1
|
Boulund U, Thorsen J, Trivedi U, Tranæs K, Jiang J, Shah SA, Stokholm J. The role of the early-life gut microbiome in childhood asthma. Gut Microbes 2025; 17:2457489. [PMID: 39882630 PMCID: PMC11784655 DOI: 10.1080/19490976.2025.2457489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/05/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Asthma is a chronic disease affecting millions of children worldwide, and in severe cases requires hospitalization. The etiology of asthma is multifactorial, caused by both genetic and environmental factors. In recent years, the role of the early-life gut microbiome in relation to asthma has become apparent, supported by an increasing number of population studies, in vivo research, and intervention trials. Numerous early-life factors, which for decades have been associated with the risk of developing childhood asthma, are now being linked to the disease through alterations of the gut microbiome. These factors include cesarean birth, antibiotic use, breastfeeding, and having siblings or pets, among others. Association studies have highlighted several specific microbes that are altered in children developing asthma, but these can vary between studies and disease phenotype. This demonstrates the importance of the gut microbial ecosystem in asthma, and the necessity of well-designed studies to validate the underlying mechanisms and guide future clinical applications. In this review, we examine the current literature on the role of the gut microbiome in childhood asthma and identify research gaps to allow for future microbial-focused therapeutic applications in asthma.
Collapse
Affiliation(s)
- Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Urvish Trivedi
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kaare Tranæs
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Jie Jiang
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Shiraz A. Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Abdelhalim A, Yilmaz O, Elshaikh Berair M, Torres T. Topical delgocitinib for the treatment of chronic hand eczema. J DERMATOL TREAT 2025; 36:2479126. [PMID: 40096745 DOI: 10.1080/09546634.2025.2479126] [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: 01/21/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE Chronic hand eczema (CHE) is a prevalent dermatological condition characterized by significant morbidity and impaired quality of life. Delgocitinib, a pan-JAK inhibitor, has emerged as a promising topical treatment for CHE, targeting key cytokine-mediated inflammatory pathways involved in the disease. Delgocitinib 20 mg/g (2%) cream was approved by the European Medicines Agency (EMA) in 2024 for moderate-to-severe CHE, and its U.S. Food and Drug Administration (FDA) review is currently in progress. MATERIALS AND METHODS PubMed, Medline and ClinicalTrials.gov were searched up to January 21, 2025, using specific search terms related to delgocitinib and chronic hand eczema. RESULTS AND CONCLUSIONS Clinical trials have demonstrated its effectiveness in improving disease severity, including eczema signs and symptoms such as pain and itching, and enhancing patient-reported outcomes compared to vehicle. Topical delgocitinib has shown a favorable safety profile, with most adverse events being mild and unrelated to treatment. Serious adverse events were rare, and treatment discontinuation due to adverse events was minimal. This narrative review synthesizes current evidence on topical delgocitinib's clinical utility and safety in CHE, positioning it as a valuable therapeutic option. Further comparative studies are needed to evaluate its efficacy against oral JAK inhibitors and other topical immunosuppressants, providing insight into optimizing treatment strategies for this chronic condition.
Collapse
Affiliation(s)
- Asaad Abdelhalim
- Allergy and Clinical Immunology Department, King Khalid Hospital, Hafar Al Batin, Saudi Arabia
| | - Orhan Yilmaz
- College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | - Tiago Torres
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| |
Collapse
|
3
|
Mahjoubi M, Rashedi R, Samieefar N, Abdollahimajd F, Rezaei N. Dermatologic presentations of hyper IgE syndrome in pediatric patients. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2025; 21:20. [PMID: 40317072 PMCID: PMC12049024 DOI: 10.1186/s13223-025-00963-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Hyper-IgE Syndrome, also known as Job's syndrome, is a rare primary immunodeficiency disorder characterized by recurrent infections and elevated levels of immunoglobulin E. While respiratory and systemic manifestations have been more emphasized, dermatological manifestations in Hyper-IgE Syndrome also play a significant role in disease presentation. METHODS This narrative review explores the dermatologic presentations of Hyper-IgE Syndrome in pediatric populations, including descriptions, associated symptoms/findings, and available treatment options. RESULTS AND CONCLUSION Neonatal rash, mucocutaneous candidiasis, noma neonatorum, psoriasis, cold staphylococcal abscesses, and candida onychomycosis are among the dermatological manifestations of Hyper-IgE Syndrome. Each manifestation has unique characteristics and treatment considerations, necessitating accurate recognition and diagnosis for effective management. Optimal treatment strategies involve a combination of supportive care, topical/systemic therapies, antifungal medications, and surgical interventions when necessary. Further research is needed to enhance our understanding of these manifestations and evaluate treatment modalities for individuals affected by Hyper-IgE Syndrome.
Collapse
Affiliation(s)
- Mohammad Mahjoubi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Clinical Research Development Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Ronak Rashedi
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Noosha Samieefar
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Pediatric Chronic Kidney Disease Research Center, Gene, Cell & Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Abdollahimajd
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Clinical Research Development Unit, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Research Center of Artificial Intelligence in Health, Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Interdisciplinarity in Neonates and Infants (NINI), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, , Tehran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Tordjman L, Mashoudy KD, Czarnowicki T. Converging paths toward unified therapeutic approaches in atopic dermatitis, vitiligo, and alopecia areata. J Allergy Clin Immunol 2025:S0091-6749(25)00456-7. [PMID: 40274075 DOI: 10.1016/j.jaci.2025.04.016] [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: 11/18/2024] [Revised: 04/01/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025]
Abstract
Emerging evidence reveals significant epidemiologic, genetic, and immunologic connections between atopic dermatitis, vitiligo, and alopecia areata, challenging previously established notions of their distinct pathogenic and molecular signatures. Exploring these commonalities not only enhances our understanding of each disease's pathogenesis, but also supports the development of unified treatment strategies for these frequently co-occurring disorders. This review examines key immune players shared across the 3 conditions, including cytokines, immune cells, and signaling pathways. Building on these insights, we also evaluate a range of therapeutic options-ranging from treatments approved by the Food and Drug Administration to those currently in clinical trials-alongside proposed future therapeutic targets. This comprehensive approach aims to advance our management of these interconnected autoimmune and inflammatory disorders with greater precision.
Collapse
Affiliation(s)
- Lea Tordjman
- University of Miami Miller School of Medicine, Miami, Fla
| | | | - Tali Czarnowicki
- Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Fla.
| |
Collapse
|
5
|
Ye J, Lai Y. Keratinocytes: new perspectives in inflammatory skin diseases. Trends Mol Med 2025:S1471-4914(25)00083-8. [PMID: 40246604 DOI: 10.1016/j.molmed.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/16/2025] [Accepted: 03/25/2025] [Indexed: 04/19/2025]
Abstract
Keratinocytes, the predominant cell type in the epidermis, are indispensable for maintaining skin barrier integrity, mediating host defense, and orchestrating immune responses. Beyond these well-established functions, emerging evidence reveals their dynamic interactions with the nervous system and their capacity to retain inflammatory memory. These discoveries position keratinocytes as key drivers of the onset, progression, and relapse of inflammatory skin diseases. In this review, we delve into the mechanisms underlying keratinocyte crosstalk with immune and neural cells, the metabolic reprogramming, including lactate and other metabolites, that may drive inflammatory memory, and the broader implications for disease pathogenesis and recurrence. Finally, we discuss the challenges to, and therapeutic potential of, targeting keratinocytes for the treatment of chronic inflammatory skin conditions.
Collapse
Affiliation(s)
- Jiafeng Ye
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai, PR China; Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, PR China
| | - Yuping Lai
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, School of Life Sciences, East China Normal University, Shanghai, PR China; Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, PR China; Liwa Institue of Skin Health, East China Normal University, Shanghai, PR China.
| |
Collapse
|
6
|
Stamatas GN, Insel R, Sørensen N, Palleja A, Moll JM, Oddos T, Chaoimh CN, O'B Hourihane J, Irvine AD. Shifts in Infant Skin Microbiome at 2 Months after Short-Term Emollient Use from Birth Are Associated with Reduced Prevalence of Atopic Dermatitis at 12 Months in a High-Risk Cohort. J Invest Dermatol 2025:S0022-202X(25)00380-X. [PMID: 40204066 DOI: 10.1016/j.jid.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025]
Affiliation(s)
| | - Richard Insel
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | - Albert Palleja
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | | | - Carol Ní Chaoimh
- Paediatrics and Child Health, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland
| | - Jonathan O'B Hourihane
- INFANT Research Centre, University College Cork, Cork, Ireland; Paediatrics and Child Health, Royal College of Surgeons, Dublin, Ireland
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Lazor JE, Bozsoki BA, Bharadwaj P. Cure for the itch: current clinical standards and therapies in allergic eczema. FRONTIERS IN ALLERGY 2025; 6:1569292. [PMID: 40248667 PMCID: PMC12003377 DOI: 10.3389/falgy.2025.1569292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/11/2025] [Indexed: 04/19/2025] Open
Abstract
Allergic Eczema (AE) is a chronic, relapsing skin condition that significantly affects the quality of life of the AE patients and their caretakers. Decades of scientific and clinical research has helped understand the highly complex underpinnings of AE presentation wherein a multitude of variables, including the conspicuous variables such as environmental allergens, immunological triggers, genetic predisposition of individuals, to more nuanced socio-economic status, play an important part. Given the complexity of the disease, it is imperative to develop biomarkers enabling early and reliable clinical identifications and help in the active management of the disease, thereby minimizing the impact and burden of the disease on the patients. In this mini review, we provide a brief overview of AE, affected demographics, variables that trigger its onset, and summarize the discovery of various clinical biomarkers such as total and specific serum IgE levels, Th2 cytokine levels, filaggrin (FLG) mutations, periostin levels in skin, etc. that have been developed over the years to further improve the state of clinical monitoring of AE presentation and progression. Lastly, we also provide an overview of the clinical interventions and therapies, such as topical agents, phototherapy, and biologics, that are available to the patients to manage AE-related complications. While we have vastly improved the standard of care and diagnosis for the AE patients, there are still many unmet needs such as developing non-invasive, effective, and reliable clinical predictors and biomarkers which can usher better personalized treatments and provide a better quality of life to affected demographics.
Collapse
|
8
|
Choi UE, Deng J, Parthasarathy V, Liao V, D'Amiano A, Taylor M, Bordeaux ZA, Kambala A, Cornman HL, Canner JK, Drucker AM, Kwatra SG. Risk factors and temporal associations of progression of the atopic march in children with early-onset atopic dermatitis. J Am Acad Dermatol 2025; 92:732-740. [PMID: 39615548 DOI: 10.1016/j.jaad.2024.10.107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND Risk factors and the temporal relationship between atopic dermatitis (AD) and atopic march remain understudied. OBJECTIVE Determine risk factors for atopic march in early-onset AD patients and the temporality between AD and atopic march. METHODS We used the MarketScan Research Database for our retrospective cohort analysis from 2010 to 2018, comparing infants diagnosed with AD before age 1 with controls without early-onset AD. Primary outcomes were hazard ratios (HRs) for the development of asthma, allergic rhinitis, and food allergy. RESULTS Compared to 55,174 controls, higher proportions of the 27,228 AD patients developed asthma (19.21% vs 8.65%, P < .001), allergic rhinitis (28.27% vs 12.62%, P < .001), food allergy (16.00% vs 2.27%, P < .001), and all atopic triad conditions (10.69% vs 0.71%, P < .001). Among AD patients, higher proportions developed the atopic triad if they were male (HR 1.66, 95% confidence interval [1.45-1.90]), had severe disease (HR 3.16, [2.77-3.60]), or had family atopy history (HR > 3.40, P < .001 for all comparisons). Among AD patients, 20.1% developed allergic rhinitis. LIMITATIONS Our study was based on health care claims data. CONCLUSION Early-onset AD is associated with higher rates of developing atopic march conditions compared to controls. Particular attention should be paid toward risk factors and atopic march screening in early-onset AD patients.
Collapse
Affiliation(s)
- Una E Choi
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Junwen Deng
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Varsha Parthasarathy
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viviane Liao
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anjali D'Amiano
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew Taylor
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Zachary A Bordeaux
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anusha Kambala
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hannah L Cornman
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Joseph K Canner
- Johns Hopkins Surgery Center for Outcomes Research, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aaron M Drucker
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada; Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
| | - Shawn G Kwatra
- Department of Dermatology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Itch Center, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
9
|
Oliva M, Sarkar MK, March ME, Saeidian AH, Mentch FD, Hsieh CL, Tang F, Uppala R, Patrick MT, Li Q, Bogle R, Kahlenberg JM, Watson D, Glessner JT, Youssefian L, Vahidnezhad H, Tsoi LC, Hakonarson H, Gudjonsson JE, Smith KM, Riley-Gillis B. Integration of GWAS, QTLs and keratinocyte functional assays reveals molecular mechanisms of atopic dermatitis. Nat Commun 2025; 16:3101. [PMID: 40164604 PMCID: PMC11958703 DOI: 10.1038/s41467-025-58310-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Atopic dermatitis is a highly heritable and common inflammatory skin condition affecting children and adults worldwide. Multi-ancestry approaches to atopic dermatitis genetic association studies are poised to boost power to detect genetic signal and identify loci contributing to atopic dermatitis risk. Here, we present a multi-ancestry GWAS meta-analysis of twelve atopic dermatitis cohorts from five ancestral populations totaling 56,146 cases and 602,280 controls. We report 101 genomic loci associated with atopic dermatitis, including 16 loci that have not been previously associated with atopic dermatitis or eczema. Fine-mapping, QTL colocalization, and cell-type enrichment analyses identified genes and cell types implicated in atopic dermatitis pathophysiology. Functional analyses in keratinocytes provide evidence for genes that could play a role in atopic dermatitis through epidermal barrier function. Our study provides insights into the etiology of atopic dermatitis by harnessing multiple genetic and functional approaches to unveil the mechanisms by which atopic dermatitis-associated variants impact genes and cell types.
Collapse
Affiliation(s)
| | | | - Michael E March
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Frank D Mentch
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | | | | | | | - Qinmengge Li
- University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Deborah Watson
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Leila Youssefian
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- City of Hope National Medical Center, Irwindale, CA, 91706, USA
| | - Hassan Vahidnezhad
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Lam C Tsoi
- University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hakon Hakonarson
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | | | | |
Collapse
|
10
|
Fukuda K, Ito Y, Amagai M. Barrier Integrity and Immunity: Exploring the Cutaneous Front Line in Health and Disease. Annu Rev Immunol 2025; 43:219-252. [PMID: 40279307 DOI: 10.1146/annurev-immunol-082323-030832] [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] [Indexed: 04/27/2025]
Abstract
Immune responses are influenced by not only immune cells but also the tissue microenvironment where these cells reside. Recent advancements in understanding the underlying molecular mechanisms and structures of the epidermal tight junctions (TJs) and stratum corneum (SC) have significantly enhanced our knowledge of skin barrier functions. TJs, located in the granular layer of the epidermis, are crucial boundary elements in the differentiation process, particularly in the transition from living cells to dead cells. The SC forms from dead keratinocytes via corneoptosis and features three distinct pH zones critical for barrier function and homeostasis. Additionally, the SC-skin microbiota interactions are crucial for modulating immune responses and protecting against pathogens. In this review, we explore how these components contribute both to healthy and disease states. By targeting the skin barrier in therapeutic strategies, we can enhance its integrity, modulate immune responses, and ultimately improve outcomes for patients with inflammatory skin conditions.
Collapse
Affiliation(s)
- Keitaro Fukuda
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan;
| | - Yoshihiro Ito
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan;
| |
Collapse
|
11
|
Tamari M, Ver Heul AM. Neuroimmune mechanisms of type 2 inflammation in the skin and lung. Allergol Int 2025; 74:177-186. [PMID: 40064568 DOI: 10.1016/j.alit.2025.02.001] [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: 01/20/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 04/01/2025] Open
Abstract
Type 2 inflammation has a major role in barrier tissues such as the skin and airways and underlies common conditions including atopic dermatitis (AD) and asthma. Cytokines including interleukin 4 (IL-4), IL-5, and IL-13 are key immune signatures of type 2 inflammation and are the targets of multiple specific therapeutics for allergic diseases. Despite shared core immune mechanisms, the distinct structures and functions of the skin and airways lead to unique therapeutic responses. It is increasingly recognized that the nervous system has a major role in sensing and directing inflammatory processes. Indeed, crosstalk between type 2 immune activation and somatosensory functions mediates tissue-specific signatures such as itching in the skin. However, neuroimmune interactions are shaped by distinct neuronal and immune landscapes, and differ between the skin and airways. In the skin, dorsal root ganglia-derived neurons mediate pruritus via type 2 cytokines and neurogenic inflammation by mast cell or basophil activation. Conversely, vagal ganglia-derived neurons regulate airway immune responses by releasing neuropeptides/neurotransmitters such as calcitonin gene-related peptides, neuromedin U, acetylcholine, and noradrenaline. Sensory neuron-derived vasoactive intestinal peptide forms a feedback loop with IL-5, amplifying eosinophilic inflammation in the airways, a mechanism that is absent in the skin. These differences influence the efficacy of cytokine-targeted therapies. For instance, IL-4/IL-13-targeted therapies like dupilumab demonstrate efficacy in AD and allergic airway diseases, whereas IL-5-targeted therapies are effective in eosinophilic asthma but not AD. Understanding these neuroimmune interactions underscores the need for tailored therapeutic approaches to address allergic diseases where barrier tissues are involved.
Collapse
Affiliation(s)
- Masato Tamari
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Aaron M Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, USA.
| |
Collapse
|
12
|
Huang F, Zhu X, Liang X. Research hotspots and trends of skin barrier in atopic dermatitis in the past 24-year: a bibliometric analysis. Front Med (Lausanne) 2025; 12:1539386. [PMID: 40144870 PMCID: PMC11936790 DOI: 10.3389/fmed.2025.1539386] [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: 12/04/2024] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Background Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin condition that imposes significant psychological and economic burdens on patients due to its recurring nature. Its etiology is multifactorial, involving interactions between genetic predispositions and environmental factors. The skin barrier serves as both a mechanical and immunological defense, and its structural damage and functional impairments significantly contribute to the pathogenesis of AD. This study aims to explore the future prospects and developmental trends of the skin barrier in the context of AD through a bibliometric analysis. Objective To analyze the research status, hot spots and development trend of skin barrier in AD. Methods Relevant studies were extracted from the Web of Science database and screened by researchers, with bibliometric analysis conducted using VOSviewer, CiteSpace, and other tools. Results A total of 4,227 publications were identified over a 24-year research period. The United States is the leading contributor, with 1,263 publications, and demonstrates extensive collaboration with numerous countries. The journal with the highest number of publications is the Journal of Allergy and Clinical Immunology. The most prolific institutions is the University of California, San Francisco. Recent years have seen high citation intensity for keywords such as "dupilumab," "barrier dysfunction," and "gut microbiota". Conclusion The mechanism of the skin barrier in AD remains an area requiring ongoing research and analysis. Although significant progress has been achieved, future research will benefit from advancements in technology.
Collapse
Affiliation(s)
- Fangchang Huang
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| | - Xin Zhu
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| | - Xinglong Liang
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| |
Collapse
|
13
|
Tanemura S, Mima Y. A Case of Cutaneous Fungal Infection Following the Administration of Dupilumab. Cureus 2025; 17:e81349. [PMID: 40291282 PMCID: PMC12034230 DOI: 10.7759/cureus.81349] [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] [Accepted: 03/28/2025] [Indexed: 04/30/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with a multifactorial etiology. Herein, we report a case of a patient with AD undergoing long-term topical treatments who developed a dermatophyte infection following the administration of dupilumab. Dupilumab is known to enhance skin barrier function and induce changes in the skin microbiome. Notably, head and neck dermatitis caused by the overgrowth of Malassezia species due to dupilumab has been widely discussed. This phenomenon is thought to result from the suppression of T helper (Th)2 cytokines by dupilumab, leading to a decrease in the proportion of Staphylococcus aureus and a relative increase in fungal populations. Additionally, feedback activation of Th17 cytokines may trigger excessive inflammation against fungi, contributing to fungal infections. IL-13 plays critical roles in fungal colony formation, and tralokinumab, an IL-13 inhibitor, has shown potential efficacy in treating this head and neck dermatitis. While the relationship between microbiome changes and biologics like lebrikizumab and nemolizumab remains unexplored, investigating the differential effects of these therapies on the cutaneous microbiome could provide deeper insights into not only the unique characteristics of each biologic agent but also the roles of Th2 cytokines such as IL-4, IL-13, and IL-31 in the pathophysiology of AD. The present case underscores the importance of the comprehensive therapeutic approach for AD that accounts for microbiome dynamics and adapts to evolving skin changes throughout the course of treatment.
Collapse
Affiliation(s)
| | - Yoshihito Mima
- Department of Dermatology, Tokyo Metropolitan Police Hospital, Tokyo, JPN
| |
Collapse
|
14
|
Alkattan A, Alzaher A, Alhabib D, Younis A, Alsalem E, Suraj N, Alsalameen E, Alrasheed N, Almuhaidib M, Ibrahim MH. An evaluation of the recently approved drugs for treating atopic dermatitis in the context of their safety and efficacy: a systematic review and meta-analysis. Expert Rev Clin Immunol 2025; 21:347-357. [PMID: 39663577 DOI: 10.1080/1744666x.2024.2435657] [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: 09/10/2024] [Revised: 10/25/2024] [Accepted: 11/13/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION The present paper aimed to conduct an updated systematic review and meta-analysis to evaluate the safety and efficacy of crisaborole, delgocitinib, and ruxolitinib in treating mild-to-moderate atopic dermatitis (AD). METHODS MEDLINE and Google Scholar databases were utilized to search articles published during the years 2015-2024. The review was limited to randomized controlled studies that measured specific outcomes for safety and efficacy aspects, including adverse events (AEs) or treatment-emergent adverse events (TEAEs) to evaluate safety and Investigator's static global assessment (ISGA) or improvement of at least 75% of Eczema Area and Severity Index (EASI-75) to evaluate efficacy. RESULTS The review included 17 articles in the analysis. The safety odds ratios (ORs) among participants using crisaborole, delgocitinib, and ruxolitinib were 1.14, 95% CI [0.97-1.36], 1.18, 95% CI [0.84-1.67], and 0.72, 95% CI [0.55-0.94], respectively, when compared to control groups. The three studied topical AD treatments were found to be significantly more effective compared to control groups (crisaborole, OR = 1.78, 95% CI [1.51-2.10], delgocitinib, OR = 6.34, 95% CI [3.57-11.27], and ruxolitinib, OR = 7.30, 95% CI [5.10-10.44]). CONCLUSION Delgocitinib and ruxolitinib demonstrated favorable safety and effectiveness profiles across various age cohorts, whereas crisaborole raised concerns over its safety and efficacy, particularly in children.
Collapse
Affiliation(s)
- Abdullah Alkattan
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Abrar Alzaher
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Dina Alhabib
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Afnan Younis
- Department of Family and Community Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Elham Alsalem
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Nadia Suraj
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Eman Alsalameen
- Department of Pharmacy, King Khalid University Hospital, Medical City King Saud University, Riyadh, Saudi Arabia
| | - Noura Alrasheed
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Moneerah Almuhaidib
- Research and Planning Unit, General Directorate of School Health, Ministry of Health, Riyadh, Saudi Arabia
| | - Mona H Ibrahim
- Department of Public Health and Community Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
15
|
Gorelick J, Nguyen A, Schneider SKR, Martel BC, Madsen DE, Armstrong AW. Biomarkers in Atopic Dermatitis: A Review of the Role of IL-13 and the Impact of Tralokinumab Treatment. Am J Clin Dermatol 2025; 26:199-211. [PMID: 39820896 PMCID: PMC11850464 DOI: 10.1007/s40257-024-00913-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/19/2025]
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory skin disease that can significantly affect quality of life. Presence, severity, and therapeutic response of AD are traditionally reported through clinical assessments including the Eczema Area and Severity Index or Investigator's Global Assessment. These clinical rating scales are visual assessments used in clinical trials to denotate AD severity. Alternatively, biomarkers open the potential to further enhance diagnosis of AD, assess disease status and severity, and potentially enable tailored treatment options for patients. Biomarkers can be classified according to their clinical use, clinical presentation, and underlying/endogenous molecular mechanisms. Specifically, interleukin (IL)-13, which has been shown to be a key biomarker in AD pathogenesis, can be used for prediction of AD development and to monitor clinical severity/response to treatment. Treatment with tralokinumab, a human monoclonal antibody that binds directly to-and subsequently blocks signaling of-IL-13, has been shown to reduce inflammation, re-balance the skin microbiome, and improve the skin barrier in patients with AD. In this review, key AD-related biomarkers, the role of IL-13 in driving AD pathogenesis, and the impact of IL-13 inhibition by tralokinumab on other AD-related biomarkers are discussed.
Collapse
Affiliation(s)
- Joe Gorelick
- California Skin Institute, San Jose-Los Gatos, CA, USA.
| | | | | | | | | | - April W Armstrong
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
16
|
Salman S, Paulet V, Hardonnière K, Kerdine‐Römer S. The role of NRF2 transcription factor in inflammatory skin diseases. Biofactors 2025; 51:e70013. [PMID: 40207460 PMCID: PMC11983367 DOI: 10.1002/biof.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The skin is the body's largest organ and performs several vital functions, such as controlling the movement of essential substances while protecting against external threats. Although mainly composed of keratinocytes (KCs), the skin also contains a complex network of immune cells that play a critical role in host defense and maintaining skin homeostasis. KCs proliferate in the basal layer of the epidermis and undergo differentiation, altering their functional and phenotypic characteristics. These differentiation steps are crucial for the stratification of the epidermis and the formation of the stratum corneum, ensuring the skin barrier's functions. Exposure to UV, environmental pollutants, or chemicals can lead to an overproduction of reactive species of oxygen (ROS), leading to oxidative stress. To ensure redox homeostasis and prevent damage resulting from the formation of ROS, the skin has an extensive network of antioxidant defense systems, mainly orchestrated by the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. Indeed, Nrf2 induces the expression of detoxification and antioxidant enzymes and suppresses inductions of pro-inflammatory cytokine genes. In this context, Nrf2 is critical in preserving skin functions such as epidermal differentiation, regulating skin immunity, and managing environmental stresses. Besides, this pathway plays an important role in the pathogenesis of common inflammatory skin diseases such as allergic contact dermatitis, atopic dermatitis, and psoriasis. Therefore, the present review highlights the crucial role of Nrf2 in KCs for maintaining skin homeostasis and regulating skin immunity, as well as its contribution to the pathophysiology of inflammatory skin diseases. Finally, a particular emphasis will be placed on the therapeutic potential of targeting the Nrf2 pathway to alleviate symptoms of these inflammatory skin disorders.
Collapse
Affiliation(s)
- Sara Salman
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Virginie Paulet
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Kévin Hardonnière
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| | - Saadia Kerdine‐Römer
- Université Paris‐Saclay, INSERM, Inflammation, Microbiome and ImmunosurveillanceOrsayFrance
| |
Collapse
|
17
|
Natsuga K. Advanced phasing techniques in congenital skin diseases. J Dermatol 2025; 52:392-399. [PMID: 39723554 PMCID: PMC11883850 DOI: 10.1111/1346-8138.17597] [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: 09/10/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Phasing, the process of determining which alleles at different loci on homologous chromosomes belong together on the same chromosome, is crucial in the diagnosis and management of autosomal recessive diseases. Advances in long-read sequencing technologies have significantly enhanced our ability to accurately determine haplotypes. This review discusses the application of low-coverage long-read sequencing, nanopore Cas9-guided long-read sequencing, and adaptive sampling in phasing, highlighting their utility in complex clinical scenarios. Through clinical vignettes, we explore the importance of phasing in gene therapy design for recessive dystrophic epidermolysis bullosa and the role of revertant mosaicism in therapeutic epidermal autografts. Despite its promise, phasing with long-read sequencing faces challenges, including low efficiency in enriching target regions and the inherent error rate of nanopore sequencing. Future developments in long-read sequencing technologies will be critical in overcoming these limitations and expanding the applicability of phasing across various clinical settings.
Collapse
Affiliation(s)
- Ken Natsuga
- Department of Dermatology, Faculty of Medicine and Graduate School of MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
18
|
Sugiura S, Yoshida H, Sugiura H, Uehara M, Sugiura Y, Maruo Y, Hayashi Y, Yamamoto T, Kato T, Fujimoto N, Udagawa J. Increased intracellular stress responses and decreased KLF2 in adult patients with atopic dermatitis. Cell Stress Chaperones 2025; 30:84-99. [PMID: 39938773 PMCID: PMC11891603 DOI: 10.1016/j.cstres.2025.02.001] [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: 09/08/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Atopic dermatitis (AD) is prone to exacerbations in response to various triggering factors and flare-ups after remission. We searched for molecules associated with relapse/exacerbation of AD among molecules with altered gene expression in the skin of patients with AD. Microarray analyses were performed on lesional and nonlesional skin of adolescent or adult patients with recalcitrant AD and healthy controls. Five chaperones involved in intracellular stress responses, namely heat shock protein family A (Hsp70) member 9 (HSPA9), heat shock protein 90 beta family member 1 (HSP90B1), calnexin (CANX), malectin (MLEC; endoplasmic reticulum-associated degradation), and heat shock protein family D (Hsp60) member 1 (HSPD1), were consistently upregulated in involved and uninvolved skin of patients with AD. Damage-associated molecular patterns were upregulated in involved skin. KLF transcription factor 2 (KLF2) was decreased in involved skin and exhibited a decreasing trend in uninvolved skin of patients with AD. CD4(+)/CD8(+) double-positive cells (1.4% of T cells) were detected in lesions with declined KLF2 levels. WNT inhibitory factor 1 (WIF1) was downregulated in involved skin. Prolactin-induced protein was upregulated in only uninvolved skin of patients with AD. We found increased intracellular stress responses and decreased expression of KLF2 in the skin of patients with AD. Multifactorial genetic diseases, such as asthma, inflammatory bowel disease, type 2 diabetes, and rheumatoid arthritis, are associated with intracellular stress. Intracellular abnormalities may also be responsible for AD. Further research on AD may incorporate enhanced intracellular stress response and the decreased expression of KLF2 into the mechanism underlying AD.
Collapse
Affiliation(s)
- Shuji Sugiura
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan; Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan.
| | - Hiderou Yoshida
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Ako, Japan
| | - Hisashi Sugiura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan; Department of Dermatology, Sugiura Dermatology Clinic, Kusatsu, Japan
| | - Masami Uehara
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Yasuo Sugiura
- International Health Care Center, National Center for Global Health and Medicine, Tokyo, Japan; Department of Pediatrics, Navitas Clinic, Tokyo, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Yuji Hayashi
- Hospital Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takefumi Yamamoto
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Japan
| | - Takeshi Kato
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
19
|
Kumar M, Choi YG, Wong T, Li PH, Chow BKC. Beyond the classic players: Mas-related G protein-coupled receptor member X2 role in pruritus and skin diseases. J Eur Acad Dermatol Venereol 2025; 39:476-486. [PMID: 39044547 PMCID: PMC11851267 DOI: 10.1111/jdv.20249] [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: 04/02/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024]
Abstract
Chronic spontaneous urticaria (CSU), atopic dermatitis (AD), psoriasis and rosacea are highly prevalent inflammatory skin conditions which impose a significant burden on patients' quality of life. Their pathophysiology is likely multifactorial, involving genetic, immune and environmental factors. Recent advancements in the field have demonstrated the key role of mast cells (MC) in the pathophysiology of these conditions. The Mas-related G protein-coupled receptor X2 (MRGPRX2) has emerged as a promising non-IgE-mediated MC activation receptor. MRGPRX2 is predominately expressed on MC and activated by endogenous and exogenous ligands, leading to MC degranulation and release of various pro-inflammatory mediators. Mounting evidence on the presence of endogenous MRGPRX2 agonists (substance P, cortistatin-14, LL37, PAMP-12 and VIP) and its high expression among patients with CSU, AD, rosacea, psoriasis and chronic pruritus emphasizes the pathogenic role of MRGPRX2 in these conditions. Despite the currently available treatments, there remains a pressing need for novel drug targets and treatment options for these chronic inflammatory skin conditions. Here, we reviewed the pathogenic role of MRGPRX2 and its potential as a novel therapeutic target and provided an update on future research directions.
Collapse
Affiliation(s)
- Mukesh Kumar
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| | - Ye Gi Choi
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| | - Trevor Wong
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
- Faculty of Health SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Philip H. Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary HospitalThe University of Hong KongPokfulamHong Kong
| | - Billy K. C. Chow
- School of Biological SciencesThe University of Hong KongPokfulamHong Kong
| |
Collapse
|
20
|
Combarros D, Brahmi R, Musaefendic E, Heit A, Kondratjeva J, Moog F, Pressanti C, Lecru LA, Arbouille S, Laffort C, Goudounèche D, Brun J, Simon M, Cadiergues MC. Reconstructed Epidermis Produced with Atopic Dog Keratinocytes Only Exhibit Skin Barrier Defects after the Addition of Proinflammatory and Allergic Cytokines. JID INNOVATIONS 2025; 5:100330. [PMID: 39811760 PMCID: PMC11730559 DOI: 10.1016/j.xjidi.2024.100330] [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: 07/09/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 01/16/2025] Open
Abstract
Our objectives were to explore epidermal barrier defects in dogs with atopic dermatitis and to determine whether the defects are genetically determined or secondary to skin inflammation. First, the expression of filaggrin, corneodesmosin, and claudin1, analyzed using indirect immunofluorescence in skin biopsies collected from 32 healthy and 32 dogs with atopic dermatitis, was weaker in the atopic skin (P = .003). Second, primary keratinocytes of atopic dogs and healthy dogs were used to produce 3-dimensional reconstructed canine epidermis. The expression of the same proteins was analyzed using indirect immunofluorescence, immunoblotting, and RT-qPCR, whereas reconstructed canine epidermis morphology was investigated by transmission electron microscopy, and the barrier was investigated by functional assays. Next, inflammatory cytokines (IL-4, IL-13, IL-31, and TNFα) were added to the culture medium. The morphology, protein expression, and barrier function of the reconstructed canine epidermis were similar whether produced with keratinocytes from healthy dogs or dogs with atopy. Addition of inflammatory cytokines impaired the protein expression and epidermal barrier of the 2 types of reconstructed canine epidermis equally. To conclude, the reduced expression of epidermal barrier proteins observed in vivo was not reproduced in vitro unless cytokines were used, suggesting that it is induced by the inflammatory milieu.
Collapse
Affiliation(s)
- Daniel Combarros
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Rahma Brahmi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Emma Musaefendic
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Alizée Heit
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Jevgenija Kondratjeva
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Fabien Moog
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Charline Pressanti
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Line A. Lecru
- Clinique vétérinaire Hermes-Plage, Marseille, France
| | | | | | - Dominique Goudounèche
- Centre de Microscopie Electronique Appliquée à la Biologie, University of Toulouse, Toulouse, France
| | - Jessie Brun
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Michel Simon
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Marie-Christine Cadiergues
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| |
Collapse
|
21
|
Mukhatayev Z, Kovenskiy A, Ren Z, Rangel SM, Katkenov N, Khuanbai Y, Shivde R, Daniel M, Dellacecca ER, Cedercreutz K, Ostapchuk Y, Nurgozhina A, Chulenbayeva L, Nurgaziyev M, Jarmukhanov Z, Nurlankyzy M, Kozhdan K, Seidulla S, Mukhanbetzhanova Z, Sergazy S, Kozhakhmetov S, Ali Y, Daftary KM, Green SJ, Kundu RV, Kushugulova A, Le Poole IC. Escherichia Abundance and Metabolism Align with Vitiligo Disease Activity. J Invest Dermatol 2025:S0022-202X(25)00119-8. [PMID: 39983982 DOI: 10.1016/j.jid.2025.01.032] [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: 11/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Vitiligo is a cutaneous autoimmune disorder characterized by progressive depigmentation due to melanocyte destruction by cytotoxic T cells. Genetic factors predispose patients to the disease and are supported by environmental factors that often initiate new disease episodes. We investigated whether disease outcomes were partially defined by pathogenic microbes that drive nutrient deficiency and inflammation. Our study presents the results of research on the diet and gut microbiome composition of patients with vitiligo and healthy controls from Kazakhstan and the United States. Dietary nutrient intake was assessed using the National Institutes of Health-generated Diet History Questionnaire. Patients with active vitiligo exhibit a limited intake of specific fatty acids, amino acids, fiber, and zinc. Disease activity was further characterized by the abundance of Odoribacter and Escherichia in the gut. Metabolic pathway analysis supported the role of the Escherichia genus in disease activity by limiting energy metabolism and amino acid biosynthetic pathways. Disease activity also aligned with elevated circulating pro-inflammatory cytokines. These findings suggest that nutritional limitations are not compensated by metabolites from the gut microbiome in active disease, potentially leaving room for inflammation and exacerbating vitiligo. The intricate relationship among diet, gut microbiome composition, and disease progression in vitiligo highlights potential avenues for targeted interventions to reduce autoimmune activity and improve patient outcomes.
Collapse
Affiliation(s)
| | - Artur Kovenskiy
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Ziyou Ren
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stephanie M Rangel
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Nurlubek Katkenov
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Rohan Shivde
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Moriel Daniel
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Emilia R Dellacecca
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | - Kamilya Kozhdan
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | - Symbat Seidulla
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Shynggyss Sergazy
- National Laboratory Astana, Nazarbayev University, Astana, Kazakhstan
| | | | - Yasmeen Ali
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Karishma M Daftary
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stefan J Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, Illinois, USA
| | - Roopal V Kundu
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - I Caroline Le Poole
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.
| |
Collapse
|
22
|
Li M, Wang J, Liu Q, Liu Y, Mi W, Li W, Li J. Beyond the dichotomy: understanding the overlap between atopic dermatitis and psoriasis. Front Immunol 2025; 16:1541776. [PMID: 39995673 PMCID: PMC11847814 DOI: 10.3389/fimmu.2025.1541776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
Atopic dermatitis and psoriasis have traditionally been considered distinct inflammatory skin diseases with unique pathogenic mechanisms. However, accumulating evidence suggests significant overlap in their immunological pathways, metabolic features, and microbiome characteristics, challenging this conventional dichotomy. This review comprehensively examines the complex relationship between psoriasis and atopic dermatitis, with particular emphasis on their shared and distinct pathogenic mechanisms. We analyze the immunological networks, metabolic pathways, and microbial factors contributing to their development and progression. The review expands upon the disease spectrum hypothesis and discusses the nomenclature for conditions exhibiting features of both diseases. We critically evaluate the clinical and histopathological characteristics of concomitant psoriasis and atopic dermatitis, highlighting recent advances in molecular diagnostics for accurate disease differentiation. Importantly, we propose standardized diagnostic criteria for psoriasis dermatitis and examine current therapeutic strategies for managing overlapping conditions. Recent developments in targeted therapies and their implications for treatment selection are thoroughly discussed. By synthesizing current evidence and identifying knowledge gaps, this review provides insights into the complex interplay between psoriasis and atopic dermatitis, aiming to guide clinical decision-making and future research directions in this evolving field.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Jiangyi Wang
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Qingfeng Liu
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Youqing Liu
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyao Mi
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyi Li
- Department of Dermatology and Venerology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Nouri Z, Biglari S, Tabatabaiefar MA, Vahidnezhad F, Hozhabrpour A, March ME, Margolis DJ, Gudjonsson JE, Hakonarson H, Vahidnezhad H. Filaggrinopathies-FLG/FLG2: Diagnostic Complexities and Immunotherapy. J Invest Dermatol 2025:S0022-202X(24)03045-8. [PMID: 39927906 DOI: 10.1016/j.jid.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
FLG and FLG2 proteins are expressed in the outer layers of the epidermis, where they are vital in epidermal differentiation and skin barrier formation. Filaggrinopathies involving dysfunctions in these proteins are associated with a spectrum of phenotypic presentations, from monogenic to multifactorial conditions. This review examines biosynthesis and function of FLG and FLG2 proteins and evaluates their molecular pathogenesis in filaggrinopathies. Moreover, genotype-phenotype correlations are assessed, emphasizing genetic diagnosis complexities and diverse immune dysregulation patterns. Finally, it examines ongoing immunotherapeutic approaches by targeting different cytokines as promising treatment options for filaggrinopathies management.
Collapse
Affiliation(s)
- Zahra Nouri
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sajjad Biglari
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Fatemeh Vahidnezhad
- Department of Computer Science and Engineering Technology, University of Maryland Eastern Shore, Princess Anne, Maryland, USA
| | - Amir Hozhabrpour
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious disease, Iran University of Medical Sciences, Tehran, Iran
| | - Michael E March
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David J Margolis
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Hassan Vahidnezhad
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
24
|
Pan Y, Hochgerner M, Cichoń MA, Benezeder T, Bieber T, Wolf P. Langerhans cells: Central players in the pathophysiology of atopic dermatitis. J Eur Acad Dermatol Venereol 2025; 39:278-289. [PMID: 39157943 PMCID: PMC11760705 DOI: 10.1111/jdv.20291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease worldwide. AD is a highly complex disease with different subtypes. Many elements of AD pathophysiology have been described, but if/how they interact with each other or which mechanisms are important in which patients is still unclear. Langerhans cells (LCs) are antigen-presenting cells (APCs) in the epidermis. Depending on the context, they can act either pro- or anti-inflammatory. Many different studies have investigated LCs in the context of AD and found them to be connected to all major mechanisms of AD pathophysiology. As APCs, LCs recruit other immune cells and shape the immune response, especially adaptive immunity via polarization of T cells. As sentinel cells, LCs are primary sensors of the skin microbiome and are important for the decision of immunity versus tolerance. LCs are also involved with the integrity of the skin barrier by influencing tight junctions. Finally, LCs are important cells in the neuro-immune crosstalk in the skin. In this review, we provide an overview about the many different roles of LCs in AD. Understanding LCs might bring us closer to a more complete understanding of this highly complex disease. Potentially, modulating LCs might offer new options for targeted therapies for AD patients.
Collapse
Affiliation(s)
- Yi Pan
- Department of Dermatology and AllergyUniversity Hospital of BonnBonnGermany
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| | - Mathias Hochgerner
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan UniversityShanghaiChina
| | | | - Theresa Benezeder
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| | - Thomas Bieber
- Department of Dermatology and AllergyUniversity Hospital of BonnBonnGermany
- CK‐CARE, Medicine CampusDavosSwitzerland
- Department of DermatologyUniversity Hospital of ZürichZürichSwitzerland
| | - Peter Wolf
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| |
Collapse
|
25
|
Kenney HM, Yoshida T, Berdyshev E, Calatroni A, Gill SR, Simpson EL, Lussier S, Boguniewicz M, Hata T, Chiesa Fuxench ZC, De Benedetto A, Ong PY, Ko J, Davidson W, David G, Schlievert PM, Leung DYM, Beck LA. CERS1 is a biomarker of Staphylococcus aureus abundance and atopic dermatitis severity. J Allergy Clin Immunol 2025; 155:479-490. [PMID: 39343173 PMCID: PMC11805642 DOI: 10.1016/j.jaci.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/15/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Atopic dermatitis (AD) is an inflammatory skin condition characterized by widely variable cutaneous Staphylococcus aureus abundance that contributes to disease severity and rapidly responds to type 2 immune blockade (ie, dupilumab). The molecular mechanisms regulating S aureus levels between AD subjects remain poorly understood. OBJECTIVE We investigated host genes that may be predictive of S aureus abundance and correspond with AD severity. METHODS We studied data derived from the National Institutes of Health/National Institute of Allergy and Infectious Diseases-funded (NCT03389893 [ADRN-09]) randomized, double-blind, placebo-controlled multicenter study of dupilumab in adults (n = 71 subjects) with moderate-to-severe AD. Bulk RNA sequencing of skin biopsy samples (n = 57 lesional, 55 nonlesional) was compared to epidermal S aureus abundance, lipidomic, and AD clinical measures. RESULTS S aureus abundance and ceramide synthase 1 (CERS1) expression positively correlated at baseline across both nonlesional (r = 0.29, P = .030) and lesional (r = 0.41, P = .0015) skin. Lesional CERS1 expression also positively correlated with AD severity (ie, SCORAD r = 0.44, P = .0006) and skin barrier dysfunction (transepidermal water loss area under the curve r = 0.31, P = .025) at baseline. CERS1 expression (forms C18:0 sphingolipids) was negatively associated with elongation of very long-chain fatty acids (ELOVL6; C16:0→C18:0) expression and corresponded with a shorter chain length sphingolipid composition. Dupilumab rapidly reduced CERS1 expression (day 7) and ablated the relationship with S aureus abundance and ELOVL6 expression by day 21. CONCLUSION CERS1 is a unique molecular biomarker of S aureus abundance and AD severity that may contribute to dysfunctional skin barrier and shorter-chain sphingolipid composition through fatty acid sequestration as a maladaptive compensatory response to reduced ELOVL6.
Collapse
Affiliation(s)
- H Mark Kenney
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY
| | - Takeshi Yoshida
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY
| | - Evgeny Berdyshev
- Department of Medicine, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | | | - Steven R Gill
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, NY
| | - Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | | | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | - Tissa Hata
- Department of Dermatology, University of California, San Diego, Calif
| | | | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY
| | - Peck Y Ong
- Department of Pediatrics, University of Southern California, Division of Clinical Immunology and Allergy Children's Hospital Los Angeles, Los Angeles, Calif
| | - Justin Ko
- Department of Dermatology, Stanford University, Stanford, Calif
| | - Wendy Davidson
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | | | - Donald Y M Leung
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | - Lisa A Beck
- Department of Pathology & Laboratory Medicine, University of Rochester Medical Center, Rochester, NY; Department of Dermatology, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|
26
|
Baker MG, Ford LS, Campbell DE, Sampson HA. Just scratching the surface: A review of pediatric skin allergies. Pediatr Allergy Immunol 2025; 36:e70038. [PMID: 39953855 DOI: 10.1111/pai.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/16/2024] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Abstract
The skin is a large and sophisticated organ populated by innate and adaptive immune effector cells. These immune cells provide a critical first line of defense against pathogens, but genetic and environmental factors can lead to inappropriate signaling that may manifest as hypersensitivity. The most common cutaneous allergic disorders in children include atopic dermatitis, urticaria/angioedema, and contact dermatitis. In this review, we will briefly review these conditions, with a focus on recent developments in our understanding of the diagnosis and management of these disorders.
Collapse
Affiliation(s)
- Mary Grace Baker
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| | - Lara S Ford
- Department of Allergy & Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Dianne E Campbell
- Department of Allergy & Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- DBV Technologies, Montrouge, France
| | - Hugh A Sampson
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| |
Collapse
|
27
|
Marsella R. Skin Barrier in Normal and Allergic Horses: What Do We Know? Vet Sci 2025; 12:91. [PMID: 40005851 PMCID: PMC11861044 DOI: 10.3390/vetsci12020091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Information on skin barrier in horses is limited. A study on the epidermal ultrastructure of normal and allergic horses documented disorganized amorphous intercellular lipids in the stratum corneum of allergic samples. These findings are similar to atopic canine and human skin. Currently, there is no published study comparing skin barrier function parameters between normal and allergic horses; thus, the functional implications of the ultrastructural changes are unknown. In normal horses, body location, gender, breed, and ambient conditions affect skin barrier parameters, such as Transepidermal Water Loss. Skin microbiome studies on normal horses have highlighted the importance of season and environmental conditions, since horses housed together share similar microbiomes. Skin dysbiosis and predominance of staphylococcus have been described in horses with pastern dermatitis. Transcriptomic studies of the epidermis of normal and allergic horses have found that lesional allergic skin has substantial transcriptomic differences when compared with healthy skin, namely downregulation of genes of tight junctions, keratins, and upregulation of serine proteases and IL-13. Keratinocytes harvested from horses with insect bite hypersensitivity show upregulation of IL-31 gene expression under stimulation. While more research is clearly needed, preliminary results seem to support skin barrier differences between normal and allergic horses.
Collapse
Affiliation(s)
- Rosanna Marsella
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA
| |
Collapse
|
28
|
Sung YY, Yuk HJ, Kim SH, Kim DS. Effects of Earthworm ( Pheretima communisima) extract on atopic dermatitis: An in vitro and in vivo study. Heliyon 2025; 11:e41140. [PMID: 39758409 PMCID: PMC11699427 DOI: 10.1016/j.heliyon.2024.e41140] [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: 08/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/07/2025] Open
Abstract
Earthworm (Pheretima communisima) is used as a traditional medicine for the management of allergic airway inflammation. Atopic dermatitis (AD) is a persistent, recurrent disorder marked by allergic inflammation and skin barrier dysfunction. However, the pharmaceutical effects of earthworms on AD have not been defined. Our study examined the anti-allergic and anti-inflammatory actions of earthworm ethanolic extract (EWE) on allergic skin inflammation in a Dermatophagoides farinae mite antigen-induced AD mice model, TNF-α/IFN-γ-treated human keratinocytes, and compound 48/80-treated mouse mast cells. Oral administration of EWE in AD mouse reduced inflammatory cell accumulation, epidermal hyperplasia, and dermatitis severity in AD skin lesions and thymic stromal lymphopoietin (TSLP) and immunoglobulin (Ig) E concentrations in serum. EWE administration in AD mice also reduced secretion of Interleukin (IL)-4, IL-13, IL-5, and IFN-γ in cultures of isolated splenic cells. Immunohistofluorescence staining of skin lesions from AD mice revealed that EWE induced expression of claudin-1, filaggrin, and SIRT1. In HaCaT keratinocytes cotreated with IFN-γ and TNF-α, EWE inhibited secretion of the chemokine Regulated on Activation, Normal T Cell Expressed and Secreted (RANTES) in a dose-dependent manner. In addition, EWE inhibited histamine release in activated MC/9 mast cells. These results show that EWE might be therapeutics for the management of AD.
Collapse
Affiliation(s)
- Yoon-Young Sung
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| | - Heung-Joo Yuk
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, 62 Daehak-ro, Dong-gu, Daejeon, 34520, South Korea
| | - Dong-Seon Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| |
Collapse
|
29
|
Huang C, Zhuo F, Guo Y, Wang S, Zhang K, Li X, Dai W, Dou X, Yu B. Skin microbiota: pathogenic roles and implications in atopic dermatitis. Front Cell Infect Microbiol 2025; 14:1518811. [PMID: 39877655 PMCID: PMC11772334 DOI: 10.3389/fcimb.2024.1518811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic and inflammatory skin disorder characterized by impaired barrier function and imbalanced immunity. Recent advances have revealed that dysbiosis of skin microbiota plays important roles in the pathogenesis and development of AD. Meanwhile, endogenous and external factors contribute to the dysbiosis of skin microbiota in AD. Additionally, various treatments, including topical treatments, phototherapy, and systemic biologics, have demonstrated positive impacts on the clinical outcomes, alongside with the modulations of cutaneous microbiota in AD patients. Importantly, therapeutics or products regulating skin microbiota homeostasis have demonstrated potential for AD treatment in early clinical studies. In this review, we underline changes of the skin microbiota correlated with AD. Meanwhile, we provide an overview of the skin microbiota regarding its roles in the pathogenesis and development of AD. Finally, we summarize therapeutic strategies restoring the skin microbial homeostasis in AD management.
Collapse
Affiliation(s)
- Cong Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Fan Zhuo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yang Guo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Siyu Wang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Dermatology, Peking University Shenzhen Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Kaoyuan Zhang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiahong Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Wenkui Dai
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xia Dou
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
30
|
Erdem Y, Özkaya E, Erbilgin Y, Saritas M, Sayitoglu M, Su Küçük Ö, Salman A, Yucelten AD, Kocatürk E, Taskapan O, Gul A. Filaggrin Gene Variants Among Patients With Atopic Dermatitis in Turkey: A Multicenter Study Investigating Its Relationship With Disease Severity and Clinical/Laboratory Findings. Dermatitis 2025. [PMID: 39772616 DOI: 10.1089/derm.2024.0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Affiliation(s)
- Yasemin Erdem
- Department of Dermatology and Venereology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Esen Özkaya
- Department of Dermatology and Venereology, İstanbul Faculty of Medicine, İstanbul University, İstanbul, Turkey
| | - Yücel Erbilgin
- Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul University, İstanbul, Turkey
| | - Merve Saritas
- Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul University, İstanbul, Turkey
| | - Müge Sayitoglu
- Aziz Sancar Institute of Experimental Medicine, Department of Genetics, İstanbul University, İstanbul, Turkey
| | - Özlem Su Küçük
- Department of Dermatology, School of Medicine, Bezmialem Vakif University, İstanbul, Turkey
| | - Andaç Salman
- Department of Dermatology, Marmara University School of Medicine, İstanbul, Turkey
- Department of Dermatology, School of Medicine, Acıbadem Mehmet Ali Aydınlar University, İstanbul, Turkey
| | - Ayse Deniz Yucelten
- Department of Dermatology, Marmara University School of Medicine, İstanbul, Turkey
| | - Emek Kocatürk
- Department of Dermatology, Koç University School of Medicine, İstanbul, Turkey
- Institute of Allergology, Charitè, Medical University of Berlin, Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Allergology and Immunology, Berlin, Germany
| | - Oktay Taskapan
- Department of Dermatology, Yeditepe University School of Medicine, İstanbul, Turkey
| | - Ahmet Gul
- Division of Rheumatology, Department of Internal Medicine, İstanbul Faculty of Medicine, University of İstanbul, Turkey
| |
Collapse
|
31
|
Nene S, Devabattula G, Vambhurkar G, Tryphena KP, Singh PK, Khatri DK, Godugu C, Srivastava S. High mobility group box 1 cytokine targeted topical delivery of resveratrol embedded nanoemulgel for the management of atopic dermatitis. Drug Deliv Transl Res 2025; 15:134-157. [PMID: 38509343 DOI: 10.1007/s13346-024-01565-2] [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] [Accepted: 02/25/2024] [Indexed: 03/22/2024]
Abstract
Resveratrol is a polyphenolic compound showing anti-inflammatory activity by inhibition of high mobility group box 1 cytokine responsible for the activation of nuclear factor-κB pathway in atopic dermatitis. To evaluate the efficacy of resveratrol through topical route we have developed resveratrol-loaded nanoemulgel for the effective management of atopic dermatitis in mice model. The resveratrol-loaded nanoemulsion (0.5%, 0.75% and 1% w/w) was optimized by spontaneous nano-emulsification. The optimized resveratrol-loaded nanoemulsions showed average globule size in the 180-230 nm range and found to be monodispersed. The resveratrol nanoemulgel was prepared with a SEPINEO™ P 600 gel base and propylene glycol. Ex vivo permeation and retention study resulted in significantly higher skin retention of resveratrol from resveratrol-loaded nanoemulgel than free resveratrol-loaded gel. Preclinical efficacy of resveratrol nanoemulgel displayed promising therapeutic outcomes where, western blotting of skin tissues disclosed a significant reduction in the relative expression of high mobility group box 1, the receptor for advanced glycation end products, toll-like receptor-4 and phosphorylated nuclear factor-κB. Further, real-time polymerase chain reaction also disclosed a significant reduction in pro-inflammatory cytokines such as thymic stromal lymphopoietin, interleukin-4, interleukin-13, interleukin-31, tumor necrosis factor-α and interleukin-6. The histopathological examination of skin sections showed improvement in the skin condition. Collectively, the findings from our study showcased the significant improvement in the atopic dermatitis skin condition in mice model after topical application of resveratrol loaded nanoemulgel.
Collapse
Affiliation(s)
- Shweta Nene
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Geetanjali Devabattula
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Kamatham Pushpa Tryphena
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
32
|
Briot J, Pons C, Foucher A, Goudounèche D, Gaudenzio N, Donovan M, Bernard D, Méchin MC, Simon M. Prolyl Endopeptidase Is Involved in Filaggrinolysis and Cornification. J Invest Dermatol 2025; 145:98-108.e15. [PMID: 38879153 DOI: 10.1016/j.jid.2024.04.028] [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: 01/22/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 08/12/2024]
Abstract
FLG is a well-known biomarker of atopic dermatitis and skin dryness. Its full proteolysis (or filaggrinolysis) produces the major constituents of the natural moisturizing factor. Some proteases/peptidases remain to be identified in this multistep process. Mining 16 omics analyses, we identified prolyl endopeptidase (PREP) as a candidate peptidase. Indirect immunofluorescence and confocal analysis demonstrated its localization in the granular and deep cornified layers, where it colocalized with FLG. Tandem mass spectroscopy and fluorescent quenching activity assays showed that PREP cleaved several synthetic peptides derived from the FLG sequence, at the carboxyl side of an internal proline. Deimination of these peptides increased PREP enzymatic efficiency. Specific inhibition of PREP in reconstructed human epidermis using benzyloxycarbonyl-pro-prolinal induced the accumulation of FLG monomers. Downregulation of PREP expression in reconstructed human epidermis using RNA interference confirmed the impact of PREP on FLG metabolism and highlighted a more general role of PREP in keratinocyte differentiation. Indeed, quantitative global proteomic, western blotting, and RT-qPCR analyses showed a strong reduction in the expression of bleomycin hydrolase, known to be involved in filaggrinolysis, and of several other actors of cornification such as loricrin. Consequently, at the functional level, the transepidermal electric resistance was drastically reduced.
Collapse
Affiliation(s)
- Julie Briot
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), University of Toulouse, INSERM UMR1291 - CNRS UMR5051, Toulouse, France
| | - Carole Pons
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), University of Toulouse, INSERM UMR1291 - CNRS UMR5051, Toulouse, France
| | - Aude Foucher
- L'Oréal Research & Innovation, Aulnay-sous-Bois, France
| | - Dominique Goudounèche
- Centre de Microscopie Electronique Appliquée à la Biologie (CMEAB), Toulouse III University, Toulouse, France
| | - Nicolas Gaudenzio
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), University of Toulouse, INSERM UMR1291 - CNRS UMR5051, Toulouse, France; Genoskin SAS, Toulouse, France
| | - Mark Donovan
- L'Oréal Research & Innovation, Aulnay-sous-Bois, France
| | | | - Marie-Claire Méchin
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), University of Toulouse, INSERM UMR1291 - CNRS UMR5051, Toulouse, France
| | - Michel Simon
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), University of Toulouse, INSERM UMR1291 - CNRS UMR5051, Toulouse, France.
| |
Collapse
|
33
|
Zhou D, Gan G, Song S, Zi C, Bao Y, Hao W, Chen Q. The Impact of Immune Cells, Metabolites, Inflammatory Factors, and Circulating Proteins on Atopic Dermatitis: Insights from a Mendelian Randomization Study. Clin Cosmet Investig Dermatol 2024; 17:2999-3011. [PMID: 39723341 PMCID: PMC11669345 DOI: 10.2147/ccid.s495217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Background The onset of atopic dermatitis (AD) is complex, and its specific pathological mechanisms have not yet been fully elucidated. Methods Using circulating multi-omics as the exposure factors and AD as the outcome, we conducted univariable MR analysis. The circulating multi-omics data included immunomics (731 immune cell types), proteomics (4907 plasma proteins), metabolomics (1400 metabolites and 486 additional metabolites), and 91 inflammatory factors. MR analysis was conducted using IVW, WM, Simple Mode, Weighted Mode, and MR-Egger methods, with IVW as the primary analysis tool. To address horizontal pleiotropy, we utilized MR-Egger intercept tests and MR-PRESSO for correction, alongside the Cochrane Q statistic for heterogeneity assessment. Sensitivity analysis was performed using a leave-one-out strategy. To control for false positives due to multiple testing, we set a standard of a 5% false discovery rate. Additionally, we conducted F-statistics on the included SNPs to eliminate the impact of weak instrumental variables. Results IL-18R1 on AD (OR = 1.12, 95% CI: 1.08-1.17, P FDR < 0.01). Mannonate levels on AD (OR = 0.88, 95% CI: 0.83-0.94, P FDR = 0.03). Retinol (Vitamin A) to linoleoyl-arachidonoyl-glycerol (18:2 to 20:4) on AD (OR = 1.12, 95% CI: 1.06-1.18, PFDR = 0.03). HVEM on CM CD4+ cells on AD (OR = 0.81, 95% CI: 0.75-0.88, P FDR < 0.01). CR2 on AD (OR = 0.81, 95% CI: 0.72-0.90, P FDR = 0.04). MANSC1 on AD (OR = 0.87, 95% CI: 0.81-0.93, P FDR = 0.04). IL18R1 (4097 inflammatory markers) on AD (OR = 1.11, 95% CI: 1.06-1.17, P FDR = 0.01). HNRNPAB on AD (OR = 1.44, 95% CI: 1.23-1.70, P FDR < 0.01). Conclusion This study further explored the correlations between multi-omics data and AD. We identified seven previously unreported circulating substances with causal relationships to AD, filling a current theoretical gap.
Collapse
Affiliation(s)
- Dongqi Zhou
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Gaofeng Gan
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Shiwei Song
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Cangyan Zi
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Yichen Bao
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Wenfeng Hao
- Sichuan Taikang Hospital, Chengdu, Sichuan Province, 610213, People’s Republic of China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610072, People’s Republic of China
| |
Collapse
|
34
|
Park S, Yang J, Sun K, Park S, Lee J, Kim S, Lyu JH, Kim H. Dictamnus dasycarpus Turcz. Root Bark Improves Skin Barrier Function and Symptoms of Atopic Dermatitis in Mice. Int J Mol Sci 2024; 25:13178. [PMID: 39684888 DOI: 10.3390/ijms252313178] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
The root bark of Dictamus dasycarpus Turcz. has been traditionally used for the topical treatment of skin disorders like pruritus. This study was designed to investigate the inflammatory and skin barrier protective effects of D. dasycarpus in mice with calcipotriol (MC903)-induced atopic dermatitis (AD). Topical skin lesions on male Balb/c mice (8 weeks old) were treated topically with an ethanolic extract of D. dasycarpus (EEDD), and skin water content, water holding capacity (WHC), histopathological abnormalities, and inflammatory cytokine and chemokine levels were investigated. Topical application of EEDD effectively alleviated skin lesion severity, improved skin water content and WHC, and ameliorated histopathological abnormalities, including hyperkeratosis, blood vessel numbers near the epidermis, spongiotic changes, and immune cell infiltration in skin tissues. EEDD also suppressed inflammatory cytokines and chemokines, such as tumor necrosis factor (TNF)-α, thymic stromal lymphopoietin (TSLP), interleukin (IL)-1β, IL-4, IL-8, and monocyte chemotactic protein (MCP)-1. In RAW264.7 cells, EEDD reduced nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) expression and suppressed the phosphorylations of extracellular signal-regulated kinase (ERK) and p38. These results suggest that the root bark of D. dasycarpus has therapeutic potential due to its anti-dermatitis and skin barrier protective effects in AD and that it could be used as an ingredient in skincare products.
Collapse
Affiliation(s)
- Sangjun Park
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jinkyu Yang
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Kyoungmin Sun
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seonah Park
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jimi Lee
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Soyeon Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ji Hyo Lyu
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Hyungwoo Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
35
|
Zhang J, Liu Y, Zhang J, Zeng F, Wu Y, Zhang X, Zhang D, Zhu M. Exploring Genetic Association of Tea Intake With Allergic Diseases Among European Population: A Bidirectional Mendelian Randomization Study. Food Sci Nutr 2024; 12:10223-10230. [PMID: 39723051 PMCID: PMC11666899 DOI: 10.1002/fsn3.4574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/27/2024] [Accepted: 10/15/2024] [Indexed: 12/28/2024] Open
Abstract
Previous observational studies focused on the association of tea intake and allergic diseases. However, it is not known whether these associations are causal. We used a bidirectional Mendelian randomization (MR) study to assess the causal relationship of tea intake with the risk of allergic diseases, such as atopic dermatitis (AD), allergic rhinitis (AR), and allergic asthma (AA). Single-nucleotide polymorphisms (SNPs) which had genetic statistical significance with tea intake were used as instrumental variables (IVs). We employed heritable IVs of tea intake from the UK Biobank, which included 447,485 samples. Sensitivity analyses were further performed using MR Egger and MR-PRESSO. Inverse variance weighted (IVW) method was used as the main approach. In this MR study, 40 independent SNPs were selected for tea intake. The MR analysis revealed that an increase in genetically predicted tea intake was associated with a lower risk of AD (OR = 0.709, 95% CI = 0.546-0.919, p = 0.009). Furthermore, we observed a causal effect of genetically predicted tea intake on the risk of AA (OR = 0.498, 95% CI = 0.320-0.776, p = 0.002). However, no significant causal relationship was found between genetically predicted tea intake and AR (OR = 1.008, 95% CI = 0.998-1.017, p = 0.115). Our MR analysis suggested that increased tea intake may reduce the risk of AD and AA in European population. This suggests that tea intake is likely a trigger or a prevention strategy for AD and AA.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Pain MedicineJi'an Central People's HospitalJi'anChina
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
| | - Yuhan Liu
- School of Basic Medical SciencesJiangxi Medical College, Nanchang UniversityNanchangChina
| | - Jiawei Zhang
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
- Department of Pain MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Fei Zeng
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
- Department of Pain MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Yuqing Wu
- Department of AnesthesiologyThe Affiliated Stomatological Hospital of Nanchang UniversityNanchangChina
| | - Xuexue Zhang
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
- Department of Pain MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Daying Zhang
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
- Department of Pain MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| | - Mengye Zhu
- Key Laboratory of Neuropathic Pain, Healthcare Commission of Jiangxi ProvinceNanchangChina
- Department of Pain MedicineThe First Affiliated Hospital, Jiangxi Medical College, Nanchang UniversityNanchangChina
| |
Collapse
|
36
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Kistler W, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024; 79:3192-3237. [PMID: 39370939 DOI: 10.1111/all.16318] [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: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Walter Kistler
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
37
|
Shim KS, Kim HJ, Ji KY, Jung DH, Park SH, Song HK, Kim T, Kim KM. Rosmarinic Acid Ameliorates Dermatophagoides farinae Extract-Induced Atopic Dermatitis-like Skin Inflammation by Activating the Nrf2/HO-1 Signaling Pathway. Int J Mol Sci 2024; 25:12737. [PMID: 39684446 DOI: 10.3390/ijms252312737] [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: 10/25/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases. AD pathogenesis is associated with increased oxidative stress, impairment of the skin barrier, and activation of the immune response. Rosmarinic acid (RA), a caffeic acid ester, is known for its anti-inflammatory and antioxidant properties. However, the effects of RA on Dermatophagoides farinae extract (DfE)-induced AD-like skin inflammation, as well as its ability to regulate oxidative stress through the Nrf2/HO-1 pathway in TNF-α/IFN-γ-treated keratinocytes, remain unclear. We investigated RA activity in a DfE-induced AD-like skin inflammation mouse model and IFN-γ/TNF-α-stimulated keratinocytes. We found that RA attenuates DfE-induced inflammation by decreasing dermatitis scores and serum inflammatory marker levels and mast cell infiltration. Additionally, RA significantly suppressed IFN-γ/TNF-α-induced chemokine production in keratinocytes and reduced Th cytokine levels in concanavalin A-stimulated splenocytes. Importantly, RA also increased Nrf2/HO-1 expression in TNF-α/IFN-γ-treated keratinocytes. In conclusion, this study demonstrated that RA effectively alleviates DfE-induced AD-like skin lesions by reducing the levels of inflammatory cytokines and chemokines. Furthermore, RA promotes Nrf2/HO-1 signaling in keratinocytes, which may help mitigate DfE-induced oxidative stress, thereby alleviating AD-like skin inflammation. These findings highlight the potential of RA as a therapeutic agent for treating AD and other skin inflammation.
Collapse
Affiliation(s)
- Ki-Shuk Shim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Hye Jin Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Kon-Young Ji
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Dong Ho Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Sun Haeng Park
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Hyun-Kyung Song
- Practical Research Division, Honam National Institute of Biological Resources, Gohadoan-gil 99, Mokpo 58762, Republic of Korea
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Ki Mo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Yuseong-daero 1672, Yuseong-gu, Daejeon 34054, Republic of Korea
| |
Collapse
|
38
|
Hughes AJ, Barbosa E, Cernova J, Thomas BR, O'Shaughnessy RFL, O'Toole EA. Loss-of-function FLG mutations are associated with reduced history of acne vulgaris in a cohort of patients with atopic eczema of Bangladeshi ancestry in East London. Clin Exp Dermatol 2024; 49:1547-1553. [PMID: 38747172 DOI: 10.1093/ced/llae185] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/19/2024] [Accepted: 05/05/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Acne vulgaris (AV) is the eighth most common nonfatal disease globally. Previous work identified an association between AV and increased filaggrin (FLG) protein expression in the follicular epidermis, but further work did not find a clear link between loss-of-function (LoF) FLG gene mutations and protection from AV. OBJECTIVES To explore any association between AV and FLG LoF mutations in a cohort of genotyped patients of Bangladeshi ancestry with atopic eczema (AE) in East London. METHODS A retrospective notes review was performed on 245 patients who had been genotyped for FLG LoF mutations and undergone -clinical assessment. A χ2-test or Fisher's exact test was used to determine differences in AV history between FLG LoF genotype groups. RESULTS We found a significant reduction in history of AV in patients with AE with FLG LoF mutations (19 of 82) relative to those without FLG mutations (47 of 129) (23% vs. 36.4%; P = 0.02). We showed a nonsignificant reduction in AV diagnosis in patients with impaired barrier function (measured by transepidermal water loss) and palmar hyperlinearity. We found that patients with severe AE were less likely to have a history of AV only if they had an existing FLG LoF mutation (P = 0.02). CONCLUSIONS In the context of AE, our work suggests that FLG LoF mutations protect patients from developing AV.
Collapse
Affiliation(s)
- Aaron J Hughes
- Queen Mary University of London Centre for Cell Biology and Cutaneous Research, London, UK
- Department of Dermatology, Barts Health NHS Trust, London, UK
| | - Elsa Barbosa
- Department of Dermatology, Barts Health NHS Trust, London, UK
| | - Jeva Cernova
- Department of Dermatology, Barts Health NHS Trust, London, UK
| | - Bjorn R Thomas
- Queen Mary University of London Centre for Cell Biology and Cutaneous Research, London, UK
| | - Ryan F L O'Shaughnessy
- Queen Mary University of London Centre for Cell Biology and Cutaneous Research, London, UK
| | - Edel A O'Toole
- Queen Mary University of London Centre for Cell Biology and Cutaneous Research, London, UK
- Department of Dermatology, Barts Health NHS Trust, London, UK
| |
Collapse
|
39
|
Yamamura K, Ohno F, Yotsumoto S, Sato Y, Kimura N, Nishio K, Inoue K, Ichiki T, Kuba-Fuyuno Y, Fujishima K, Ito T, Kido-Nakahara M, Tsuji G, Nakahara T. Extracellular ATP Contributes to Barrier Function and Inflammation in Atopic Dermatitis: Potential for Topical Treatment of Atopic Dermatitis by Targeting Extracellular ATP. Int J Mol Sci 2024; 25:12294. [PMID: 39596359 PMCID: PMC11595171 DOI: 10.3390/ijms252212294] [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: 10/29/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Atopic dermatitis (AD) is characterized by chronic inflammation, barrier dysfunction, and pruritus, exacerbated by external stimuli, such as scratching. This study investigates the role of extracellular adenosine triphosphate (ATP) in the pathophysiology of AD and assesses the therapeutic potential of clodronate, an ATP release inhibitor. Our research demonstrates that extracellular ATP impairs skin barrier function by reducing the filaggrin expression in the keratinocytes, a critical protein for barrier integrity. Furthermore, ATP release, triggered by IL-4 and mechanical stimuli, amplifies inflammation by promoting cytokine and chemokine production by the immune cells. Clodronate, by inhibiting ATP release, restores the filaggrin levels in the keratinocytes, reduces TARC production in the dendritic cells, and alleviates AD symptoms in a mouse model. These findings suggest that targeting extracellular ATP could offer a novel therapeutic approach to improving skin barrier function and reducing inflammation in AD. Future studies should explore the long-term efficacy and safety of ATP-targeted therapies in clinical settings.
Collapse
Affiliation(s)
- Kazuhiko Yamamura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Fumitaka Ohno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Shu Yotsumoto
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Yuki Sato
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Nanae Kimura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Kiichiro Nishio
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Keiichi Inoue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Toshio Ichiki
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Yoko Kuba-Fuyuno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Kei Fujishima
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
| | - Gaku Tsuji
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.Y.)
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, Fukuoka 812-8582, Japan
| |
Collapse
|
40
|
Yew YW, Loh M, Brown SJ. Understanding Atopic Dermatitis in Asian and European Population Cohorts Using Complementary Omics Techniques. J Invest Dermatol 2024:S0022-202X(24)02171-7. [PMID: 39503693 DOI: 10.1016/j.jid.2024.08.036] [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: 05/16/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 11/08/2024]
Abstract
Atopic dermatitis is highly heterogeneous with respect to pathogenesis, clinical manifestations, and treatment response. There is evidence that ancestry and skin type each contribute to this heterogeneity, indicating the need to improve understanding of disease mechanisms in diverse populations. Methods to integrate multiomics studies have been well-described, but this review focuses on the importance and the strategies needed to integrate data across different ancestral groups, focusing, because of data availability, on Asian and European populations. Skin scientists and clinicians will each benefit from an understanding of how the multiple complimentary layers of omics data may inform future clinical management, from insight into disease pathogenesis and treatment targets.
Collapse
Affiliation(s)
- Yik Weng Yew
- National Skin Centre, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| | - Marie Loh
- National Skin Centre, Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore; Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom; Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Sara J Brown
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom; Department of Dermatology, NHS Lothian, Edinburgh, United Kingdom
| |
Collapse
|
41
|
Tang X, Li M. The role of the skin in the atopic march. Int Immunol 2024; 36:567-577. [PMID: 39271155 DOI: 10.1093/intimm/dxae053] [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: 06/04/2024] [Accepted: 09/12/2024] [Indexed: 09/15/2024] Open
Abstract
Atopic diseases, including atopic dermatitis (AD), food allergy (FA), asthma, and allergic rhinitis (AR) are closely related to inflammatory diseases involving different body sites (i.e. the skin, airway, and digestive tract) with characteristic features including specific IgE to allergens (so-called "atopy") and Th2 cell-mediated inflammation. It has been recognized that AD often precedes the development of other atopic diseases. The progression from AD during infancy to FA or asthma/AR in later childhood is referred to as the "atopic march" (AM). Clinical, genetic, and experimental studies have provided evidence that allergen sensitization occurring through AD skin could be the origin of the AM. Here, we provide an updated review focusing on the role of the skin in the AM, from genetic mutations and environmental factors associated with epidermal barrier dysfunction in AD and the AM to immunological mechanisms for skin sensitization, particularly recent progress on the function of key cytokines produced by epidermal keratinocytes or by immune cells infiltrating the skin during AD. We also highlight the importance of developing strategies that target AD skin to prevent and attenuate the AM.
Collapse
Affiliation(s)
- Xin Tang
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, Inserm U 1258, Université de Strasbourg, Illkirch 67404, France
- Department of Dermatology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 40000, People's Republic of China
| | - Mei Li
- Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR 7104, Inserm U 1258, Université de Strasbourg, Illkirch 67404, France
| |
Collapse
|
42
|
Ito T, Nakamura Y. The skin barrier and microbiome in infantile atopic dermatitis development: can skincare prevent onset? Int Immunol 2024; 36:579-584. [PMID: 38887075 DOI: 10.1093/intimm/dxae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024] Open
Abstract
Atopic dermatitis (AD), a prevalent Th2-dominant skin disease, involves complex genetic and environmental factors, including mutations in the Filaggrin gene and dysbiosis of skin microbiota characterized by an increased abundance of Staphylococcus aureus. Our recent findings emphasize the pivotal role of the skin barrier's integrity and microbial composition in infantile AD and allergic diseases. Early skin dysbiosis predisposes infants to AD, suggesting targeted skincare practices as a preventive strategy. The effects of skincare interventions, particularly the application of moisturizers with the appropriate molar concentration of ceramides, cholesterol, and fatty acids, play a crucial role in restoring the skin barrier. Notably, our study revealed that appropriate skincare can reduce Streptococcus abundance while supporting Cutibacterium acnes presence, thus directly linking skincare practices to microbial modulation in neonatal skin. Despite the mixed outcomes of previous Randomized Controlled Trials on the efficacy of moisturizers in AD prevention, our research points to the potential of skincare intervention as a primary preventive method against AD by minimizing the impact of genetic and environmental factors. Furthermore, our research supports the notion that early aggressive management of eczema may reduce the incidence of food allergies, highlighting the necessity for multifaceted prevention strategies that address both the skin barrier and immune sensitization. By focusing on repairing the skin barrier and adjusting the skin's microbiome from birth, we propose a novel perspective on preventing infantile AD and allergic diseases, opening new avenues for future studies, and practices in allergy prevention.
Collapse
Affiliation(s)
- Tomoka Ito
- Department of Dermatology, The University of Osaka, Graduate School of Medicine, Osaka 565-0871, Japan
| | - Yuumi Nakamura
- Department of Dermatology, The University of Osaka, Graduate School of Medicine, Osaka 565-0871, Japan
- Cutaneous Allergy and Host Defense, Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
| |
Collapse
|
43
|
Kiss O, Bahri R, Watson REB, Chike C, Langton AK, Newton VL, Bell M, Griffiths CEM, Bulfone-Paus S, Pilkington SM. The impact of irritant challenge on the skin barrier and myeloid-resident immune cells in women who are postmenopausal is modulated by hormone replacement therapy. Br J Dermatol 2024; 191:746-759. [PMID: 38819239 DOI: 10.1093/bjd/ljae226] [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: 03/06/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND Sex hormone changes during menopausal transition contribute to declining skin health. However, how menopause and its treatment by hormone replacement therapy (HRT) impact the skin barrier and immune system is unclear. OBJECTIVES To examine how menopause and HRT affect the skin barrier and immune cell composition in postmenopausal women following irritant challenge. METHODS Two cohorts of postmenopausal women were recruited to the study. The first cohort consisted of 10 untreated women [HRT-; mean (SEM) age 56.5 (1.6) years (range 48-63)] and the second was composed of 8 women receiving HRT [HRT+; mean (SEM) age 54.0 (2.1) years (range 48-63)]. Skin irritation was induced by applying topical sodium lauryl sulfate (SLS) 1.25% to occluded buttock skin for 48 h. Clinical assessment was conducted after 24 h, followed by biopsy of both SLS-challenged and unchallenged skin for analysis of skin barrier proteins and immune cell distribution using immunofluorescence. RESULTS Clinically, there were no significant differences in skin irritant responses between those taking or not taking HRT (including increased skin redness and blood flow). In response to SLS challenge a significant increase in transepidermal water loss (P < 0.05), filaggrin deposition and cytokeratin 10 (K10)+ cell layers (P < 0.01) was observed in individuals receiving HRT compared with the HRT- group. Following SLS challenge in individuals taking HRT, a significant (P < 0.01) reduction in CD207+ cells in the epidermis was observed, accompanied by an increase of CD207+ cells in the dermis, indicative of migrating Langerhans cells (LCs). Significantly fewer migrating LCs were found in those who were not receiving HRT (P < 0.01). Furthermore, the numbers of dermal dendritic cells (DCs), macrophages, and CD11c+CD206- and CD68+CD206- subsets were found to be significantly (P < 0.05) higher in those taking HRT following SLS challenge. CONCLUSIONS Individuals receiving HRT displayed enhanced skin barrier response to SLS challenge with thicker filaggrin and increased K10+ epidermal cell layers. Following challenge, HRT users exhibited elevated LC, inflammatory DC and macrophage counts in the dermis. These may render skin both more prone to inflammation and more capable of resolving it, while also promoting skin repair.
Collapse
Affiliation(s)
- Orsolya Kiss
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Rajia Bahri
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation and Manchester Collaborative Centre for Inflammation Research, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Rachel E B Watson
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), National Skin Centre and Skin Research Institute of Singapore (SRIS), Republic of Singapore
| | - Chidera Chike
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Abigail K Langton
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Mike Bell
- No7 Beauty Company, Walgreens Boots Alliance, Nottingham, UK
| | - Christopher E M Griffiths
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Department of Dermatology, King's College Hospital NHS Foundation Trust, King's College London, London, UK
| | - Silvia Bulfone-Paus
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation and Manchester Collaborative Centre for Inflammation Research, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Suzanne M Pilkington
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, The University of Manchester and Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
44
|
Gautam Y, Satish L, Ramirez S, Grashel B, Biagini JM, Martin LJ, Rothenberg ME, Khurana Hershey GK, Mersha TB. Joint genotype and ancestry analysis identify novel loci associated with atopic dermatitis in African American population. HGG ADVANCES 2024; 5:100350. [PMID: 39245941 PMCID: PMC11470243 DOI: 10.1016/j.xhgg.2024.100350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic itchy inflammatory disease of the skin. Genetic studies have identified multiple risk factors linked to the disease; however, most of the studies have been derived from European and East Asian populations. The admixed African American (AA) genome may provide an opportunity to discovery ancestry-specific loci involved in AD susceptibility. Herein, we present joint analysis of ancestry and genotype effects followed by validation using differential gene expression analysis on AD using 726 AD-affected individuals and 999 non-AD control individuals from the AA population, genotyped using Multi-Ethnic Global Array (MEGA) followed by imputation using the Consortium on Asthma among African Ancestry Populations in the Americas (CAAPA) reference panel. The joint analysis identified two novel AD-susceptibility loci, rs2195989 in gene ANGPT1 (8q23.1) and rs62538818 in the intergenic region between genes LURAP1L and MPDZ (9p23). Admixture mapping (AM) results showed potential genomic inflation, and we implemented genomic control and identified five ancestry-of-origin loci with European ancestry effects. The multi-omics functional prioritization of variants in AM signals prioritized the loci SLAIN2, RNF39, and FOXA2. Genome-wide association study (GWAS) identified variants significantly associated with AD in the AA population, including SGK1 (rs113357522, odds ratio [OR] = 2.81), EFR3A (rs16904552, OR = 1.725), and MMP14 (rs911912, OR = 1.791). GWAS variants were common in the AA but rare in the European population, which suggests an African-ancestry-specific risk of AD. Four genes (ANGPT1, LURAP1L, EFR3A, and SGK1) were further validated using qPCR from AD and healthy skin. This study highlighted the importance of genetic studies on admixed populations, as well as local ancestry and genotype-ancestry joint effects to identify risk loci for AD.
Collapse
Affiliation(s)
- Yadu Gautam
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Latha Satish
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Stephen Ramirez
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Brittany Grashel
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Jocelyn M Biagini
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Lisa J Martin
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Tesfaye B Mersha
- Division of Asthma Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
45
|
Delanghe L, De Boeck I, Van Malderen J, Allonsius CN, Van Rillaer T, Bron PA, Claes I, Hagendorens M, Lebeer S, Leysen J. Mild atopic dermatitis is characterized by increase in non-staphylococcus pathobionts and loss of specific species. Sci Rep 2024; 14:23659. [PMID: 39390034 PMCID: PMC11467409 DOI: 10.1038/s41598-024-74513-2] [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: 01/11/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Atopic dermatitis is the most common inflammatory skin condition with a severe negative impact on patients' quality of life. The etiology of AD is complex and depends on age, genetics, the immune system, environmental factors, and the skin microbiome, with a key role for pathogenic Staphylococcus aureus in the development of severe AD. However, the composition of the skin microbiome in mild AD is understudied. Here, using metagenomic shallow shotgun sequencing, we showed that mild AD lesions did not show a significant difference in the diversity of the skin microbiome compared to samples from non-AD patients and that the relative abundance of S. aureus did not differ in these mild AD lesions. However, when we assessed other taxa, Mycobacterium ostraviense, Pedobacter panaciterrae_A and four Streptomyces species were identified with higher abundances in mild AD lesions and species of 15 genera were decreased in abundance. The highest fold decreases were observed for Paracoccus marcusii, Microbacterium lacticum, Micrococcus luteus, and Moraxella sp002478835. These microbiome compositional insights are a first step towards novel microbiome-based diagnostics and therapeutics for early intervention at the stage of mild AD and provide a path forward for the functional study of species involved in this often-overlooked patient population.
Collapse
Affiliation(s)
- Lize Delanghe
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | - Ilke De Boeck
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | - Joke Van Malderen
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | - Camille Nina Allonsius
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | - Tim Van Rillaer
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | - Peter A Bron
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium
| | | | - Margo Hagendorens
- University Hospital Antwerp, Department of Pediatrics, University of Antwerp, Wilrijkstraat 10, Edegem, B-2650, Belgium
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, Antwerpen, B-2020, Belgium.
| | - Julie Leysen
- University Hospital Antwerp, Department of Dermatology, University of Antwerp, Wilrijkstraat 10, Edegem, B-2650, Belgium
| |
Collapse
|
46
|
Almalki B, Alghamdi Y, Aman A, Alamri S, Alshareef A, Alraddadi A. Autosomal recessive congenital ichthyosis caused by a novel variant in cornifelin gene: A case report. JAAD Case Rep 2024; 52:25-27. [PMID: 39282523 PMCID: PMC11401099 DOI: 10.1016/j.jdcr.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Affiliation(s)
- Basel Almalki
- Dermatology Department, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Yara Alghamdi
- Department of Dermatology, King Abdullah Medical Complex, Jeddah, Saudi Arabia
| | - Abdullah Aman
- Dermatology Department, King Fahad General Hospital, Jeddah, Saudi Arabia
| | - Samer Alamri
- Dermatology Department, King Fahad General Hospital, Jeddah, Saudi Arabia
- Dermatology Department, King Abdulaziz Medical City, Ministry of National Guard, Jeddah, Saudi Arabia
| | - Alhussain Alshareef
- Dermatology Department, King Fahad General Hospital, Jeddah, Saudi Arabia
- Dermatology Department, King Abdulaziz Medical City, Ministry of National Guard, Jeddah, Saudi Arabia
| | - Ali Alraddadi
- Dermatology Department, King Fahad General Hospital, Jeddah, Saudi Arabia
- Dermatology Department, King Abdulaziz Medical City, Ministry of National Guard, Jeddah, Saudi Arabia
| |
Collapse
|
47
|
Trogen B, Verma M, Sicherer SH, Cox A. The Role of Food Allergy in Atopic Dermatitis. Dermatol Clin 2024; 42:527-535. [PMID: 39278706 DOI: 10.1016/j.det.2024.04.004] [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] [Indexed: 09/18/2024]
Abstract
Atopic dermatitis (AD) and food allergies are 2 atopic conditions that tend to develop early in life. Their interrelationship has been a topic of controversy and many studies. The presence of atopic dermatitis in infancy and early childhood, particularly if severe, is a risk factor for the development of immunoglobulin E (IgE) -mediated food allergies. While it is common for children with AD to demonstrate extensive sensitization to foods, serum IgE testing is not always indicative of clinical allergy.
Collapse
Affiliation(s)
- Brit Trogen
- Division of Pediatric Allergy, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA
| | - Megha Verma
- Department of Internal Medicine, Mount Sinai Morningside/West, 1111 Amsterdam Avenue, New York, NY 10025, USA
| | - Scott H Sicherer
- Division of Pediatric Allergy, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA
| | - Amanda Cox
- Division of Pediatric Allergy, Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, New York, NY 10029, USA.
| |
Collapse
|
48
|
Wittmann M, Smith IL, Brown ST, Berekméri A, Vargas-Palacios A, Sunderland L, Barker A, Cowdell F, Ersser S, Gilberts R, Green C, Hampton P, Smith C, Nixon J. Alitretinoin versus phototherapy as the first-line treatment in adults with severe chronic hand eczema: the ALPHA RCT. Health Technol Assess 2024; 28:1-123. [PMID: 39364555 PMCID: PMC11472215 DOI: 10.3310/twqc0141] [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] [Indexed: 10/05/2024] Open
Abstract
Background Hand eczema is common and a cause of morbidity and occupational disability. When education, irritant/contact allergen avoidance, moisturisation and topical corticosteroids are insufficient to control chronic hand eczema, ultraviolet therapy or systemic immune-modifying drugs are used. There is no treatment pathway generally accepted by UK dermatologists. Primary objective Compare alitretinoin and ultraviolet therapy as first-line therapy in terms of disease activity at 12 weeks post planned start of treatment. Design Prospective, multicentre, open-label, two-arm parallel group, adaptive randomised controlled trial with one planned interim analysis, and an economic evaluation. Setting UK secondary care dermatology outpatient clinics. Participants Patients with severe chronic hand eczema unresponsive to at least 4 weeks of treatment with potent topical corticosteroids. Primary end point Natural logarithm of the Hand Eczema Severity Index + 1, 12 weeks post planned start of treatment. Randomisation Participants randomised 1 : 1 by minimisation to alitretinoin or ultraviolet therapy for 12 to 24 weeks. Blinding Blinded primary end-point assessor. Results Intention-to-treat population: 441 (100.0%) participants; 220 (49.9%) alitretinoin and 221 (50.1%) ultraviolet therapy. At least one dose was received by 212 (96.4%) alitretinoin and 196 (88.7%) ultraviolet therapy participants. Primary outcome The unadjusted median (interquartile range) relative change in hand eczema severity index at 12 weeks was 30% (10-70%) of that at baseline for alitretinoin compared with 50% (20-100%) for ultraviolet therapy. There was a statistically significant benefit of alitretinoin compared with ultraviolet therapy at 12 weeks, with an estimated fold change or relative difference (95% confidence interval) = 0.66 (0.52 to 0.82), p = 0.0003 at 12 weeks. There was no evidence of a difference at 24 or 52 weeks, with the estimated fold change (95% confidence interval) equal to 0.92 (0.798 to 1.08) and 1.27 (0.97 to 1.67), respectively. Primary analysis results were consistent for secondary end points Fifty-nine per cent allocated to alitretinoin and 61% allocated to ultraviolet therapy achieved a clear/almost clear assessment during the trial period. Differential treatment compliance observed: 145 (65.9%) alitretinoin and 53 (24.0%) ultraviolet therapy participants confirmed compliance (≥ 80% received, no treatment breaks > 7 days during first 12 weeks). High levels of missing data were observed. Safety One hundred and thirty-five reportable adverse events across 79 participants, 55 (25.0%) alitretinoin and 24 (10.9%) ultraviolet therapy. Four serious adverse events (two alitretinoin, two ultraviolet therapy). Four pregnancies reported (three alitretinoin, one ultraviolet therapy). No new safety signals were detected. Conclusion As a first-line therapy, alitretinoin showed more rapid improvement and superiority to ultraviolet therapy at week 12. This difference was not observed at later time points. Alitretinoin is cost-effective at weeks 12 and 52. Ultraviolet therapy is cost-effective after 10 years, with a high degree of uncertainty. Hand eczema severity index may be a useful primary outcome measure for hand eczema trials; ALPHA results will inform future trials. Limitations Treatment compliance was poor for ultraviolet therapy. Regular twice weekly treatment was not received by most patients. Assessment of long-term effects of randomised treatments was complicated by use of second-line treatments post treatment phase. Further work Further analysis of substudies and pilot data will provide valuable information for future studies. A clear need for better therapeutic approaches for severe chronic hand eczema remains. Future studies will need to further address long-term benefits of treatments given. Trial registration This trial is registered as ISRCTN80206075. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 12/186/01) and is published in full in Health Technology Assessment; Vol. 28, No. 59. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, Yorkshire, UK
- University Medical Centre of Johannes Gutenberg University Mainz, Mainz, Germany
| | - Isabelle L Smith
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Sarah Tess Brown
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Anna Berekméri
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, Yorkshire, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit (LMBRU), Chapel Allerton Hospital, Leeds, UK
| | - Armando Vargas-Palacios
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | | | | | - Fiona Cowdell
- Faculty of Health Education and Life Sciences, Birmingham City University, Birmingham, UK
| | - Steven Ersser
- Department of Nursing Science, Faculty of Health and Social Science, Bournemouth University, Bournemouth, UK
| | - Rachael Gilberts
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
| | - Cathy Green
- Department of Dermatology, Ninewells Hospital, Dundee, Tayside, UK
| | - Philip Hampton
- Department of Dermatology, Newcastle Hospital, Newcastle, Tyne and Wear, UK
| | - Catherine Smith
- St John's Institute of Dermatology, Guy's and St Thomas's NHS Foundation Trust, London, UK
| | - Jane Nixon
- Clinical Trials Research Unit, Leeds Institute of Clinical Trials Research, University of Leeds, Leeds, UK
- Academic Unit of Health Economics, Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
49
|
Zhu R, Yao X, Li W. Langerhans cells and skin immune diseases. Eur J Immunol 2024; 54:e2250280. [PMID: 39030782 DOI: 10.1002/eji.202250280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Langerhans cells (LCs) are the key antigen-presenting cells in the epidermis in normal conditions and respond differentially to environmental and/or endogenous stimuli, exerting either proinflammatory or anti-inflammatory effects. Current knowledge about LCs mainly originates from studies utilizing mouse models, whereas with the development of single-cell techniques, there has been significant progress for human LCs, which has updated our understanding of the phenotype, ontogeny, differentiation regulation, and function of LCs. In this review, we delineated the progress of human LCs and summarized LCs' function in inflammatory skin diseases, providing new ideas for precise regulation of LC function in the prevention and treatment of skin diseases.
Collapse
Affiliation(s)
- Ronghui Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
- Department of Dermatology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Hubei Province & Key Laboratory of Skin Infection and Immunity, Wuhan, P. R. China
| | - Xu Yao
- Department, of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, P. R. China
| | - Wei Li
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, P. R. China
| |
Collapse
|
50
|
Davis KL, Claudio-Etienne E, Frischmeyer-Guerrerio PA. Atopic dermatitis and food allergy: More than sensitization. Mucosal Immunol 2024; 17:1128-1140. [PMID: 38906220 PMCID: PMC11471387 DOI: 10.1016/j.mucimm.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/01/2024] [Accepted: 06/13/2024] [Indexed: 06/23/2024]
Abstract
The increased risk of food allergy in infants with atopic dermatitis (AD) has long been recognized; an epidemiologic phenomenon termed "the atopic march." Current literature supports the hypothesis that food antigen exposure through the disrupted skin barrier in AD leads to food antigen-specific immunoglobulin E production and food sensitization. However, there is growing evidence that inflammation in the skin drives intestinal remodeling via circulating inflammatory signals, microbiome alterations, metabolites, and the nervous system. We explore how this skin-gut axis helps to explain the link between AD and food allergy beyond sensitization.
Collapse
Affiliation(s)
- Katelin L Davis
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Comparative Biomedical Scientist Training Program, The Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, The National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Comparative Pathobiology Department, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Estefania Claudio-Etienne
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, The National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|