| | - Lucio S Gonçalves
- Post graduation Program in Dentistry, School of Dentistry, Estácio de Sá University, Rio de Janeiro, Brazil
| | - Fabio Vidal
- Post graduation Program in Dentistry, School of Dentistry, Estácio de Sá University, Rio de Janeiro, Brazil
| | - Katia RN dos Santos
- Paulo de Goes Microbiology Institute, Federal University Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Knaysi G, Smith AR, Wilson JM, Wisniewski JA. The Skin as a Route of Allergen Exposure: Part II. Allergens and Role of the Microbiome and Environmental Exposures. Curr Allergy Asthma Rep 2017; 17:7. [PMID: 28210979 DOI: 10.1007/s11882-017-0675-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW This second part of the article aims to highlight recent contributions in the literature that enhance our understanding of the cutaneous immune response to allergen. RECENT FINDINGS Several properties of allergens facilitate barrier disruption and cutaneous sensitization. There is a strong epidemiologic relationship between the microbiome, both the gut and skin, and atopic dermatitis (AD). The mechanisms connecting these two entities remain enigmatic; however, recent murine models show that commensal skin bacteria play an active role in supporting skin barrier homeostasis and defense against microbial penetration. Likewise, the association between the lack of colonization with Staph species and AD development suggests a potentially functional role for these organisms in regulating the skin barrier and response to environmental allergens. In undisrupted skin, evidence suggests that the cutaneous route may promote allergen tolerance. Properties of environmental allergens and commensal bacteria add to the complex landscape of skin immunity. Further investigation is needed to elucidate how these properties regulate the cutaneous immune response to allergen.
Collapse
Affiliation(s)
- George Knaysi
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Geisel School of Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, USA
| | - Anna R Smith
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Jeffrey M Wilson
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Julia A Wisniewski
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
31
|
Liang Y, Chang C, Lu Q. The Genetics and Epigenetics of Atopic Dermatitis-Filaggrin and Other Polymorphisms. Clin Rev Allergy Immunol 2017; 51:315-328. [PMID: 26385242 DOI: 10.1007/s12016-015-8508-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease caused by a combination of genetic and environmental factors. Genetic evidences depict a complex network comprising by epidermal barrier dysfunctions and dysregulation of innate and adaptive immunity in the pathogenesis of AD. Mutations in the human filaggrin gene (FLG) are the most significant and well-replicated genetic mutation associated with AD, and other mutations associated with epidermal barriers such as SPINK5, FLG-2, SPRR3, and CLDN1 have all been linked to AD. Gene variants may also contribute to the abnormal innate and adaptive responses found in AD, including mutations in PRRs and AMPs, TSLP and TSLPR, IL-1 family cytokines and receptors genes, vitamin D pathway genes, FCER1A, and Th2 and other cytokines genes. GWAS and Immunochip analysis have identified a total of 19 susceptibility loci for AD. Candidate genes at these susceptibility loci identified by GWAS and Immunochip analysis also suggest roles for epidermal barrier functions, innate and adaptive immunity, interleukin-1 family signaling, regulatory T cells, the vitamin D pathway, and the nerve growth factor pathway in the pathogenesis of AD. Increasing evidences show the modern lifestyle (i.e., the hygiene hypothesis, Western diet) and other environmental factors such as pollution and environmental tobacco smoke (ETS) lead to the increasing prevalence of AD with the development of industrialization. Epigenetic alterations in response to these environmental factors, including DNA methylation and microRNA related to immune system and skin barriers, have been found to contribute to the pathogenesis of AD. Genetic variants and epigenetic alteration might be the key tools for the molecular taxonomy of AD and provide the background for the personalized management.
Collapse
Affiliation(s)
- Yunsheng Liang
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan, 410011, China
| | - Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, 95616, USA
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics & Department of Dermatology, The Second Xiangya Hospital, Central South University, 139 Renmin Middle Rd, Changsha, Hunan, 410011, China.
| |
Collapse
|
32
|
Ong PY, Leung DYM. Bacterial and Viral Infections in Atopic Dermatitis: a Comprehensive Review. Clin Rev Allergy Immunol 2017; 51:329-337. [PMID: 27377298 DOI: 10.1007/s12016-016-8548-5] [Citation(s) in RCA: 170] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Atopic dermatitis (AD) is the most common allergic skin disease in the general population. It is a chronic inflammatory skin disease complicated by recurrent bacterial and viral infections that, when left untreated, can lead to significant complications. The current article will review immunologic and molecular mechanisms underlying the propensity of AD patients to microbial infections. These infections include Staphylococcus aureus (S. aureus) skin infections, eczema herpeticum, eczema vaccinatum, and eczema coxsackium. Previous studies have shown that skin barrier defects, a decrease in antimicrobial peptides, increased skin pH, or Th2 cytokines such as IL-4 and IL-13 are potential contributing factors for the increased risk of skin infections in AD. In addition, bacterial virulence such as methicillin-resistant S. aureus (MRSA) produces significantly higher number of superantigens that increase their potential in causing infection and more severe cutaneous inflammation in AD patients. More recent studies suggest that skin microbiome including Staphylococcus epidermidis or other coagulase-negative staphylococci may play an important role in controlling S. aureus skin infections in AD. Other studies also suggest that genetic variants in the innate immune response may predispose AD patients to increased risk of viral skin infections. These genetic variants include thymic stromal lymphopoietin (TSLP), type I interferon (α, ß, ω), type II interferon (γ), and molecular pathways that lead to the production of interferons (interferon regulatory factor 2). A common staphylococcal toxin, α-toxin, may also play a role in enhancing herpes simplex virus skin infections in AD. Further understanding of these disease processes may have important clinical implications for the prevention and treatment of skin infections in this common skin disease.
Collapse
Affiliation(s)
- Peck Y Ong
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Los Angeles, USA
- Department of Pediatrics, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Donald Y M Leung
- Division of Pediatric Allergy-Immunology, National Jewish Health, 1400 Jackson Street (Room K926i), Denver, 80206, CO, USA.
- Department of Pediatrics, University of Colorado Denver Health Sciences Center, Denver, CO, USA.
| |
Collapse
|
33
|
Li MMH, Bozzacco L, Hoffmann HH, Breton G, Loschko J, Xiao JW, Monette S, Rice CM, MacDonald MR. Interferon regulatory factor 2 protects mice from lethal viral neuroinvasion. J Exp Med 2016; 213:2931-2947. [PMID: 27899441 PMCID: PMC5154937 DOI: 10.1084/jem.20160303] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/09/2016] [Accepted: 11/01/2016] [Indexed: 01/08/2023] Open
Abstract
Li et al. describe a novel role for IRF2, previously known as a negative regulator of type I IFN signaling, in protection of mice from lethal viral neuroinvasion by facilitating the proper localization of B cells and antibodies to the central nervous system. The host responds to virus infection by activating type I interferon (IFN) signaling leading to expression of IFN-stimulated genes (ISGs). Dysregulation of the IFN response results in inflammatory diseases and chronic infections. In this study, we demonstrate that IFN regulatory factor 2 (IRF2), an ISG and a negative regulator of IFN signaling, influences alphavirus neuroinvasion and pathogenesis. A Sindbis virus strain that in wild-type (WT) mice only causes disease when injected into the brain leads to lethal encephalitis in Irf2−/− mice after peripheral inoculation. Irf2−/− mice fail to control virus replication and recruit immune infiltrates into the brain. Reduced B cells and virus-specific IgG are observed in the Irf2−/− mouse brains despite the presence of peripheral neutralizing antibodies, suggesting a defect in B cell trafficking to the central nervous system (CNS). B cell–deficient μMT mice are significantly more susceptible to viral infection, yet WT B cells and serum are unable to rescue the Irf2−/− mice. Collectively, our data demonstrate that proper localization of B cells and local production of antibodies in the CNS are required for protection. The work advances our understanding of host mechanisms that affect viral neuroinvasion and their contribution to immunity against CNS infections.
Collapse
Affiliation(s)
- Melody M H Li
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Leonia Bozzacco
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Gaëlle Breton
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Jakob Loschko
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065
| | - Jing W Xiao
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Sébastien Monette
- Tri-Institutional Laboratory of Comparative Pathology, Memorial Sloan-Kettering Cancer Center, The Rockefeller University, Weill Cornell Medical College, New York, NY 10065
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065
| |
Collapse
|
34
|
Brar K, Leung DYM. Recent considerations in the use of recombinant interferon gamma for biological therapy of atopic dermatitis. Expert Opin Biol Ther 2016; 16:507-14. [PMID: 26694988 DOI: 10.1517/14712598.2016.1135898] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Atopic dermatitis (AD) is the most common inflammatory skin disease in the general population. There are different endophenotypes of AD that likely have a unique immune and molecular basis, such as those who are predisposed to eczema herpeticum, or Staphylococcus aureus infections. AREAS COVERED In this review, we highlight the endophenotypes of AD where reduced interferon gamma expression may be playing a role. Additionally, we review the potential role of recombinant interferon gamma therapy in the treatment of atopic dermatitis and the particular phenotypes that may benefit from this treatment. EXPERT OPINION Recombinant interferon gamma treatment will likely benefit the pediatric population with AD, as well as those with susceptibilities for skin infections. Future studies are needed to elucidate whether IFN-γ may reduce the prevalence of skin infection in AD.
Collapse
Affiliation(s)
- Kanwaljit Brar
- a Division of Pediatric Allergy-Immunology, Department of Pediatrics , National Jewish Health , Denver , CO , USA
| | - Donald Y M Leung
- a Division of Pediatric Allergy-Immunology, Department of Pediatrics , National Jewish Health , Denver , CO , USA.,b Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology , The State Key Clinical Specialty in Allergy, The Second Affiliated Hospital of Guangzhou Medical University , Guangzhou , China
| |
Collapse
|
35
|
Gao L, Bin L, Rafaels NM, Huang L, Potee J, Ruczinski I, Beaty TH, Paller AS, Schneider LC, Gallo R, Hanifin JM, Beck LA, Geha RS, Mathias RA, Barnes KC, Leung DYM. Targeted deep sequencing identifies rare loss-of-function variants in IFNGR1 for risk of atopic dermatitis complicated by eczema herpeticum. J Allergy Clin Immunol 2015; 136:1591-1600. [PMID: 26343451 PMCID: PMC4679503 DOI: 10.1016/j.jaci.2015.06.047] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 05/13/2015] [Accepted: 06/23/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND A subset of atopic dermatitis is associated with increased susceptibility to eczema herpeticum (ADEH+). We previously reported that common single nucleotide polymorphisms (SNPs) in the IFN-γ (IFNG) and IFN-γ receptor 1 (IFNGR1) genes were associated with the ADEH+ phenotype. OBJECTIVE We sought to interrogate the role of rare variants in interferon pathway genes for the risk of ADEH+. METHODS We performed targeted sequencing of interferon pathway genes (IFNG, IFNGR1, IFNAR1, and IL12RB1) in 228 European American patients with AD selected according to their eczema herpeticum status, and severity was measured by using the Eczema Area and Severity Index. Replication genotyping was performed in independent samples of 219 European American and 333 African American subjects. Functional investigation of loss-of-function variants was conducted by using site-directed mutagenesis. RESULTS We identified 494 single nucleotide variants encompassing 105 kb of sequence, including 145 common, 349 (70.6%) rare (minor allele frequency <5%), and 86 (17.4%) novel variants, of which 2.8% were coding synonymous, 93.3% were noncoding (64.6% intronic), and 3.8% were missense. We identified 6 rare IFNGR1 missense variants, including 3 damaging variants (Val14Met [V14M], Val61Ile, and Tyr397Cys [Y397C]) conferring a higher risk for ADEH+ (P = .031). Variants V14M and Y397C were confirmed to be deleterious, leading to partial IFNGR1 deficiency. Seven common IFNGR1 SNPs, along with common protective haplotypes (2-7 SNPs), conferred a reduced risk of ADEH+ (P = .015-.002 and P = .0015-.0004, respectively), and both SNP and haplotype associations were replicated in an independent African American sample (P = .004-.0001 and P = .001-.0001, respectively). CONCLUSION Our results provide evidence that both genetic variants in the gene encoding IFNGR1 are implicated in susceptibility to the ADEH+ phenotype.
Collapse
Affiliation(s)
- Li Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Lianghua Bin
- Department of Pediatrics, National Jewish Health, 1400 Jackson St, Denver, CO
| | - Nicholas M Rafaels
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Lili Huang
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Joseph Potee
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Ingo Ruczinski
- Department of Biostatistics, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD
| | - Terri H. Beaty
- Department of Epidemiology, Bloomberg School of Public Health, The Johns Hopkins University, Baltimore, MD
| | - Amy S. Paller
- Northwestern University and Children’s Memorial Hospital, Chicago, IL
| | | | - Rich Gallo
- Division of Dermatology, University of California San Diego, San Diego, CA
| | - Jon M. Hanifin
- Department of Dermatology, Oregon Health & Science University, Portland, OR
| | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY
| | - Raif S. Geha
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Rasika A. Mathias
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Kathleen C. Barnes
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, Baltimore, MD
| | - Donald Y. M. Leung
- Department of Pediatrics, National Jewish Health, 1400 Jackson St, Denver, CO
| |
Collapse
|
36
|
Czarnowicki T, Gonzalez J, Shemer A, Malajian D, Xu H, Zheng X, Khattri S, Gilleaudeau P, Sullivan-Whalen M, Suárez-Fariñas M, Krueger JG, Guttman-Yassky E. Severe atopic dermatitis is characterized by selective expansion of circulating TH2/TC2 and TH22/TC22, but not TH17/TC17, cells within the skin-homing T-cell population. J Allergy Clin Immunol 2015; 136:104-115.e7. [PMID: 25748064 DOI: 10.1016/j.jaci.2015.01.020] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/15/2015] [Accepted: 01/23/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND Past studies of blood T-cell phenotyping in patients with atopic dermatitis (AD) have provided controversial results and were mostly performed before the identification of TH9, TH17, and TH22 T-cell populations in human subjects. OBJECTIVE We sought to quantify TH1, TH2, TH9, TH17, and TH22 T-cell populations and corresponding CD8(+) T-cell subsets in both cutaneous lymphocyte antigen (CLA)-positive and CLA(-) T-cell subsets in patients with AD and control subjects. METHODS We studied 42 adults with severe AD (mean SCORAD score, 65) and 25 healthy subjects using an 11-color flow cytometric antibody panel. Frequencies of IFN-γ-, IL-22-, IL-13-, IL-17-, and IL-9-producing CD4(+) and CD8(+) T cells were compared in CLA(-) and CLA(+) populations. RESULTS We measured increased TH2/TC2/IL-13(+) and TH22/TC22/IL-22(+) populations (P < .1) in patients with severe AD versus control subjects, with significant differences in CLA(+) T-cell numbers (P < .01). A significantly lower frequency of CLA(+) IFN-γ-producing cells was observed in patients with AD, with no significant differences in CLA(-) T-cell numbers. The CLA(+) TH1/TH2 and TC1/TC2 ratio was highly imbalanced in patients with AD (10 vs 3 [P = .005] and 19 vs 7 [P < .001], respectively). Positive correlations were found between frequencies of IL-13- and IL-22-producing CD4(+) and CD8(+) T cells (r = 0.5 and 0.8, respectively; P < .0001), and frequencies of IL-13-producing CLA(+) cells were also correlated with IgE levels and SCORAD scores. Patients with AD with skin infections had higher CD4(+) IL-22(+) and IL-17(+) cell frequencies, which were highly significant among CLA(-) cells (IL-22: 3.7 vs 1.7 [P < .001] and IL-17: 1.7 vs 0.6 [P < .001]), with less significant effects among CLA(+) T cells (IL-22: 11 vs 7.5, P = .04). CONCLUSIONS Severe AD is accompanied by expansion of skin-homing TH2/TC2 and TH22/TC22 subsets with lower TH1/TC1 frequencies. These data create a critical basis for studying alterations in immune activation in adults and pediatric patients with AD.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY.
| | - Juana Gonzalez
- Translational Technology Core Laboratory, Rockefeller University, New York, NY
| | - Avner Shemer
- Department of Dermatology, Tel-Hashomer Hospital, Tel Aviv, Israel
| | - Dana Malajian
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Columbia University College of Physicians and Surgeons, New York, NY
| | - Hui Xu
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Xiuzhong Zheng
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Saakshi Khattri
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | | | | | - Mayte Suárez-Fariñas
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Center for Clinical and Translational Science, Rockefeller University, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, Rockefeller University, New York, NY; Dermatology Department at the Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
37
|
Sanati G, Aryan Z, Barbadi M, Rezaei N. Innate lymphoid cells are pivotal actors in allergic, inflammatory and autoimmune diseases. Expert Rev Clin Immunol 2015; 11:885-95. [DOI: 10.1586/1744666x.2015.1050382] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
Affiliation(s)
- W. Peng
- Department of Dermatology and Allergy; University of Bonn; Bonn Germany
| | - N. Novak
- Department of Dermatology and Allergy; University of Bonn; Bonn Germany
| |
Collapse
|
39
|
Kawasaki A, Furukawa H, Nishida N, Warabi E, Kondo Y, Ito S, Matsumoto I, Kusaoi M, Amano H, Suda A, Nagaoka S, Setoguchi K, Nagai T, Hirohata S, Shimada K, Sugii S, Okamoto A, Chiba N, Suematsu E, Ohno S, Katayama M, Okamoto A, Kono H, Tokunaga K, Takasaki Y, Hashimoto H, Sumida T, Tohma S, Tsuchiya N. Association of functional polymorphisms in interferon regulatory factor 2 (IRF2) with susceptibility to systemic lupus erythematosus: a case-control association study. PLoS One 2014; 9:e109764. [PMID: 25285625 PMCID: PMC4186848 DOI: 10.1371/journal.pone.0109764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 09/08/2014] [Indexed: 02/02/2023] Open
Abstract
Interferon regulatory factor 2 (IRF2) negatively regulates type I interferon (IFN) responses, while it plays a role in induction of Th1 differentiation. Previous linkage and association studies in European-American populations suggested genetic role of IRF2 in systemic lupus erythematosus (SLE); however, this observation has not yet been confirmed. No studies have been reported in the Asian populations. Here we investigated whether IRF2 polymorphisms contribute to susceptibility to SLE in a Japanese population. Association study of 46 IRF2 tag single nucleotide polymorphisms (SNPs) detected association of an intronic SNP, rs13146124, with SLE. When the association was analyzed in 834 Japanese patients with SLE and 817 healthy controls, rs13146124 T was significantly increased in SLE compared with healthy controls (dominant model, P = 5.4×10−4, Bonferroni-corrected P [Pc] = 0.026, odds ratio [OR] 1.48, 95% confidence interval [CI] 1.18–1.85). To find causal SNPs, resequencing was performed by next-generation sequencing. Twelve polymorphisms in linkage disequilibrium with rs13146124 (r2: 0.30–1.00) were identified, among which significant association was observed for rs66801661 (allele model, P = 7.7×10−4, Pc = 0.037, OR 1.53, 95%CI 1.19–1.96) and rs62339994 (dominant model, P = 9.0×10−4, Pc = 0.043, OR 1.46, 95%CI 1.17–1.82). The haplotype carrying both of the risk alleles (rs66801661A–rs62339994A) was significantly increased in SLE (P = 9.9×10−4), while the haplotype constituted by both of the non-risk alleles (rs66801661G–rs62339994G) was decreased (P = 0.0020). A reporter assay was carried out to examine the effect of the IRF2 haplotypes on the transcriptional activity, and association of the IRF2 risk haplotype with higher transcriptional activity was detected in Jurkat T cells under IFNγ stimulation (Tukey's test, P = 1.2×10−4). In conclusion, our observations supported the association of IRF2 with susceptibility to SLE, and the risk haplotype was suggested to be associated with transcriptional activation of IRF2.
Collapse
Affiliation(s)
- Aya Kawasaki
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiroshi Furukawa
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Kanagawa, Japan
| | - Nao Nishida
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| | - Eiji Warabi
- Environmental Molecular Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yuya Kondo
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoshi Ito
- Department of Rheumatology, Niigata Rheumatic Center, Shibata, Niigata, Japan
| | - Isao Matsumoto
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Hirofumi Amano
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Akiko Suda
- Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Shouhei Nagaoka
- Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Kanagawa, Japan
| | - Keigo Setoguchi
- Allergy and Immunological Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Tatsuo Nagai
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Shunsei Hirohata
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Kota Shimada
- Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Shoji Sugii
- Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Tokyo, Japan
| | - Akira Okamoto
- Department of Rheumatology, Himeji Medical Center, National Hospital Organization, Himeji, Hyogo, Japan
| | - Noriyuki Chiba
- Department of Rheumatology, Morioka Hospital, National Hospital Organization, Morioka, Iwate, Japan
| | - Eiichi Suematsu
- Department of Internal Medicine and Rheumatology, Clinical Research Institute, Kyushu Medical Center, National Hospital Organization, Fukuoka, Japan
| | - Shigeru Ohno
- Center for Rheumatic Diseases, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Masao Katayama
- Department of Internal Medicine, Nagoya Medical Center, National Hospital Organization, Nagoya, Aichi, Japan
| | - Akiko Okamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Hajime Kono
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Yoshinari Takasaki
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Takayuki Sumida
- Department of Internal Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, Sagamihara Hospital, National Hospital Organization, Sagamihara, Kanagawa, Japan
| | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
40
|
Variants in the interferon regulatory factor-2 gene are not associated with pancreatitis in Japan. Pancreas 2014; 43:1125-6. [PMID: 25207663 DOI: 10.1097/mpa.0000000000000207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
41
|
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with specific genetic and immunological mechanisms. The rapid development of new techniques in molecular biology had ushered in new discoveries on the role of cytokines, chemokines, and immune cells in the pathogenesis of AD. New polymorphisms of AD are continually being reported in different populations. The physical and immunological barrier of normal intact skin is an important part of the innate immune system that protects the host against microbials and allergens that are associated with AD. Defects in the filaggrin gene FLG may play a role in facilitating exposure to allergens and microbial pathogens, which may induce Th2 polarization. Meanwhile, Th22 cells also play roles in skin barrier impairment through IL-22, and AD is often considered to be a Th2/Th22-dominant allergic disease. Mast cells and eosinophils are also involved in the inflammation via Th2 cytokines. Release of pruritogenic substances by mast cells induces scratching that further disrupts the skin barrier. Th1 and Th17 cells are mainly involved in chronic phase of AD. Keratinocytes also produce proinflammatory cytokines such as thymic stromal lymphopoietin (TSLP), which can further affect Th cells balance. The immunological characteristics of AD may differ for various endotypes and phenotypes. Due to the heterogeneity of the disease, and the redundancies of these mechanisms, our knowledge of the pathophysiology of the disease is still incomplete, which is reflected by the absence of a cure for the disease.
Collapse
Affiliation(s)
- Zhanglei Mu
- Department of Dermatology, Peking University People's Hospital, No11, Xizhimen South Street, Beijing, 100044, China
| | | | | | | | | |
Collapse
|
42
|
Bin L, Edwards MG, Heiser R, Streib JE, Richers B, Hall CF, Leung DYM. Identification of novel gene signatures in patients with atopic dermatitis complicated by eczema herpeticum. J Allergy Clin Immunol 2014; 134:848-55. [PMID: 25159465 PMCID: PMC4186924 DOI: 10.1016/j.jaci.2014.07.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND A subset of patients with atopic dermatitis (AD) is prone to disseminated herpes simplex virus (HSV) infection (ie, atopic dermatitis with a history of eczema herpeticum [ADEH+]). Biomarkers that identify ADEH+ are lacking. OBJECTIVE We sought to search for novel ADEH+ gene signatures in PBMCs. METHODS An RNA-sequencing approach was applied to evaluate global transcriptional changes by using PBMCs from patients with ADEH+ and patients with atopic dermatitis without a history of eczema herpeticum (ADEH-). Candidate genes were confirmed by means of quantitative PCR or ELISA. RESULTS PBMCs from patients with ADEH+ had distinct changes to the transcriptome when compared with those from patients with ADEH- after HSV-1 stimulation: 792 genes were differentially expressed at a false discovery rate of less than 0.05 (ANOVA), and 15 type I and type III interferon genes were among the top 20 most downregulated genes in patients with ADEH+. We further validated that IFN-α and IL-29 mRNA and protein levels were significantly decreased in HSV-1-stimulated PBMCs from patients with ADEH+ compared with those from patients with ADEH- and healthy subjects. Ingenuity Pathway Analysis demonstrated that the upstream regulators of type I and type III interferons, interferon regulatory factor (IRF) 3 and IRF7, were significantly inhibited in patients with ADEH+ based on the downregulation of their target genes. Furthermore, we found that gene expression of IRF3 and IRF7 was significantly decreased in HSV-1-stimulated PBMCs from patients with ADEH+. CONCLUSIONS PBMCs from patients with ADEH+ have a distinct immune response after HSV-1 exposure compared with those from patients with ADEH-. Inhibition of the IRF3 and IRF7 innate immune pathways in patients with ADEH+ might be an important mechanism for increased susceptibility to disseminated viral infection.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Michael G Edwards
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colo
| | - Ryan Heiser
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Joanne E Streib
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Clifton F Hall
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colo; Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo.
| |
Collapse
|
43
|
Clinical Management of Atopic Dermatitis: Practical Highlights and Updates from the Atopic Dermatitis Practice Parameter 2012. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2014; 2:361-9; quiz 370. [DOI: 10.1016/j.jaip.2014.02.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/25/2014] [Accepted: 02/26/2014] [Indexed: 11/20/2022]
|
44
|
Eyerich K, Novak N. Immunology of atopic eczema: overcoming the Th1/Th2 paradigm. Allergy 2013; 68:974-82. [PMID: 23889510 DOI: 10.1111/all.12184] [Citation(s) in RCA: 203] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2013] [Indexed: 12/12/2022]
Abstract
Atopic eczema (AE) is a challenge for modern medicine, because it is prevalent, severely affects quality of life of patients and their families, and causes high socioeconomic costs. The pathogenesis of AE is complex. While initial studies suggested a Th2 deviation as primary reason for the disease, numerous studies addressed a genetically predetermined impaired epidermal barrier as leading cause in a subgroup of patients. Recently, immune changes beyond the initial Th2 concept were defined in AE, with a role for specialized dendritic cells as well as newly identified T helper cell subsets such as Th17 and Th22 cells. Furthermore, trigger factors are expanded beyond classical Th2 allergens such as pollen or house dust mites to microbial products as well as self-antigens. This review pieces together our current understanding of immune as well as barrier abnormalities into the pathogenesis mosaic of AE.
Collapse
Affiliation(s)
| | - N. Novak
- Department of Dermatology and Allergy; University of Bonn Medical Center; Bonn; Germany
| |
Collapse
|
45
|
Dreyfus DH. Herpesviruses and the microbiome. J Allergy Clin Immunol 2013; 132:1278-86. [PMID: 23611298 DOI: 10.1016/j.jaci.2013.02.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 02/07/2013] [Accepted: 02/25/2013] [Indexed: 12/15/2022]
Abstract
The focus of this article will be to examine the role of common herpesviruses as a component of the microbiome of atopic patients and to review clinical observations suggesting that atopic patients might be predisposed to more severe and atypical herpes-related illness because their immune response is biased toward a TH2 cytokine profile. Human populations are infected with 8 herpesviruses, including herpes simplex virus HSV1 and HSV2 (also termed HHV1 and HHV2), varicella zoster virus (VZV or HHV3), EBV (HHV4), cytomegalovirus (HHV5), HHV6, HHV7, and Kaposi sarcoma-associated herpesvirus (termed KSV or HHV8). Herpesviruses are highly adapted to lifelong infection of their human hosts and thus can be considered a component of the human "microbiome" in addition to their role in illness triggered by primary infection. HSV1 and HSV2 infection and reactivation can present with more severe cutaneous symptoms termed eczema herpeticum in the atopic population, similar to the more severe eczema vaccinatum, and drug reaction with eosinophilia and systemic symptoms syndrome (DRESS) is associated with reactivation of HSV6 and possibly other herpesviruses in both atopic and nonatopic patients. In this review evidence is reviewed that primary infection with herpesviruses may have an atypical presentation in the atopic patient and conversely that childhood infection might alter the atopic phenotype. Reactivation of latent herpesviruses can directly alter host cytokine profiles through viral expression of cytokine-like proteins, such as IL-10 (EBV) or IL-6 (cytomegalovirus and HHV8), viral encoded and secreted siRNA and microRNAs, and modulation of expression of host transcription pathways, such as nuclear factor κB. Physicians caring for allergic and atopic populations should be aware of common and uncommon presentations of herpes-related disease in atopic patients to provide accurate diagnosis and avoid unnecessary laboratory testing or incorrect diagnosis of other conditions, such as drug allergy or autoimmune disease. Antiviral therapy and vaccines should be administered promptly when indicated clinically.
Collapse
Affiliation(s)
- David H Dreyfus
- Department of Pediatrics, Clinical Faculty, Yale School of Medicine, New Haven, and the Center for Allergy, Asthma, and Immunology, Waterbury, Conn.
| |
Collapse
|
46
|
Bin L, Kim BE, Brauweiler A, Goleva E, Streib J, Ji Y, Schlievert PM, Leung DYM. Staphylococcus aureus α-toxin modulates skin host response to viral infection. J Allergy Clin Immunol 2012; 130:683-691.e2. [PMID: 22840852 DOI: 10.1016/j.jaci.2012.06.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 06/13/2012] [Indexed: 10/28/2022]
Abstract
BACKGROUND Patients with atopic dermatitis (AD) with a history of eczema herpeticum have increased staphylococcal colonization and infections. However, whether Staphylococcus aureus alters the outcome of skin viral infection has not been determined. OBJECTIVE We investigated whether S aureus toxins modulated host response to herpes simplex virus (HSV) 1 and vaccinia virus (VV) infections in normal human keratinocytes (NHKs) and in murine infection models. METHODS NHKs were treated with S aureus toxins before incubation of viruses. BALB/c mice were inoculated with S aureus 2 days before VV scarification. Viral loads of HSV-1 and VV were evaluated by using real-time PCR, a viral plaque-forming assay, and immunofluorescence staining. Small interfering RNA duplexes were used to knockdown the gene expression of the cellular receptor of α-toxin, a disintegrin and metalloprotease 10 (ADAM10). ADAM10 protein and α-toxin heptamers were detected by using Western blot assays. RESULTS We demonstrate that sublytic staphylococcal α-toxin increases viral loads of HSV-1 and VV in NHKs. Furthermore, we demonstrate in vivo that the VV load is significantly greater (P < .05) in murine skin inoculated with an α-toxin-producing S aureus strain compared with murine skin inoculated with the isogenic α-toxin-deleted strain. The viral enhancing effect of α-toxin is mediated by ADAM10 and is associated with its pore-forming property. Moreover, we demonstrate that α-toxin promotes viral entry in NHKs. CONCLUSION The current study introduces the novel concept that staphylococcal α-toxin promotes viral skin infection and provides a mechanism by which S aureus infection might predispose the host toward disseminated viral infections.
Collapse
Affiliation(s)
- Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | | | | | | | | | | | | | | |
Collapse
|