1
|
Emmert H, Rademacher F, Hübenthal M, Gläser R, Norsgaard H, Weidinger S, Harder J. Type 2 Cytokine-Dependent Skin Barrier Regulation in Personalized 2-Dimensional and 3-Dimensional Skin Models of Atopic Dermatitis: A Pilot Study. JID INNOVATIONS 2025; 5:100309. [PMID: 39385749 PMCID: PMC11460444 DOI: 10.1016/j.xjidi.2024.100309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 10/12/2024] Open
Abstract
Keratinocytes (KCs) from healthy donors stimulated with type 2 cytokines are often used to experimentally study atopic dermatitis (AD) inflammatory responses. Owing to potential intrinsic alterations, it seems favorable to use KCs from patients with AD. KCs isolated from hair follicles offer a noninvasive approach to investigate AD-derived KCs. To evaluate whether such AD-derived KCs are suitable to mimic AD inflammatory responses, we compared hair follicle-derived KCs from healthy donors with those from patients with AD in a type 2 cytokine environment. Stimulation of AD-derived KCs with IL-4 and IL-13 induced higher expression changes of AD-associated markers than that of healthy KCs. The combination of IL-4 and IL-13 generally induced highest expression changes, but IL-13 alone also induced significant changes of AD-specific markers. Similar to the 2-dimensional cultures, IL-4/IL-13 stimulation of 3-dimensional skin models generated with AD-derived KCs modulated the expression of several AD-relevant factors. Whole-transcriptome analysis revealed that IL-4 and IL-13 acted similarly on these 3-dimensional skin models. Histologically, IL-13 alone and in combination with IL-4 increased epidermal spongiosis, a histological hallmark of AD skin. Taken together, our pilot study suggests that hair follicle-derived KCs from patients with AD represent a useful model system to study AD-related inflammation in a personalized in vitro model.
Collapse
Affiliation(s)
- Hila Emmert
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Franziska Rademacher
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Hübenthal
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Regine Gläser
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hanne Norsgaard
- Department of Translational Sciences, Research & Early Development, LEO Pharma, Ballerup, Denmark
| | - Stephan Weidinger
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
2
|
Chu H, Jung YW, Kim SM, Bae BG, Bae JM, Kim LK, Lee KH, Park CO. Exploring the Interplay: Obesity, Lipid Metabolism, and Clinical Manifestations in Atopic Dermatitis. Ann Dermatol 2024; 36:310-317. [PMID: 39343758 PMCID: PMC11439982 DOI: 10.5021/ad.23.151] [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: 12/08/2023] [Revised: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The interest toward the association between atopic dermatitis (AD) and obesity is increasing, yet the possibility of abnormal lipid metabolism has never been investigated before. OBJECTIVE To identify the characteristics of patients with AD who are obese and analyze the serum lipid profiles of these patients. METHODS This observational study included 167 patients diagnosed with AD and underwent evaluations for serum lipid panels between July 2017 and October 2017. The patients' body mass index was used to determine obesity and the serum lipid panels were analyzed between patients who are obese and non-obese. Also, the disease severity and subjective symptoms were evaluated and serum total immunoglobulin E (IgE) and specific IgE levels were assessed. RESULTS Of the 167 patients with AD, there were 36 obese and 131 non-obese subjects. Obese patients were found to have a significantly higher disease severity, as well as higher serum triglyceride and low-density lipoprotein-cholesterol levels and lower high-density lipoprotein-cholesterol levels, which was most pronounced in adult males. Obesity AD patients also had a significantly higher serum total IgE, itch intensity score, and number of allergens sensitized. CONCLUSION Patients with AD who are obese should be monitored for possible abnormalities in lipid metabolism. Obesity may be a factor that contributes to a higher disease severity of AD.
Collapse
Affiliation(s)
- Howard Chu
- Department of Dermatology, Severance Hospital and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yeon Woo Jung
- Department of Dermatology, Severance Hospital and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Su Min Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Byung Gi Bae
- Department of Dermatology, Severance Hospital and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Min Bae
- Department of Dermatology, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang Hoon Lee
- Department of Dermatology, Severance Hospital and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Chang Ook Park
- Department of Dermatology, Severance Hospital and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
3
|
Borrego-Ruiz A, Borrego JJ. Microbial Dysbiosis in the Skin Microbiome and Its Psychological Consequences. Microorganisms 2024; 12:1908. [PMID: 39338582 PMCID: PMC11433878 DOI: 10.3390/microorganisms12091908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
The homeostasis of the skin microbiome can be disrupted by both extrinsic and intrinsic factors, leading to a state of dysbiosis. This imbalance has been observed at the onset of persistent skin diseases that are closely linked to mental health conditions like anxiety and depression. This narrative review explores recent findings on the relationship between the skin microbiome and the pathophysiology of specific skin disorders, including acne vulgaris, atopic dermatitis, psoriasis, and wound infections. Additionally, it examines the psychological impact of these skin disorders, emphasizing their effect on patients' quality of life and their association with significant psychological consequences, such as anxiety, depression, stress, and suicidal ideation in the most severe cases.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| | - Juan J Borrego
- Departamento de Microbiología, Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
4
|
Kenney HM, Battaglia J, Herman K, Beck LA. Atopic dermatitis and IgE-mediated food allergy: Common biologic targets for therapy and prevention. Ann Allergy Asthma Immunol 2024; 133:262-277. [PMID: 38908432 DOI: 10.1016/j.anai.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE To highlight common mechanistic targets for the treatment of atopic dermatitis (AD) and IgE-mediated food allergy (IgE-FA) with potential to be effective for both diseases and prevent atopic progression. DATA SOURCES Data sources were PubMed searches or National Clinical Trials (NCT)-registered clinical trials related to AD, IgE-FA, and other atopic conditions, especially focused on the pediatric population. STUDY SELECTIONS Human seminal studies and/or articles published in the past decade were emphasized with reference to preclinical models when relevant. NCT-registered clinical trials were filtered by inclusion of pediatric subjects younger than 18 years with special focus on children younger than 12 years as a critical period when AD and IgE-FA diseases may often be concurrent. RESULTS AD and IgE-FA share several pathophysiologic features, including epithelial barrier dysfunction, innate and adaptive immune abnormalities, and microbial dysbiosis, which may be critical for the clinical progression between these diseases. Revolutionary advances in targeted biologic therapies have shown the benefit of inhibiting type 2 immune responses, using dupilumab (anti-interleukin-4Rα) or omalizumab (anti-IgE), to potentially reduce symptom burden for both diseases in pediatric populations. Although the potential for biologics to promote disease remission (AD) or sustained unresponsiveness (IgE-FA) remains unclear, the refinement of biomarkers to predict infants at risk for atopic disorders provides promise for prevention through timely intervention. CONCLUSION AD and IgE-FA exhibit common features that may be leveraged to develop biologic therapeutic strategies to treat both conditions and even prevent atopic progression. Future studies should be designed with consistent age stratification in the pediatric population and standardized regimens of adjuvant oral immunotherapy or dose escalation (IgE-FA) to improve cross-study interpretation.
Collapse
Affiliation(s)
- H Mark Kenney
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jennifer Battaglia
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Katherine Herman
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Division of Allergy and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
5
|
Quílez C, Bebiano LB, Jones E, Maver U, Meesters L, Parzymies P, Petiot E, Rikken G, Risueño I, Zaidi H, Zidarič T, Bekeschus S, H van den Bogaard E, Caley M, Colley H, López NG, Letsiou S, Marquette C, Maver T, Pereira RF, Tobin DJ, Velasco D. Targeting the Complexity of In Vitro Skin Models: A Review of Cutting-Edge Developments. J Invest Dermatol 2024:S0022-202X(24)01499-4. [PMID: 39127929 DOI: 10.1016/j.jid.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 08/12/2024]
Abstract
Skin in vitro models offer much promise for research, testing drugs, cosmetics, and medical devices, reducing animal testing and extensive clinical trials. There are several in vitro approaches to mimicking human skin behavior, ranging from simple cell monolayer to complex organotypic and bioengineered 3-dimensional models. Some have been approved for preclinical studies in cosmetics, pharmaceuticals, and chemicals. However, development of physiologically reliable in vitro human skin models remains in its infancy. This review reports on advances in in vitro complex skin models to study skin homeostasis, aging, and skin disease.
Collapse
Affiliation(s)
- Cristina Quílez
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Luís B Bebiano
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Eleri Jones
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia; Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Luca Meesters
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Piotr Parzymies
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Emma Petiot
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Gijs Rikken
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ignacio Risueño
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Hamza Zaidi
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Tanja Zidarič
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Sander Bekeschus
- Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Rostock, Germany; ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | | | - Matthew Caley
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Helen Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Nuria Gago López
- Melanoma group, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Athens, Greece; Department of Food Science and Technology, University of West Attica, Athens, Greece
| | - Christophe Marquette
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Tina Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia; Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rúben F Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Desmond J Tobin
- Charles Institute of Dermatology, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Diego Velasco
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.
| |
Collapse
|
6
|
Adhikary PP, Idowu T, Tan Z, Hoang C, Shanta S, Dumbani M, Mappalakayil L, Awasthi B, Bermudez M, Weiner J, Beule D, Wolber G, Page BD, Hedtrich S. Disrupting TSLP-TSLP receptor interactions via putative small molecule inhibitors yields a novel and efficient treatment option for atopic diseases. EMBO Mol Med 2024; 16:1630-1656. [PMID: 38877290 PMCID: PMC11250841 DOI: 10.1038/s44321-024-00085-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/16/2024] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a key player in atopic diseases, which has sparked great interest in therapeutically targeting TSLP. Yet, no small-molecule TSLP inhibitors exist due to the challenges of disrupting the protein-protein interaction between TSLP and its receptor. Here, we report the development of small-molecule TSLP receptor inhibitors using virtual screening and docking of >1,000,000 compounds followed by iterative chemical synthesis. BP79 emerged as our lead compound that effectively abrogates TSLP-triggered cytokines at low micromolar concentrations. For in-depth analysis, we developed a human atopic disease drug discovery platform using multi-organ chips. Here, topical application of BP79 onto atopic skin models that were co-cultivated with lung models and Th2 cells effectively suppressed immune cell infiltration and IL-13, IL-4, TSLP, and periostin secretion, while upregulating skin barrier proteins. RNA-Seq analysis corroborate these findings and indicate protective downstream effects on the lungs. To the best of our knowledge, this represents the first report of a potent putative small molecule TSLPR inhibitor which has the potential to expand the therapeutic and preventive options in atopic diseases.
Collapse
Affiliation(s)
- Partho Protim Adhikary
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Temilolu Idowu
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Zheng Tan
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Christopher Hoang
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Selina Shanta
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Malti Dumbani
- Institute of Pharmacy, Freie Universität of Berlin, Berlin, Germany
| | - Leah Mappalakayil
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Bhuwan Awasthi
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Marcel Bermudez
- Institute of Pharmacy, Freie Universität of Berlin, Berlin, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - January Weiner
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität of Berlin, Berlin, Germany
| | - Brent Dg Page
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.
| |
Collapse
|
7
|
David E, Hawkins K, Shokrian N, Del Duca E, Guttman-Yassky E. Monoclonal antibodies for moderate-to-severe atopic dermatitis: a look at phase III and beyond. Expert Opin Biol Ther 2024; 24:471-489. [PMID: 38888099 DOI: 10.1080/14712598.2024.2368192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION The understanding of atopic dermatitis (AD) pathogenesis has rapidly expanded in recent years, catalyzing the development of new targeted monoclonal antibody treatments for AD. AREAS COVERED This review aims to summarize the latest clinical and molecular data about monoclonal antibodies that are in later stages of development for AD, either in Phase 3 trials or in the pharmacopoeia for up to 5 years, highlighting the biologic underpinning of each drug's mechanism of action and the potential modulation of the AD immune profile. EXPERT OPINION The therapeutic pipeline of AD treatments is speedily progressing, introducing the potential for a personalized medical approach in the near future. Understanding how targeting pathogenic players in AD modifies disease progression and symptomatology is key in improving therapeutic choices for patients and identifying ideal patient candidates.
Collapse
Affiliation(s)
- Eden David
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
| | - Kelly Hawkins
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Albert Einstein College of Medicine, New York, NY, USA
| | - Neda Shokrian
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Department of Dermatology, Albert Einstein College of Medicine, New York, NY, USA
| | - Ester Del Duca
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn school of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
8
|
Graff P, Woerz D, Wilzopolski J, Voss A, Sarrazin J, Blimkie TM, Weiner J, Kershaw O, Panwar P, Hackett T, Lau S, Brömme D, Beule D, Lee YA, Hancock REW, Gruber AD, Bäumer W, Hedtrich S. Extracellular Matrix Remodeling in Atopic Dermatitis Harnesses the Onset of an Asthmatic Phenotype and Is a Potential Contributor to the Atopic March. J Invest Dermatol 2024; 144:1010-1021.e23. [PMID: 37838332 DOI: 10.1016/j.jid.2023.09.278] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/30/2023] [Accepted: 09/02/2023] [Indexed: 10/16/2023]
Abstract
The development of atopic dermatitis in infancy, and subsequent allergies, such as asthma in later childhood, is known as the atopic march. The mechanism is largely unknown, however the course of disease indicates an inter-epithelial crosstalk, through the onset of inflammation in the skin and progression to other mucosal epithelia. In this study, we investigated if and how skin-lung epithelial crosstalk contributes to the development of the atopic march. First, we emulated inter-epithelial crosstalk through indirect coculture of bioengineered atopic-like skin disease models and three-dimensional bronchial epithelial models triggering an asthma-like phenotype in the latter. A subsequent secretome analysis identified thrombospondin-1, CD44, complement factor C3, fibronectin, and syndecan-4 as potentially relevant skin-derived mediators. Because these mediators are extracellular matrix-related proteins, we then studied the involvement of the extracellular matrix, unveiling distinct proteomic, transcriptomic, and ultrastructural differences in atopic samples. The latter indicated extracellular matrix remodeling triggering the release of the above-mentioned mediators. In vivo mouse data showed that exposure to these mediators dysregulated activated circadian clock genes which are increasingly discussed in the context of atopic diseases and asthma development. Our data point toward the existence of a skin-lung axis that could contribute to the atopic march driven by skin extracellular matrix remodeling.
Collapse
Affiliation(s)
- Patrick Graff
- Institute for Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Germany
| | - Dana Woerz
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Jenny Wilzopolski
- Institute of Pharmacology and Toxicology, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Anne Voss
- Institute of Veterinary Pathology, Freie Universität Berlin, Germany
| | - Jana Sarrazin
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Travis M Blimkie
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, British Columbia, Canada
| | - January Weiner
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Olivia Kershaw
- Institute of Veterinary Pathology, Freie Universität Berlin, Germany
| | - Preety Panwar
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia; Centre for Blood Research, University of British Columbia, British Columbia, Canada
| | - Tillie Hackett
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia; Centre for Heart Lung Innovation, St Paul's Hospital, British Columbia, Canada
| | - Susanne Lau
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin, Berlin, Germany
| | - Dieter Brömme
- Department of Oral Biological & Medical Sciences, Faculty of Dentistry, University of British Columbia; Centre for Blood Research, University of British Columbia, British Columbia, Canada
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany
| | - Young-Ae Lee
- Max Delbrück Center for Molecular Medicine, Berlin, Germany, Clinic for Pediatric Allergy, Experimental and Clinical Research Center of Charité Universitätsmedizin Berlin and Max Delbrück Center, Berlin, Germany
| | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, British Columbia, Canada
| | - Achim D Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Germany
| | - Wolfgang Bäumer
- Institute of Pharmacology and Toxicology, Department of Veterinary Medicine, Freie Universität Berlin, Germany
| | - Sarah Hedtrich
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Germany; Max Delbrück Center for Molecular Medicine, Berlin, Germany, Clinic for Pediatric Allergy, Experimental and Clinical Research Center of Charité Universitätsmedizin Berlin and Max Delbrück Center, Berlin, Germany; Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada; Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
9
|
Virolainen SJ, Satish L, Biagini JM, Chaib H, Chang WC, Dexheimer PJ, Dixon MR, Dunn K, Fletcher D, Forney C, Granitto M, Hestand MS, Hurd M, Kauffman K, Lawson L, Martin LJ, Peña LD, Phelan KJ, Shook M, Weirauch MT, Khurana Hershey GK, Kottyan LC. Filaggrin loss-of-function variants are associated with atopic dermatitis phenotypes in a diverse, early-life prospective cohort. JCI Insight 2024; 9:e178258. [PMID: 38564302 PMCID: PMC11141906 DOI: 10.1172/jci.insight.178258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
Loss-of-function (LoF) variants in the filaggrin (FLG) gene are the strongest known genetic risk factor for atopic dermatitis (AD), but the impact of these variants on AD outcomes is poorly understood. We comprehensively identified genetic variants through targeted region sequencing of FLG in children participating in the Mechanisms of Progression of Atopic Dermatitis to Asthma in Children cohort. Twenty FLG LoF variants were identified, including 1 novel variant and 9 variants not previously associated with AD. FLG LoF variants were found in the cohort. Among these children, the presence of 1 or more FLG LoF variants was associated with moderate/severe AD compared with those with mild AD. Children with FLG LoF variants had a higher SCORing for Atopic Dermatitis (SCORAD) and higher likelihood of food allergy within the first 2.5 years of life. LoF variants were associated with higher transepidermal water loss (TEWL) in both lesional and nonlesional skin. Collectively, our study identifies established and potentially novel AD-associated FLG LoF variants and associates FLG LoF variants with higher TEWL in lesional and nonlesional skin.
Collapse
Affiliation(s)
- Samuel J. Virolainen
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Immunology Graduate Program and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Latha Satish
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Jocelyn M. Biagini
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Hassan Chaib
- Division of Human Genetics and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Wan Chi Chang
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Phillip J. Dexheimer
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | | | - Katelyn Dunn
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | | | - Carmy Forney
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Marissa Granitto
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | | | - Makenna Hurd
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Kenneth Kauffman
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Lucinda Lawson
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Lisa J. Martin
- Division of Human Genetics and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Loren D.M. Peña
- Division of Human Genetics and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Kieran J. Phelan
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Molly Shook
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Matthew T. Weirauch
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Immunology Graduate Program and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Divisions of Developmental Biology and Bioinformatics and Allergy and Immunology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| | - Gurjit K. Khurana Hershey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Asthma Research, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Leah C. Kottyan
- Division of Human Genetics and
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
- Immunology Graduate Program and
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Divisions of Developmental Biology and Bioinformatics and Allergy and Immunology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA
| |
Collapse
|
10
|
Dong S, Li D, Shi D. Skin barrier-inflammatory pathway is a driver of the psoriasis-atopic dermatitis transition. Front Med (Lausanne) 2024; 11:1335551. [PMID: 38606161 PMCID: PMC11007107 DOI: 10.3389/fmed.2024.1335551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/13/2024] [Indexed: 04/13/2024] Open
Abstract
As chronic inflammatory conditions driven by immune dysregulation are influenced by genetics and environment factors, psoriasis and atopic dermatitis (AD) have traditionally been considered to be distinct diseases characterized by different T cell responses. Psoriasis, associated with type 17 helper T (Th17)-mediated inflammation, presents as well-defined scaly plaques with minimal pruritus. AD, primarily linked to Th2-mediated inflammation, presents with poorly defined erythema, dry skin, and intense itching. However, psoriasis and AD may overlap or transition into one another spontaneously, independent of biological agent usage. Emerging evidence suggests that defects in skin barrier-related molecules interact with the polarization of T cells, which forms a skin barrier-inflammatory loop with them. This loop contributes to the chronicity of the primary disease or the transition between psoriasis and AD. This review aimed to elucidate the mechanisms underlying skin barrier defects in driving the overlap between psoriasis and AD. In this review, the importance of repairing the skin barrier was underscored, and the significance of tailoring biologic treatments based on individual immune status instead of solely adhering to the treatment guidelines for AD or psoriasis was emphasized.
Collapse
Affiliation(s)
- Sitan Dong
- College of Clinical Medicine, Jining Medical University, Jining, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Dongmei Shi
- Department of Dermatology/Laboratory of Medical Mycology, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
11
|
Chen HW, Liu FC, Kuo HM, Tang SH, Niu GH, Zhang MM, Tsou LK, Sung PJ, Wen ZH. Immunomodulatory and anti-angiogenesis effects of excavatolide B and its derivatives in alleviating atopic dermatitis. Biomed Pharmacother 2024; 172:116279. [PMID: 38368838 DOI: 10.1016/j.biopha.2024.116279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition primarily driven by T helper 2 (Th2) cytokines, resulting in skin barrier defects, angiogenesis, and inflammatory responses. The marine natural product excavatolide B (EXCB), isolated from the Formosan Gorgonian coral Briareum stechei, exhibits anti-inflammatory and analgesic properties. To enhance solubility, EXCB is chemically modified into the derivatives EXCB-61 salt and EXCB-79. The study aims to investigate the therapeutic effects of these compounds on dinitrochlorbenzene (DNCB)-induced skin damage and to elucidate the underlying anti-inflammatory and anti-angiogenesis mechanism. In vitro, using lipopolysaccharide (LPS)-induced RAW 264.7 cells, all compounds at 10 μM significantly inhibited expression of inflammatory proteins (inducible nitric oxide synthase and cyclooxygenase-2), vascular endothelial growth factor (VEGF), and cytokines (interleukin (IL)-1β, IL-6, and IL-17A). In vivo, topical application of these compounds on DNCB-induced AD mice alleviated skin symptoms, reduced serum levels of IgE, IL-4, IL-13, IL-17, and interferon-γ, and moderated histological phenomena such as hyperplasia, inflammatory cell infiltration, and angiogenesis. The three compounds restored the expression of skin barrier-related proteins (loricrin, filaggrin, and claudin-1) and reduced the expression of angiogenesis-related proteins (VEGF and platelet endothelial cell adhesion molecule-CD31) in the tissues. This is the first study to indicate that EXCB, EXCB-61 salt, and EXCB-79 can treat AD disease by reducing inflammation and angiogenesis. Hence, they may be considered potential candidates for the development of new drugs for AD.
Collapse
Affiliation(s)
- Hsiu-Wen Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Shih-Hsuan Tang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan.
| | - Ping-Jyun Sung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|
12
|
Wang X, Mao D, Jia J, Zhang J. Benvitimod Inhibits IL-4- and IL-13-Induced Tight Junction Impairment by Activating AHR/ARNT Pathway and Inhibiting STAT6 Phosphorylation in Human Keratinocytes. J Invest Dermatol 2024; 144:509-519.e7. [PMID: 37734479 DOI: 10.1016/j.jid.2023.07.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 07/05/2023] [Accepted: 07/22/2023] [Indexed: 09/23/2023]
Abstract
Tight junctions are involved in skin barrier functions. In this study, the expression of CLDN1, CLDN4, and OCLN was found to decrease in skin lesions of atopic dermatitis by bioinformatics analysis. Immunohistochemistry staining in skin specimens from 12 patients with atopic dermatitis and 12 healthy controls also showed decreased CLDN1, CLDN4, and OCLN expression in atopic dermatitis lesions. In vitro studies showed that IL-4 and IL-13 downregulated CLDN1, CLDN4, and OCLN expression in HaCaT cells as well as CLDN4 and OCLN expression in human primary keratinocytes. This effect, which was mediated through the Jak-signal transducer and activator of transcription 6 signaling pathway, increased paracellular flux of 4-kDa dextran. Benvitimod, a new drug for atopic dermatitis, upregulated CLDN4 and OCLN through the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator pathway. Benvitimod induced nuclear translocation of NRF2 and reduced production of ROS in keratinocytes, thus inhibiting IL-4-/IL-13-induced CLDN1 downregulation and signal transducer and activator of transcription 6 phosphorylation. These results indicate that T helper 2 cytokines are involved in tight junction impairment, and benvitimod can inhibit these effects.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Dandan Mao
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Jun Jia
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
13
|
Shin HY, Jin HJ, Tae HJ, Oh HG, Hwang JH. Establishment of an experimental model of ovalbumin-induced atopic dermatitis in canines. Front Vet Sci 2024; 11:1296138. [PMID: 38304543 PMCID: PMC10830637 DOI: 10.3389/fvets.2024.1296138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
Introduction A reliable standard model is required to evaluate the efficacy of new drugs for companion animals, especially dogs. Canine atopic dermatitis (cAD), also known as allergic inflammatory skin disease, is a common condition. Currently, the house dust mite animal model is used in the research of cAD; however, this model exhibits significant individual variation and is difficult to standardize. In this study, we used ovalbumin as an antigen to sensitize and stimulate dogs, thereby establishing a stable model mimicking the T-helper 2 (Th2) response seen in cAD. Our objective was to create a cAD model that could be employed to evaluate the efficacy of novel drugs and mimic the Th2 dominant allergic response observed in the pathogenesis of atopic dermatitis of dogs. Methods In this study, six beagles were used. Normal saline was applied to two animals, and ovalbumin to four, on their dorsal skin. Results The ovalbumin-treated groups exhibited clinical cAD symptoms, such as pruritus and erythema. Moreover, plasma levels of the cAD markers immunoglobulin E and CCL17 chemokine were higher in the ovalbumin-treated group than in the vehicle control group. The skin thickness of the epidermis was significantly increased in the ovalbumin-treated group, with infiltration of inflammatory cells observed in the thickened dermis region. In conclusion, treatment of canine skin with an optimal concentration of ovalbumin induced typical cAD-like symptoms, and histological and molecular analyses confirmed an enhanced Th2-related immune response. Conclusion Therefore, we successfully established a suitable Th2-dominant response mimicking cAD, which will facilitate targeted research of atopic dermatitis in dogs.
Collapse
Affiliation(s)
- Ha-Young Shin
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Republic of Korea
- Companion Animal New Drug Development Center, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Hyung Jun Jin
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Republic of Korea
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do, Republic of Korea
| | - Hyun-Jin Tae
- Companion Animal New Drug Development Center, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Hong-Geun Oh
- R&D Division, HUVET Co., Ltd., Iksan-si, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Korea Institute of Toxicology, Jeongeup, Republic of Korea
- College of Veterinary Medicine and Biosafety Research Institute, Jeonbuk National University, Iksan, Jeollabuk-do, Republic of Korea
| |
Collapse
|
14
|
Bolsoni J, Liu D, Mohabatpour F, Ebner R, Sadhnani G, Tafech B, Leung J, Shanta S, An K, Morin T, Chen Y, Arguello A, Choate K, Jan E, Ross CJ, Brambilla D, Witzigmann D, Kulkarni J, Cullis PR, Hedtrich S. Lipid Nanoparticle-Mediated Hit-and-Run Approaches Yield Efficient and Safe In Situ Gene Editing in Human Skin. ACS NANO 2023; 17:22046-22059. [PMID: 37918441 PMCID: PMC10655174 DOI: 10.1021/acsnano.3c08644] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Despite exciting advances in gene editing, the efficient delivery of genetic tools to extrahepatic tissues remains challenging. This holds particularly true for the skin, which poses a highly restrictive delivery barrier. In this study, we ran a head-to-head comparison between Cas9 mRNA or ribonucleoprotein (RNP)-loaded lipid nanoparticles (LNPs) to deliver gene editing tools into epidermal layers of human skin, aiming for in situ gene editing. We observed distinct LNP composition and cell-specific effects such as an extended presence of RNP in slow-cycling epithelial cells for up to 72 h. While obtaining similar gene editing rates using Cas9 RNP and mRNA with MC3-based LNPs (10-16%), mRNA-loaded LNPs proved to be more cytotoxic. Interestingly, ionizable lipids with a pKa ∼ 7.1 yielded superior gene editing rates (55%-72%) in two-dimensional (2D) epithelial cells while no single guide RNA-dependent off-target effects were detectable. Unexpectedly, these high 2D editing efficacies did not translate to actual skin tissue where overall gene editing rates between 5%-12% were achieved after a single application and irrespective of the LNP composition. Finally, we successfully base-corrected a disease-causing mutation with an efficacy of ∼5% in autosomal recessive congenital ichthyosis patient cells, showcasing the potential of this strategy for the treatment of monogenic skin diseases. Taken together, this study demonstrates the feasibility of an in situ correction of disease-causing mutations in the skin that could provide effective treatment and potentially even a cure for rare, monogenic, and common skin diseases.
Collapse
Affiliation(s)
- Juliana Bolsoni
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Danny Liu
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Fatemeh Mohabatpour
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Ronja Ebner
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Gaurav Sadhnani
- Berlin
Institute of Health @ Charité Universitätsmedizin, Berlin 10117, Germany
| | - Belal Tafech
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Jerry Leung
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Selina Shanta
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Kevin An
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Tessa Morin
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Yihang Chen
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Alfonso Arguello
- University
of Montréal, Faculty of Pharmacy, Montréal H3T 1J4, Quebec, Canada
| | - Keith Choate
- Departments
of Dermatology, Genetics, and Pathology, Yale University School of Medicine, New Haven 06510, Connecticut, United States
| | - Eric Jan
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Colin J.D. Ross
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
| | - Davide Brambilla
- University
of Montréal, Faculty of Pharmacy, Montréal H3T 1J4, Quebec, Canada
| | - Dominik Witzigmann
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Jayesh Kulkarni
- NanoVation
Therapeutics, 2405 Wesbrook
Mall, Vancouver V6T 1Z3, BC, Canada
| | - Pieter R. Cullis
- Department
of Biochemistry and Molecular Biology, University
of British Columbia, 2350 Health Sciences Mall, Vancouver V6T 1Z3, BC, Canada
| | - Sarah Hedtrich
- Faculty
of Pharmaceutical Sciences, University of
British Columbia, 2405 Wesbrook Mall, Vancouver V6T 1Z3, BC, Canada
- Berlin
Institute of Health @ Charité Universitätsmedizin, Berlin 10117, Germany
- Department
of Infectious Diseases and Respiratory Medicine, Charité -
Universitätsmedizin Berlin, corporate
member of Freie Universität Berlin and Humboldt Universität, Berlin 10117, Germany
- Max-Delbrück
Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin 13125, Germany
| |
Collapse
|
15
|
Simpson EL, Guttman-Yassky E, Eichenfield LF, Boguniewicz M, Bieber T, Schneider S, Guana A, Silverberg JI. Tralokinumab therapy for moderate-to-severe atopic dermatitis: Clinical outcomes with targeted IL-13 inhibition. Allergy 2023; 78:2875-2891. [PMID: 37455359 DOI: 10.1111/all.15811] [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: 01/19/2023] [Revised: 05/15/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory, intensely pruritic skin disorder associated with significant patient burden. Interleukin (IL)-13 is a cytokine that acts as a driver of immune dysregulation, skin-barrier dysfunction, and microbiome dysbiosis that characterizes AD, and is consistently overexpressed in AD skin. Tralokinumab is a fully human immunoglobulin (Ig) G4 monoclonal antibody that binds specifically to IL-13 with high affinity, thereby inhibiting subsequent downstream IL-13 signaling. Three pivotal phase 3 clinical trials demonstrated that tralokinumab 300 mg every other week, as monotherapy or in combination with topical corticosteroids as needed, provides significant improvements in signs and symptoms of moderate-to-severe AD, as measured by Investigator's Global Assessment 0/1 (clear/almost clear) and Eczema Area and Severity Index-75 at Week 16. Improvements were observed soon after tralokinumab initiation and were maintained over 52 weeks of therapy. Tralokinumab significantly improved patient-reported outcomes such as itch and sleep, and demonstrated a safety profile comparable with placebo; conjunctivitis during tralokinumab therapy was generally mild. Similar results were observed in a phase 3 adolescent trial. The role of IL-13 in the pathophysiology of AD justifies a targeted approach and a wealth of clinical data supports tralokinumab as a new therapeutic option for people with moderate-to-severe AD.
Collapse
Affiliation(s)
- Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Oregon, USA
| | - Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego and Rady Children's Hospital San Diego, San Diego, California, USA
| | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colorado, USA
| | - Thomas Bieber
- Department of Dermatology and Allergy, Christine Kühne-Center for Allergy Research and Education (CK-CARE), University Hospital Bonn, Bonn, Germany
| | | | | | - Jonathan I Silverberg
- Department of Dermatology, George Washington University School of Medicine, Washington, DC, USA
| |
Collapse
|
16
|
Kolkhir P, Akdis CA, Akdis M, Bachert C, Bieber T, Canonica GW, Guttman-Yassky E, Metz M, Mullol J, Palomares O, Renz H, Ständer S, Zuberbier T, Maurer M. Type 2 chronic inflammatory diseases: targets, therapies and unmet needs. Nat Rev Drug Discov 2023; 22:743-767. [PMID: 37528191 DOI: 10.1038/s41573-023-00750-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 08/03/2023]
Abstract
Over the past two decades, significant progress in understanding of the pathogenesis of type 2 chronic inflammatory diseases has enabled the identification of compounds for more than 20 novel targets, which are approved or at various stages of development, finally facilitating a more targeted approach for the treatment of these disorders. Most of these newly identified pathogenic drivers of type 2 inflammation and their corresponding treatments are related to mast cells, eosinophils, T cells, B cells, epithelial cells and sensory nerves. Epithelial barrier defects and dysbiotic microbiomes represent exciting future drug targets for chronic type 2 inflammatory conditions. Here, we review common targets, current treatments and emerging therapies for the treatment of five major type 2 chronic inflammatory diseases - atopic dermatitis, chronic prurigo, chronic urticaria, asthma and chronic rhinosinusitis with nasal polyps - with a high need for targeted therapies. Unmet needs and future directions in the field are discussed.
Collapse
Affiliation(s)
- Pavel Kolkhir
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos, University of Zürich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) Davos, University of Zürich, Davos, Switzerland
| | - Claus Bachert
- Department of Otorhinolaryngology - Head and Neck Surgery, University Hospital of Münster, Münster, Germany
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Division of ENT diseases, Karolinska Hospital, Stockholm, Sweden
| | - Thomas Bieber
- Department of Dermatology and Allergy, University Hospital, Bonn, Germany
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
- Davos Biosciences, Davos, Switzerland
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Asthma & Allergy Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Martin Metz
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Joaquim Mullol
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clínic Barcelona, FRCB-IDIBAPS, Universitat de Barcelona, CIBERES, Barcelona, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Harald Renz
- Institute of Laboratory Medicine, member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps-University Marburg, Marburg, Germany
- Kilimanjaro Christian Medical University College (KCMUCo), Moshi, Tanzania
| | - Sonja Ständer
- Section Pruritus Medicine, Department of Dermatology and Center for Chronic Pruritus, University Hospital Münster, Münster, Germany
| | - Torsten Zuberbier
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Marcus Maurer
- Institute of Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany.
| |
Collapse
|
17
|
Hatano Y, Elias PM. "Outside-to-inside," "inside-to-outside," and "intrinsic" endogenous pathogenic mechanisms in atopic dermatitis: keratinocytes as the key functional cells involved in both permeability barrier dysfunction and immunological alterations. Front Immunol 2023; 14:1239251. [PMID: 37638035 PMCID: PMC10451066 DOI: 10.3389/fimmu.2023.1239251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Permeability barrier disruption has been shown to induce immunological alterations (i.e., an "outside-to-inside" pathogenic mechanism). Conversely, several inflammatory and immunological mechanisms reportedly interrupt permeability barrier homeostasis (i.e., an "inside-to-outside" pathogenic mechanism). It is now widely recognized that alterations of even a single molecule in keratinocytes can lead to not only permeability barrier dysfunction but also to immunological alterations. Such a simultaneous, bidirectional functional change by keratinocytes is herein named an "intrinsic" pathogenic mechanism. Molecules and/or pathways involved in this mechanism could be important not only as factors in disease pathogenesis but also as potential therapeutic targets for inflammatory cutaneous diseases, such as atopic dermatitis, psoriasis, and prurigo nodularis. Elevation of skin surface pH following permeability barrier abrogation comprises one of the key pathogenic phenomena of the "outside-to-inside" mechanism. Not only type 2 cytokines (e.g., IL-4, IL-13, IL-31) but also type 1 (e.g. IFN-γ), and type 3 (e.g., IL-17, IL-22) as well as several other inflammatory factors (e.g. histamine) can disrupt permeability barrier homeostasis and are all considered part of the "inside-to-outside" mechanism. Finally, examples of molecules relevant to the "intrinsic" pathogenic mechanism include keratin 1, filaggrin, and peroxisome proliferator-activated receptor-α (PPARα).
Collapse
Affiliation(s)
- Yutaka Hatano
- Department of Dermatology, Faculty of Medicine, Oita University, Oita, Japan
| | - Peter M. Elias
- Department of Dermatology, University of California, San Francisco and Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
18
|
Hartmann J, Moitinho-Silva L, Sander N, Harder I, Häsler R, Rodriguez E, Haufe E, Kleinheinz A, Abraham S, Heratizadeh A, Weisshaar E, Schäkel K, Handrick C, Augustin M, Wollenberg A, Staubach-Renz P, Ertner K, Sticherling M, Schwarz B, Quist S, Wiemers F, Schenck F, Wildberger J, Tittmann L, Lieb W, Schmitt J, Werfel T, Weidinger S. Dupilumab but not cyclosporine treatment shifts the microbiome toward a healthy skin flora in patients with moderate-to-severe atopic dermatitis. Allergy 2023; 78:2290-2300. [PMID: 37032440 DOI: 10.1111/all.15742] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) patients display an altered skin microbiome which may not only be an indicator but also a driver of inflammation. We aimed to investigate associations among AD patients' skin microbiome, clinical data, and response to systemic therapy in patients of the TREATgermany registry. METHODS Skin swabs of 157 patients were profiled with 16S rRNA gene amplicon sequencing before and after 3 months of treatment with dupilumab or cyclosporine. For comparison, 16s microbiome data from 258 population-based healthy controls were used. Disease severity was assessed using established instruments such as the Eczema Area and Severity Index (EASI). RESULTS We confirmed the previously shown correlation of Staphylococcus aureus abundance and bacterial alpha diversity with AD severity as measured by EASI. Therapy with Dupilumab shifted the bacterial community toward the pattern seen in healthy controls. The relative abundance of Staphylococci and in particular S. aureus significantly decreased on both lesional and non-lesional skin, whereas the abundance of Staphylococcus hominis increased. These changes were largely independent from the degree of clinical improvement and were not observed for cyclosporine. CONCLUSIONS Systemic treatment with dupilumab but not cyclosporine tends to restore a healthy skin microbiome largely independent of the clinical response indicating potential effects of IL-4RA blockade on the microbiome.
Collapse
Affiliation(s)
- Jan Hartmann
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Lucas Moitinho-Silva
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Nicole Sander
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Inken Harder
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Robert Häsler
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Elke Rodriguez
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Eva Haufe
- Center of Evidence-Based Healthcare, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | - Susanne Abraham
- Department of Dermatology, Medical Faculty Carl Gustav Carus, University Allergy Center, TU Dresden, Dresden, Germany
| | - Annice Heratizadeh
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Elke Weisshaar
- Division of Occupational Dermatology, Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | | | - Matthias Augustin
- Institute for Health Services Research in Dermatology Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Andreas Wollenberg
- Clinics and Outpatient Clinics for Dermatology and Allergy, LMU Munich, Munich, Germany
| | - Petra Staubach-Renz
- Department of Dermatology and Allergy, University Medical Center Mainz, Mainz, Germany
| | | | - Michael Sticherling
- Department of Dermatology, University Hospital, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | | | - Sven Quist
- Dermatology Clinic, Helix Medical Excellence Center Mainz, Mainz, Germany
| | | | | | - Julia Wildberger
- Practice Dr. med. Julia Wildberger, Hautmedizin Bad Soden, Bad Soden, Germany
| | - Lukas Tittmann
- Biobank PopGen and Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Jochen Schmitt
- Center of Evidence-Based Healthcare, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Stephan Weidinger
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
19
|
De La Cruz NC, Möckel M, Niehues H, Rübsam M, Malter W, Zinser M, Krummenacher C, Knebel-Mörsdorf D. Ex Vivo Infection of Human Skin Models with Herpes Simplex Virus 1: Accessibility of the Receptor Nectin-1 during Formation or Impairment of Epidermal Barriers Is Restricted by Tight Junctions. J Virol 2023; 97:e0026223. [PMID: 37289055 PMCID: PMC10308952 DOI: 10.1128/jvi.00262-23] [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: 02/17/2023] [Accepted: 05/18/2023] [Indexed: 06/09/2023] Open
Abstract
Herpes simplex virus 1 (HSV-1) must overcome epidermal barriers to reach its receptors on keratinocytes and initiate infection in human skin. The cell-adhesion molecule nectin-1, which is expressed in human epidermis, acts as an efficient receptor for HSV-1 but is not within reach of the virus upon exposure of human skin under nonpathological conditions. Atopic dermatitis skin, however, can provide an entry portal for HSV-1 emphasizing the role of impaired barrier functions. Here, we explored how epidermal barriers impact HSV-1 invasion in human epidermis and influence the accessibility of nectin-1 for the virus. Using human epidermal equivalents, we observed a correlation of the number of infected cells with tight-junction formation, suggesting that mature tight junctions prior to formation of the stratum corneum prevent viral access to nectin-1. Consequently, impaired epidermal barriers driven by Th2-inflammatory cytokines interleukin 4 (IL-4) and IL-13 as well as the genetic predisposition of nonlesional atopic dermatitis keratinocytes correlated with enhanced infection supporting the impact of functional tight junctions for preventing infection in human epidermis. Comparable to E-cadherin, nectin-1 was distributed throughout the epidermal layers and localized just underneath the tight-junctions. While nectin-1 was evenly distributed on primary human keratinocytes in culture, the receptor was enriched at lateral surfaces of basal and suprabasal cells during differentiation. Nectin-1 showed no major redistribution in the thickened atopic dermatitis and IL-4/IL-13-treated human epidermis in which HSV-1 can invade. However, nectin-1 localization toward tight junction components changed, suggesting that defective tight-junction barriers make nectin-1 accessible for HSV-1 which enables facilitated viral penetration. IMPORTANCE Herpes simplex virus 1 (HSV-1) is a widely distributed human pathogen which productively infects epithelia. The open question is which barriers of the highly protected epithelia must the virus overcome to reach its receptor nectin-1. Here, we used human epidermal equivalents to understand how physical barrier formation and nectin-1 distribution contribute to successful viral invasion. Inflammation-induced barrier defects led to facilitated viral penetration strengthening the role of functional tight-junctions in hindering viral access to nectin-1 that is localized just underneath tight junctions and distributed throughout all layers. We also found nectin-1 ubiquitously localized in the epidermis of atopic dermatitis and IL-4/IL-13-treated human skin implying that impaired tight-junctions in combination with a defective cornified layer allow the accessibility of nectin-1 to HSV-1. Our results support that successful invasion of HSV-1 in human skin relies on defective epidermal barriers, which not only include a dysfunctional cornified layer but also depend on impaired tight junctions.
Collapse
Affiliation(s)
- Nydia C. De La Cruz
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Maureen Möckel
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Hanna Niehues
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Matthias Rübsam
- Department Cell Biology of the Skin, University Hospital Cologne, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Response in Aging-associated Diseases, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Wolfram Malter
- Department of Gynecology and Obstetrics, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max Zinser
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Claude Krummenacher
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, New Jersey, USA
| | - Dagmar Knebel-Mörsdorf
- Center for Biochemistry, University Hospital Cologne, University of Cologne, Cologne, Germany
- Department of Pediatrics, University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Loewa A, Feng JJ, Hedtrich S. Human disease models in drug development. NATURE REVIEWS BIOENGINEERING 2023; 1:1-15. [PMID: 37359774 PMCID: PMC10173243 DOI: 10.1038/s44222-023-00063-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 06/20/2023]
Abstract
Biomedical research is undergoing a paradigm shift towards approaches centred on human disease models owing to the notoriously high failure rates of the current drug development process. Major drivers for this transition are the limitations of animal models, which, despite remaining the gold standard in basic and preclinical research, suffer from interspecies differences and poor prediction of human physiological and pathological conditions. To bridge this translational gap, bioengineered human disease models with high clinical mimicry are being developed. In this Review, we discuss preclinical and clinical studies that benefited from these models, focusing on organoids, bioengineered tissue models and organs-on-chips. Furthermore, we provide a high-level design framework to facilitate clinical translation and accelerate drug development using bioengineered human disease models.
Collapse
Affiliation(s)
- Anna Loewa
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - James J. Feng
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
- Department of Mathematics, University of British Columbia, Vancouver, BC Canada
| | - Sarah Hedtrich
- Department of Infectious Diseases and Respiratory Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Center of Biological Design, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC Canada
- Max-Delbrück Center for Molecular Medicine (MCD), Helmholtz Association, Berlin, Germany
| |
Collapse
|
21
|
Th2 Cytokines Affect the Innate Immune Barrier without Impairing the Physical Barrier in a 3D Model of Normal Human Skin. J Clin Med 2023; 12:jcm12051941. [PMID: 36902728 PMCID: PMC10003590 DOI: 10.3390/jcm12051941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
(1) Background: Atopic dermatitis is one of the most common inflammatory skin diseases characterized by T helper (Th) 2 and Th22 cells producing interleukin (IL)-4/IL-13 and IL-22, respectively. The specific contribution of each cytokine to the impairment of the physical and the immune barrier via Toll-like receptors (TLRs) is poorly addressed concerning the epidermal compartment of the skin. (2) Methods: The effect of IL-4, IL-13, IL-22, and the master cytokine IL-23 is evaluated in a 3D model of normal human skin biopsies (n = 7) at the air-liquid interface for 24 and 48 h. We investigated by immunofluorescence the expressions of (i) claudin-1, zonula occludens (ZO)-1 filaggrin, involucrin for the physical barrier and (ii) TLR2, 4, 7, 9, human beta-defensin 2 (hBD-2) for the immune barrier. (3) Results: Th2 cytokines induce spongiosis and fail in impairing tight junction composition, while IL-22 reduces and IL-23 induces claudin-1 expression. IL-4 and IL-13 affect the TLR-mediated barrier largely than IL-22 and IL-23. IL-4 early inhibits hBD-2 expression, while IL-22 and IL-23 induce its distribution. (4) Conclusions: This experimental approach looks to the pathogenesis of AD through molecular epidermal proteins rather than cytokines only and paves the way for tailored patient therapy.
Collapse
|
22
|
The dynamic balance of the skin microbiome across the lifespan. Biochem Soc Trans 2023; 51:71-86. [PMID: 36606709 PMCID: PMC9988004 DOI: 10.1042/bst20220216] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
For decades research has centered on identifying the ideal balanced skin microbiome that prevents disease and on developing therapeutics to foster this balance. However, this single idealized balance may not exist. The skin microbiome changes across the lifespan. This is reflected in the dynamic shifts of the skin microbiome's diverse, inter-connected community of microorganisms with age. While there are core skin microbial taxa, the precise community composition for any individual person is determined by local skin physiology, genetics, microbe-host interactions, and microbe-microbe interactions. As a key interface with the environment, the skin surface and its appendages are also constantly exchanging microbes with close personal contacts and the environment. Hormone fluctuations and immune system maturation also drive age-dependent changes in skin physiology that support different microbial community structures over time. Here, we review recent insights into the factors that shape the skin microbiome throughout life. Collectively, the works summarized within this review highlight how, depending on where we are in lifespan, our skin supports robust microbial communities, while still maintaining microbial features unique to us. This review will also highlight how disruptions to this dynamic microbial balance can influence risk for dermatological diseases as well as impact lifelong health.
Collapse
|
23
|
Koch M, Kockmann T, Rodriguez E, Wehkamp U, Hiebert P, Ben-Yehuda Greenwald M, Stölzl D, Beer HD, Tschachler E, Weidinger S, Werner S, Auf dem Keller U. Quantitative Proteomics Identifies Reduced NRF2 Activity and Mitochondrial Dysfunction in Atopic Dermatitis. J Invest Dermatol 2023; 143:220-231.e7. [PMID: 36108803 DOI: 10.1016/j.jid.2022.08.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 01/25/2023]
Abstract
Atopic dermatitis is the most common inflammatory skin disease and is characterized by a deficient epidermal barrier and cutaneous inflammation. Genetic studies suggest a key role of keratinocytes in atopic dermatitis pathogenesis, but the alterations in the proteome that occur in the full epidermis have not been defined. Using a pressure-cycling technology and data-independent acquisition approach, we performed quantitative proteomics of epidermis from healthy volunteers and lesional and nonlesional patient skin. Results were validated by targeted proteomics using parallel reaction monitoring mass spectrometry and immunofluorescence staining. Proteins that were differentially abundant in the epidermis of patients with atopic dermatitis versus in healthy control reflect the strong inflammation in lesional skin and the defect in keratinocyte differentiation and epidermal stratification that already characterizes nonlesional skin. Most importantly, they reveal impaired activation of the NRF2-antioxidant pathway and reduced abundance of mitochondrial proteins involved in key metabolic pathways in the affected epidermis. Analysis of primary human keratinocytes with small interfering RNA‒mediated NRF2 knockdown revealed that the impaired NRF2 activation and mitochondrial abnormalities are partially interlinked. These results provide insight into the molecular alterations in the epidermis of patients with atopic dermatitis and identify potential targets for pharmaceutical intervention.
Collapse
Affiliation(s)
- Michael Koch
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Tobias Kockmann
- Functional Genomics Center Zurich, University of Zurich/ETH Zürich, Zürich, Switzerland
| | - Elke Rodriguez
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Ulrike Wehkamp
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Paul Hiebert
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | | | - Dora Stölzl
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Stephan Weidinger
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Ulrich Auf dem Keller
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
24
|
Joshi AA, Vocanson M, Nicolas JF, Wolf P, Patra V. Microbial derived antimicrobial peptides as potential therapeutics in atopic dermatitis. Front Immunol 2023; 14:1125635. [PMID: 36761743 PMCID: PMC9907850 DOI: 10.3389/fimmu.2023.1125635] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease that significantly affects the patient's quality of life. A disrupted skin barrier, type 2 cytokine-dominated inflammation, and microbial dysbiosis with increased Staphylococcus aureus colonization are critical components of AD pathogenesis. Patients with AD exhibit decreased expression of antimicrobial peptides (AMPs) which is linked to increased colonization by Staphylococcus aureus. The skin microbiome itself is a source of several AMPs. These host- and microbiome-derived AMPs define the microbial landscape of the skin based on their differential antimicrobial activity against a range of skin microbes or their quorum sensing inhibitory properties. These are particularly important in preventing and limiting dysbiotic colonization with Staphylococcus aureus. In addition, AMPs are critical for immune homeostasis. In this article, we share our perspectives about the implications of microbial derived AMPs in AD patients and their potential effects on overlapping factors involved in AD. We argue and discuss the potential of bacterial AMPs as therapeutics in AD.
Collapse
Affiliation(s)
- Aaroh Anand Joshi
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR 5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Jean-Francois Nicolas
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR 5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France,Department of Allergology & Clinical Immunology, Lyon-Sud University Hospital, Lyon, France
| | - Peter Wolf
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria,BioTechMed Graz, Graz, Austria
| | - Vijaykumar Patra
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria,Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR 5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France,*Correspondence: Vijaykumar Patra,
| |
Collapse
|
25
|
Čelakovská J, Čermákova E, Vaňková R, Boudkova P, Andrýs C, Krejsek J. Sensitisation to molecular components of mites in atopic dermatitis patients. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2022.2100323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- J. Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - E. Čermákova
- Department of Medical Biophysics, Medical Faculty of Charles University, Hradec Králové, Czech republic
| | - R. Vaňková
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - P. Boudkova
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - C. Andrýs
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - J. Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
26
|
Herpes Simplex Virus 1 Can Bypass Impaired Epidermal Barriers upon Ex Vivo Infection of Skin from Atopic Dermatitis Patients. J Virol 2022; 96:e0086422. [PMID: 35969080 PMCID: PMC9472615 DOI: 10.1128/jvi.00864-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To infect its human host, herpes simplex virus 1 (HSV-1) must overcome the protective barriers of skin and mucosa. Here, we addressed whether pathological skin conditions can facilitate viral entry via the skin surface and used ex vivo infection studies to explore viral invasion in atopic dermatitis (AD) skin characterized by disturbed barrier functions. Our focus was on the visualization of the onset of infection in single cells to determine the primary entry portals in the epidermis. After ex vivo infection of lesional AD skin, we observed infected cells in suprabasal layers indicating successful invasion in the epidermis via the skin surface which was never detected in control skin where only sample edges allowed viral access. The redistribution of filaggrin, loricrin, and tight-junction components in the lesional skin samples suggested multiple defective mechanical barriers. To dissect the parameters that contribute to HSV-1 invasion, we induced an AD-like phenotype by adding the Th2 cytokines interleukin 4 (IL-4) and IL-13 to healthy human skin samples. Strikingly, we detected infected cells in the epidermis, implying that the IL-4/IL-13-driven inflammation is sufficient to induce modifications allowing HSV-1 to penetrate the skin surface. In summary, not only did lesional AD skin facilitate HSV-1 penetration but IL-4/IL-13 responses alone allowed virus invasion. Our results suggest that the defective epidermal barriers of AD skin and the inflammation-induced altered barriers in healthy skin can make receptors accessible for HSV-1. IMPORTANCE Herpes simplex virus 1 (HSV-1) can target skin to establish primary infection in the epithelium. While the human skin provides effective barriers against viral invasion under healthy conditions, a prominent example of successful invasion is the disseminated HSV-1 infection in the skin of atopic dermatitis (AD) patients. AD is characterized by impaired epidermal barrier functions, chronic inflammation, and dysbiosis of skin microbiota. We addressed the initial invasion process of HSV-1 in atopic dermatitis skin to understand whether the physical barrier functions are sufficiently disturbed to allow the virus to invade skin and reach its receptors on skin cells. Our results demonstrate that HSV-1 can indeed penetrate and initiate infection in atopic dermatitis skin. Since treatment of skin with IL-4 and IL-13 already resulted in successful invasion, we assume that inflammation-induced barrier defects play an important role for the facilitated access of HSV-1 to its target cells.
Collapse
|
27
|
Leyva-Castillo JM, Sun L, Wu SY, Rockowitz S, Sliz P, Geha R. Single-cell transcriptome profile of mouse skin undergoing antigen-driven allergic inflammation recapitulates findings in atopic dermatitis skin lesions. J Allergy Clin Immunol 2022; 150:373-384. [PMID: 35300986 PMCID: PMC9378429 DOI: 10.1016/j.jaci.2022.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 02/05/2022] [Accepted: 03/03/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Allergic skin inflammation elicited in mice by epicutaneous (EC) sensitization with antigen shares characteristics with human atopic dermatitis (AD). OBJECTIVE We characterized gene expression by single cells in mouse skin undergoing antigen-driven allergic inflammation and compared the results with findings in AD skin lesions. METHODS Mice were EC sensitized by application of ovalbumin (OVA) or saline to tape-stripped skin. Single-cell RNA sequencing was performed on skin cells 12 days later. Flow cytometry analysis was performed to validate results. RESULTS Sequencing identified 7 nonhematopoietic and 6 hematopoietic cell subsets in EC-sensitized mouse skin. OVA sensitization resulted in the expansion in the skin of T cells, dendritic cells, macrophages, mast cells/basophils, fibroblasts, and myocytes cell clusters, and in upregulation of TH2 cytokine gene expression in CD4+ T cells and mast cells/basophils. Genes differentially expressed in OVA-sensitized skin included genes important for inflammation in dendritic cells and macrophages, collagen deposition, and leukocyte migration in fibroblasts, chemotaxis in endothelial cells and skin barrier integrity, and differentiation in KCs-findings that recapitulate those in AD skin lesions. Unexpectedly, mast cells/basophils, rather than T cells, were the major source of Il4 and ll13 in OVA-sensitized mouse skin. In addition, our results suggest novel pathways in fibroblast and endothelial cells that may contribute to allergic skin inflammation. CONCLUSION The gene expression profile of single cells in mouse skin undergoing antigen-driven shares many features with that in AD skin lesions and unveils novel pathways that may be involved in allergic skin inflammation.
Collapse
Affiliation(s)
- Juan Manuel Leyva-Castillo
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA.,Corresponding authors: Juan-Manuel Leyva-Castillo, PhD. Boston Children’s Hospital, Division of Immunology, One Blackfan Circle, Boston, Massachusetts 02115, USA. Phone: 617-919-2465, Fax: 617-730-0528, Raif S. Geha, MD. Boston Children’s Hospital, Division of Immunology, One Blackfan Circle, Boston, Massachusetts 02115, USA. Phone: 617-919-2482, Fax: 617-730-0528,
| | - Liang Sun
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, USA
| | - Shih-Ying Wu
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, USA
| | - Shira Rockowitz
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, USA
| | - Piotr Sliz
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, USA.,Division of Molecular Medicine, Boston Children’s Hospital, Boston, USA
| | - Raif Geha
- Division of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, MA.,Corresponding authors: Juan-Manuel Leyva-Castillo, PhD. Boston Children’s Hospital, Division of Immunology, One Blackfan Circle, Boston, Massachusetts 02115, USA. Phone: 617-919-2465, Fax: 617-730-0528, Raif S. Geha, MD. Boston Children’s Hospital, Division of Immunology, One Blackfan Circle, Boston, Massachusetts 02115, USA. Phone: 617-919-2482, Fax: 617-730-0528,
| |
Collapse
|
28
|
Phang SJ, Basak S, Teh HX, Packirisamy G, Fauzi MB, Kuppusamy UR, Neo YP, Looi ML. Advancements in Extracellular Matrix-Based Biomaterials and Biofabrication of 3D Organotypic Skin Models. ACS Biomater Sci Eng 2022; 8:3220-3241. [PMID: 35861577 DOI: 10.1021/acsbiomaterials.2c00342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Over the last decades, three-dimensional (3D) organotypic skin models have received enormous attention as alternative models to in vivo animal models and in vitro two-dimensional assays. To date, most organotypic skin models have an epidermal layer of keratinocytes and a dermal layer of fibroblasts embedded in an extracellular matrix (ECM)-based biomaterial. The ECM provides mechanical support and biochemical signals to the cells. Without advancements in ECM-based biomaterials and biofabrication technologies, it would have been impossible to create organotypic skin models that mimic native human skin. In this review, the use of ECM-based biomaterials in the reconstruction of skin models, as well as the study of complete ECM-based biomaterials, such as fibroblasts-derived ECM and decellularized ECM as a better biomaterial, will be highlighted. We also discuss the benefits and drawbacks of several biofabrication processes used in the fabrication of ECM-based biomaterials, such as conventional static culture, electrospinning, 3D bioprinting, and skin-on-a-chip. Advancements and future possibilities in modifying ECM-based biomaterials to recreate disease-like skin models will also be highlighted, given the importance of organotypic skin models in disease modeling. Overall, this review provides an overview of the present variety of ECM-based biomaterials and biofabrication technologies available. An enhanced organotypic skin model is expected to be produced in the near future by combining knowledge from previous experiences and current research.
Collapse
Affiliation(s)
- Shou Jin Phang
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Soumyadeep Basak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247 667, Uttarakhand, India
| | - Huey Xhin Teh
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Gopinath Packirisamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247 667, Uttarakhand, India
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yun Ping Neo
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Mee Lee Looi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Ma X, Ru Y, Luo Y, Kuai L, Chen QL, Bai Y, Liu YQ, Chen J, Luo Y, Song JK, Zhou M, Li B. Post-Translational Modifications in Atopic Dermatitis: Current Research and Clinical Relevance. Front Cell Dev Biol 2022; 10:942838. [PMID: 35874824 PMCID: PMC9301047 DOI: 10.3389/fcell.2022.942838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/16/2022] [Indexed: 11/20/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic and relapsing cutaneous disorder characterized by compromised immune system, excessive inflammation, and skin barrier disruption. Post-translational modifications (PTMs) are covalent and enzymatic modifications of proteins after their translation, which have been reported to play roles in inflammatory and allergic diseases. However, less attention has been paid to the effect of PTMs on AD. This review summarized the knowledge of six major classes (including phosphorylation, acetylation, ubiquitination, SUMOylation, glycosylation, o-glycosylation, and glycation) of PTMs in AD pathogenesis and discussed the opportunities for disease management.
Collapse
Affiliation(s)
- Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| | - Qi-Long Chen
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yun Bai
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Ye-Qiang Liu
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Jia Chen
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Yue Luo
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Jian-Kun Song
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
| | - Mi Zhou
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mi Zhou, ; Bin Li,
| | - Bin Li
- Shanghai Skin Disease Hospital of Tongji University, Shanghai, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Mi Zhou, ; Bin Li,
| |
Collapse
|
30
|
Yoshitake R, Nakai H, Ebina M, Kawasaki K, Murosaki S, Hirose Y. Beneficial Effect of Heat-Killed Lactiplantibacillus plantarum L-137 on Skin Functions in Healthy Participants: A Randomized, Placebo-Controlled, Double-Blind Study. Front Med (Lausanne) 2022; 9:912280. [PMID: 35872749 PMCID: PMC9299260 DOI: 10.3389/fmed.2022.912280] [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: 04/05/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
To determine whether consuming heat-killed Lactiplantibacillus plantarum L-137 (HK L-137) influences skin functions, we performed a randomized, placebo-controlled, double-blind study in healthy participants who were conscious of dry skin. A total of 80 healthy participants (20 men, 60 women; mean age, 47.3 years) were assigned to receive a tablet containing HK L-137 or a placebo tablet daily for 12 weeks. Every 4 weeks, the skin water content and transepidermal water loss (TEWL) were measured at the forearm and face, and participants completed two skin-related questionnaires, the Dermatology Life Quality Index and a self-evaluation. The HK L-137 group tended to show greater increases from baseline of water content at the forearm and larger decreases of TEWL at the face. The total scores of both questionnaires improved significantly more in the HK L-137 group. Water content and TEWL improved significantly in participants in the HK L-137 group who were above the median age of study participants or had relatively dry skin. These findings suggest that daily HK L-137 intake can improve dry skin, thereby contributing to skin satisfaction.
Collapse
Affiliation(s)
| | | | | | | | | | - Yoshitaka Hirose
- Research and Development Institute, House Wellness Foods Corp., Itami, Japan
| |
Collapse
|
31
|
Marsella R, Ahrens K, Wilkes R. Differences in Behavior between Normal and Atopic Keratinocytes in Culture: Pilot Studies. Vet Sci 2022; 9:vetsci9070329. [PMID: 35878346 PMCID: PMC9319359 DOI: 10.3390/vetsci9070329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Skin barrier dysfunction is important in atopic dermatitis and can be secondary to inflammation. Observation of keratinocytes in culture may show intrinsic differences. TransEpithelial Electrical Resistance (TEER) measures epithelial permeability. We cultured normal and atopic keratinocytes and found that TEER of atopic keratinocytes was significantly lower (p < 0.0001) than that of normals. Atopic keratinocytes grew upwards, first creating isolated dome-like structures and later horizontally into a monolayer. At time of confluence (D0), atopic keratinocytes were more differentiated, with higher filaggrin gene expression than normals. No differences existed between groups for TJ proteins (claudin, occludin, and Zonula Occludens-1) on D0 and D6. On D6, claudin and occludin were higher than D0, in normal (p = 0.0296 and p = 0.0011) and atopic keratinocytes (p = 0.0348 and 0.0491). Immunofluorescent staining showed nuclear location of filaggrin on D0 and cytoplasmic on D6. ANOVA showed increased cell size from D0 to D6 in both groups (effect of time, p = 0.0076) but no differences between groups. Significant subject effect (p = 0.0022) was found, indicating that cell size was subject-dependent but not disease-dependent. No difference for continuity for TJ protein existed between groups. These observations suggest that decreased TEER in atopics is not linked to TJ differences but is possibly linked to different growth behavior.
Collapse
|
32
|
Park I, Nam H, Goo D, Wickramasuriya SS, Zimmerman N, Smith AH, Rehberger TG, Lillehoj HS. Gut Microbiota-Derived Indole-3-Carboxylate Influences Mucosal Integrity and Immunity Through the Activation of the Aryl Hydrocarbon Receptors and Nutrient Transporters in Broiler Chickens Challenged With Eimeria maxima. Front Immunol 2022; 13:867754. [PMID: 35812452 PMCID: PMC9259858 DOI: 10.3389/fimmu.2022.867754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/26/2022] [Indexed: 11/23/2022] Open
Abstract
Two studies were conducted to evaluate the effects of indole-3-carboxylate (ICOOH) as a postbiotic on maintaining intestinal homeostasis against avian coccidiosis. In the first study, an in vitro culture system was used to investigate the effects of ICOOH on the proinflammatory cytokine response of chicken macrophage cells (CMCs), gut integrity of chicken intestinal epithelial cells (IECs), differentiation of quail muscle cells (QMCs), and primary chicken embryonic muscle cells (PMCs) and anti-parasitic effect against Eimeria maxima. Cells to be tested were seeded in the 24-well plates and treated with ICOOH at concentrations of 0.1, 1.0, and 10.0 µg. CMCs were first stimulated by lipopolysaccharide (LPS) to induce an innate immune response, and QMCs and PMCs were treated with 0.5% and 2% fetal bovine serum, respectively, before they were treated with ICOOH. After 18 h of incubation, cells were harvested, and RT-PCR was performed to measure gene expression of proinflammatory cytokines of CMCs, tight junction (TJ) proteins of IECs, and muscle cell growth markers of QMCs and PMCs. In the second study, in vivo trials were carried out to study the effect of dietary ICOOH on disease parameters in broiler chickens infected with E. maxima. One hundred twenty male broiler chickens (0-day-old) were allocated into the following four treatment groups: 1) basal diet without infection (CON), 2) basal diet with E. maxima (NC), 3) ICOOH at 10.0 mg/kg feed with E. maxima (HI), and 4) ICOOH at 1.0 mg/kg feed with E. maxima (LO). Body weights (BWs) were measured on 0, 7, 14, 20, and 22 days. All groups except the CON chickens were orally infected with E. maxima on day 14. Jejunal samples were collected for lesion score and the transcriptomic analysis of cytokines and TJ proteins. In vitro, ICOOH increased the expression of TJ proteins in IECs and decreased IL-1β and IL-8 transcripts in the LPS-stimulated CMCs. In vivo, chickens on the HI diet showed reduced jejunal IL-1β, IFN-γ, and IL-10 expression and increased expression of genes activated by aryl hydrocarbon receptors and nutrient transporters in E. maxima-infected chickens. In conclusion, these results demonstrate the beneficial effects of dietary ICOOH on intestinal immune responses and barrier integrity in broiler chickens challenged with E. maxima. Furthermore, the present finding supports the notion to use microbial metabolites as novel feed additives to enhance resilience in animal agriculture.
Collapse
Affiliation(s)
- Inkyung Park
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Hyoyoun Nam
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Doyun Goo
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Samiru S. Wickramasuriya
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
| | - Noah Zimmerman
- Arm & Hammer Animal and Food Production, Waukesha, WI, United States
| | | | | | - Hyun S. Lillehoj
- Animal Bioscience and Biotechnology Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, United States Department of Agriculture, Beltsville, MD, United States
- *Correspondence: Hyun S. Lillehoj,
| |
Collapse
|
33
|
Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, Rossi AB. Type 2 Inflammation Contributes to Skin Barrier Dysfunction in Atopic Dermatitis. JID INNOVATIONS 2022; 2:100131. [PMID: 36059592 PMCID: PMC9428921 DOI: 10.1016/j.xjidi.2022.100131] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
Skin barrier dysfunction, a defining feature of atopic dermatitis (AD), arises from multiple interacting systems. In AD, skin inflammation is caused by host-environment interactions involving keratinocytes as well as tissue-resident immune cells such as type 2 innate lymphoid cells, basophils, mast cells, and T helper type 2 cells, which produce type 2 cytokines, including IL-4, IL-5, IL-13, and IL-31. Type 2 inflammation broadly impacts the expression of genes relevant for barrier function, such as intracellular structural proteins, extracellular lipids, and junctional proteins, and enhances Staphylococcus aureus skin colonization. Systemic anti‒type 2 inflammation therapies may improve dysfunctional skin barrier in AD.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- CLDN, claudin
- FFA, free fatty acid
- ILC2, type 2 innate lymphoid cell
- Jaki, Jak inhibitor
- K, keratin
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NMF, natural moisturizing factor
- PAR, protease-activated receptor
- PDE-4, phosphodiesterase-4
- SC, stratum corneum
- SG, stratum granulosum
- TCI, topical calcineurin inhibitor
- TCS, topical corticosteroid
- TEWL, transepidermal water loss
- TJ, tight junction
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor alpha
- TYK, tyrosine kinase
- Th, T helper
- ZO, zona occludens
- hBD, human β-defensin
Collapse
Affiliation(s)
- Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA,Correspondence: Lisa A. Beck, Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, New York 14642, USA.
| | - Michael J. Cork
- Sheffield Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease (IICD), The University of Sheffield, The Medical School, Sheffield, United Kingdom
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
34
|
Graff P, Hönzke S, Joshi AA, Yealland G, Fleige E, Unbehauen M, Schäfer-Korting M, Hocke A, Haag R, Hedtrich S. Preclinical Testing of Dendritic Core-Multishell Nanoparticles in Inflammatory Skin Equivalents. Mol Pharm 2022; 19:1795-1802. [PMID: 35266720 DOI: 10.1021/acs.molpharmaceut.1c00734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Human skin equivalents emerged as novel tools in preclinical dermatological research. It is being claimed that they may bridge the translational gap between preclinical and clinical research, yet only a few studies have investigated their suitability for preclinical drug testing so far. Therefore, we investigated if inflammatory skin equivalents, which emulate hallmarks of atopic dermatitis (AD), are suitable to assess the anti-inflammatory effects of dexamethasone (DXM) in a cream formulation or loaded onto dendritic core-multishell nanoparticles. Topical DXM application resulted in significantly decreased expression of the proinflammatory cytokine TSLP, increased expression of the skin barrier protein involucrin, and facilitated glucocorticoid receptor translocation in a dose-dependent manner. Further, DXM treatment inhibited gene expression of extracellular matrix components, potentially indicative of the known skin atrophy-inducing side effects of glucocorticoids. Overall, we were able to successfully assess the anti-inflammatory effects of DXM and the superiority of the nanoparticle formulation. Nevertheless the identification of robust readout parameters proved challenging and requires careful study design.
Collapse
Affiliation(s)
- Patrick Graff
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Biological Design, 13125 Berlin, Germany
| | - Stefan Hönzke
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany.,Research Unit for Photodermatology, Department of Dermatology and Venereology, Medical University of Graz, 8036 Graz, Austria
| | - Aaroh Anand Joshi
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany.,Department of Nephrology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Guy Yealland
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Emanuel Fleige
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Michael Unbehauen
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Monika Schäfer-Korting
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany
| | - Andreas Hocke
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Sarah Hedtrich
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.,Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Center of Biological Design, 13125 Berlin, Germany.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
35
|
Babity S, Couture F, Campos EVR, Hedtrich S, Hagen R, Fehr D, Bonmarin M, Brambilla D. A Naked Eye-Invisible Ratiometric Fluorescent Microneedle Tattoo for Real-Time Monitoring of Inflammatory Skin Conditions. Adv Healthc Mater 2022; 11:e2102070. [PMID: 34921529 DOI: 10.1002/adhm.202102070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/13/2021] [Indexed: 01/05/2023]
Abstract
The field of portable healthcare monitoring devices has an urgent need for the development of real-time, noninvasive sensing and detection methods for various physiological analytes. Currently, transdermal sensing techniques are severely limited in scope (i.e., measurement of heart rate or sweat composition), or else tend to be invasive, often needing to be performed in a clinical setting. This study proposes a minimally invasive alternative strategy, consisting of using dissolving polymeric microneedles to deliver naked eye-invisible functional fluorescent ratiometric microneedle tattoos directly to the skin for real-time monitoring and quantification of physiological and pathological parameters. Reactive oxygen species are overexpressed in the skin in association with various pathological conditions. Here, one demonstrates for the first time the microneedle-based delivery to the skin of active fluorescent sensors in the form of an invisible, ratiometric microneedle tattoo capable of sensing reactive oxygen species in a reconstructed human-based skin disease model, as well as an in vivo model of UV-induced dermal inflammation. One also elaborates a universal ratiometric quantification concept coupled with a custom-built, multiwavelength portable fluorescence detection system. Fully realized, this approach presents an opportunity for the minimally invasive monitoring of a broad range of physiological parameters through the skin.
Collapse
Affiliation(s)
- Samuel Babity
- Faculté de Pharmacie Université de Montréal C.P. 6128, Succursale Centre‐ville, Montréal Québec H3C 3J7 Canada
| | - Frédéric Couture
- TransBIOTech 201 Monseigneur‐Bourget Lévis Québec G6V 6Z9 Canada
- Nutraceuticals and Functional Foods Institute (INAF) Université Laval, Québec Québec G1K 7P4 Canada
- Centre intégré de santé et de services sociaux de Chaudière‐Appalaches Lévis Québec G6E 3E2 Canada
| | - Estefânia V. R. Campos
- Faculty of Pharmaceutical Sciences University of British Columbia 2405 Wesbrook Mall Vancouver British Columbia V6T 1Z3 Canada
- Human and Natural Sciences Center Federal University of ABC Santo Andre SP 09210‐580 Brazil
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences University of British Columbia 2405 Wesbrook Mall Vancouver British Columbia V6T 1Z3 Canada
| | - Raphael Hagen
- School of Engineering Zurich University of Applied Sciences Technikumstrasse 9 Winterthur 8400 Switzerland
| | - Daniel Fehr
- School of Engineering Zurich University of Applied Sciences Technikumstrasse 9 Winterthur 8400 Switzerland
| | - Mathias Bonmarin
- School of Engineering Zurich University of Applied Sciences Technikumstrasse 9 Winterthur 8400 Switzerland
| | - Davide Brambilla
- Faculté de Pharmacie Université de Montréal C.P. 6128, Succursale Centre‐ville, Montréal Québec H3C 3J7 Canada
| |
Collapse
|
36
|
Fania L, Moretta G, Antonelli F, Scala E, Abeni D, Albanesi C, Madonna S. Multiple Roles for Cytokines in Atopic Dermatitis: From Pathogenic Mediators to Endotype-Specific Biomarkers to Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23052684. [PMID: 35269828 PMCID: PMC8910412 DOI: 10.3390/ijms23052684] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/21/2022] [Accepted: 02/25/2022] [Indexed: 12/20/2022] Open
Abstract
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases, which generally presents with intense itching and recurrent eczematous lesions. AD affects up to 20% of children and 10% of adults in high-income countries. The prevalence and incidence of AD have increased in recent years. The onset of AD mostly occurs in childhood, although in some cases AD may persist in adult life or even manifest in middle age (adult-onset AD). AD pathophysiology is made of a complex net, in which genetic background, skin barrier dysfunction, innate and adaptive immune responses, as well as itch contribute to disease development, progression, and chronicization. One of the most important features of AD is skin dehydration, which is mainly caused by filaggrin mutations that determine trans-epidermal water loss, pH alterations, and antigen penetration. In accordance with the “outside-inside” theory of AD pathogenesis, in a context of an altered epidermal barrier, antigens encounter epidermal antigen presentation cells (APCs), such as epidermal Langerhans cells and inflammatory epidermal dendritic cells, leading to their maturation and Th-2 cell-mediated inflammation. APCs also bear trimeric high-affinity receptors for immunoglobulin E (IgE), which induce IgE-mediated sensitizations as part of pathogenic mechanisms leading to AD. In this review, we discuss the role of cytokines in the pathogenesis of AD, considering patients with various clinical AD phenotypes. Moreover, we describe the cytokine patterns in patients with AD at different phases of the disease evolution, as well as in relation to different phenotypes/endotypes, including age, race, and intrinsic/extrinsic subtypes. We also discuss the outcomes of current biologics for AD, which corroborate the presence of multiple cytokine axes involved in the background of AD. A deep insight into the correlation between cytokine patterns and the related clinical forms of AD is a crucial step towards increasingly personalized, and therefore more efficient therapy.
Collapse
Affiliation(s)
- Luca Fania
- Integrated Center for Research in Atopic Dermatitis (CRI-DA), IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy; (L.F.); (G.M.); (F.A.); (E.S.)
| | - Gaia Moretta
- Integrated Center for Research in Atopic Dermatitis (CRI-DA), IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy; (L.F.); (G.M.); (F.A.); (E.S.)
| | - Flaminia Antonelli
- Integrated Center for Research in Atopic Dermatitis (CRI-DA), IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy; (L.F.); (G.M.); (F.A.); (E.S.)
| | - Enrico Scala
- Integrated Center for Research in Atopic Dermatitis (CRI-DA), IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy; (L.F.); (G.M.); (F.A.); (E.S.)
| | - Damiano Abeni
- Clinical Epidemiology Unit, IDI-IRCCS, 00167 Rome, Italy;
| | - Cristina Albanesi
- Laboratory of Experimental Immunology, IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy;
- Correspondence:
| | - Stefania Madonna
- Laboratory of Experimental Immunology, IDI-IRCCS, Via Monti di Creta, 104, 00167 Rome, Italy;
| |
Collapse
|
37
|
Dai X, Muto J, Shiraishi K, Utsunomiya R, Mori H, Murakami M, Sayama K. TSLP impairs epidermal barrier integrity by stimulating the formation of nuclear IL-33/phosphorylated STAT3 complex in human keratinocytes. J Invest Dermatol 2022; 142:2100-2108.e5. [DOI: 10.1016/j.jid.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
|
38
|
Klinngam W, Rungkamoltip P, Thongin S, Joothamongkhon J, Khumkhrong P, Khongkow M, Namdee K, Tepaamorndech S, Chaikul P, Kanlayavattanakul M, Lourith N, Piboonprai K, Ruktanonchai U, Asawapirom U, Iempridee T. Polymethoxyflavones from Kaempferia parviflora ameliorate skin aging in primary human dermal fibroblasts and ex vivo human skin. Biomed Pharmacother 2021; 145:112461. [PMID: 34839253 DOI: 10.1016/j.biopha.2021.112461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/12/2022] Open
Abstract
Skin aging is accompanied by an increase in the number of senescent cells, resulting in various pathological outcomes. These include inflammation, impaired barrier function, and susceptibility to skin disorders such as cancer. Kaempferia parviflora (Thai black ginger), a medicinal plant native to Thailand, has been shown to counteract inflammation, cancer, and senescence. This study demonstrates that polymethoxyflavones (5,7-dimethoxyflavone, 5,7,4'-trimethoxyflavone, and 3,5,7,3',4'-pentamethoxyflavone) purified from K. parviflora rhizomes suppressed cellular senescence, reactive oxygen species, and the senescence-associated secretory phenotype in primary human dermal fibroblasts. In addition, they increased tropocollagen synthesis and alleviated free radical-induced cellular and mitochondrial damage. Moreover, the compounds mitigated chronological aging in a human ex vivo skin model by attenuating senescence and restoring expression of essential components of the extracellular matrix, including collagen type I, fibrillin-1, and hyaluronic acid. Finally, we report that polymethoxyflavones enhanced epidermal thickness and epidermal-dermal stability, while blocking age-related inflammation in skin explants. Our findings support the use of polymethoxyflavones from K. parviflora as natural anti-aging agents, highlighting their potential as active ingredients in cosmeceutical and nutraceutical products.
Collapse
Affiliation(s)
- Wannita Klinngam
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Phetploy Rungkamoltip
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Saowarose Thongin
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Jaruwan Joothamongkhon
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Phattharachanok Khumkhrong
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Mattaka Khongkow
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Katawut Namdee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Surapun Tepaamorndech
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Puxvadee Chaikul
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Mayuree Kanlayavattanakul
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Nattaya Lourith
- Phytocosmetics and Cosmeceuticals Research Group, School of Cosmetic Science, Mae Fah Luang University, Chiang Rai, Thailand; School of Cosmetic Science, Mae Fah Luang University, Chiang Rai 57100, Thailand
| | - Kitiya Piboonprai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; Laboratory of Host Defense, The World Premier International Research Center Initiative (WPI), Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Uracha Ruktanonchai
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Udom Asawapirom
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand
| | - Tawin Iempridee
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand.
| |
Collapse
|
39
|
Moon S, Kim DH, Shin JU. In Vitro Models Mimicking Immune Response in the Skin. Yonsei Med J 2021; 62:969-980. [PMID: 34672130 PMCID: PMC8542468 DOI: 10.3349/ymj.2021.62.11.969] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022] Open
Abstract
The skin is the first line of defense of our body, and it is composed of the epidermis and dermis with diverse immune cells. Various in vitro models have been investigated to recapitulate the immunological functions of the skin and to model inflammatory skin diseases. The simplest model is a two-dimensional (2D) co-culture system, which helps understand the direct and indirect cell-to-cell interactions between immune and structural cells; however, it has limitations when observing three-dimensional (3D) interactions or reproducing skin barriers. Conversely, 3D skin constructs can mimic the human skin characteristics in terms of epidermal and dermal structures, barrier functions, cell migration, and cell-to-cell interaction in the 3D space. Recently, as the importance of neuro-immune-cutaneous interactions in the inflammatory response is emerging, 3D skin constructs containing both immune cells and neurons are being developed. A microfluidic culture device called "skin-on-a-chip," which simulates the structures and functions of the human skin with perfusion, was also developed to mimic immune cell migration through the vascular system. This review summarizes the in vitro skin models with immune components, focusing on two highly prevalent chronic inflammatory skin diseases: atopic dermatitis and psoriasis. The development of these models will be valuable in studying the pathophysiology of skin diseases and evaluating the efficacy and toxicity of new drugs.
Collapse
Affiliation(s)
- Sujin Moon
- CHA University College of Medicine, Seongnam, Korea
| | - Dong Hyun Kim
- CHA University College of Medicine, Seongnam, Korea
- Department of Dermatology, CHA Bundang Medical Center, Seongnam, Korea
| | - Jung U Shin
- CHA University College of Medicine, Seongnam, Korea
- Department of Dermatology, CHA Bundang Medical Center, Seongnam, Korea.
| |
Collapse
|
40
|
Oh JS, Lee SJ, Choung SY. Lithospermum erythrorhizon Alleviates Atopic Dermatitis-like Skin Lesions by Restoring Immune Balance and Skin Barrier Function in 2.4-Dinitrochlorobenzene-Induced NC/Nga Mice. Nutrients 2021; 13:nu13093209. [PMID: 34579088 PMCID: PMC8470668 DOI: 10.3390/nu13093209] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023] Open
Abstract
The incidence of atopic dermatitis (AD), a disease characterized by an abnormal immune balance and skin barrier function, has increased rapidly in developed countries. This study investigated the anti-atopic effect of Lithospermum erythrorhizon (LE) using NC/Nga mice induced by 2,4-dinitrochlorobenzene. LE reduced AD clinical symptoms, including inflammatory cell infiltration, epidermal thickness, ear thickness, and scratching behavior, in the mice. Additionally, LE reduced serum IgE and histamine levels, and restored the T helper (Th) 1/Th2 immune balance through regulation of the IgG1/IgG2a ratio. LE also reduced the levels of AD-related cytokines and chemokines, including interleukin (IL)-1β, IL-4, IL-6, tumor necrosis factor-α (TNF-α), thymic stromal lymphopoietin, thymus and activation-regulated chemokine, macrophage-derived chemokine, regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 in the serum. Moreover, LE modulated AD-related cytokines and chemokines expressed and secreted by Th1, Th2, Th17, and Th22 cells in the dorsal skin and splenocytes. Furthermore, LE restored skin barrier function by increasing pro-filaggrin gene expression and levels of skin barrier-related proteins filaggrin, involucrin, loricrin, occludin, and zonula occludens-1. These results suggest that LE is a potential therapeutic agent that can alleviate AD by modulating Th1/Th2 immune balance and restoring skin barrier function.
Collapse
Affiliation(s)
- Jin-Su Oh
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | | | - Se-Young Choung
- Department of Life and Nanopharmaceutical Sciences, Graduate School, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Department of Preventive Pharmacy and Toxicology, College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence:
| |
Collapse
|
41
|
Suwanchote S, Waitayangkoon P, Chancheewa B, Inthanachai T, Niwetbowornchai N, Edwards SW, Virakul S, Thammahong A, Kiatsurayanon C, Rerknimitr P, Chiewchengchol D. Role of antimicrobial peptides in atopic dermatitis. Int J Dermatol 2021; 61:532-540. [PMID: 34432296 DOI: 10.1111/ijd.15814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 01/04/2023]
Abstract
Host defense peptides (HDPs) or antimicrobial peptides (AMPs) are short cationic amphipathic peptides of divergent sequences, which are part of the innate immune system and produced by various types of cells and tissues. The predominant role of HDPs is to respond to and protect humans against infection and inflammation. Common human HDPs include defensins, cathelicidin, psoriasin, dermcidin, and ribonucleases, but these peptides may be dysregulated in the skin of patients with atopic dermatitis (AD). Current evidence suggests that the antimicrobial properties and immunomodulatory effects of HDPs are involved in AD pathogenesis, making HDPs research a promising area for predicting disease severity and developing novel treatments for AD. In this review, we describe a potential role for human HDPs in the development, exacerbation, and progression of AD and propose their potential therapeutic benefits.
Collapse
Affiliation(s)
- Supaporn Suwanchote
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Palapun Waitayangkoon
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Bussabong Chancheewa
- Division of Dermatology, Skin and Allergy Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thananya Inthanachai
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nattarika Niwetbowornchai
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Steven W Edwards
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Sita Virakul
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Arsa Thammahong
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chanisa Kiatsurayanon
- Department of Medical Services, Institute of Dermatology, Ministry of Public Health, Bangkok, Thailand
| | - Pawinee Rerknimitr
- Division of Dermatology, Skin and Allergy Research Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Direkrit Chiewchengchol
- Center of Excellence in Immunology and Immune-mediated diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
42
|
Gao W, Gong J, Mu M, Zhu Y, Wang W, Chen W, Han G, Hu H, Bao P. The Pathogenesis of Eosinophilic Asthma: A Positive Feedback Mechanism That Promotes Th2 Immune Response via Filaggrin Deficiency. Front Immunol 2021; 12:672312. [PMID: 34484176 PMCID: PMC8414997 DOI: 10.3389/fimmu.2021.672312] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 07/14/2021] [Indexed: 01/16/2023] Open
Abstract
Eosinophilic asthma (EA) is a common subtype of asthma and often progresses to severe disease. In order to understand its pathogenesis, targeted next-generation gene sequencing was performed on 77 Chinese EA patients and 431 Chinese healthy controls to obtain differential genomic variations. Among the 41 Single Nucleotide Polymorphisms (SNPs) screened for mutation sites in more than 3 patients, filaggrin gene FLG rs192116923 T>G and FLG rs75235053 C>G were newly found to be associated with EA patients with atopic dermatitis (AD) (P <0.001) and severe EA (P=0.032), respectively. Filaggrin has been shown to be mainly expressed in epithelial cells and plays an important role in formation of an effective skin barrier. Bioinformatic analysis indicated FLG rs192116923 T>G may increase the binding of Smad3 to transmit TGF-β1 signaling, and thereby inhibit filaggrin expression, and FLG rs75235053 C>G may add new splicing sites to reduce filaggrin monomers. It has been known that the level of Th2 cytokine IL-4 is increased in EA patients, and IL-4 increases airway epithelial permeability and enhances inflammatory response through some unclear mechanisms. To figure out whether filaggrin is involved in immune responses in asthma, we have treated human respiratory epithelial cell line BEAS-2B cells with IL-4 and found that the expression levels of filaggrin and E-cadherin decreased significantly in a time and dose-dependent manner, suggesting that IL-4 increased airway epithelial permeability by reducing filaggrin and adhesion molecule. In addition, in our study, IL-4 increased the expression of epithel-derived inflammatory cytokines IL-33 and TSLP which further enhanced the Th2 inflammatory response. To investigate the role of filaggrin in development of EA, knockdown filaggrin with siRNA revealed a decrease in E-cadherin levels, which were further down-regulated by IL-4 stimulation. Knockdown of filaggrin alone did not affect the levels of IL-33 and TSLP, but further exacerbated the decrease of IL-33/TSLP caused by IL-4, suggesting that filaggrin may involve in IL-4R signaling pathway to regulate the level of IL-33/TSLP. In conclusion, in the Th2 cytokine milieu of asthma, FLG deficient mutation in airway epithelial cells may increase the epithelial permeability and the expression of IL-33/TSLP which positively feedback the Th2 inflammation response.
Collapse
Affiliation(s)
- Wei Gao
- Respiratory and Critical Care Unit, 1st Medical Center of Chinese Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jiuyu Gong
- Department of Internal Medicine, Hubei Province Corps Hospital of The Chinese Armed Police Force (CAPF), Wuhan, China
| | - Mi Mu
- Pulmonary and Critical Care Medicine College of Chinese PLA General Hospital, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yujin Zhu
- Respiratory and Critical Care Unit, 1st Medical Center of Chinese Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Internal Medicine, Tianjin Municipal Corps Hospital of CAPF, Tianjin, China
| | - Wenjuan Wang
- Department of Dermatology, 1st Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wen Chen
- Department of Pathology, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guojing Han
- Respiratory and Critical Care Unit, 1st Medical Center of Chinese Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Hong Hu
- Respiratory and Critical Care Unit, 1st Medical Center of Chinese Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Pengtao Bao
- Pulmonary and Critical Care Medicine College of Chinese PLA General Hospital, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
43
|
Piazzon MC, Mladineo I, Dirks RP, Santidrián Yebra-Pimentel E, Hrabar J, Sitjà-Bobadilla A. Ceratothoa oestroides Infection in European Sea Bass: Revealing a Long Misunderstood Relationship. Front Immunol 2021; 12:645607. [PMID: 33777043 PMCID: PMC7991915 DOI: 10.3389/fimmu.2021.645607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/05/2021] [Indexed: 12/16/2022] Open
Abstract
Ceratothoa oestroides (Cymothoidea, Isopoda) is a generalist crustacean parasite that negatively affects the economic sustainability of European sea bass (Dicentrarchus labrax) aquaculture in the North-East Mediterranean. While mortalities are observed in fry and fingerlings, infection in juvenile and adult fish result in approximately 20% growth delay. A transcriptomic analysis (PCR array, RNA-Seq) was performed on organs (tongue, spleen, head kidney, and liver) from infected vs. Ceratothoa-free sea bass fingerlings. Activation of local and systemic immune responses was detected, particularly in the spleen, characterized by the upregulation of cytokines (also in the tongue), a general reshaping of the immunoglobulin (Ig) response and suppression of T-cell mediated responses. Interestingly, starvation and iron transport and metabolism genes were strongly downregulated, suggesting that the parasite feeding strategy is not likely hematophagous. The regulation of genes related to growth impairment and starvation supported the growth delay observed in infected animals. Most differentially expressed (DE) transcripts were exclusive of a specific organ; however, only in the tongue, the difference between infected and uninfected fish was significant. At the attachment/feeding site, the pathways involved in muscle contraction and intercellular junction were the most upregulated, whereas the pathways involved in fibrosis (extracellular matrix organization, collagen formation, and biosynthesis) were downregulated. These results suggest that parasite-inflicted damage is successfully mitigated by the host and characterized by regenerative processes that prevail over the reparative ones.
Collapse
Affiliation(s)
- M Carla Piazzon
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal - Consejo Superior de Investigaciones Científicas (IATS-CSIC), Castellón, Spain
| | - Ivona Mladineo
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia.,Biology Centre of the Czech Academy of Sciences, Institute of Parasitology, Ceske Budejovice, Czechia
| | - Ron P Dirks
- Future Genomics Technology, Leiden, Netherlands
| | | | - Jerko Hrabar
- Laboratory for Aquaculture, Institute of Oceanography and Fisheries, Split, Croatia
| | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Institute of Aquaculture Torre de la Sal - Consejo Superior de Investigaciones Científicas (IATS-CSIC), Castellón, Spain
| |
Collapse
|
44
|
|
45
|
Abstract
Atopic dermatitis (AD) is a prevalent inflammatory skin disease. IL-13 contributes significantly to the pathogenesis of AD in several ways, and beneficial results have been demonstrated with anti-IL-13 therapies. Currently, the only monoclonal antibody (mAb) approved for AD treatment is dupilumab, an antagonist of the IL-4 receptor alpha (IL-4Rα) subunit common to IL-4 and IL-13 receptors, but clinical trials evaluating anti-IL-13 mAbs are providing promising results. The topics of this review will be mAbs targeting IL-13 for the treatment of AD such as dupilumab, tralokinumab and lebrikizumab, small molecules targeting the IL-13 pathway, and a brief explanation of therapies targeting IL-13 for the treatment of other skin diseases.
Collapse
Affiliation(s)
- Carla Tubau
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona Mas Casanovas 90, Barcelona 08041, Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona Mas Casanovas 90, Barcelona 08041, Spain
| |
Collapse
|
46
|
Hennies HC, Poumay Y. Skin Disease Models In Vitro and Inflammatory Mechanisms: Predictability for Drug Development. Handb Exp Pharmacol 2021; 265:187-218. [PMID: 33387068 DOI: 10.1007/164_2020_428] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Investigative skin biology, analysis of human skin diseases, and numerous clinical and pharmaceutical applications rely on skin models characterized by reproducibility and predictability. Traditionally, such models include animal models, mainly rodents, and cellular models. While animal models are highly useful in many studies, they are being replaced by human cellular models in more and more approaches amid recent technological development due to ethical considerations. The culture of keratinocytes and fibroblasts has been used in cell biology for many years. However, only the development of co-culture and three-dimensional epidermis and full-skin models have fundamentally contributed to our understanding of cell-cell interaction and cell signalling in the skin, keratinocyte adhesion and differentiation, and mechanisms of skin barrier function. The modelling of skin diseases has highlighted properties of the skin important for its integrity and cutaneous development. Examples of monogenic as well as complex diseases including atopic dermatitis and psoriasis have demonstrated the role of skin models to identify pathomechanisms and drug targets. Recent investigations have indicated that 3D skin models are well suitable for drug testing and preclinical studies of topical therapies. The analysis of skin diseases has recognized the importance of inflammatory mechanisms and immune responses and thus other cell types such as dendritic cells and T cells in the skin. Current developments include the production of more complete skin models comprising a range of different cell types. Organ models and even multi-organ systems are being developed for the analysis of higher levels of cellular interaction and drug responses and are among the most recent innovations in skin modelling. They promise improved robustness and flexibility and aim at a body-on-a-chip solution for comprehensive pharmaceutical in vitro studies.
Collapse
Affiliation(s)
- Hans Christian Hennies
- Department of Biological and Geographical Sciences, University of Huddersfield, Huddersfield, UK. .,Cologne Center for Genomics, University Hospital Cologne, Cologne, Germany.
| | - Yves Poumay
- Faculty of Medicine, Namur Research Institute for Life Sciences, University of Namur, Namur, Belgium
| |
Collapse
|
47
|
Tubau C, Puig L. Therapeutic targeting of the IL-13 pathway in skin inflammation. Expert Rev Clin Immunol 2020; 17:15-25. [PMID: 33275064 DOI: 10.1080/1744666x.2020.1858802] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Atopic dermatitis (AD) is a heterogeneous, chronic, inflammatory skin disease with a non-negligible prevalence at present. Its pathogenesis is complex, but mainly characterized by constitutive T helper type 2 (Th2)-cell activation. Systemic therapies for moderate-to-severe AD can be associated with adverse events that encumber their satisfactory long-term use. Several drugs targeting relevant molecules in the immunopathogenesis of AD have been approved or are under clinical development for the treatment of moderate to severe AD. To elaborate this review, literature searches were performed in PubMed on 29 August 2020.Areas covered: This narrative literature review is focused on the pivotal role of IL-13 in the immunopathogenesis of AD and other skin diseases.Expert opinion: Dupilumab has demonstrated the central role of IL-13 and IL-4 in the pathogenesis of AD, asthma, and other diseases in the atopic spectrum. In addition, phase III randomized clinical trials (RCTs) evaluating specific blockade of IL-13 with tralokinumab for treatment of AD also demonstrated favorable results, and phase III RCT evaluating lebrikizumab are ongoing. The role of IL-13 in other skin diseases should be further investigated.
Collapse
Affiliation(s)
- Carla Tubau
- Dermatology Department, Hospital De La Santa Creu I Sant Pau, Universitat Autònoma De Barcelona, Barcelona, Spain
| | - Lluís Puig
- Dermatology Department, Hospital De La Santa Creu I Sant Pau, Universitat Autònoma De Barcelona, Barcelona, Spain
| |
Collapse
|
48
|
Leyva-Castillo JM, Das M, Artru E, Yoon J, Galand C, Geha RS. Mast cell-derived IL-13 downregulates IL-12 production by skin dendritic cells to inhibit the T H1 cell response to cutaneous antigen exposure. J Allergy Clin Immunol 2020; 147:2305-2315.e3. [PMID: 33316284 DOI: 10.1016/j.jaci.2020.11.036] [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: 07/09/2020] [Revised: 11/06/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is characterized by a skin barrier defect aggravated by mechanical injury inflicted by scratching, a TH2 cell-dominated immune response, and susceptibility to viral skin infections that are normally restrained by a TH1 cell response. The signals leading to a TH2 cell-dominated immune response in AD are not completely understood. OBJECTIVE Our aim was to determine the role of IL-13 in initiation of the TH cell response to cutaneously encountered antigens. METHODS Wild-type, Il13-/-, Il1rl1-/-, and Il4ra-/- mice, as well as mice with selective deficiency of IL-13 in mast cells (MCs) were studied; in addition, dendritic cells (DCs) purified from the draining lymph nodes of tape-stripped and ovalbumin (OVA)-sensitized skin were examined for their ability to polarize naive OVA-TCR transgenic CD4+ T cells. Cytokine expression was examined by reverse-transcriptase quantitative PCR, intracellular flow cytometry, and ELISA. Contact hypersensitivity to dinitrofluorobenzene was examined. RESULTS Tape stripping caused IL-33-driven upregulation of Il13 expression by skin MCs. MC-derived IL-13 acted on DCs from draining lymph nodes of OVA-sensitized skin to selectively suppress their ability to polarize naive OVA-TCR transgenic CD4+ T cells into IFN-γ-secreting cells. MC-derived IL-13 inhibited the TH1 cell response in contact hypersensitivity to dinitrofluorobenzene. IL-13 suppressed IL-12 production by mouse skin-derived DCs in vitro and in vivo. Scratching upregulated IL13 expression in human skin, and IL-13 suppressed the capacity of LPS-stimulated human skin DCs to express IL-12 and promote IFN-γ secretion by CD4+ T cells. CONCLUSION Release of IL-13 by cutaneous MCs in response to mechanical skin injury inhibits the TH1 cell response to cutaneous antigen exposure in AD.
Collapse
Affiliation(s)
| | - Mrinmoy Das
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School
| | - Emilie Artru
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School
| | - Juhan Yoon
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School
| | - Claire Galand
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School.
| |
Collapse
|
49
|
Ghamrawi R, Bell KA, Balogh EA, Strowd LC, Feldman SR. Current and emerging biologics for the treatment of pediatric atopic dermatitis. Expert Opin Biol Ther 2020; 20:1435-1445. [PMID: 33078990 DOI: 10.1080/14712598.2021.1840548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by erythematous lesions, pruritus, and a skin barrier defect. Long-term treatment in children is challenging, as there is only one Food and Drug Administration-approved systemic medication. Current treatments may have limited efficacy or serious side effects in children. With a deeper understanding of AD pathogenesis and the advent of target-specific medications, several biologics are undergoing clinical trials for future use in pediatric AD. AREAS COVERED This article reviews the current and emerging biologic therapies for treatment of pediatric AD. It allows for comprehensive comparison of medications and their clinical trials to help providers optimize patient treatment plans while providing expert insight into upcoming advancements in the treatment of pediatric AD. EXPERT OPINION Treating pediatric AD is complicated given the variety of disease severity, psychosocial impact, and relative lack of approved medications for severe disease. Given the safety data on dupilumab, newer biologics will likely be second-line. We do not yet understand the long-term impact of newer biologics on an immature immune system, nor do we fully understand their risks and toxicities. We should proceed optimistically, yet cautiously, with the study of biologics in children.
Collapse
Affiliation(s)
- Rima Ghamrawi
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Katheryn A Bell
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Esther A Balogh
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Lindsay C Strowd
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Pathology, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Social Sciences & Health Policy, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Dermatology, University of Southern Denmark , Odense, Denmark
| |
Collapse
|
50
|
Clayton K, Vallejo A, Sirvent S, Davies J, Porter G, Reading I, Lim F, Ardern‐Jones M, Polak M. Machine learning applied to atopic dermatitis transcriptome reveals distinct therapy‐dependent modification of the keratinocyte immunophenotype*. Br J Dermatol 2020; 184:913-922. [DOI: 10.1111/bjd.19431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/12/2022]
Affiliation(s)
- K. Clayton
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
| | - A. Vallejo
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
| | - S. Sirvent
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
| | - J. Davies
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
| | - G. Porter
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
| | - I.C. Reading
- Department of Primary Care and Population Sciences (Faculty of Medicine) University of Southampton Southampton Hants UK
- NIHR Research Design Service South CentralSouthampton Hants UK
| | - F. Lim
- Unilever Research Colworth Science ParkSharnbrook, Bedfordshire UK
| | - M.R. Ardern‐Jones
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
- Department of Dermatology University Hospital Southampton NHS Foundation TrustSouthampton Hants UK
| | - M.E. Polak
- Department of Clinical and Experimental Sciences (Sir Henry Wellcome Laboratories, Faculty of Medicine) University of Southampton Southampton Hants UK
- Institute for Life Sciences University of SouthamptonSouthampton Hants UK
| |
Collapse
|