1
|
Tamminga SM, Van Der Wal MM, Saager ES, Van Der Gang LF, Boesjes CM, Hendriks A, Pannekoek Y, De Bruin MS, Van Wijk F, Van Sorge NM. Single-cell sequencing of human Langerhans cells identifies altered gene expression profiles in patients with atopic dermatitis. Immunohorizons 2025; 9:vlae009. [PMID: 39849992 DOI: 10.1093/immhor/vlae009] [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: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 01/25/2025] Open
Abstract
Atopic dermatitis (AD) is characterized by dysregulated T cell immunity and skin microbiome dysbiosis with predominance of Staphylococcus aureus, which is associated with exacerbating AD skin inflammation. Specific glycosylation patterns of S. aureus cell wall structures amplify skin inflammation through interaction with Langerhans cells (LCs). Nevertheless, the role of LCs in AD remains poorly characterized. Here, we performed single cell RNA sequencing of primary epidermal LCs and dermal T cells, isolated from skin biopsies of AD patients and healthy control subjects, alongside specific glycoanalysis of S. aureus strains isolated from the AD lesions. Our findings revealed 4 LC subpopulations ie, 2 steady-state clusters [LC1 and LC1H] and 2 proinflammatory/matured subsets [LC2 and migratory LCs]. The latter 2 subsets were enriched in AD skin. AD LCs showed enhanced expression of C-type lectin receptors, the high-affinity IgE receptor, and activation of prostaglandin and leukotriene biosynthesis pathways, upregulated transcriptional signatures related to T cell activation pathways, and increased expression of CCL17 compared with healthy LCs. Correspondingly, T helper 2 and T regulatory cell populations were increased in AD lesions. Complementary, we performed bulk RNA sequencing of primary LCs stimulated with the S. aureus strains isolated from the AD lesions, which showed upregulation of T helper 2-related pathways. Our study provides proof-of-concept for a role of LCs in connecting the S. aureus-T cell axis in the AD inflammatory cycle.
Collapse
Affiliation(s)
- Sara M Tamminga
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - M Marlot Van Der Wal
- Center for Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Elise S Saager
- Center for Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lian F Van Der Gang
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Celeste M Boesjes
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Astrid Hendriks
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Yvonne Pannekoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Marjolein S De Bruin
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Femke Van Wijk
- Center for Translational Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nina M Van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| |
Collapse
|
2
|
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
|
3
|
Welponer T, Puttinger C, Korte EWH, van der Werf S, Prodinger C, Bolling MC, Laimer M. Systematic review on antipruritic therapies for patients with Epidermolysis bullosa. J DERMATOL TREAT 2024; 35:2381762. [PMID: 39069296 DOI: 10.1080/09546634.2024.2381762] [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: 05/29/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Itch is one of the most burdensome symptoms in epidermolysis bullosa (EB), indicating a hitherto unmet therapeutic need. This review leverages existing data on efficacy of itch treatment in EB to support sound decision making. METHODS A systematic literature search was performed on 29 March 2022. Studies written later than 1991 and reporting outcomes in patients with EB treated for itch were considered. RESULTS Of the 3,099 articles screened, 21 studies met eligibility criteria, comprising 353 patients (65.9%) diagnosed for recessive dystrophic EB. Only two studies (9.5%) evaluated itch as primary endpoint, of which solely one revealed a significant relief of self-reported itch upon topical skin care. In those studies assessing itch as secondary endpoint (19/21, 90.5%), only 36.8% studies (n = 7/19) revealed a statistically significant itch reduction of up to 42%. Methodological limitations (heterogeneity of outcomes, inconsistent data assessment) in addition to limited superiority over control were implicated to account for low treatment efficacy observed in most studies. CONCLUSION Current data quality impairs comparative efficacy analyses of itch treatments in EB. Large scale randomized clinical trials and more personalized approaches applying validated measurement instruments for core outcomes are needed to substantiate evidence-based treatment approaches for EB-associated itch.
Collapse
Affiliation(s)
- Tobias Welponer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Christian Puttinger
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Eva W H Korte
- Department of Dermatology, UMCG Center of Expertise for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sjoukje van der Werf
- Central Medical Library, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Christine Prodinger
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Marieke C Bolling
- Department of Dermatology, UMCG Center of Expertise for Blistering Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin Laimer
- Department of Dermatology and Allergology and EB House Austria, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
4
|
Nevid M, Boguniewicz M. Current and Emerging Biologics for Atopic Dermatitis. Immunol Allergy Clin North Am 2024; 44:577-594. [PMID: 39389711 DOI: 10.1016/j.iac.2024.08.001] [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] [Indexed: 10/12/2024]
Abstract
Atopic dermatitis (AD) is a common chronic pruritic inflammatory skin disease that affects all ages and is recognized as a global health problem. Pathophysiology is complex with skin barrier abnormalities, immune dysregulation, and microbial dysbiosis all implicated. Markers of immune and inflammatory activation in the circulation provide a rationale for systemic therapy. Type 2 immune polarization is central, though other cytokine pathways including Th22 and Th17/IL-23 have been described, suggesting additional therapeutic targets in a subset of patients. Dupilumab and tralokinumab are monoclonal antibodies currently approved for moderate-to-severe AD with lebrikizumab and nemolizumab in late stages of development.
Collapse
Affiliation(s)
- Michael Nevid
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, 1400 Jackson Street, J312, Denver, CO 80206, USA
| | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, 1400 Jackson Street, J310, Denver, CO 80206, USA.
| |
Collapse
|
5
|
Brooks SG, Lopez LM, Mashoudy KD, Yosipovitch G, Czarnowicki T. Addressing Unmet Needs in Atopic Dermatitis: Evaluating Disease-Modifying Capabilities of Current and Emerging Therapies. Dermatitis 2024. [PMID: 39465269 DOI: 10.1089/derm.2024.0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Atopic dermatitis (AD) is a highly burdensome inflammatory skin condition affecting nearly one-quarter of the pediatric population and often continuing into adulthood. Despite recent advancements in systemic therapies providing temporary symptom relief over the past decade, AD frequently remains difficult to control, necessitating increased dosages or alternative treatments due to recurrent disease. This review synthesizes current literature to identify unmet needs of treating AD beyond medication-related limitations and evaluates existing therapies for their efficacy in modifying underlying disease mechanisms. Key findings include variability in AD pathophysiology and phenotypes across different age groups and ethnicities, indicating a need for research into endotype-specific treatments. The literature also comprises evidence suggesting that select current drugs, such as targeted biologics and Janus Kinase (JAK) inhibitors, may offer long-term disease-modifying benefits. Future management strategies should explore novel approaches, including manipulation of the microbiome, immune response, and neural function, as these may lead to additional improvements in AD treatment and long-term symptom relief.
Collapse
Affiliation(s)
- Sarah G Brooks
- From the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Coral Gables, Florida, USA
| | - Lourdes M Lopez
- From the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Coral Gables, Florida, USA
| | - Kayla D Mashoudy
- From the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Coral Gables, Florida, USA
| | - Gil Yosipovitch
- From the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Coral Gables, Florida, USA
| | - Tali Czarnowicki
- From the Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Coral Gables, Florida, USA
| |
Collapse
|
6
|
Zeldin J, Ratley G, Shobnam N, Myles IA. The clinical, mechanistic, and social impacts of air pollution on atopic dermatitis. J Allergy Clin Immunol 2024; 154:861-873. [PMID: 39151477 PMCID: PMC11456380 DOI: 10.1016/j.jaci.2024.07.027] [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: 03/15/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/19/2024]
Abstract
Atopic dermatitis (AD) is a complex disease characterized by dry, pruritic skin and significant atopic and psychological sequelae. Although AD has always been viewed as multifactorial, early research was dominated by overlapping genetic determinist views of either innate barrier defects leading to inflammation or innate inflammation eroding skin barrier function. Since 1970, however, the incidence of AD in the United States has increased at a pace that far exceeds genetic drift, thus suggesting a modern, environmental etiology. Another implicated factor is Staphylococcus aureus; however, a highly contagious microorganism is unlikely to be the primary etiology of a noncommunicable disease. Recently, the roles of the skin and gut microbiomes have received greater attention as potentially targetable drivers of AD. Here too, however, dysbiosis on a population scale would require induction by an environmental factor. In this review, we describe the evidence supporting the environmental hypothesis of AD etiology and detail the molecular mechanisms of each of the AD-relevant toxins. We also outline how a pollution-focused paradigm demands earnest engagement with environmental injustice if the field is to meaningfully address racial and geographic disparities. Identifying specific toxins and their mechanisms can also inform in-home and national mitigation strategies.
Collapse
Affiliation(s)
- Jordan Zeldin
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Md
| | - Grace Ratley
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Md
| | - Nadia Shobnam
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Md
| | - Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, Epithelial Therapeutics Unit, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
7
|
Pažur K, Francuzik W, El-Mahmoud H, Kraft M, Worm M. Proteomic, miRNA and bacterial biomarker patterns in atopic dermatitis patients and their course upon anti-IL-4Rα therapy. J Eur Acad Dermatol Venereol 2024; 38:1749-1759. [PMID: 38379385 DOI: 10.1111/jdv.19911] [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: 08/09/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Identification of biomarkers is required for a systems medicine approach and personalized treatment in atopic dermatitis (AD). These biomarkers may not only aid in diagnosing but also might be suitable to predict the effectiveness of targeted treatment. OBJECTIVE We aimed to identify proteomic, microbial and miRNA biomarkers in AD patients and investigated their course in relation to the clinical response upon anti-IL-4Rα therapy. METHODS Proteomic and miRNA screening was performed in AD patients in comparison to healthy controls. Differentially regulated serum proteins, miRNA and selected skin microbiota were measured consecutively in 50 AD patients before and upon systemic dupilumab treatment. A random forest classifier was used to predict the outcome of dupilumab therapy based on the initial biomarker patterns. RESULTS We identified 27 proteomic candidates, miRNA and three microbial strains to be dysregulated in AD. CCL17, CCL13, CCL22, E-selectin and BDNF were differently regulated and significantly associated with treatment response. In contrast, neither the microbial composition nor the miRNA pattern was associated with treatment response upon dupilumab treatment. CONCLUSION AD patients display defined dysregulations regarding their systemic proteomic serum profile, miRNA patterns and their skin microbiome. The proteomic profile and selected skin bacteria changed profoundly upon anti-IL-4Rα therapy which was associated with an overall clinical response. This was not seen in miRNA-related biomarkers. Our findings support the hypothesis that biomarker profiles reflect treatment responses and may in the future be used to develop a personalized medicine approach for the treatment of AD patients.
Collapse
Affiliation(s)
- K Pažur
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - W Francuzik
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - H El-Mahmoud
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - M Kraft
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - M Worm
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
8
|
Choi JY, Kim H, Min KH, Song WH, Yu DS, Lee M, Lee YB. Bacteria, Fungi, and Scalp Psoriasis: Understanding the Role of the Microbiome in Disease Severity. J Clin Med 2024; 13:4846. [PMID: 39200988 PMCID: PMC11355785 DOI: 10.3390/jcm13164846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Psoriasis is a chronic skin condition affected by genetic and environmental factors. Changes in the skin microbiome may affect the immune system and skin barrier functions, thereby contributing to the development and progression of psoriasis. The scalp, which is a common site for psoriasis, is often resistant to therapy. Although several studies have investigated the scalp microbiome, analyses focusing on both bacteria and fungi remain scarce. Methods: We examined the scalp microbiomes of 11 patients with psoriasis complicated with scalp lesions and categorized them according to their Psoriasis Area Severity Index (PASI) scores. The bacterial and fungal data were analyzed using QIIME2 pipeline version 2021.04 and the UNITE database version 8.3, respectively. Results: The Shannon indices for mild (2 patients), moderate (4 patients), and severe (5 patients) groups were 0.97, 1.38, and 1.88, respectively. A significant correlation was observed between increased mycobiome diversity and disease severity (p = 4.07 × 10-5, Spearman's correlation: 0.9269). Compared with the mild and moderate groups, the severe group exhibited a higher abundance of Malassezia globosa. Pseudomonas and Staphylococcus were, respectively, more prevalent in the moderate and severe groups than in the mild group. Conclusions: This study highlights the potential role of increased fungal diversity and specific microbial compositions in the severity of scalp psoriasis, suggesting a possible avenue for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Jin-Young Choi
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyunseong Kim
- Department of Life Science, Dongguk University—Seoul, Goyang 10326, Republic of Korea
- Department of Tropical Medicine, Institute of Tropical Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Kyung-Hyun Min
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Woo-Hyun Song
- Department of Life Science, Dongguk University—Seoul, Goyang 10326, Republic of Korea
| | - Dong-Soo Yu
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University—Seoul, Goyang 10326, Republic of Korea
| | - Young-Bok Lee
- Department of Dermatology, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Dermatology, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu 07345, Republic of Korea
| |
Collapse
|
9
|
De Pessemier B, López CD, Taelman S, Verdonck M, Chen Y, Stockman A, Lambert J, Van de Wiele T, Callewaert C. Comparative Whole Metagenome Analysis in Lesional and Nonlesional Scalp Areas of Patients with Psoriasis Capitis and Healthy Individuals. J Invest Dermatol 2024:S0022-202X(24)01984-5. [PMID: 39128495 DOI: 10.1016/j.jid.2024.07.020] [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: 04/12/2024] [Revised: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024]
Abstract
Psoriasis is an immune-mediated inflammatory disorder, where the majority of the patients suffer from psoriasis capitis or scalp psoriasis. Current therapeutics remain ineffective to treat scalp lesions. In this study, we present a whole-metagenome characterization of the scalp microbiome in psoriasis capitis. We investigated how changes in the homeostatic cutaneous microbiome correlate with the condition and identified metagenomic biomarkers (taxonomic, functional, virulence factors, antimicrobial resistance genes) that could partly explain its emergence. Within this study, 83 top and back scalp samples from healthy individuals and 64 lesional and nonlesional scalp samples from subjects with untreated psoriasis capitis were analyzed. Using qPCR targeting the 16S and 18S ribosomal RNA genes, we found a significant decrease in microbial load within scalp regions affected by psoriasis compared with that in their nonlesional counterparts. Metagenomic analysis revealed that psoriatic lesions displayed significant lower Cutibacterium species (including C. modestum, C. namnetense, C. granulosum, C. porci), along with an elevation in Staphylococcus aureus. A heightened relative presence of efflux pump protein-encoding genes was detected, suggesting potential antimicrobial resistance mechanisms. These mechanisms are known to specifically target human antimicrobial peptides (including cathelicidin LL-37), which are frequently encountered within psoriasis lesions. These shifts in microbial community dynamics may contribute to psoriasis disease pathogenesis.
Collapse
Affiliation(s)
- Britta De Pessemier
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Celia Díez López
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Steff Taelman
- Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium; BIOLIZARD, Ghent, Belgium
| | - Merel Verdonck
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Yang Chen
- Department of Dermatology, School of Medicine, University of California San Diego, California, USA; Department of Pediatrics, School of Medicine, University of California San Diego, California, USA; Biomedical Sciences Graduate Program, University of California San Diego, California, USA
| | | | - Jo Lambert
- Department of Head & Skin, Ghent University, Ghent, Belgium
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium
| | - Chris Callewaert
- Center for Microbial Ecology and Technology, Ghent University, Ghent, Belgium.
| |
Collapse
|
10
|
Dekkers C, de Bruin-Weller M. The pleiotropic role of interleukin-13 in the pathogenesis of atopic dermatitis. Br J Dermatol 2024; 191:316-317. [PMID: 38659397 DOI: 10.1093/bjd/ljae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 04/26/2024]
Affiliation(s)
- Coco Dekkers
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, the Netherlands
| | - Marjolein de Bruin-Weller
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, the Netherlands
| |
Collapse
|
11
|
Hülpüsch C, Rohayem R, Reiger M, Traidl-Hoffmann C. Exploring the skin microbiome in atopic dermatitis pathogenesis and disease modification. J Allergy Clin Immunol 2024; 154:31-41. [PMID: 38761999 DOI: 10.1016/j.jaci.2024.04.029] [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: 06/07/2023] [Revised: 03/04/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
Inflammatory skin diseases such as atopic eczema (atopic dermatitis [AD]) affect children and adults globally. In AD, the skin barrier is impaired on multiple levels. Underlying factors include genetic, chemical, immunologic, and microbial components. Increased skin pH in AD is part of the altered microbial microenvironment that promotes overgrowth of the skin microbiome with Staphylococcus aureus. The secretion of virulence factors, such as toxins and proteases, by S aureus further aggravates the skin barrier deficiency and additionally disrupts the balance of an already skewed immune response. Skin commensal bacteria, however, can inhibit the growth and pathogenicity of S aureus through quorum sensing. Therefore, restoring a healthy skin microbiome could contribute to remission induction in AD. This review discusses direct and indirect approaches to targeting the skin microbiome through modulation of the skin pH; UV treatment; and use of prebiotics, probiotics, and postbiotics. Furthermore, exploratory techniques such as skin microbiome transplantation, ozone therapy, and phage therapy are discussed. Finally, we summarize the latest findings on disease and microbiome modification through targeted immunomodulatory systemic treatments and biologics. We believe that targeting the skin microbiome should be considered a crucial component of successful AD treatment in the future.
Collapse
Affiliation(s)
- Claudia Hülpüsch
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland
| | - Robin Rohayem
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; Dermatology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Chair of Environmental Medicine, Technical University of Munich, Munich, Germany; Institute of Environmental Medicine, Helmholtz Center Munich-German Research Center for Environmental Health, Augsburg, Germany; Christine-Kühne Center for Allergy Research and Education, Davos, Switzerland; ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany.
| |
Collapse
|
12
|
Rothenberg-Lausell C, Bar J, Dahabreh D, Renert-Yuval Y, Del Duca E, Guttman-Yassky E. Biologic and small-molecule therapy for treating moderate to severe atopic dermatitis: Mechanistic considerations. J Allergy Clin Immunol 2024; 154:20-30. [PMID: 38670231 DOI: 10.1016/j.jaci.2024.04.009] [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: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Atopic dermatitis (AD) is a complex and heterogeneous skin disease for which achieving complete clinical clearance for most patients has proven challenging through single cytokine inhibition. Current studies integrate biomarkers and evaluate their role in AD, aiming to advance our understanding of the diverse molecular profiles implicated. Although traditionally characterized as a TH2-driven disease, extensive research has recently revealed the involvement of TH1, TH17, and TH22 immune pathways as well as the interplay of pivotal immune molecules, such as OX40, OX40 ligand (OX40L), thymic stromal lymphopoietin, and IL-33. This review explores the mechanistic effects of treatments for AD, focusing on mAbs and Janus kinase inhibitors. It describes how these treatments modulate immune pathways and examines their impact on key inflammatory and barrier biomarkers.
Collapse
Affiliation(s)
- Camille Rothenberg-Lausell
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Jonathan Bar
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dante Dahabreh
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yael Renert-Yuval
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; Pediatric Dermatology Unit, Schneider Children's Medical Center of Israel and the Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ester Del Duca
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Dermatology, University of La Sapienza, Rome, Italy
| | - Emma Guttman-Yassky
- Department of Dermatology and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
13
|
Salle R, Del Giudice P, Skayem C, Hua C, Chosidow O. Secondary Bacterial Infections in Patients with Atopic Dermatitis or Other Common Dermatoses. Am J Clin Dermatol 2024; 25:623-637. [PMID: 38578398 DOI: 10.1007/s40257-024-00856-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
Secondary bacterial infections of common dermatoses such as atopic dermatitis, ectoparasitosis, and varicella zoster virus infections are frequent, with Staphylococcus aureus and Streptococcus pyogenes being the bacteria most involved. There are also Gram-negative infections secondary to common dermatoses such as foot dyshidrotic eczema and tinea pedis. Factors favoring secondary bacterial infections in atopic dermatitis, ectoparasitosis, and varicella zoster virus infections mainly include an epidermal barrier alteration as well as itch. Mite-bacteria interaction is also involved in scabies and some environmental factors can promote Gram-negative bacterial infections of the feet. Furthermore, the bacterial ecology of these superinfections may depend on the geographical origin of the patients, especially in ectoparasitosis. Bacterial superinfections can also have different clinical aspects depending on the underlying dermatoses. Subsequently, the choice of class, course, and duration of antibiotic treatment depends on the severity of the infection and the suspected bacteria, primarily targeting S. aureus. Prevention of these secondary bacterial infections depends first and foremost on the management of the underlying skin disorder. At the same time, educating the patient on maintaining good skin hygiene and reporting changes in the primary lesions is crucial. In the case of recurrent secondary infections, decolonization of S. aureus is deemed necessary, particularly in atopic dermatitis.
Collapse
Affiliation(s)
- Romain Salle
- Service de Dermatologie Générale et Oncologique, UVSQ, EA4340-BECCOH, AP-HP, Hôpital Ambroise-Paré, Université Paris-Saclay, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France.
| | - Pascal Del Giudice
- Unité D'Infectiologie et Dermatologie, Centre Hospitalier Intercommunal de Fréjus-Saint-Raphaël, Fréjus, France
| | - Charbel Skayem
- Service de Dermatologie Générale et Oncologique, UVSQ, EA4340-BECCOH, AP-HP, Hôpital Ambroise-Paré, Université Paris-Saclay, 9 Avenue Charles de Gaulle, 92100, Boulogne-Billancourt, France
| | - Camille Hua
- AP-HP, Service de Dermatologie, Hôpital Henri Mondor, Créteil, France
| | - Olivier Chosidow
- Consultation Dermatoses Faciales, Service d'ORL, AP-HP, Hôpital Universitaire Pitié-Salpêtrière, Paris, France
- UPEC Créteil, Créteil, France
| |
Collapse
|
14
|
Jackson ND, Dyjack N, Goleva E, Bin L, Montgomery MT, Rios C, Everman JL, Taylor P, Bronchick C, Richers BN, Leung DY, Seibold MA. Atopic Dermatitis Complicated by Recurrent Eczema Herpeticum Is Characterized by Multiple, Concurrent Epidermal Inflammatory Endotypes. JID INNOVATIONS 2024; 4:100279. [PMID: 39006317 PMCID: PMC11239700 DOI: 10.1016/j.xjidi.2024.100279] [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: 11/15/2023] [Revised: 03/11/2024] [Accepted: 03/14/2024] [Indexed: 07/16/2024] Open
Abstract
A subgroup of patients with atopic dermatitis (AD) suffers from recurrent, disseminated herpes simplex virus skin infection, termed eczema herpeticum. To determine the transcriptional mechanisms of the skin and immune system pathobiology that underlie development of AD with eczema herpeticum (ADEH), we performed RNA-sequencing analysis of nonlesional skin (epidermis, dermis) from AD patients with and without a history of ADEH (ADEH+, n = 15; ADEH-, n = 13) along with healthy controls (n = 15). We also performed RNA sequencing on participants' plasmacytoid dendritic cells infected in vitro with herpes simplex virus 1. ADEH+ patients exhibited dysregulated gene expression, limited in the dermis (14 differentially expressed genes) and more widespread in the epidermis (129 differentially expressed genes). ADEH+-upregulated epidermal differentially expressed genes were enriched in type 2 cytokine (IL4R , CCL22, CRLF2, IL7R), interferon (CXCL10, ICAM1, IFI44, IRF7), and IL-36γ (IL36G) inflammatory gene pathways. All ADEH+ participants exhibited type 2 cytokine and inteferon endotypes, and 87% were IL36G-high. In contrast, these endotypes were more variably expressed among ADEH- participants. ADEH+ skin also had dysregulated epidermal differentiation complex gene expression of the late-cornified envelope, S100A, and small proline-rich gene families, which are involved in skin barrier function and antimicrobial activities. Plasmacytoid dendritic cell transcriptional responses to herpes simplex virus 1 infection were unaltered by ADEH status. The study concluded that the pathobiology underlying ADEH+ risk is associated with a unique, multifaceted epidermal inflammation that accompanies dysregulation of epidermal differentiation complex genes. These findings will help direct future studies that define how these inflammatory patterns may drive risk of eczema herpeticum in AD.
Collapse
Affiliation(s)
- Nathan D. Jackson
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Nathan Dyjack
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Elena Goleva
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Lianghua Bin
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Michael T. Montgomery
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Cydney Rios
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Jamie L. Everman
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Patricia Taylor
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | | | | | - Donald Y.M. Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado, USA
| | - Max A. Seibold
- Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
15
|
Berni Canani R, Caminati M, Carucci L, Eguiluz-Gracia I. Skin, gut, and lung barrier: Physiological interface and target of intervention for preventing and treating allergic diseases. Allergy 2024; 79:1485-1500. [PMID: 38439599 DOI: 10.1111/all.16092] [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: 09/05/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/06/2024]
Abstract
The epithelial barriers of the skin, gut, and respiratory tract are critical interfaces between the environment and the host, and they orchestrate both homeostatic and pathogenic immune responses. The mechanisms underlying epithelial barrier dysfunction in allergic and inflammatory conditions, such as atopic dermatitis, food allergy, eosinophilic oesophagitis, allergic rhinitis, chronic rhinosinusitis, and asthma, are complex and influenced by the exposome, microbiome, individual genetics, and epigenetics. Here, we review the role of the epithelial barriers of the skin, digestive tract, and airways in maintaining homeostasis, how they influence the occurrence and progression of allergic and inflammatory conditions, how current treatments target the epithelium to improve symptoms of these disorders, and what the unmet needs are in the identification and treatment of epithelial disorders.
Collapse
Affiliation(s)
- Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Marco Caminati
- Allergy Unit and Asthma Centre, Verona Integrated University Hospital and Department of Medicine, University of Verona, Verona, Italy
| | - Laura Carucci
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Ibon Eguiluz-Gracia
- Allergy Unit, Hospital Regional Universitario de Malága, Malaga, Spain
- Allergy Group, Biomedical Research Institute of Malaga (IBIMA)-BIONAND Platform, RICORS Inflammatory Diseases, Malaga, Spain
| |
Collapse
|
16
|
Tham EH, Chia M, Riggioni C, Nagarajan N, Common JE, Kong HH. The skin microbiome in pediatric atopic dermatitis and food allergy. Allergy 2024; 79:1470-1484. [PMID: 38308490 PMCID: PMC11142881 DOI: 10.1111/all.16044] [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: 10/04/2023] [Revised: 01/03/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
The skin microbiome is an extensive community of bacteria, fungi, mites, viruses and archaea colonizing the skin. Fluctuations in the composition of the skin microbiome have been observed in atopic dermatitis (AD) and food allergy (FA), particularly in early life, established disease, and associated with therapeutics. However, AD is a multifactorial disease characterized by skin barrier aberrations modulated by genetics, immunology, and environmental influences, thus the skin microbiome is not the sole feature of this disease. Future research should focus on mechanistic understanding of how early-life skin microbial shifts may influence AD and FA onset, to guide potential early intervention strategies or as microbial biomarkers to identify high-risk infants who may benefit from possible microbiome-based biotherapeutic strategies. Harnessing skin microbes as AD biotherapeutics is an emerging field, but more work is needed to investigate whether this approach can lead to sustained clinical responses.
Collapse
Affiliation(s)
- Elizabeth Huiwen Tham
- Khoo Teck Puat-National University Children’s Medical Institute, National University Health System (NUHS), Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Minghao Chia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Carmen Riggioni
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore
| | - Niranjan Nagarajan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore
| | - John E.A. Common
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Heidi H. Kong
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Hu YQ, Zhang JZ, Zhao Y. Efficacy and Safety of Dupilumab in Chinese Elderly Patients with Atopic Dermatitis: A Real-World Study. Dermatology 2024; 240:589-596. [PMID: 38797168 DOI: 10.1159/000539355] [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: 09/12/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
INTRODUCTION With the aging of the population in China, the prevalence of atopic dermatitis (AD) is high in elderly patients. These patients usually have more comorbidities and they need more effective and safer treatments. Dupilumab is an anti-interleukin-4 (IL-4) receptor monoclonal antibody which was approved for the treatment of moderate-to-severe AD. METHODS Elderly patients (60 years or older) with moderate-to-severe AD who treated with dupilumab were included. Eczema Area and Severity Index (EASI) score, Peak Pruritus Numerical Rating Scale (PP-NRS), EASI-50, EASI-75, and EASI-50 were evaluated. The efficacy in subgroups was also investigated. RESULTS Fifty-eight patients were enrolled. The EASI score and PP-NRS score were significantly reduced at weeks 4, 16, 28, and 52. 91.2% and 79.4% of the patients achieved EASI-50 and EASI-75 at week 16, respectively. 95.8% and 87.5% patients achieved EASI-50 and EASI-75 at week 52, respectively. Adverse events were reported in 10 (17.2%) patients, and no severe adverse event was reported. Male, older age, and moderate AD (EASI <21) were related to better efficacy. CONCLUSIONS This study demonstrated that dupilumab is effective and safe in elderly patients with AD.
Collapse
Affiliation(s)
- Yu-Qing Hu
- Department of Dermatology, Peking University People's Hospital, Beijing, China,
| | - Jian-Zhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Yan Zhao
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
18
|
Leyva-Castillo JM, Vega-Mendoza D, Strakosha M, Deng L, Choi S, Miyake K, Karasuyama H, Chiu IM, Phipatanakul W, Geha RS. Basophils are important for development of allergic skin inflammation. J Allergy Clin Immunol 2024; 153:1344-1354.e5. [PMID: 38336257 PMCID: PMC11070311 DOI: 10.1016/j.jaci.2024.01.022] [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: 05/03/2023] [Revised: 12/18/2023] [Accepted: 01/24/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Atopic dermatitis skin lesions exhibit increased infiltration by basophils. Basophils produce IL-4, which plays an important role in the pathogenesis of atopic dermatitis. OBJECTIVE We sought to determine the role of basophils in a mouse model of antigen-driven allergic skin inflammation. METHODS Wild-type mice, mice with selective and inducible depletion of basophils, and mice expressing Il4-driven enhanced green fluorescent protein were subjected to epicutaneous sensitization with ovalbumin or saline. Sensitized skin was examined by histology for epidermal thickening. Cells were analyzed for surface markers and intracellular expression of enhanced green fluorescent protein by flow cytometry. Gene expression was evaluated by real-time reverse transcription-quantitative PCR. RESULTS Basophils were important for epidermal hyperplasia, dermal infiltration by CD4+ T cells, mast cells, and eosinophils in ovalbumin-sensitized mouse skin and for the local and systemic TH2 response to epicutaneous sensitization. Moreover, basophils were the major source of IL-4 in epicutaneous-sensitized mouse skin and promote the ability of dendritic cells to drive TH2 polarization of naive T cells. CONCLUSION Basophils play an important role in the development of allergic skin inflammation induced by cutaneous exposure to antigen in mice.
Collapse
Affiliation(s)
- Juan-Manuel Leyva-Castillo
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Daniela Vega-Mendoza
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Maria Strakosha
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, Mass
| | - Samantha Choi
- Department of Immunology, Harvard Medical School, Boston, Mass
| | - Kensuke Miyake
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hajime Karasuyama
- Inflammation, Infection and Immunity Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, Mass
| | - Wanda Phipatanakul
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, and the Department of Pediatrics, Harvard Medical School, Boston, Mass
| |
Collapse
|
19
|
Gallo RL, Horswill AR. Staphylococcus aureus: The Bug Behind the Itch in Atopic Dermatitis. J Invest Dermatol 2024; 144:950-953. [PMID: 38430083 DOI: 10.1016/j.jid.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 03/03/2024]
Abstract
Pruritus or itch is a defining symptom of atopic dermatitis (AD). The origins of itch are complex, and it is considered both a defense mechanism and a cause of disease that leads to inflammation and psychological stress. Considerable progress has been made in understanding the processes that trigger itch, particularly the pruritoceptive origins that are generated in the skin. This perspective review discusses the implications of a recent observation that the V8 protease expressed by Staphylococcus aureus can directly trigger sensory neurons in the skin through activation of protease-activated receptor 1. This may be a key to understanding why itch is so common in AD because S. aureus commonly overgrows in this disease owing to deficient antimicrobial defense from both the epidermis and the cutaneous microbiome. Increased understanding of the role of microbes in AD provides increased opportunities for safely improving the treatment of this disorder.
Collapse
Affiliation(s)
- Richard L Gallo
- Department of Dermatology, University of California San Diego, La Jolla, California, USA.
| | - Alexander R Horswill
- Department of Immunology & Microbiology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
20
|
Schmuth M, Eckmann S, Moosbrugger-Martinz V, Ortner-Tobider D, Blunder S, Trafoier T, Gruber R, Elias PM. Skin Barrier in Atopic Dermatitis. J Invest Dermatol 2024; 144:989-1000.e1. [PMID: 38643989 DOI: 10.1016/j.jid.2024.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 04/23/2024]
Abstract
A compromised permeability barrier is a hallmark of atopic dermatitis (AD). Localized to the outermost skin layer, the stratum corneum (SC) is critically dependent on terminal differentiation of epidermal keratinocytes, which transform into protein-rich corneocytes surrounded by extracellular lamellae of unique epidermal lipids, conferring permeability barrier function. These structures are disrupted in AD. A leaky barrier is prone to environmental insult, which in AD elicits type 2-dominant inflammation, in turn resulting in a vicious cycle further impairing the SC structure. Therapies directed at enforcing SC structure and anti-inflammatory strategies administered by topical and systemic route as well as UV therapy have differential effects on the permeability barrier. The expanding armamentarium of therapeutic modalities for AD treatment warrants optimization of their effects on permeability barrier function.
Collapse
Affiliation(s)
- Matthias Schmuth
- Dermatology, Venerology and Allergy, Medical University Innsbruck, Innsbruck, Austria; Institute for Pediatric Dermatology and Rare Diseases, Karl Landsteiner Society, Innsbruck, Austria.
| | - Sonja Eckmann
- Dermatology, Venerology and Allergy, Medical University Innsbruck, Innsbruck, Austria
| | | | | | - Stefan Blunder
- Dermatology, Venerology and Allergy, Medical University Innsbruck, Innsbruck, Austria
| | - Thomas Trafoier
- Dermatology, Venerology and Allergy, Medical University Innsbruck, Innsbruck, Austria
| | - Robert Gruber
- Dermatology, Venerology and Allergy, Medical University Innsbruck, Innsbruck, Austria; Institute for Pediatric Dermatology and Rare Diseases, Karl Landsteiner Society, Innsbruck, Austria
| | - Peter M Elias
- Dermatology, Veteran Affairs Health Care System, San Francisco, California, USA; University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Yang L, Li D, Sun S, Liu D, Wang Y, Liu X, Zhou B, Nie W, Li L, Wang Y, Sha S, Li Y, Shen C, Tao J. Dupilumab therapy improves gut microbiome dysbiosis and tryptophan metabolism in Chinese patients with atopic dermatitis. Int Immunopharmacol 2024; 131:111867. [PMID: 38493690 DOI: 10.1016/j.intimp.2024.111867] [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: 09/23/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Dupilumab has demonstrate its potential to orchestrate inflammatory skin microenvironment, enhance skin barrier and shift skin microbiome dysbiosis, collectively contributing to clinical improvement in patients with atopic dermatitis (AD). As the second genome of human body, growing evidence suggests that the gut microbiome might relate to the host response to treatments. Little is known about the association between dupilumab treatment and gut microbiome in AD patients. OBJECTIVE We aimed to characterize the gut microbiome among Chinese subjects with or without AD and determine the potential effect of dupilumab on the gut microbiome. RESULTS The 16 s rRNA gene sequencing was conducted on 48 healthy controls (HC), 44 AD patients and 27 AD patients who received dupilumab for 16 weeks. Prior to treatment, we identified the changed beta-diversity, increased Firmicutes/Bacteroidetes ratio, decreased Bifidobacterium and expanded Faecalibacterium among the AD patients compared to HC. After 16 weeks of dupilumab treatment, gut microbiome dysbiosis of the AD patients improved with reversed beta-diversity, closer bacterial connections, increased colonization of Bifidobacterium, Ruminococcus gnavus, and Coprococcus, which were negatively correlated with disease severity indicators. This shift was largely independent of the degree of clinical improvement. Bacterial function analysis revealed further metabolic alterations following dupilumab treatment, including up-regulated expression of genes involved in the indole pathway of tryptophan metabolism, corroborated by quantitative UHPLC-MS/MS analysis. CONCLUSION Dupilumab treatment tends to help shift the gut microbial dysbiosis in AD patients to a healthier state, along with improved intestinal tryptophan metabolism, suggesting the gut flora and its metabolites may mediate part of the synergistic therapeutic effects on the host.
Collapse
Affiliation(s)
- Liu Yang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Danqi Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Shuomin Sun
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Danping Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaohuan Liu
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha 410007, Hunan, China
| | - Bin Zhou
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Wenjia Nie
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Lu Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Yifei Wang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Shanshan Sha
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Yan Li
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Chen Shen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China
| | - Juan Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Engineering Research Center for Skin Repair and Theranostics, Wuhan 430022, China.
| |
Collapse
|
22
|
Clowry J, Dempsey DJ, Claxton TJ, Towell AM, Turley MB, Sutton M, Geoghegan JA, Kezic S, Jakasa I, White A, Irvine AD, McLoughlin RM. Distinct T cell signatures are associated with Staphylococcus aureus skin infection in pediatric atopic dermatitis. JCI Insight 2024; 9:e178789. [PMID: 38716729 PMCID: PMC11141913 DOI: 10.1172/jci.insight.178789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/03/2024] [Indexed: 06/02/2024] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.
Collapse
Affiliation(s)
- Julianne Clowry
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Dempsey
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Tracey J. Claxton
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling M. Towell
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Mary B. Turley
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Sutton
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sanja Kezic
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Ivone Jakasa
- Laboratory for Analytical Chemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Arthur White
- School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - Alan D. Irvine
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Greenzaid JD, Chan LJ, Chandani BM, Kiritsis NR, Feldman SR. Microbiome modulators for atopic eczema: a systematic review of experimental and investigational therapeutics. Expert Opin Investig Drugs 2024; 33:415-430. [PMID: 38441984 DOI: 10.1080/13543784.2024.2326625] [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: 09/28/2023] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a common inflammatory cutaneous disease that arises due to dysregulation of the Th2 immune response, impaired skin barrier integrity, and dysbiosis of the skin and gut microbiota. An abundance of Staphylococcus aureus biofilms in AD lesions increases the Th2 immune response, and gut bacteria release breakdown products such as Short Chain Fatty Acids that regulate the systemic immune response. AREAS COVERED We aim to evaluate therapies that modulate the microbiome in humans and discuss the clinical implications of these treatments. We performed a review of the literature in which 2,673 records were screened, and describe the findings of 108 studies that were included after full-text review. All included studies discussed the effects of therapies on the human microbiome and AD severity. Oral probiotics, topical probiotics, biologics, and investigational therapies were included in our analysis. EXPERT OPINION Oral probiotics demonstrate mixed efficacy at relieving AD symptoms. Topical probiotics reduce S. aureus abundance in AD lesional skin, yet for moderate-severe disease, these therapies may not reduce AD severity scores to the standard of biologics. Dupilumab and tralokinumab target key inflammatory pathways in AD and modulate the skin microbiome, further improving disease severity.
Collapse
Affiliation(s)
- Jonathan D Greenzaid
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Lina J Chan
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Brittany M Chandani
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Nicholas R Kiritsis
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Social Sciences & Health Policy, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
24
|
Wang M, Gao XH, Zhang L. A Review of Dupilumab in the Treatment of Atopic Dermatitis in Infants and Children. Drug Des Devel Ther 2024; 18:941-951. [PMID: 38560522 PMCID: PMC10981892 DOI: 10.2147/dddt.s457761] [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: 01/04/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024] Open
Abstract
Atopic dermatitis (AD), a common pruritic and chronic inflammatory skin disease, has a major impact on a patient's quality of life. It is characterized by dry, itchy, and eczema-like rashes. AD is more prevalent in young children and has been linked to a variety of other allergy disorders. Traditional drug therapy has certain limitations for treating young children with AD. However, biologics have good clinical application prospects in the medical treatment of young patients. Dupilumab, a fully human monoclonal antibody, specifically binds to the IL-4 Rα subunit, inhibiting IL-4 and IL-13 signaling and blocking the occurrence of type 2 inflammatory response. It has a good effect on treating infants and children with moderate-to-severe AD. This review explores the safety and efficacy of dupilumab in the treatment of AD in infants and children and the impact of early intervention on AD progression, with the aim of informing clinical practice in the use of dupilumab for the treatment of young patients with AD.
Collapse
Affiliation(s)
- Mingyue Wang
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
- NHC Key Laboratory of Immunodermatology, China Medical University, Shenyang, People’s Republic of China
- Key Laboratory of Immunodermatology, China Medical University, Ministry of Education, Shenyang, People’s Republic of China
- National and Local Joint Engineering Research Center of Immunodermatological Theranostics, Shenyang, People’s Republic of China
| | - Xing-Hua Gao
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
- NHC Key Laboratory of Immunodermatology, China Medical University, Shenyang, People’s Republic of China
- Key Laboratory of Immunodermatology, China Medical University, Ministry of Education, Shenyang, People’s Republic of China
- National and Local Joint Engineering Research Center of Immunodermatological Theranostics, Shenyang, People’s Republic of China
| | - Li Zhang
- Department of Dermatology, the First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| |
Collapse
|
25
|
Paller AS, Siegfried EC, Cork MJ, Arkwright PD, Eichenfield LF, Ramien M, Khokhar FA, Chen Z, Zhang A, Cyr SL. Infections in Children Aged 6 Months to 5 Years Treated with Dupilumab in a Placebo-Controlled Clinical Trial of Moderate-to-Severe Atopic Dermatitis. Paediatr Drugs 2024; 26:163-173. [PMID: 38267692 PMCID: PMC10890978 DOI: 10.1007/s40272-023-00611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Patients with atopic dermatitis (AD), particularly infants and young children, are at greater risk of developing skin infections. In this study, we assessed infection rates in AD patients aged 6 months to 5 years treated with dupilumab. METHODS In LIBERTY AD PRESCHOOL, a double-blind, placebo-controlled, phase III clinical trial, children aged 6 months to 5 years with moderate-to-severe AD were randomized 1:1 to subcutaneous dupilumab or placebo, with concomitant low-potency topical corticosteroids, every 4 weeks for 16 weeks. Exposure-adjusted infection rates were used to compare treatment groups. RESULTS The analysis included 162 patients, of whom 83 received dupilumab and 79 received placebo. Total infection rates were not significantly different between the dupilumab and placebo groups (rate ratio [RR] 0.75, 95% CI 0.48-1.19; p = 0.223). Non-herpetic adjudicated skin infections and bacterial infections were significantly less frequent with dupilumab versus placebo (non-herpetic skin infections: RR 0.46, 95% CI 0.21-0.99; p = 0.047; bacterial infections: RR 0.09, 95% CI 0.01-0.67; p = 0.019), and the number of patients using systemic anti-infective medication was significantly lower in the dupilumab group (RR 0.52, 95% CI 0.30-0.89; p = 0.019). There were no significant differences in the number of herpetic infections between the dupilumab and placebo groups (RR 1.17, 95% CI 0.31-4.35; p = 0.817). The number of patients with two or more infection events was significantly higher in the placebo group (RR 0.29, 95% CI 0.12-0.68; p = 0.004), and no severe or serious infections (including eczema herpeticum) were observed among patients receiving dupilumab. CONCLUSIONS These data suggest that dupilumab treatment in infants and children younger than 6 years with AD does not increase overall risk of infections and is associated with a reduced risk of bacterial and non-herpetic skin infections compared with placebo, resulting in a reduced need for anti-infective medication. TRIAL REGISTRATION The trial was registered with ClinicalTrials.gov with ID number NCT03346434 on November 17, 2017. INFOGRAPHIC.
Collapse
Affiliation(s)
- Amy S Paller
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Ann and Robert H. Lurie Children's Hospital, Chicago, IL, USA
| | - Elaine C Siegfried
- Saint Louis University School of Medicine, St. Louis, MO, USA
- Cardinal Glennon Children's Hospital, St. Louis, MO, USA
| | - Michael J Cork
- Sheffield Dermatology Research, University of Sheffield, Sheffield, UK
- Sheffield Children's Hospital, Sheffield, UK
| | - Peter D Arkwright
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Michele Ramien
- Division of Dermatology, Department of Medicine, University of Calgary, Calgary, AB, Canada
- Section of Community Pediatrics, Department of Pediatrics, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| | | | - Zhen Chen
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | | | - Sonya L Cyr
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA.
- Regeneron Pharmaceuticals Inc., 1 Rockwood Road, Sleepy Hollow, NY, 10591, USA.
| |
Collapse
|
26
|
Zhang XE, Zheng P, Ye SZ, Ma X, Liu E, Pang YB, He QY, Zhang YX, Li WQ, Zeng JH, Guo J. Microbiome: Role in Inflammatory Skin Diseases. J Inflamm Res 2024; 17:1057-1082. [PMID: 38375021 PMCID: PMC10876011 DOI: 10.2147/jir.s441100] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
As the body's largest organ, the skin harbors a highly diverse microbiota, playing a crucial role in resisting foreign pathogens, nurturing the immune system, and metabolizing natural products. The dysregulation of human skin microbiota is implicated in immune dysregulation and inflammatory responses. This review delineates the microbial alterations and immune dysregulation features in common Inflammatory Skin Diseases (ISDs) such as psoriasis, rosacea, atopic dermatitis(AD), seborrheic dermatitis(SD), diaper dermatitis(DD), and Malassezia folliculitis(MF).The skin microbiota, a complex and evolving community, undergoes changes in composition and function that can compromise the skin microbial barrier. These alterations induce water loss and abnormal lipid metabolism, contributing to the onset of ISDs. Additionally, microorganisms release toxins, like Staphylococcus aureus secreted α toxins and proteases, which may dissolve the stratum corneum, impairing skin barrier function and allowing entry into the bloodstream. Microbes entering the bloodstream activate molecular signals, leading to immune disorders and subsequent skin inflammatory responses. For instance, Malassezia stimulates dendritic cells(DCs) to release IL-12 and IL-23, differentiating into a Th17 cell population and producing proinflammatory mediators such as IL-17, IL-22, TNF-α, and IFN-α.This review offers new insights into the role of the human skin microbiota in ISDs, paving the way for future skin microbiome-specific targeted therapies.
Collapse
Affiliation(s)
- Xue-Er Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Pai Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Sheng-Zhen Ye
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 6610072, People’s Republic of China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People’s Republic of China
| | - E Liu
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Yao-Bin Pang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Qing-Ying He
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Yu-Xiao Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Wen-Quan Li
- Chengdu University of Traditional Chinese Medicine, Chengdu, 6610075, People’s Republic of China
| | - Jin-Hao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, People’s Republic of China
| | - Jing Guo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 6610072, People’s Republic of China
| |
Collapse
|
27
|
Nawrot D, Ambrożkiewicz-Mosler W, Doležal M, Bouz G. Antistaphylococcal discovery pipeline; where are we now? Eur J Med Chem 2024; 266:116077. [PMID: 38219657 DOI: 10.1016/j.ejmech.2023.116077] [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: 10/03/2023] [Revised: 12/01/2023] [Accepted: 12/18/2023] [Indexed: 01/16/2024]
Abstract
The serious spread of antibiotic-resistant Staphylococcal aureus strains is alarming. This is reflected by the measures governments and health-related bodies are offering to ease antibiotic drug development. Finding new active agents, preferably with novel mechanism of action, or even finding new targets for drug development are essential. In this review, we summarize the current status of novel antistaphylococcal agents undergoing clinical trials. We mainly discuss antistaphylococcal small molecules and peptides in the text with a special focus on their chemistry, while antistaphylococcal immunotherapy (antibodies) are mentioned in a summative table. This review shall serve as a summary that influences future synthetic efforts in the antistaphyloccocals development field.
Collapse
Affiliation(s)
- Daria Nawrot
- Faculty of Pharmacy in Hradec Králové, Charles University, 50005, Hradec Králové, Czech Republic.
| | | | - Martin Doležal
- Faculty of Pharmacy in Hradec Králové, Charles University, 50005, Hradec Králové, Czech Republic
| | - Ghada Bouz
- Faculty of Pharmacy in Hradec Králové, Charles University, 50005, Hradec Králové, Czech Republic.
| |
Collapse
|
28
|
Cork MJ, Danby SG, Rossi AB, Bansal A. Dupilumab Treatment in Pediatric Patients Aged 6-11 Years with Severe Atopic Dermatitis Whose Disease Is Not Adequately Controlled: A Review. Drug Des Devel Ther 2024; 18:277-289. [PMID: 38333897 PMCID: PMC10849907 DOI: 10.2147/dddt.s426947] [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: 07/26/2023] [Accepted: 01/03/2024] [Indexed: 02/10/2024] Open
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease in children. Children with severe AD have a multidimensional disease burden characterized by skin lesions, itching, frequent infections, sleep deprivation, and a high rate of comorbidities. These impact the mental health and overall quality of life of not only the children but also of their parents and caregivers. There are few effective available treatment options for young children with severe AD that are suitable for long-term use. Due to their adverse effects, practice guidelines consider systemic agents inappropriate for this age group, although they are still used off-label in extreme cases. The biologic dupilumab has recently been approved for children aged 6-11 years with severe (EU) and moderate-to-severe (USA) AD, offering hope to this population of patients with a high unmet clinical need. The purpose of this review is to describe the unmet needs of AD patients aged 6-11 years prior to dupilumab approval and to summarize existing clinical data supporting dupilumab's safety and efficacy in these children.
Collapse
Affiliation(s)
- Michael J Cork
- Sheffield Dermatology Research, University of Sheffield, Sheffield, UK
- Sheffield Children’s Hospital, Sheffield, UK
| | - Simon G Danby
- Sheffield Dermatology Research, University of Sheffield, Sheffield, UK
- Sheffield Children’s Hospital, Sheffield, UK
| | | | | |
Collapse
|
29
|
Umemoto N, Kakurai M, Matsumoto T, Mizuno K, Cho O, Sugita T, Demitsu T. Dupilumab Alters Both the Bacterial and Fungal Skin Microbiomes of Patients with Atopic Dermatitis. Microorganisms 2024; 12:224. [PMID: 38276210 PMCID: PMC10820602 DOI: 10.3390/microorganisms12010224] [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: 11/25/2023] [Revised: 01/08/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The skin microbiome at lesion sites in patients with atopic dermatitis (AD) is characterized by dysbiosis. Although the administration of dupilumab, an IL-4Rα inhibitor, improves dysbiosis in the bacterial microbiome, information regarding the fungal microbiome remains limited. This study administered dupilumab to 30 patients with moderate-to-severe AD and analyzed changes in both fungal and bacterial skin microbiomes over a 12-week period. Malassezia restricta and M. globosa dominated the fungal microbiome, whereas non-Malassezia yeast species increased in abundance, leading to greater microbial diversity. A qPCR analysis revealed a decrease in Malassezia colonization following administration, with a higher reduction rate observed where the pretreatment degree of colonization was higher. A correlation was found between the group classified by the Eczema Area and Severity Index, the group categorized by the concentration of Thymus and activation-regulated chemokine, and the degree of skin colonization by Malassezia. Furthermore, an analysis of the bacterial microbiome also confirmed a decrease in the degree of skin colonization by the exacerbating factor Staphylococcus aureus and an increase in the microbial diversity of the bacterial microbiome. Our study is the first to show that dupilumab changes the community structure of the bacterial microbiome and affects the fungal microbiome in patients with AD.
Collapse
Affiliation(s)
- Naoka Umemoto
- Department of Dermatology, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan; (N.U.); (M.K.); (T.M.); (K.M.); (T.D.)
| | - Maki Kakurai
- Department of Dermatology, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan; (N.U.); (M.K.); (T.M.); (K.M.); (T.D.)
| | - Takanao Matsumoto
- Department of Dermatology, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan; (N.U.); (M.K.); (T.M.); (K.M.); (T.D.)
| | - Kenta Mizuno
- Department of Dermatology, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan; (N.U.); (M.K.); (T.M.); (K.M.); (T.D.)
| | - Otomi Cho
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose 204-8588, Japan;
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose 204-8588, Japan;
| | - Toshio Demitsu
- Department of Dermatology, Jichi Medical University Saitama Medical Center, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan; (N.U.); (M.K.); (T.M.); (K.M.); (T.D.)
| |
Collapse
|
30
|
Patra V, Woltsche N, Cerpes U, Bokanovic D, Repelnig M, Joshi A, Perchthaler I, Fischl M, Vocanson M, Bordag N, Durdevic M, Woltsche J, Quehenberger F, Legat F, Wedrich A, Horwath-Winter J, Wolf P. Persistent Neutrophil Infiltration and Unique Ocular Surface Microbiome Typify Dupilumab-Associated Conjunctivitis in Patients with Atopic Dermatitis. OPHTHALMOLOGY SCIENCE 2024; 4:100340. [PMID: 37869024 PMCID: PMC10585475 DOI: 10.1016/j.xops.2023.100340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 10/24/2023]
Abstract
Objective To elucidate the pathogenesis of dupilumab (Dupixent®)-associated ocular surface disease (DAOSD). Design Prospective single-center cohort study. Subjects Twenty patients with moderate-to-severe atopic dermatitis (AD) who received dupilumab and 10 age- and sex-matched healthy controls were enrolled in the study. Methods The study cohort underwent a thorough slit-lamp and entire-body dermatologic examination. Conjunctival swabs and smears were collected at baseline, 4 and 16 weeks after treatment initiation, and during the conjunctivitis episode. To analyse the ocular surface microbiome, 16S ribosomal RNA sequencing was performed, smears were hematoxylin and eosin stained, and serum cytokines were measured by using a multiplex immunobead assay. Main Outcome Measures Composition of ocular surface microbiome and cellular component as well as serum cytokine levels. Results Six of the 20 patients with AD developed DAOSD after dupilumab initiation; these patients responded after a delay to treatment as quantified by Eczema Area and Severity Index and Investigator's Global Assessment score. Conjunctival smears showed massive neutrophilic infiltration and serum analysis revealed increased systemic levels of neutrophil-priming proinflammatory cytokines, in particular interleukin-1β and tumor necrosis factor α, in patients with DAOSD compared with those without it. The ocular surface microbiome of patients with DAOSD was characterized by a diverse and persistent microbial colonization, particularly by Acetobacter aceti. In contrast, microbial diversity decreased in patients with AD without DAOSD after the initiation of dupilumab treatment, especially the abundance of Staphylococcus aureus. In vitro experiments substantiated the potential role of the microbiome, showing increased growth of A. aceti and decreased growth of S. aureus in presence of dupilumab. Conclusions Persistent neutrophilic infiltration and a unique microbial landscape on the ocular surface associated with elevated levels of systemic proinflammatory cytokines typify DAOSD. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- VijayKumar Patra
- Department of Dermatology, Medical University of Graz, Graz, Austria
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Nora Woltsche
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | - Urban Cerpes
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | | | - Maria Repelnig
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Aaroh Joshi
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | | | - Manuela Fischl
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Natalie Bordag
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Marija Durdevic
- Computational Bioanalytics, Center for Medical Research, Medical University of Graz, Graz, Austria
- Institute of Pathology, Medical University of Graz, Graz, Austria
- Theodor Escherich Laboratory for Medical Microbiome Research, Medical University of Graz, Graz, Austria
| | - Johannes Woltsche
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | | | - Franz Legat
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Andreas Wedrich
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | | | - Peter Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| |
Collapse
|
31
|
Stolarczyk A, Perez-Nazario N, Knowlden SA, Chinchilli E, Grier A, Paller A, Gill SR, De Benedetto A, Yoshida T, Beck LA. Bleach baths enhance skin barrier, reduce itch but do not normalize skin dysbiosis in atopic dermatitis. Arch Dermatol Res 2023; 315:2883-2892. [PMID: 37755506 PMCID: PMC10615920 DOI: 10.1007/s00403-023-02723-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/02/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Studies have demonstrated that bleach baths improve atopic dermatitis (AD) severity; however, the effects on itch, skin barrier, and cutaneous microbial composition are less clear. We examined whether bleach baths reduce itch, normalize skin barrier function, reduce S. aureus absolute abundance, and increase microbial diversity in adults with AD who were colonized with S. aureus on their non-lesional skin. This was an open label, non-randomized, controlled trial performed at a single academic center. Fifteen AD and five non-atopic healthy controls (NA) were instructed to take two bleach baths (0.005% NaClO; 5-10 min duration) per week for a total of 12 weeks as add-on therapy. Adults 18 to 65 years (inclusive) with mild to severe AD were recruited with EASI score > 6.0, S. aureus culture positivity, access to a bathtub, and ability and willingness to maintain current topical or systemic treatments. They were evaluated at baseline (before bleach baths), 6 weeks, and 12 weeks after the intervention of twice-weekly bleach baths. Efficacy measurements included EASI as well as 5-D Pruritus and ItchyQoL™. Transepidermal water loss (TEWL) and stratum corneum (SC) integrity assay were performed to assess the skin barrier. Skin dysbiosis was measured by S. aureus cultivation, S. aureus abundance (qPCR of thermonuclease gene), and V1-V3 16S rRNA gene sequencing on non-lesional and lesional AD skin. After 12 weeks of bleach baths, 8/15 (53.3%) AD subjects achieved an EASI50 and a significant reduction in itch as measured by 5-D pruritus and Itchy QoL. Eighty-seven percent reported improvements in sleep quality. At study entry, AD subjects had higher non-lesional TEWL values than NA subjects, and only AD subjects experienced a reduction with bleach baths (p = 0.006). Similarly, SC integrity improved as early as 6 weeks after bleach baths in AD subjects. Notably, bleach baths had no significant effect on S. aureus culture-positivity, qPCR absolute abundance, or microbial diversity. The addition of twice-weekly bleach baths improves investigator-assessed AD severity, patient-reported pruritus and sleep as well as physiological measures of skin barrier function in adult AD subjects while having no effect on qualitative and quantitative measures of cutaneous S. aureus. Trial Registration: ClinicalTrials.gov Identifier: NCT01996150, Date of registration: November 27th, 2013.
Collapse
Affiliation(s)
- Ania Stolarczyk
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Nelissa Perez-Nazario
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Sara A Knowlden
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Ellen Chinchilli
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Alex Grier
- University of Rochester Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Amy Paller
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Steven R Gill
- University of Rochester Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Takeshi Yoshida
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, NY, 14642, USA.
| |
Collapse
|
32
|
Dewi DAR, Perdiyana A, Wiliantari NM, Nadhira F, Arkania N, Salsabila CA, Allun CV, Allatib A, Dewantara K. Managing the Skin Microbiome as a New Bacteriotherapy for Inflammatory Atopic Dermatitis. Cureus 2023; 15:e48803. [PMID: 38024036 PMCID: PMC10645580 DOI: 10.7759/cureus.48803] [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: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
The microbiome, comprising various bacteria, assumes a significant role in the immune system's maturation and maintaining bodily homeostasis. Alterations in the microbial composition can contribute to the initiation and progression of inflammation. Recent studies reveal that changes in microbial composition and function, known as dysbiosis in the skin and gut, have been associated with altered immunological responses and skin barrier disruption. These changes are implicated in the development of several skin diseases, such as atopic dermatitis (AD). This review examines research demonstrating the potential of microbiome repair as a therapeutic approach to reduce the effect of inflammatory processes in the skin during atopic dermatitis. This way, corticosteroids in atopic dermatitis therapy can be reduced or even replaced with treatments focusing on controlling the skin microbiome. This study used scientific literature from recognized platforms, including PubMed, Scopus, Google Scholar, and ScienceDirect, covering publications from 2013 to 2023. The primary aim of this study was to assess the efficacy of skin microbiome management in treating atopic dermatitis. This study concludes that physicians must comprehensively understand the microbiome's involvement in atopic dermatitis, including its pathophysiological implications and its relevance to therapeutic interventions.
Collapse
Affiliation(s)
- Dian Andriani Ratna Dewi
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
- Department of Dermatovenereology, Gatot Soebroto Central Army Hospital, Central Jakarta, IDN
| | - Angki Perdiyana
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
| | - Ni M Wiliantari
- Department of Dermatovenereology, Ratna Dewi Principal Clinic, Bekasi, IDN
| | - Farrasila Nadhira
- Department of Dermatovenereology, Ratna Dewi Principal Clinic, Bekasi, IDN
| | - Nabila Arkania
- Department of Dermatovenereology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University, Special Region of Yogyakarta, IDN
| | - Cut A Salsabila
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
| | - Clara V Allun
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
| | - Arohid Allatib
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
| | - Kelvin Dewantara
- Department of Dermatovenereology, Faculty of Military Medicine, The Republic of Indonesia Defense University, Bogor, IDN
| |
Collapse
|
33
|
Simpson EL, Schlievert PM, Yoshida T, Lussier S, Boguniewicz M, Hata T, Fuxench Z, De Benedetto A, Ong PY, Ko J, Calatroni A, Rudman Spergel AK, Plaut M, Quataert SA, Kilgore SH, Peterson L, Gill AL, David G, Mosmann T, Gill SR, Leung DYM, Beck LA. Rapid reduction in Staphylococcus aureus in atopic dermatitis subjects following dupilumab treatment. J Allergy Clin Immunol 2023; 152:1179-1195. [PMID: 37315812 PMCID: PMC10716365 DOI: 10.1016/j.jaci.2023.05.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is an inflammatory disorder characterized by dominant type 2 inflammation leading to chronic pruritic skin lesions, allergic comorbidities, and Staphylococcus aureus skin colonization and infections. S aureus is thought to play a role in AD severity. OBJECTIVES This study characterized the changes in the host-microbial interface in subjects with AD following type 2 blockade with dupilumab. METHODS Participants (n = 71) with moderate-severe AD were enrolled in a randomized (dupilumab vs placebo; 2:1), double-blind study at Atopic Dermatitis Research Network centers. Bioassays were performed at multiple time points: S aureus and virulence factor quantification, 16s ribosomal RNA microbiome, serum biomarkers, skin transcriptomic analyses, and peripheral blood T-cell phenotyping. RESULTS At baseline, 100% of participants were S aureus colonized on the skin surface. Dupilumab treatment resulted in significant reductions in S aureus after only 3 days (compared to placebo), which was 11 days before clinical improvement. Participants with the greatest S aureus reductions had the best clinical outcomes, and these reductions correlated with reductions in serum CCL17 and disease severity. Reductions (10-fold) in S aureus cytotoxins (day 7), perturbations in TH17-cell subsets (day 14), and increased expression of genes relevant for IL-17, neutrophil, and complement pathways (day 7) were also observed. CONCLUSIONS Blockade of IL-4 and IL-13 signaling, very rapidly (day 3) reduces S aureus abundance in subjects with AD, and this reduction correlates with reductions in the type 2 biomarker, CCL17, and measures of AD severity (excluding itch). Immunoprofiling and/or transcriptomics suggest a role for TH17 cells, neutrophils, and complement activation as potential mechanisms to explain these findings.
Collapse
Affiliation(s)
- Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | | | - Takeshi Yoshida
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | - 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
| | - Zelma Fuxench
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pa
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Peck Y Ong
- Department of Pediatrics, University Southern California, Los Angeles, Calif
| | - Justin Ko
- Department of Dermatology, Stanford University, Stanford, Calif
| | | | - Amanda K Rudman Spergel
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Marshall Plaut
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Sally A Quataert
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Samuel H Kilgore
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa
| | - Liam Peterson
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Ann L Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | | | - Tim Mosmann
- Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - Steven R Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY
| | - 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 Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
34
|
Leyva-Castillo JM, McGurk A, Strakosha M, Vega-Mendoza D, Smith SEM, Stafstrom K, Elkins M, Chou J, Wang YH, Geha RS. IL-4 receptor alpha blockade dampens allergic inflammation and upregulates IL-17A expression to promote Saureus clearance in antigen sensitized mouse skin. J Allergy Clin Immunol 2023; 152:907-915. [PMID: 37315811 PMCID: PMC10592541 DOI: 10.1016/j.jaci.2023.05.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Skin colonization with Staphylococcus aureus aggravates atopic dermatitis and exaggerates allergic skin inflammation in mice. IL-4 receptor α (IL-4Rα) blockade is beneficial in atopic dermatitis and reduces Saureus skin colonization through unknown mechanisms. The cytokine IL-17A restrains Saureus growth. OBJECTIVES This study sought to examine the effect of IL-4Rα blockade on Saureus colonization at sites of allergic skin inflammation in mice and determine the mechanism involved. METHODS BALB/c mice were epicutaneously sensitized with ovalbumin (OVA). Immediately after, PSVue 794-labeled S aureus strain SF8300 or saline was applied and a single dose of anti-IL-4Rα blocking antibody, a mixture of anti-IL-4Rα and anti-IL-17A blocking antibodies, or IgG isotype controls were administered intradermally. Saureus load was assessed 2 days later by in vivo imaging and enumeration of colony forming units. Skin cellular infiltration was examined by flow cytometry and gene expression by quantitative PCR and transcriptome analysis. RESULTS IL-4Rα blockade decreased allergic skin inflammation in OVA-sensitized skin, as well as in OVA-sensitized and Saureus-exposed skin, evidenced by significantly decreased epidermal thickening and reduced dermal infiltration by eosinophils and mast cells. This was accompanied by increased cutaneous expression of Il17a and IL-17A-driven antimicrobial genes with no change in Il4 and Il13 expression. IL-4Rα blockade significantly decreased Saureus load in OVA-sensitized and S aureus-exposed skin. IL-17A blockade reversed the beneficial effect of IL-4Rα blockade on Saureus clearance and reduced the cutaneous expression of IL-17A-driven antimicrobial genes. CONCLUSIONS IL-4Rα blockade promotes Saureus clearance from sites of allergic skin inflammation in part by enhancing IL-17A expression.
Collapse
Affiliation(s)
| | - Alex McGurk
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Maria Strakosha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Daniela Vega-Mendoza
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Sophia E M Smith
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Kelsey Stafstrom
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Megan Elkins
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass
| | | | - Raif S Geha
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, Mass.
| |
Collapse
|
35
|
Çetinarslan T, Kümper L, Fölster-Holst R. The immunological and structural epidermal barrier dysfunction and skin microbiome in atopic dermatitis-an update. Front Mol Biosci 2023; 10:1159404. [PMID: 37654796 PMCID: PMC10467310 DOI: 10.3389/fmolb.2023.1159404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 08/04/2023] [Indexed: 09/02/2023] Open
Abstract
Atopic dermatitis (AD) is a common, chronic and relapsing inflammatory skin disease with various clinical presentations and combinations of symptoms. The pathophysiology of AD is complex and multifactorial. There are several factors involved in the etiopathogenesis of AD including structural and immunological epidermal barrier defect, imbalance of the skin microbiome, genetic background and environmental factors. Alterations in structural proteins, lipids, proteases, and their inhibitors, lead to the impairment of the stratum corneum which is associated with the increased skin penetration and transepidermal water loss. The elevated serum immunoglobulin E levels and blood eosinophilia have been shown in the majority of AD patients. Type 2 T-helper cell immune pathway with increased expression of interleukin (IL)-4, IL-5, and IL-13, has an important role in the etiopathogenesis of AD. Both T cells and keratinocytes contribute to epidermal barrier impairment in AD via a dynamic interaction of cytokines and chemokines. The skin microbiome is another factor of relevance in the etiopathogenesis of AD. It has been shown that during AD flares, Staphylococcus aureus (S. aureus) colonization increased, while Staphylococcus epidermidis (S. epidermidis) decreased. On the contrary, S. epidermidis and species of Streptococcus, Corynebacterium and Propionibacterium increased during the remision phases. However, it is not clear whether skin dysbiosis is one of the symptoms or one of the causes of AD. There are several therapeutic options, targeting these pathways which play a critical role in the etiopathogenesis of AD. Although topical steroids are the mainstay of the treatment of AD, new biological therapies including IL-4, IL-13, and IL-31 inhibitors, as well as Janus kinase inhibitors (JAKi), increasingly gain more importance with new advances in the therapy of AD. In this review, we summarize the role of immunological and structural epidermal barrier dysfunction, immune abnormalities, impairment of lipids, filaggrin mutation and skin microbiome in the etiopathogenesis of AD, as well as the therapeutic options for AD and their effects on these abnormalities in AD skin.
Collapse
Affiliation(s)
- Tubanur Çetinarslan
- Department of Dermatology and Venereology, Manisa Celal Bayar University, Manisa, Türkiye
| | - Lisa Kümper
- MEDICE Arzneimittel Pütter GmbH and Co. KG, Iserlohn, Germany
| | - Regina Fölster-Holst
- Department of Dermatology-Venereology and Allergology, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
36
|
Hartmann J, Moitinho-Silva L, Sander N, Harder I, Häsler R, Rodriguez E, Haufe E, Kleinheinz A, Abraham S, Heratizadeh A, Weisshaar E, Schäkel K, Handrick C, Augustin M, Wollenberg A, Staubach-Renz P, Ertner K, Sticherling M, Schwarz B, Quist S, Wiemers F, Schenck F, Wildberger J, Tittmann L, Lieb W, Schmitt J, Werfel T, Weidinger S. Dupilumab but not cyclosporine treatment shifts the microbiome toward a healthy skin flora in patients with moderate-to-severe atopic dermatitis. Allergy 2023; 78:2290-2300. [PMID: 37032440 DOI: 10.1111/all.15742] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) patients display an altered skin microbiome which may not only be an indicator but also a driver of inflammation. We aimed to investigate associations among AD patients' skin microbiome, clinical data, and response to systemic therapy in patients of the TREATgermany registry. METHODS Skin swabs of 157 patients were profiled with 16S rRNA gene amplicon sequencing before and after 3 months of treatment with dupilumab or cyclosporine. For comparison, 16s microbiome data from 258 population-based healthy controls were used. Disease severity was assessed using established instruments such as the Eczema Area and Severity Index (EASI). RESULTS We confirmed the previously shown correlation of Staphylococcus aureus abundance and bacterial alpha diversity with AD severity as measured by EASI. Therapy with Dupilumab shifted the bacterial community toward the pattern seen in healthy controls. The relative abundance of Staphylococci and in particular S. aureus significantly decreased on both lesional and non-lesional skin, whereas the abundance of Staphylococcus hominis increased. These changes were largely independent from the degree of clinical improvement and were not observed for cyclosporine. CONCLUSIONS Systemic treatment with dupilumab but not cyclosporine tends to restore a healthy skin microbiome largely independent of the clinical response indicating potential effects of IL-4RA blockade on the microbiome.
Collapse
Affiliation(s)
- Jan Hartmann
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Lucas Moitinho-Silva
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Nicole Sander
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Inken Harder
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Robert Häsler
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Elke Rodriguez
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Eva Haufe
- Center of Evidence-Based Healthcare, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | - Susanne Abraham
- Department of Dermatology, Medical Faculty Carl Gustav Carus, University Allergy Center, TU Dresden, Dresden, Germany
| | - Annice Heratizadeh
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Elke Weisshaar
- Division of Occupational Dermatology, Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Ruprecht-Karls University Heidelberg, Heidelberg, Germany
| | | | - Matthias Augustin
- Institute for Health Services Research in Dermatology Hamburg, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Andreas Wollenberg
- Clinics and Outpatient Clinics for Dermatology and Allergy, LMU Munich, Munich, Germany
| | - Petra Staubach-Renz
- Department of Dermatology and Allergy, University Medical Center Mainz, Mainz, Germany
| | | | - Michael Sticherling
- Department of Dermatology, University Hospital, Friedrich Alexander University Erlangen-Nuernberg, Erlangen, Germany
| | | | - Sven Quist
- Dermatology Clinic, Helix Medical Excellence Center Mainz, Mainz, Germany
| | | | | | - Julia Wildberger
- Practice Dr. med. Julia Wildberger, Hautmedizin Bad Soden, Bad Soden, Germany
| | - Lukas Tittmann
- Biobank PopGen and Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Wolfgang Lieb
- Institute of Epidemiology, Kiel University, Kiel, Germany
| | - Jochen Schmitt
- Center of Evidence-Based Healthcare, University Hospital and Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Stephan Weidinger
- Center for Inflammatory Skin Diseases, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
37
|
Simpson EL, De Benedetto A, Boguniewicz M, Ong PY, Lussier S, Villarreal M, Schneider LC, Paller AS, Guttman-Yassky E, Hanifin JM, Spergel JM, Barnes KC, David G, Austin B, Leung DYM, Beck LA. Phenotypic and Endotypic Determinants of Atopic Dermatitis Severity From the Atopic Dermatitis Research Network (ADRN) Registry. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2504-2515. [PMID: 37182563 PMCID: PMC10524351 DOI: 10.1016/j.jaip.2023.04.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 04/07/2023] [Accepted: 04/28/2023] [Indexed: 05/16/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin condition with a highly variable clinical phenotype. OBJECTIVE This study aimed to identify historical and clinical features and biomarkers associated with AD severity. METHODS A US registry of extensively phenotyped AD participants (aged 0.73-80 years) were enrolled at 9 academic centers. Information on family and personal medical history, examination, skin swabs (culture), and serum biomarkers was collected to evaluate their association with AD severity. RESULTS Participants with AD (N = 2862) whose disease was categorized as mild (11.6%), moderate (58.0%), or severe (30.4%) based on Rajka-Langeland scoring were enrolled. The trend test, when adjusting for gender, race, and age, demonstrated that severity was strongly (P ≤ .04) associated with a personal/family history of allergic disorders, history of alopecia, exposure to passive smoke, ocular herpes infection, skin bacterial and viral infections, and history of arrhythmia. Features observed more frequently (P ≤ .002), as a function of severity, included skin infections (impetigo, human papillomavirus, and molluscum contagiosum virus), Staphylococcus aureus colonization, excoriations, hyperlinear palms, ichthyosis, blepharitis, conjunctivitis, ectropion, and wheezing. Serum IgE, allergen and food (≤6 years) Phadiatop, and eosinophilia were strongly linked to severity (P < .001). CONCLUSIONS In a diverse US AD population, severity was associated with a history of atopic disorders, skin and extracutaneous bacterial and viral infections (by history and physical examination), higher IgE, eosinophilia and allergen sensitization, atopic skin manifestations (ie, excoriation, hyperlinear palms, and ichthyosis), and atopic ocular features (ie, blepharitis, conjunctivitis, and ectropion) as well as asthma findings (ie, wheezing). Data from our prospective registry significantly advance our understanding of AD phenotypes and endotypes, which is critical to achieve optimal management.
Collapse
Affiliation(s)
- Eric L Simpson
- Department of Dermatology, Oregon Health & Science University, Portland, Ore
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Mark Boguniewicz
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health and University of Colorado School of Medicine, Denver, Colo
| | - Peck Y Ong
- Division of Clinical Immunology and Allergy, Department of Pediatrics, Children's Hospital Los Angeles, University Southern California, Los Angeles, Calif
| | | | | | - Lynda C Schneider
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Amy S Paller
- Departments of Dermatology and Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Emma Guttman-Yassky
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jon M Hanifin
- Department of Dermatology, Oregon Health & Science University, Portland, Ore
| | - Jonathan M Spergel
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | - Kathleen C Barnes
- Department of Medicine and Epidemiology, University of Colorado Anschutz Medical Campus, Denver, Colo
| | | | | | - 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 Dermatology, Medicine and Pathology, University of Rochester Medical Center, Rochester, NY.
| |
Collapse
|
38
|
Xu Z, Yan J, Wen W, Zhang N, Bachert C. Pathophysiology and management of Staphylococcus aureus in nasal polyp disease. Expert Rev Clin Immunol 2023; 19:981-992. [PMID: 37409375 DOI: 10.1080/1744666x.2023.2233700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/03/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Staphylococcus aureus (S. aureus) is a common pathogen that frequently colonizes the sinonasal cavity. Recent studies demonstrated the essential role of Staphylococcus aureus in the pathophysiology of uncontrolled severe chronic rhinosinusitis with nasal polyps (NP) by initiating an immune response to the germ and its products, resulting in type 2 inflammation. AREAS COVERED This review aims to summarize the evidence for the role of S. aureus in the development of NP disease including S. aureus-related virulence factors, the pathophysiologic mechanisms used by S. aureus, and the synergistic effects of S. aureus and other pathogens. It also describes the current management of S. aureus associated with NPs as well as potential therapeutic strategies that are used in clinical practice. EXPERT OPINION S. aureus is able to damage the nasal mucosal epithelial barrier, impair the clearance of the host immune system, and trigger adaptive and innate immune reactions which lead to the formation of inflammation and nasal polyp growth. Further studies should focus on the development of novel therapeutic strategies, such as biologics, bacteriophages, probiotics, and nanomedicine, which could be used to treat S. aureus and its immunological consequences in the future.
Collapse
Affiliation(s)
- Zhaofeng Xu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Jieying Yan
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Weiping Wen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Nan Zhang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
| | - Claus Bachert
- Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
- Upper Airway Research Laboratory, Ghent University, Ghent, Belgium
- Division of ENT Diseases, Stockholm, Sweden
- Clinic for ENT Diseases and Head and Neck Surgery, University Clinic Münster, Münster, Germany
| |
Collapse
|
39
|
Rapin A, Rehbinder EM, Macowan M, Pattaroni C, Lødrup Carlsen KC, Harris NL, Jonassen CM, Landrø L, Lossius AH, Nordlund B, Rudi K, Skjerven HO, Cathrine Staff A, Söderhäll C, Ubags N, Vettukattil R, Marsland BJ. The skin microbiome in the first year of life and its association with atopic dermatitis. Allergy 2023; 78:1949-1963. [PMID: 36779606 DOI: 10.1111/all.15671] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 02/14/2023]
Abstract
BACKGROUND Early-life microbial colonization of the skin may modulate the immune system and impact the development of atopic dermatitis (AD) and allergic diseases later in life. To address this question, we assessed the association between the skin microbiome and AD, skin barrier integrity and allergic diseases in the first year of life. We further explored the evolution of the skin microbiome with age and its possible determinants, including delivery mode. METHODS Skin microbiome was sampled from the lateral upper arm on the first day of life, and at 3, 6, and 12 months of age. Bacterial communities were assessed by 16S rRNA gene amplicon sequencing in 346 infants from the PreventADALL population-based birth cohort study, representing 970 samples. Clinical investigations included skin examination and skin barrier function measured as trans-epidermal water loss (TEWL) at the site and time of microbiome sampling at 3, 6, and 12 months. Parental background information was recorded in electronic questionnaires, and delivery mode (including vaginal delivery (VD), VD in water, elective caesarean section (CS) and emergency CS) was obtained from maternal hospital charts. RESULTS Strong temporal variations in skin bacterial community composition were found in the first year of life, with distinct patterns associated with different ages. Confirming our hypothesis, skin bacterial community composition in the first year of life was associated with skin barrier integrity and later onsets of AD. Delivery mode had a strong impact on the microbiome composition at birth, with each mode leading to distinct patterns of colonization. Other possible determinants of the skin microbiome were identified, including environmental and parental factors as well as breastfeeding. CONCLUSION Skin microbiome composition during infancy is defined by age, transiently influenced by delivery mode as well as environmental, parental factors and breastfeeding. The microbiome is also associated with skin barrier integrity and the onset of AD.
Collapse
Affiliation(s)
- Alexis Rapin
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Service de Pneumologie, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Epalinges, Switzerland
| | - Eva Maria Rehbinder
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Matthew Macowan
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Céline Pattaroni
- Service de Pneumologie, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Epalinges, Switzerland
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Karin C Lødrup Carlsen
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Nicola L Harris
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| | - Christine M Jonassen
- Genetic Unit, Centre for Laboratory Medicine, Østfold Hospital Trust, Kalnes, Norway
| | - Linn Landrø
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Astrid H Lossius
- Department of Dermatology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Björn Nordlund
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Håvard O Skjerven
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Anne Cathrine Staff
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Cilla Söderhäll
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Niki Ubags
- Service de Pneumologie, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Epalinges, Switzerland
| | - Riyas Vettukattil
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Benjamin J Marsland
- Service de Pneumologie, Département de Médecine, Centre Hospitalier Universitaire Vaudois (CHUV), Epalinges, Switzerland
- Department of Immunology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
De Tomassi A, Reiter A, Reiger M, Rauer L, Rohayem R, Ck-Care Study Group, Traidl-Hoffmann C, Neumann AU, Hülpüsch C. Combining 16S Sequencing and qPCR Quantification Reveals Staphylococcus aureus Driven Bacterial Overgrowth in the Skin of Severe Atopic Dermatitis Patients. Biomolecules 2023; 13:1030. [PMID: 37509067 PMCID: PMC10377005 DOI: 10.3390/biom13071030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/11/2023] [Accepted: 06/18/2023] [Indexed: 07/30/2023] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease with a microbiome dysbiosis towards a high relative abundance of Staphylococcus aureus. However, information is missing on the actual bacterial load on AD skin, which may affect the cell number driven release of pathogenic factors. Here, we combined the relative abundance results obtained by next-generation sequencing (NGS, 16S V1-V3) with bacterial quantification by targeted qPCR (total bacterial load = 16S, S. aureus = nuc gene). Skin swabs were sampled cross-sectionally (n = 135 AD patients; n = 20 healthy) and longitudinally (n = 6 AD patients; n = 6 healthy). NGS and qPCR yielded highly inter-correlated S. aureus relative abundances and S. aureus cell numbers. Additionally, intra-individual differences between body sides, skin status, and consecutive timepoints were also observed. Interestingly, a significantly higher total bacterial load, in addition to higher S. aureus relative abundance and cell numbers, was observed in AD patients in both lesional and non-lesional skin, as compared to healthy controls. Moreover, in the lesional skin of AD patients, higher S. aureus cell numbers significantly correlated with the higher total bacterial load. Furthermore, significantly more severe AD patients presented with higher S. aureus cell number and total bacterial load compared to patients with mild or moderate AD. Our results indicate that severe AD patients exhibit S. aureus driven increased bacterial skin colonization. Overall, bacterial quantification gives important insights in addition to microbiome composition by sequencing.
Collapse
Affiliation(s)
- Amedeo De Tomassi
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Anna Reiter
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Luise Rauer
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, 86156 Augsburg, Germany
- Environmental Medicine, Technical University of Munich, 86156 Augsburg, Germany
| | - Robin Rohayem
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
| | - Ck-Care Study Group
- CK CARE, Christine-Kühne Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, 86156 Augsburg, Germany
- Environmental Medicine, Technical University of Munich, 86156 Augsburg, Germany
- CK CARE, Christine-Kühne Center for Allergy Research and Education, 7265 Davos, Switzerland
- ZIEL-Institute for Food and Health, Technical University of Munich, 85354 Freising, Germany
| | - Avidan U Neumann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
- Institute of Environmental Medicine, Helmholtz Zentrum München, 86156 Augsburg, Germany
- CK CARE, Christine-Kühne Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Claudia Hülpüsch
- Environmental Medicine, Faculty of Medicine, University of Augsburg, 86156 Augsburg, Germany
- Environmental Medicine, Technical University of Munich, 86156 Augsburg, Germany
- CK CARE, Christine-Kühne Center for Allergy Research and Education, 7265 Davos, Switzerland
| |
Collapse
|
41
|
Conte AL, Brunetti F, Marazzato M, Longhi C, Maurizi L, Raponi G, Palamara AT, Grassi S, Conte MP. Atopic dermatitis-derived Staphylococcus aureus strains: what makes them special in the interplay with the host. Front Cell Infect Microbiol 2023; 13:1194254. [PMID: 37389215 PMCID: PMC10303148 DOI: 10.3389/fcimb.2023.1194254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/22/2023] [Indexed: 07/01/2023] Open
Abstract
Background Atopic dermatitis (AD) is a chronic inflammatory skin condition whose pathogenesis involves genetic predisposition, epidermal barrier dysfunction, alterations in the immune responses and microbial dysbiosis. Clinical studies have shown a link between Staphylococcus aureus and the pathogenesis of AD, although the origins and genetic diversity of S. aureus colonizing patients with AD is poorly understood. The aim of the study was to investigate if specific clones might be associated with the disease. Methods WGS analyses were performed on 38 S. aureus strains, deriving from AD patients and healthy carriers. Genotypes (i.e. MLST, spa-, agr- and SCCmec-typing), genomic content (e.g. virulome and resistome), and the pan-genome structure of strains have been investigated. Phenotypic analyses were performed to determine the antibiotic susceptibility, the biofilm production and the invasiveness within the investigated S. aureus population. Results Strains isolated from AD patients revealed a high degree of genetic heterogeneity and a shared set of virulence factors and antimicrobial resistance genes, suggesting that no genotype and genomic content are uniquely associated with AD. The same strains were characterized by a lower variability in terms of gene content, indicating that the inflammatory conditions could exert a selective pressure leading to the optimization of the gene repertoire. Furthermore, genes related to specific mechanisms, like post-translational modification, protein turnover and chaperones as well as intracellular trafficking, secretion and vesicular transport, were significantly more enriched in AD strains. Phenotypic analysis revealed that all of our AD strains were strong or moderate biofilm producers, while less than half showed invasive capabilities. Conclusions We conclude that in AD skin, the functional role played by S. aureus may depend on differential gene expression patterns and/or on post-translational modification mechanisms rather than being associated with peculiar genetic features.
Collapse
Affiliation(s)
- Antonietta Lucia Conte
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Francesca Brunetti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Massimiliano Marazzato
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Catia Longhi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Linda Maurizi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giammarco Raponi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia- Cenci Bolognetti Foundation, Rome, Italy
| | - Sara Grassi
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Maria Pia Conte
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
42
|
Wiggins S, Levit NA. Dupilumab Treatment is Not Associated with Increased Risk of Overall Skin Infections [Letter]. Immunotargets Ther 2023; 12:77-78. [PMID: 37256021 PMCID: PMC10226537 DOI: 10.2147/itt.s421440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023] Open
Affiliation(s)
- Simmi Wiggins
- Sanofi, 410 Thames Valley Park Dr, Reading, RG6 1PT, UK
| | - Noah A Levit
- Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| |
Collapse
|
43
|
Nakatsuji T, Brinton SL, Cavagnero KJ, O'Neill AM, Chen Y, Dokoshi T, Butcher AM, Osuoji OC, Shafiq F, Espinoza JL, Dupont CL, Hata TR, Gallo RL. Competition between skin antimicrobial peptides and commensal bacteria in type 2 inflammation enables survival of S. aureus. Cell Rep 2023; 42:112494. [PMID: 37167061 DOI: 10.1016/j.celrep.2023.112494] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/17/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023] Open
Abstract
During inflammation, the skin deploys antimicrobial peptides (AMPs) yet during allergic inflammation it becomes more susceptible to Staphylococcus aureus. To understand this contradiction, single-cell sequencing of Il4ra-/- mice combined with skin microbiome analysis reveals that lower production of AMPs from interleukin-4 receptor α (IL-4Rα) activation selectively inhibits survival of antibiotic-producing strains of coagulase-negative Staphylococcus (CoNS). Diminished AMPs under conditions of T helper type 2 (Th2) inflammation enable expansion of CoNS strains without antibiotic activity and increase Staphylococcus aureus (S. aureus), recapitulating the microbiome on humans with atopic dermatitis. This response is rescued in Camp-/- mice or after topical steroids, since further inhibition of AMPs enables survival of antibiotic-producing CoNS strains. In conditions of Th17 inflammation, a higher expression of host AMPs is sufficient to directly inhibit S. aureus survival. These results show that antimicrobials produced by the host and commensal bacteria each act to control S. aureus on the skin.
Collapse
Affiliation(s)
- Teruaki Nakatsuji
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Samantha L Brinton
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Kellen J Cavagnero
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alan M O'Neill
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Yang Chen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Anna M Butcher
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Olive C Osuoji
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Faiza Shafiq
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Josh L Espinoza
- Genomic Medicine, J. Craig Venter Institute, La Jolla, CA 92037, USA
| | | | - Tissa R Hata
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
44
|
Butala S, Castelo-Soccio L, Seshadri R, Simpson EL, O'Shea JJ, Bieber T, Paller AS. Biologic Versus Small Molecule Therapy for Treating Moderate to Severe Atopic Dermatitis: Clinical Considerations. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1361-1373. [PMID: 36948491 PMCID: PMC10164714 DOI: 10.1016/j.jaip.2023.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/24/2023]
Abstract
The U.S. Food and Drug Administration approval of dupilumab for moderate-to-severe atopic dermatitis shifted the paradigm from use of broad, systemic immunosuppressants to a safer, targeted treatment and led to the emergence of newer interleukin (IL)-4/IL-13 directed biologics and small molecule therapies, namely Janus kinase (JAK) inhibitors (JAKi). Tralokinumab and emerging (not yet approved) lebrikizumab, which both target IL-13, are alternative biologics to dupilumab. The emerging anti-IL-31 receptor nemolizumab is likely to be used second-line to other biologics, primarily for pruritus. Three JAKi are currently in use for treating atopic dermatitis, 2 of which, abrocitinib and upadacitinib, are U.S. Food and Drug Administration-approved. This review provides an in-depth, practical discussion on use of these biologics and JAKi that are approved or have completed phase 3 clinical trials in pediatric patients and adults, comparing the groups of medications based on available efficacy and safety data.
Collapse
Affiliation(s)
- Sneha Butala
- Departments of Dermatology and Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Ill
| | - Leslie Castelo-Soccio
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Rishi Seshadri
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | - Eric L Simpson
- Department of Dermatology, Oregon Health and Science University, Portland, Ore
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Thomas Bieber
- Department of Dermatology and Allergy, University of Bonn, Bonn, Germany
| | - Amy S Paller
- Departments of Dermatology and Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Ill.
| |
Collapse
|
45
|
van den Bogaard EH, Elias PM, Goleva E, Berdyshev E, Smits JPH, Danby SG, Cork MJ, Leung DYM. Targeting Skin Barrier Function in Atopic Dermatitis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1335-1346. [PMID: 36805053 PMCID: PMC11346348 DOI: 10.1016/j.jaip.2023.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/22/2023]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease in the general population. Skin barrier dysfunction is the central abnormality leading to AD. The cause of skin barrier dysfunction is complex and rooted in genetic mutations, interactions between the immune pathway activation and epithelial cells, altered host defense mechanisms, as well as environmental influences that cause epithelial cell activation and release of alarmins (such as thymic stromal lymphopoietin) that can activate the type 2 immune pathway, including generation of interleukins 4 and 13, which induces defects in the skin barrier and increased allergic inflammation. These inflammatory pathways are further influenced by environmental factors including the microbiome (especially Staphylococcus aureus), air pollution, stress, and other factors. As such, AD is a syndrome involving multiple phenotypes, all of which have in common skin barrier dysfunction as a key contributing factor. Understanding mechanisms leading to skin barrier dysfunction in AD is pointing to the development of new topical and systemic treatments in AD that helps keep skin borders secure and effectively treat the disease.
Collapse
Affiliation(s)
- Ellen H van den Bogaard
- Department of Dermatology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter M Elias
- Department of Dermatology, University of California San Francisco and VA Medical Center, San Francisco, Calif
| | - Elena Goleva
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, National Jewish Health, Denver, Colo
| | - Evgeny Berdyshev
- Department of Pulmonology, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colo
| | - Jos P H Smits
- Department of Dermatology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Simon G Danby
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School at The University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Michael J Cork
- Department of Infection, Immunity and Cardiovascular Disease, The Medical School at The University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Donald Y M Leung
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, National Jewish Health, Denver, Colo.
| |
Collapse
|
46
|
Rauer L, Reiger M, Bhattacharyya M, Brunner PM, Krueger JG, Guttman-Yassky E, Traidl-Hoffmann C, Neumann AU. Skin microbiome and its association with host cofactors in determining atopic dermatitis severity. J Eur Acad Dermatol Venereol 2023; 37:772-782. [PMID: 36433676 DOI: 10.1111/jdv.18776] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a heterogeneous, chronic inflammatory skin disease linked to skin microbiome dysbiosis with reduced bacterial diversity and elevated relative abundance of Staphylococcus aureus (S. aureus). OBJECTIVES We aimed to characterize the yet incompletely understood association between the skin microbiome and patients' demographic and clinical cofactors in relation to AD severity. METHODS The skin microbiome in 48 adult moderate-to-severe AD patients was investigated using next-generation deep sequencing (16S rRNA gene, V1-V3 region) followed by denoising (DADA2) to obtain amplicon sequence variant (ASV) composition. RESULTS In lesional skin, AD severity was associated with S. aureus relative abundance (rS = 0.53, p < 0.001) and slightly better with the microbiome diversity measure Evenness (rS = -0.58, p < 0.001), but not with Richness. Multiple regression confirmed the association of AD severity with microbiome diversity, including Shannon (in lesional skin, p < 0.001), Evenness (in non-lesional skin, p = 0.015) or S. aureus relative abundance (p < 0.012), and with patient's IgE levels (p < 0.001), race (p < 0.032), age (p < 0.034) and sex (p = 0.012). The lesional model explained 62% of the variation in AD severity, and the non-lesional model 50% of the variation. CONCLUSIONS Our results specify the frequently reported "reduced diversity" of the AD-related skin microbiome to reduced Evenness, which was in turn mainly driven by S. aureus relative abundance, rather than to a reduced microbiome Richness. Finding associations between AD severity, the skin microbiome and patient's cofactors is a key aspect in developing new personalized AD treatments, particularly those targeting the AD microbiome.
Collapse
Affiliation(s)
- Luise Rauer
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Institute for Medical Information Processing, Biometry and Epidemiology (IBE), LMU, Munich, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Madhumita Bhattacharyya
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany
| | - Patrick M Brunner
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE Center for Allergy Research and Education, Davos, Switzerland.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Avidan U Neumann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
47
|
Youn C, Dikeman DA, Chang E, Liu H, Nolan SJ, Alphonse MP, Joyce DP, Liu Q, Meixiong J, Dong X, Miller LS, Archer NK. Crisaborole efficacy in murine models of skin inflammation and Staphylococcus aureus infection. Exp Dermatol 2023; 32:425-435. [PMID: 36461082 PMCID: PMC10066830 DOI: 10.1111/exd.14722] [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: 04/19/2022] [Revised: 09/09/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Phosphodiesterase 4 (PDE4) is highly expressed in keratinocytes and immune cells and promotes pro-inflammatory responses upon activation. The activity of PDE4 has been attributed to various inflammatory conditions, leading to the development and approval of PDE4 inhibitors as host-directed therapeutics in humans. For example, the topical PDE4 inhibitor, crisaborole, is approved for the treatment of mild-to-moderate atopic dermatitis and has shown efficacy in patients with psoriasis. However, the role of crisaborole in regulating the immunopathogenesis of inflammatory skin diseases and infection is not entirely known. Therefore, we evaluated the effects of crisaborole in multiple mouse models, including psoriasis-like dermatitis, AD-like skin inflammation with and without filaggrin mutations, and Staphylococcus aureus skin infection. We discovered that crisaborole dampens myeloid cells and itch in the skin during psoriasis-like dermatitis. Furthermore, crisaborole was effective in reducing skin inflammation in the context of filaggrin deficiency. Importantly, crisaborole reduced S. aureus skin colonization during AD-like skin inflammation. However, crisaborole was not efficacious in treating S. aureus skin infections, even as adjunctive therapy to antibiotics. Taken together, we found that crisaborole reduced itch during psoriasis-like dermatitis and decreased S. aureus skin colonization upon AD-like skin inflammation, which act as additional mechanisms by which crisaborole dampens the immunopathogenesis in mouse models of inflammatory skin diseases. Further examination is warranted to translate these preclinical findings to human disease.
Collapse
Affiliation(s)
- Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dustin A. Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evelyn Chang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabrina J. Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James Meixiong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xinzhong Dong
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
48
|
Beck LA, Bieber T, Weidinger S, Tauber M, Saeki H, Irvine AD, Eichenfield LF, Werfel T, Arlert P, Jiang L, Røpke M, Paller AS. Tralokinumab treatment improves the skin microbiota by increasing the microbial diversity in adults with moderate-to-severe atopic dermatitis: Analysis of microbial diversity in ECZTRA 1, a randomized controlled trial. J Am Acad Dermatol 2023; 88:816-823. [PMID: 36473633 DOI: 10.1016/j.jaad.2022.11.047] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is characterized by microbial dysbiosis, immune dysregulation, and an impaired skin barrier. Microbial dysbiosis in AD involves a reduction in diversity primarily driven by an increased abundance of Staphylococcus aureus. Tralokinumab, an approved treatment for adults with moderate-to-severe AD, improves the skin barrier and immune abnormalities by specifically targeting the interleukin 13 cytokine, but its impact on the skin microbiome is unknown. OBJECTIVE To investigate how tralokinumab affects the skin microbiome by examining the lesional skin of adults with moderate-to-severe AD from the phase 3 ECZTRA 1 trial (NCT03131648). METHODS Microbiome profiling, S aureus abundance, and biomarker data were assessed in a subset of ECZTRA 1 participants (S aureus abundance at baseline and week 16; microbiome profiling at baseline, and week 8/16; and serum sampling before dose and week 4/8/16/28/52). RESULTS Tralokinumab treatment led to increased microbial diversity, reduced S aureus abundance, and increased abundance of the commensal coagulase-negative Staphylococci. LIMITATIONS Limitations include a lack of S aureus abundance data at week 8, sampling site variation between participants, and possible influence from concomitant systemic antiinfectives. CONCLUSION Our findings indicate specific targeting of the interleukin 13 cytokine with tralokinumab can directly and/or indirectly improve microbial dysbiosis seen in AD skin.
Collapse
Affiliation(s)
- Lisa A Beck
- Department of Dermatology, Medicine and Pathology, University of Rochester Medical Center, Rochester, New York.
| | - Thomas Bieber
- Department of Dermatology and Allergy, University Hospital, Bonn, Germany; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Marie Tauber
- Dermatology and Allergology Department, Toulouse University Hospital and Inserm UMR1291 - CNRS UMR5051, Toulouse, France
| | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Alan D Irvine
- Clinical Medicine, Trinity College Dublin, Ireland; Department of Dermatology, Children's Health Ireland, Dublin, Ireland
| | - Lawrence F Eichenfield
- Departments of Dermatology and Pediatrics, University of California San Diego School of Medicine, La Jolla, California
| | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | | | - Li Jiang
- LEO Pharma A/S, Ballerup, Denmark
| | | | - Amy S Paller
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
49
|
Facheris P, Jeffery J, Del Duca E, Guttman-Yassky E. The translational revolution in atopic dermatitis: the paradigm shift from pathogenesis to treatment. Cell Mol Immunol 2023; 20:448-474. [PMID: 36928371 DOI: 10.1038/s41423-023-00992-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin disease, and it is considered a complex and heterogeneous condition. Different phenotypes of AD, defined according to the patient age at onset, race, and ethnic background; disease duration; and other disease characteristics, have been recently described, underlying the need for a personalized treatment approach. Recent advancements in understanding AD pathogenesis resulted in a real translational revolution and led to the exponential expansion of the therapeutic pipeline. The study of biomarkers in clinical studies of emerging treatments is helping clarify the role of each cytokine and immune pathway in AD and will allow addressing the unique immune fingerprints of each AD subset. Personalized medicine will be the ultimate goal of this targeted translational research. In this review, we discuss the changes in the concepts of both the pathogenesis of and treatment approach to AD, highlight the scientific rationale behind each targeted treatment and report the most recent clinical efficacy data.
Collapse
Affiliation(s)
- Paola Facheris
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Humanitas Clinical and Research Center, Department of Dermatology, Rozzano, Milano, Italy
| | - Jane Jeffery
- Duke University School of Medicine, Durham, NC, USA
| | - Ester Del Duca
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
50
|
Chen M, Gao K, Ali K, Shan J, Qiu Y, Xie T, Yu Y, Wu L. Case report: Dupilumab leads to an increased chance of head and neck Staphylococcus aureus infection in atopic dermatitis patients. Front Med (Lausanne) 2023; 10:1027589. [PMID: 36968828 PMCID: PMC10031056 DOI: 10.3389/fmed.2023.1027589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/16/2023] [Indexed: 03/10/2023] Open
Abstract
Dupilumab was the first biological medication licensed to treat atopic dermatitis (AD), and it has shown remarkable effectiveness and safety in the treatment of moderate-to-severe atopic dermatitis. There are limited drug-related adverse events associated with dupilumab in atopic dermatitis (AD) treatment. Here, we present two cases of local Staphylococcus aureus infection during the treatment of atopic dermatitis with dupilumab.
Collapse
Affiliation(s)
- Min Chen
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kai Gao
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kamran Ali
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jinpeng Shan
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - YunMi Qiu
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Tianci Xie
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yiling Yu
- Department of Dermatology, The Fourth School of Medicine Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liming Wu
- Department of Dermatology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Liming Wu
| |
Collapse
|