1
|
Prakoeswa CRS, Damayanti, Anggraeni S, Umborowati MA, Sari M, Hendaria MP, Thahir TF. The Role of Moisturizer Containing Anti-inflammatory on Skin Hydration in Mild-Moderate Atopic Dermatitis Patients. Dermatol Res Pract 2024; 2024:3586393. [PMID: 39741562 PMCID: PMC11685320 DOI: 10.1155/drp/3586393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 10/09/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic, inflammatory skin condition characterized by eczema lesions and dry, itchy skin. Recent guidelines for the management of AD emphasize the importance of using moisturizers in the management of AD. This study is a double-blind clinical trial to determine the effectiveness of moisturizers containing anti-inflammatory ingredients compared with moisturizers without anti-inflammatory ingredients for skin hydration in mild to moderate adult AD patients for 14 days at the Dermatology and Venereology Outpatient Clinic at Dr. Soetomo General Academic Hospital. There was a significant difference (p < 0.05) at the baseline and day 14 skin hydration values in the experiment group with anti-inflammatory ingredients (35.97 ± 6.04-66.06 ± 15.84) and the control group without anti-inflammatory ingredients (40.74 ± 10.94-56.12 ± 8.34). After comparison, there was a significant difference (p < 0.05) in the skin hydration value between the experiment group and the control group on the 14th day. There was also a significant difference in the improvement of skin hydration outcomes between both groups (p < 0.05). The severity of the disease using Scoring Atopic Dermatitis (SCORAD) showed a significant difference (p < 0.05) between the experiment group and the control group after 2 weeks of intervention. The addition of anti-inflammatory ingredients in the moisturizer, namely, shea butter, bacterial lysate, allantoin, bisabolol, Phragmites kharka extract, Poria cocos, and Mirabilis jalapa in a moisturizer containing occlusive (dimethicone), humectants (glycerin, saccharide, butylene glycol, and hyaluronic acid), and emollient (shea butter and squalane) was shown to be significantly better in improving skin hydration in patients with mild to moderate AD.
Collapse
Affiliation(s)
- Cita Rosita Sigit Prakoeswa
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Damayanti
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sylvia Anggraeni
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Menul Ayu Umborowati
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Maylita Sari
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Made Putri Hendaria
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Tanziela Firdausi Thahir
- Department of Dermatology, Venereology and Aesthetic, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Department of Dermatology, Venereology and Aesthetic, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
2
|
Yue C, Zhou H, Wang X, Yu J, Hu Y, Zhou P, Zhao F, Zeng F, Li G, Li Y, Feng Y, Sun X, Huang S, He M, Wu W, Huang N, Li J. Atopic dermatitis: pathogenesis and therapeutic intervention. MedComm (Beijing) 2024; 5:e70029. [PMID: 39654684 PMCID: PMC11625510 DOI: 10.1002/mco2.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
The skin serves as the first protective barrier for nonspecific immunity and encompasses a vast network of skin-associated immune cells. Atopic dermatitis (AD) is a prevalent inflammatory skin disease that affects individuals of all ages and races, with a complex pathogenesis intricately linked to genetic, environmental factors, skin barrier dysfunction as well as immune dysfunction. Individuals diagnosed with AD frequently exhibit genetic predispositions, characterized by mutations that impact the structural integrity of the skin barrier. This barrier dysfunction leads to the release of alarmins, activating the type 2 immune pathway and recruiting various immune cells to the skin, where they coordinate cutaneous immune responses. In this review, we summarize experimental models of AD and provide an overview of its pathogenesis and the therapeutic interventions. We focus on elucidating the intricate interplay between the immune system of the skin and the complex regulatory mechanisms, as well as commonly used treatments for AD, aiming to systematically understand the cellular and molecular crosstalk in AD-affected skin. Our overarching objective is to provide novel insights and inform potential clinical interventions to reduce the incidence and impact of AD.
Collapse
Affiliation(s)
- Chengcheng Yue
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Jiadong Yu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Yawen Hu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Fulei Zhao
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Fanlian Zeng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Guolin Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Ya Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Yuting Feng
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Xiaochi Sun
- Department of CardiologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Shishi Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Mingxiang He
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Wenling Wu
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Nongyu Huang
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer CenterWest China HospitalSichuan UniversitySichuan University and Collaborative Innovation Center for BiotherapyChengduSichuanChina
| |
Collapse
|
3
|
Livshits G, Kalinkovich A. Resolution of Chronic Inflammation, Restoration of Epigenetic Disturbances and Correction of Dysbiosis as an Adjunctive Approach to the Treatment of Atopic Dermatitis. Cells 2024; 13:1899. [PMID: 39594647 PMCID: PMC11593003 DOI: 10.3390/cells13221899] [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: 10/08/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with multifactorial and unclear pathogenesis. Its development is characterized by two key elements: epigenetic dysregulation of molecular pathways involved in AD pathogenesis and disrupted skin and gut microbiota (dysbiosis) that jointly trigger and maintain chronic inflammation, a core AD characteristic. Current data suggest that failed inflammation resolution is the main pathogenic mechanism underlying AD development. Inflammation resolution is provided by specialized pro-resolving mediators (SPMs) derived from dietary polyunsaturated fatty acids acting through cognate receptors. SPM levels are reduced in AD patients. Administration of SPMs or their stable, small-molecule mimetics and receptor agonists, as well as supplementation with probiotics/prebiotics, demonstrate beneficial effects in AD animal models. Epidrugs, compounds capable of restoring disrupted epigenetic mechanisms associated with the disease, improve impaired skin barrier function in AD models. Based on these findings, we propose a novel, multilevel AD treatment strategy aimed at resolving chronic inflammation by application of SPM mimetics and receptor agonists, probiotics/prebiotics, and epi-drugs. This approach can be used in conjunction with current AD therapy, resulting in AD alleviation.
Collapse
Affiliation(s)
- Gregory Livshits
- Department of Morphological Sciences, Adelson School of Medicine, Ariel University, Ariel 4077625, Israel
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel;
| | - Alexander Kalinkovich
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel Aviv 6927846, Israel;
| |
Collapse
|
4
|
Lee E, Kim JH, Lee SY, Lee SH, Park YM, Oh HY, Yeom J, Ahn HS, Yoo HJ, Kim BS, Yun SM, Choi EJ, Song KB, Park MJ, Ahn K, Kim KW, Shin YH, Suh DI, Song JY, Hong SJ. Developmental trajectories of atopic dermatitis with multiomics approaches in the infant gut: COCOA birth cohort. J Allergy Clin Immunol 2024:S0091-6749(24)01187-4. [PMID: 39547281 DOI: 10.1016/j.jaci.2024.10.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/07/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND An understanding of the phenotypes and endotypes of atopic dermatitis (AD) is essential for developing precision therapies. Recent studies have demonstrated evidence for the gut-skin axis in AD. OBJECTIVE We sought to determine the natural course and clinical characteristics of AD phenotypes and investigate their mechanisms on the basis of multiomics analyses. METHODS Latent class trajectory analysis was used to classify AD phenotypes in 2247 children who were followed until age 9 years from the COhort for Childhood Origin of Asthma and allergic diseases birth cohort study. Multiomics analyses (microbiome, metabolites, and gut epithelial cell transcriptome) using stool samples collected at age 6 months were performed to elucidate the underlying mechanisms of AD phenotypes. RESULTS Five AD phenotypes were classified as follows: never/infrequent, early-onset transient, intermediate transient, late-onset, and early-onset persistent. Early-onset persistent and late-onset phenotypes showed increased risks of food allergy and wheezing treatment ever, with bronchial hyperresponsiveness evident only in the early-onset persistent phenotype. Multiomics analyses revealed a significantly lower relative abundance of Ruminococcus gnavus and a decreased gut acetate level in the early-onset persistent phenotype, with potential associations to ACSS2, Janus kinase-signal transducer and activator of transcription signaling, and systemic TH2 inflammation. The early-onset transient phenotype was associated with adenosine monophosphate-activated protein kinase (AMPK) and/or chemokine signaling regulation, whereas the late-onset phenotype was linked with IL-17 and barrier dysfunction. CONCLUSIONS Multiomics profiling in early life may offer insights into different mechanisms underlying AD phenotypes in children.
Collapse
Affiliation(s)
- Eun Lee
- Department of Pediatrics, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Jeong-Hyun Kim
- Department of Medicine, University of Ulsan Collage of Medicine, Seoul, Korea
| | | | - Si Hyeon Lee
- Department of Medicine, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Yoon Mee Park
- Department of Medicine, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Hea Young Oh
- Department of Medicine, University of Ulsan Collage of Medicine, Seoul, Korea
| | - Jeonghun Yeom
- Prometabio Research Institute, Prometabio Co, Ltd, Seoul, Korea
| | | | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, Seoul, Korea
| | - Bong-Soo Kim
- Department of Life Science, Multidisciplinary Genome Institute, Hallym University, Chuncheon, Korea
| | - Sun Mi Yun
- Microbiome Division, Macrogen, Inc, Seoul, Korea
| | - Eom Ji Choi
- Department of Pediatrics, CHA Gangnam Medical Center, Seoul, Korea
| | - Kun Baek Song
- Department of Pediatrics, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | - Min Jee Park
- Department of Pediatrics, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Kangmo Ahn
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Won Kim
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Youn Ho Shin
- Department of Pediatrics, Kyunghee University of Medicine, Seoul, Korea
| | - Dong In Suh
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Joo Young Song
- Department of Pediatrics, CHA University Ilsan Medical Center, Goyang, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
van den Brink NJM, Pardow F, Meesters LD, van Vlijmen-Willems I, Rodijk-Olthuis D, Niehues H, Jansen PAM, Roelofs SH, Brewer MG, van den Bogaard EH, Smits JPH. Electrical Impedance Spectroscopy Quantifies Skin Barrier Function in Organotypic In Vitro Epidermis Models. J Invest Dermatol 2024; 144:2488-2500.e4. [PMID: 38642800 DOI: 10.1016/j.jid.2024.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/16/2024] [Accepted: 03/02/2024] [Indexed: 04/22/2024]
Abstract
Three-dimensional human epidermal equivalents (HEEs) are a state-of-the-art organotypic culture model in preclinical investigative dermatology and regulatory toxicology. In this study, we investigated the utility of electrical impedance spectroscopy (EIS) for noninvasive measurement of HEE epidermal barrier function. Our setup comprised a custom-made lid fit with 12 electrode pairs aligned on the standard 24-transwell cell culture system. Serial EIS measurements for 7 consecutive days did not impact epidermal morphology, and readouts showed comparable trends with HEEs measured only once. We determined 2 frequency ranges in the resulting impedance spectra: a lower frequency range termed EISdiff correlated with keratinocyte terminal differentiation independent of epidermal thickness and a higher frequency range termed EISSC correlated with stratum corneum thickness. HEEs generated from CRISPR/Cas9-engineered keratinocytes that lack key differentiation genes FLG, TFAP2A, AHR, or CLDN1 confirmed that keratinocyte terminal differentiation is the major parameter defining EISdiff. Exposure to proinflammatory psoriasis- or atopic dermatitis-associated cytokine cocktails lowered the expression of keratinocyte differentiation markers and reduced EISdiff. This cytokine-associated decrease in EISdiff was normalized after stimulation with therapeutic molecules. In conclusion, EIS provides a noninvasive system to consecutively and quantitatively assess HEE barrier function and to sensitively and objectively measure barrier development, defects, and repair.
Collapse
Affiliation(s)
| | - Felicitas Pardow
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands; Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Luca D Meesters
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands; Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | | | | | - Hanna Niehues
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
| | | | | | - Matthew G Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Jos P H Smits
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands; Department of Dermatology, Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Rousel J, Mergen C, Bergmans ME, Klarenbeek NB, der Kolk TNV, van Doorn MBA, Bouwstra JA, Rissmann R. Lesional Psoriasis is Associated With Alterations in the Stratum Corneum Ceramide Profile and Concomitant Decreases in Barrier Function. Exp Dermatol 2024; 33:e15185. [PMID: 39382258 DOI: 10.1111/exd.15185] [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: 05/22/2024] [Revised: 09/09/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Psoriasis is an inflammatory skin disease associated with an impaired skin barrier. The skin barrier function is dependent on the extracellular lipid matrix which surrounds the corneocytes in the stratum corneum. Ceramides comprise essential components of this matrix. Alterations in the stratum corneum ceramide profile have been directly linked to barrier dysfunction and might be an underlying factor of the barrier impairment in psoriasis. In this study, we investigated the ceramide profile and barrier function in psoriasis. Lesional and non-lesional skin of 26 patients and 10 healthy controls were analysed using in-depth ceramide lipidomics by liquid chromatography-mass spectrometry. Barrier function was assessed by measuring transepidermal water loss. Lesional skin showed a significant decrease in the abundance of total ceramides with significant alterations in the ceramide subclass composition compared to control and non-lesional skin. Additionally, the percentage of monounsaturated ceramides was significantly increased, and the average ceramide chain length significantly decreased in lesional skin. Altogether, this resulted in a markedly different profile compared to controls for lesional skin, but not for non-lesional skin. Importantly, the reduced barrier function in lesional psoriasis correlated to alterations in the ceramide profile, highlighting their interdependence. By assessing the parameters 2 weeks apart, we are able to highlight the reproducibility of these findings, which further affirms this connection. To conclude, we show that changes in the ceramide profile and barrier impairment are observed in, and limited to, lesional psoriatic skin. Their direct correlation provides a further mechanistic basis for the concomitantly observed impairment of barrier dysfunction.
Collapse
Affiliation(s)
- Jannik Rousel
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherine Mergen
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Menthe E Bergmans
- Centre for Human Drug Research, Leiden, The Netherlands
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Martijn B A van Doorn
- Centre for Human Drug Research, Leiden, The Netherlands
- Department of Dermatology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Joke A Bouwstra
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Robert Rissmann
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
7
|
Tsai YY, Chen YJ, Chang LS, Wu CC. Skin colonization by Staphylococcus aureus in hemodialysis patients with pruritus and the effect of Staphylococcus aureus-secreted α-toxin on filaggrin expression. J Dermatol 2024; 51:1318-1328. [PMID: 38894607 DOI: 10.1111/1346-8138.17326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/18/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024]
Abstract
Staphylococcus aureus (S. aureus) commonly reside on human skin in residents in long-term care facilities, yet its colonization and impact on the skin of hemodialysis (HD) patients have yet to be studied. The aim of the present study was to investigate the colonization of S. aureus on the skin of pruritic and non-pruritic HD patients, and the influence of S. aureus and S. aureus-secreted α-toxin on skin barrier function-related protein expression. In this study, a higher relative S. aureus count in pruritic HD patients compared to non-pruritic HD patients and healthy subjects were revealed by real-time polymerase chain reaction. S. aureus and α-toxin decreased mRNA and protein expression levels of aryl hydrocarbon receptor (AHR), ovo-like transcriptional repressor 1 (OVOL1), and filaggrin (FLG) in keratinocytes. In addition, anti-alpha-hemolysin (anti-hla) was used as an α-toxin neutralizer, and it successfully abrogated S. aureus-induced AHR, OVOL1, and FLG mRNA and protein expression downregulation. Mechanistically, α-toxin could decrease FLG activity by preventing the recruitment of AHR to the FLG promoter region. In conclusion, pruritic HD patients had higher S. aureus colonization, with S. aureus-secreted α-toxin suppressing FLG expression through the AHR-FLG axis.
Collapse
Affiliation(s)
- Yen-Yu Tsai
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ying-Jung Chen
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Cheng-Ching Wu
- Division of Cardiology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, E-Da Cancer Hospital, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Ferrarese L, Koch M, Baumann A, Bento-Lopes L, Wüst D, Berest I, Kopf M, Werner S. Inflammatory Mediators Suppress FGFR2 Expression in Human Keratinocytes to Promote Inflammation. Mol Cell Biol 2024; 44:489-504. [PMID: 39340759 PMCID: PMC11529413 DOI: 10.1080/10985549.2024.2399766] [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: 05/30/2024] [Revised: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Fibroblast growth factors (FGFs) are key orchestrators of development, tissue homeostasis and repair. FGF receptor (FGFR) deficiency in mouse keratinocytes causes an inflammatory skin phenotype with similarities to atopic dermatitis, but the human relevance is unclear. Therefore, we generated human keratinocytes with a CRISPR/Cas9-induced knockout of FGFR2. Loss of this receptor promoted the expression of interferon-stimulated genes and pro-inflammatory cytokines under homeostatic conditions and in particular in response to different inflammatory mediators. Expression of FGFR2 itself was strongly downregulated in cultured human keratinocytes exposed to various pro-inflammatory stimuli. This is relevant in vivo, because bioinformatics analysis of bulk and single-cell RNA-seq data showed strongly reduced expression of FGFR2 in lesional skin of atopic dermatitis patients, which likely aggravates the inflammatory phenotype. These results reveal a key function of FGFR2 in human keratinocytes in the suppression of inflammation and suggest a role of FGFR2 downregulation in the pathogenesis of atopic dermatitis and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Luca Ferrarese
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Michael Koch
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Artemis Baumann
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Liliana Bento-Lopes
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Daria Wüst
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Ivan Berest
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
9
|
Panganiban RA, Nadeau KC, Lu Q. Pyroptosis, gasdermins and allergic diseases. Allergy 2024; 79:2380-2395. [PMID: 39003568 PMCID: PMC11368650 DOI: 10.1111/all.16236] [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: 03/05/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Pyroptosis is an inflammatory form of programmed cell death that is distinct from necrosis and apoptosis. Pyroptosis is primarily mediated by the gasdermin family of proteins (GSDMA-E and PVJK), which, when activated by proteolytic cleavage, form pores in the plasma membrane, leading to cell death. While much of the past research on pyroptosis has focused on its role in cancer, metabolic disorders, and infectious diseases, recent experimental and observational studies have begun to implicate pyroptosis in allergic diseases. These studies suggest that gasdermin-mediated pyroptosis contributes to the development of allergic conditions and could offer novel targets for therapy. Here, we review our current understanding of pyroptosis with an emphasis on the role of gasdermins as executioners of pyroptosis and potential mediators to allergic disease. We highlight new discoveries that establish a mechanistic link between the biochemical actions of gasdermins and the onset of allergic diseases. Additionally, we discuss how pyroptosis and gasdermins might contribute to the dysfunction of epithelial barrier, a key factor believed to initiate the progression of various allergic diseases.
Collapse
Affiliation(s)
- Ronald Allan Panganiban
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Yamamoto-Hanada K, Ohya Y. Management of Infant Atopic Eczema to Prevent Severe Eczema and Food Allergy. Clin Exp Allergy 2024; 54:669-681. [PMID: 38866599 DOI: 10.1111/cea.14515] [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: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024]
Abstract
Early intervention and active management of infant atopic eczema may play a crucial role in limiting eczema severity and preventing the onset of immediate-type food allergy. Eczema management involves education, skincare and medications targeting skin inflammation and barrier repair. Topical corticosteroids are the mainstay of anti-inflammatory therapy, with nonsteroidal options available for some infants. Proactive therapy, addressing subclinical inflammation, is useful for preventing eczema flares, especially in infants with recurrent eczema flares despite reactive therapy. In clinical practice, holistic consideration of overall infant and family health is essential. Providing advice on maternal stress management, nutritional guidance and recommendations for proper sleep and lifestyle is crucial for the well-being of children and their families, not limited to eczema treatment alone.
Collapse
Affiliation(s)
| | - Yukihiro Ohya
- Allergy Center, National Center for Child Health and Development, Tokyo, Japan
- Department of Occupational and Environmental Health, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| |
Collapse
|
11
|
Kenney HM, Battaglia J, Herman K, Beck LA. Atopic dermatitis and IgE-mediated food allergy: Common biologic targets for therapy and prevention. Ann Allergy Asthma Immunol 2024; 133:262-277. [PMID: 38908432 DOI: 10.1016/j.anai.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE To highlight common mechanistic targets for the treatment of atopic dermatitis (AD) and IgE-mediated food allergy (IgE-FA) with potential to be effective for both diseases and prevent atopic progression. DATA SOURCES Data sources were PubMed searches or National Clinical Trials (NCT)-registered clinical trials related to AD, IgE-FA, and other atopic conditions, especially focused on the pediatric population. STUDY SELECTIONS Human seminal studies and/or articles published in the past decade were emphasized with reference to preclinical models when relevant. NCT-registered clinical trials were filtered by inclusion of pediatric subjects younger than 18 years with special focus on children younger than 12 years as a critical period when AD and IgE-FA diseases may often be concurrent. RESULTS AD and IgE-FA share several pathophysiologic features, including epithelial barrier dysfunction, innate and adaptive immune abnormalities, and microbial dysbiosis, which may be critical for the clinical progression between these diseases. Revolutionary advances in targeted biologic therapies have shown the benefit of inhibiting type 2 immune responses, using dupilumab (anti-interleukin-4Rα) or omalizumab (anti-IgE), to potentially reduce symptom burden for both diseases in pediatric populations. Although the potential for biologics to promote disease remission (AD) or sustained unresponsiveness (IgE-FA) remains unclear, the refinement of biomarkers to predict infants at risk for atopic disorders provides promise for prevention through timely intervention. CONCLUSION AD and IgE-FA exhibit common features that may be leveraged to develop biologic therapeutic strategies to treat both conditions and even prevent atopic progression. Future studies should be designed with consistent age stratification in the pediatric population and standardized regimens of adjuvant oral immunotherapy or dose escalation (IgE-FA) to improve cross-study interpretation.
Collapse
Affiliation(s)
- H Mark Kenney
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jennifer Battaglia
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - Katherine Herman
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; Division of Allergy and Immunology, University of Rochester Medical Center, Rochester, New York
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
12
|
Pan Y, Hochgerner M, Cichoń MA, Benezeder T, Bieber T, Wolf P. Langerhans cells: Central players in the pathophysiology of atopic dermatitis. J Eur Acad Dermatol Venereol 2024. [PMID: 39157943 DOI: 10.1111/jdv.20291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease worldwide. AD is a highly complex disease with different subtypes. Many elements of AD pathophysiology have been described, but if/how they interact with each other or which mechanisms are important in which patients is still unclear. Langerhans cells (LCs) are antigen-presenting cells (APCs) in the epidermis. Depending on the context, they can act either pro- or anti-inflammatory. Many different studies have investigated LCs in the context of AD and found them to be connected to all major mechanisms of AD pathophysiology. As APCs, LCs recruit other immune cells and shape the immune response, especially adaptive immunity via polarization of T cells. As sentinel cells, LCs are primary sensors of the skin microbiome and are important for the decision of immunity versus tolerance. LCs are also involved with the integrity of the skin barrier by influencing tight junctions. Finally, LCs are important cells in the neuro-immune crosstalk in the skin. In this review, we provide an overview about the many different roles of LCs in AD. Understanding LCs might bring us closer to a more complete understanding of this highly complex disease. Potentially, modulating LCs might offer new options for targeted therapies for AD patients.
Collapse
Affiliation(s)
- Yi Pan
- Department of Dermatology and Allergy, University Hospital of Bonn, Bonn, Germany
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Mathias Hochgerner
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | | | - Theresa Benezeder
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Thomas Bieber
- Department of Dermatology and Allergy, University Hospital of Bonn, Bonn, Germany
- CK-CARE, Medicine Campus, Davos, Switzerland
- Department of Dermatology, University Hospital of Zürich, Zürich, Switzerland
| | - Peter Wolf
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| |
Collapse
|
13
|
Braun C, Badiou C, Guironnet-Paquet A, Iwata M, Lenief V, Mosnier A, Beauclair C, Renucci E, Bouschon P, Cuzin R, Briend Y, Patra V, Patot S, Scharschmidt TC, van Wamel W, Lemmens N, Nakajima S, Vandenesh F, Nicolas JF, Lina G, Nosbaum A, Vocanson M. Staphylococcus aureus-specific skin resident memory T cells protect against bacteria colonization but exacerbate atopic dermatitis-like flares in mice. J Allergy Clin Immunol 2024; 154:355-374. [PMID: 38734386 DOI: 10.1016/j.jaci.2024.03.032] [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: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/26/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The contribution of Staphylococcus aureus to the exacerbation of atopic dermatitis (AD) is widely documented, but its role as a primary trigger of AD skin symptoms remains poorly explored. OBJECTIVES This study sought to reappraise the main bacterial factors and underlying immune mechanisms by which S aureus triggers AD-like inflammation. METHODS This study capitalized on a preclinical model, in which different clinical isolates were applied in the absence of any prior experimental skin injury. RESULTS The development of S aureus-induced dermatitis depended on the nature of the S aureus strain, its viability, the concentration of the applied bacterial suspension, the production of secreted and nonsecreted factors, as well as the activation of accessory gene regulatory quorum sensing system. In addition, the rising dermatitis, which exhibited the well-documented AD cytokine signature, was significantly inhibited in inflammasome adaptor apoptosis-associated speck-like protein containing a CARD domain- and monocyte/macrophage-deficient animals, but not in T- and B-cell-deficient mice, suggesting a major role for the innate response in the induction of skin inflammation. However, bacterial exposure generated a robust adaptive immune response against S aureus, and an accumulation of S aureus-specific γδ and CD4+ tissue resident memory T cells at the site of previous dermatitis. The latter both contributed to worsen the flares of AD-like dermatitis on new bacteria exposures, but also, protected the mice from persistent bacterial colonization. CONCLUSIONS These data highlight the induction of unique AD-like inflammation, with the generation of proinflammatory but protective tissue resident memory T cells in a context of natural exposure to pathogenic S aureus strains.
Collapse
Affiliation(s)
- Camille Braun
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Pédiatrie, Pneumologie, Allergologie, Mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Cédric Badiou
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Aurélie Guironnet-Paquet
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Etablissement Français du Sang Auvergne Rhône-Alpes, Apheresis Unit, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Masashi Iwata
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Vanina Lenief
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Amandine Mosnier
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Charlotte Beauclair
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Emilie Renucci
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Pauline Bouschon
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Roxane Cuzin
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Yoann Briend
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Vijaykumar Patra
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Sabine Patot
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Willem van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicole Lemmens
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - François Vandenesh
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Jean-François Nicolas
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Gérard Lina
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Audrey Nosbaum
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
14
|
Rousel J, Mergen C, Bergmans ME, Bruijnincx LJ, de Kam ML, Klarenbeek NB, Niemeyer-van der Kolk T, van Doorn MBA, Bouwstra JA, Rissmann R. Guselkumab treatment normalizes the stratum corneum ceramide profile and alleviates barrier dysfunction in psoriasis: results of a randomized controlled trial. J Lipid Res 2024; 65:100591. [PMID: 38992724 PMCID: PMC11342092 DOI: 10.1016/j.jlr.2024.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/15/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
The epidermal inflammation associated with psoriasis drives skin barrier perturbations. The skin barrier is primarily located in stratum corneum (SC). Its function depends on the SC lipid matrix of which ceramides constitute important components. Changes in the ceramide profile directly correlate to barrier function. In this study, we characterized the dynamics of the barrier function and ceramide profile of psoriatic skin during anti-Interleukin-23 therapy with guselkumab. We conducted a double-blind, randomized controlled trial in which 26 mild-to-severe plaque psoriasis patients were randomization 3:1-100 mg guselkumab or placebo for 16 weeks and barrier dynamics monitored throughout. Barrier function was measured by trans-epidermal water loss measurements. Untargeted ceramide profiling was performed using liquid chromatography-mass spectrometry after SC was harvested using tape-stripping. The barrier function and ceramide profile of lesional skin normalized to that of controls during treatment with guselkumab, but not placebo. This resulted in significant differences compared to placebo at the end of the treatment. Changes in the lesional ceramide profile during treatment correlated with barrier function and target lesion severity. Nonlesional skin remained similar throughout treatment. Guselkumab therapy restored the skin barrier in psoriasis. Concomitant correlations between skin barrier function, the ceramide profile, and disease severity demonstrate their interdependency.
Collapse
Affiliation(s)
- Jannik Rousel
- Centre for Human Drug Research, Leiden, The Netherlands; Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Catherine Mergen
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Menthe E Bergmans
- Centre for Human Drug Research, Leiden, The Netherlands; Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | - Martijn B A van Doorn
- Centre for Human Drug Research, Leiden, The Netherlands; Department of Dermatology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Joke A Bouwstra
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Robert Rissmann
- Centre for Human Drug Research, Leiden, The Netherlands; Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands; Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
15
|
Nicolaou A, Kendall AC. Bioactive lipids in the skin barrier mediate its functionality in health and disease. Pharmacol Ther 2024; 260:108681. [PMID: 38897295 DOI: 10.1016/j.pharmthera.2024.108681] [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: 02/05/2024] [Revised: 05/11/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Our skin protects us from external threats including ultraviolet radiation, pathogens and chemicals, and prevents excessive trans-epidermal water loss. These varied activities are reliant on a vast array of lipids, many of which are unique to skin, and that support physical, microbiological and immunological barriers. The cutaneous physical barrier is dependent on a specific lipid matrix that surrounds terminally-differentiated keratinocytes in the stratum corneum. Sebum- and keratinocyte-derived lipids cover the skin's surface and support and regulate the skin microbiota. Meanwhile, lipids signal between resident and infiltrating cutaneous immune cells, driving inflammation and its resolution in response to pathogens and other threats. Lipids of particular importance include ceramides, which are crucial for stratum corneum lipid matrix formation and therefore physical barrier functionality, fatty acids, which contribute to the acidic pH of the skin surface and regulate the microbiota, as well as the stratum corneum lipid matrix, and bioactive metabolites of these fatty acids, involved in cell signalling, inflammation, and numerous other cutaneous processes. These diverse and complex lipids maintain homeostasis in healthy skin, and are implicated in many cutaneous diseases, as well as unrelated systemic conditions with skin manifestations, and processes such as ageing. Lipids also contribute to the gut-skin axis, signalling between the two barrier sites. Therefore, skin lipids provide a valuable resource for exploration of healthy cutaneous processes, local and systemic disease development and progression, and accessible biomarker discovery for systemic disease, as well as an opportunity to fully understand the relationship between the host and the skin microbiota. Investigation of skin lipids could provide diagnostic and prognostic biomarkers, and help identify new targets for interventions. Development and improvement of existing in vitro and in silico approaches to explore the cutaneous lipidome, as well as advances in skin lipidomics technologies, will facilitate ongoing progress in skin lipid research.
Collapse
Affiliation(s)
- Anna Nicolaou
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK; Lydia Becker Institute of Immunology and Inflammation; Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK.
| | - Alexandra C Kendall
- Laboratory for Lipidomics and Lipid Biology, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9NT, UK
| |
Collapse
|
16
|
Meng Y, Liu Y, Guo J, Guo X, Han X, Zhang L, Di T, Zhao J, Wang Y, Li P. Qing-Re-Chu-shi decoction ameliorates 2,4-dinitrochlorobenzene-induced atopic dermatitis in NC/Nga mice through anti-inflammation and immunoregulatory mechanisms. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117702. [PMID: 38176665 DOI: 10.1016/j.jep.2024.117702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing-Re-Chu-Shi Decoction (QRCSD), a traditional Chinese herbal formula, has been employed as a complementary and alternative therapy for inflammatory skin diseases. However, its active constituents and the mechanistic basis of its action on atopic dermatitis remain in adequately understood. AIM OF THE STUDY Atopic dermatitis (AD) is an allergic dermatitis marked by eczematous lesions and pruritus. The study aimed to elucidate the underlying effects of QRCSD on AD and to identify the components responsible for its therapeutic efficacy in a mouse model. MATERIALS AND METHODS Network pharmacology and UPLC-mass analysis were used to anticipate the pharmacological mechanisms and to identify active components of QRCSD, respectively. A DNCB-induced AD-like model was established in NC/Nga mice. QRCSD or prednisolone (as a positive control) was administered via gavage every other day from day14 to day 21. Dermatitis severity score, scratching behavior, skin barrier function, spleen index, Th1/Th2 lymphocyte ratio, and serum IgE levels were evaluated. Protein arrays, including 40 inflammatory cytokines, were performed on skin lesions, followed by confirmation experiments of Western blotting in dorsal skin lesions. RESULTS The construction of a QRCSD-AD-Network and topological analysis firstly proposed potential targets of QRCSD acting on AD. Animal experiments demonstrated that oral administration of QRCSD ameliorated AD-like lesions, reduced epidermal thickness and mast cell count, decreased serum IgE levels, augmented tight junction protein (Claudin 1, Occludin) levels, and regulated the Th1/Th2 balance in the spleen, as well as spleen index. Elevated levels of interleukin (IL)-4, IL-5, IL-6, IL-17, and Eotaxin were revealed in AD-like skin lesions by protein arrays. Western blotting confirmed that the phosphorylation levels of ERK, P38, JNK, STAT3 and P65 were downregulated, and IL-6 expression was also reduced following QRCSD treatment. CONCLUSIONS The study enhances the understanding of the anti-inflammatory and immunomodulatory effects of QRCSD, showcasing its significant protective role against atopic dermatitis. Treatment with QRCSD may be considered as a viable candidate for complementary and alternative therapy in managing atopic dermatitis.
Collapse
Affiliation(s)
- YuJiao Meng
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Jianning Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Xiaoyao Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Xuyang Han
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Lu Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Tingting Di
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing Key Laboratory of Clinic and Basic Research with Traditional Chinese Medicine on Psoriasis (BZ0375), Beijing, 100010, China; Beijing University of Chinese Medicine, Beijing, 100105, China.
| |
Collapse
|
17
|
Luo M, Zheng Y, Zhuo Q, Lin L, Han Y. The causal effects of atopic dermatitis on the risk of skin cancers: A two-sample Mendelian randomization study. J Eur Acad Dermatol Venereol 2024; 38:703-709. [PMID: 38009387 DOI: 10.1111/jdv.19674] [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: 07/14/2023] [Accepted: 10/20/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Observational and epidemiological studies show conflicting results on the relationship between atopic dermatitis and skin cancer. Additionally, observational studies are susceptible to the reverse causation and confounders, thus, may not interpret true causal relationships. The causal effects of atopic dermatitis on the risk of skin cancers remains unclear. OBJECTIVES To investigate the causal relationship between atopic dermatitis and skin cancer including cutaneous malignant melanoma, cutaneous squamous cell carcinoma, basal cell carcinoma and actinic keratosis. METHODS We performed a two-sample Mendelian randomization analysis based on summary datasets of public genome-wide association studies of European ancestry. The inverse variance-weighted approach was applied as the main analysis. MR-Egger and weighted median methods were used to complement the inverse variance-weighted results. A series of sensitivity analyses were used to ensure the robustness of the causality estimates. RESULTS Inverse variance-weighted method showed that genetically predicted dermatitis patients were significantly associated with an increased incidence of basal cell carcinoma (OR, 1.20; 95% CI, 1.10-1.31; p = 4.07E-05) and cutaneous squamous cell carcinoma (OR, 1.14; 95% CI, 1.10-1.19; p = 1.05E-11). However, we did not find a significant causality for atopic dermatitis on melanoma neither did we find actinic keratosis. Subsequent sensitive analyses supported these results. CONCLUSIONS Our study identified the causality between atopic dermatitis basal cell carcinoma and squamous cell carcinoma. Accordingly, regular skin cancer screening is recommended for patients with atopic dermatitis.
Collapse
Affiliation(s)
- Min Luo
- Department of Dermatology, The Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yaxuan Zheng
- Department of Dermatology, The Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qianwei Zhuo
- Department of Dermatology, The Union Hospital, Fujian Medical University, Fuzhou, China
| | - Lihang Lin
- Department of Dermatology, The Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yue Han
- Department of Dermatology, The Union Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
18
|
van den Brink NJM, Pardow F, Meesters LD, van Vlijmen-Willems I, Rodijk-Olthuis D, Niehues H, Jansen PAM, Roelofs SH, Brewer MG, van den Bogaard EH, Smits JPH. Electrical Impedance Spectroscopy Quantifies Skin Barrier Function in Organotypic In Vitro Epidermis Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.18.585587. [PMID: 38562885 PMCID: PMC10983962 DOI: 10.1101/2024.03.18.585587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
3 D human epidermal equivalents (HEEs) are a state-of-the-art organotypic culture model in pre-clinical investigative dermatology and regulatory toxicology. Here, we investigated the utility of electrical impedance spectroscopy (EIS) for non-invasive measurement of HEE epidermal barrier function. Our setup comprised a custom-made lid fit with 12 electrode pairs aligned on the standard 24-transwell cell culture system. Serial EIS measurements for seven consecutive days did not impact epidermal morphology and readouts showed comparable trends to HEEs measured only once. We determined two frequency ranges in the resulting impedance spectra: a lower frequency range termed EISdiff correlated with keratinocyte terminal differentiation independent of epidermal thickness and a higher frequency range termed EISSC correlated with stratum corneum thickness. HEEs generated from CRISPR/Cas9 engineered keratinocytes that lack key differentiation genes FLG, TFAP2A, AHR or CLDN1 confirmed that keratinocyte terminal differentiation is the major parameter defining EISdiff. Exposure to pro-inflammatory psoriasis- or atopic dermatitis-associated cytokine cocktails lowered the expression of keratinocyte differentiation markers and reduced EISdiff. This cytokine-associated decrease in EISdiff was normalized after stimulation with therapeutic molecules. In conclusion, EIS provides a non-invasive system to consecutively and quantitatively assess HEE barrier function and to sensitively and objectively measure barrier development, defects and repair.
Collapse
Affiliation(s)
| | - F Pardow
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - L D Meesters
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | | | - D Rodijk-Olthuis
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
| | - H Niehues
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
| | - P A M Jansen
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
| | | | - M G Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - J P H Smits
- Department of Dermatology, Radboudumc, Nijmegen, The Netherlands
- Department of Dermatology, Heinrich Heine University, University Hospital Düsseldorf , Düsseldorf, Germany
| |
Collapse
|
19
|
Chu DK, Schneider L, Asiniwasis RN, Boguniewicz M, De Benedetto A, Ellison K, Frazier WT, Greenhawt M, Huynh J, Kim E, LeBovidge J, Lind ML, Lio P, Martin SA, O'Brien M, Ong PY, Silverberg JI, Spergel JM, Wang J, Wheeler KE, Guyatt GH, Capozza K, Begolka WS, Chu AWL, Zhao IX, Chen L, Oykhman P, Bakaa L, Golden D, Shaker M, Bernstein JA, Greenhawt M, Horner CC, Lieberman J, Stukus D, Rank MA, Wang J, Ellis A, Abrams E, Ledford D, Chu DK. Atopic dermatitis (eczema) guidelines: 2023 American Academy of Allergy, Asthma and Immunology/American College of Allergy, Asthma and Immunology Joint Task Force on Practice Parameters GRADE- and Institute of Medicine-based recommendations. Ann Allergy Asthma Immunol 2024; 132:274-312. [PMID: 38108679 DOI: 10.1016/j.anai.2023.11.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Guidance addressing atopic dermatitis (AD) management, last issued in 2012 by the American Academy of Allergy, Asthma and Immunology/American College of Allergy, Asthma and Immunology Joint Task Force, requires updating as a result of new treatments and improved guideline and evidence synthesis methodology. OBJECTIVE To produce evidence-based guidelines that support patients, clinicians, and other decision-makers in the optimal treatment of AD. METHODS A multidisciplinary guideline panel consisting of patients and caregivers, AD experts (dermatology and allergy/immunology), primary care practitioners (family medicine, pediatrics, internal medicine), and allied health professionals (psychology, pharmacy, nursing) convened, prioritized equity, diversity, and inclusiveness, and implemented management strategies to minimize influence of conflicts of interest. The Evidence in Allergy Group supported guideline development by performing systematic evidence reviews, facilitating guideline processes, and holding focus groups with patient and family partners. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach informed rating the certainty of evidence and strength of recommendations. Evidence-to-decision frameworks, subjected to public comment, translated evidence to recommendations using trustworthy guideline principles. RESULTS The panel agreed on 25 recommendations to gain and maintain control of AD for patients with mild, moderate, and severe AD. The eAppendix provides practical information and implementation considerations in 1-2 page patient-friendly handouts. CONCLUSION These evidence-based recommendations address optimal use of (1) topical treatments (barrier moisturization devices, corticosteroids, calcineurin inhibitors, PDE4 inhibitors [crisaborole], topical JAK inhibitors, occlusive [wet wrap] therapy, adjunctive antimicrobials, application frequency, maintenance therapy), (2) dilute bleach baths, (3) dietary avoidance/elimination, (4) allergen immunotherapy, and (5) systemic treatments (biologics/monoclonal antibodies, small molecule immunosuppressants [cyclosporine, methotrexate, azathioprine, mycophenolate, JAK inhibitors], and systemic corticosteroids) and UV phototherapy (light therapy).
Collapse
Affiliation(s)
- Derek K Chu
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada.
| | - Lynda Schneider
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts.
| | | | - Mark Boguniewicz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; Division of Pediatric Allergy and Clinical Immunology, National Jewish Health, Denver, Colorado
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York
| | | | - Winfred T Frazier
- Department of Family Medicine, UPMC St. Margaret, Pittsburgh, Pennsylvania
| | - Matthew Greenhawt
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; Section of Allergy and Immunology, Children's Hospital Colorado, Aurora, Colorado
| | - Joey Huynh
- Sepulveda VA Medical Center, North Hills, California
| | | | - Jennifer LeBovidge
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Mary Laura Lind
- School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Arizona
| | - Peter Lio
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stephen A Martin
- University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Monica O'Brien
- Tufts University School of Medicine, Boston, Massachusetts
| | - Peck Y Ong
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, USC Keck School of Medicine, Los Angeles, California
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Jonathan M Spergel
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Julie Wang
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Kathryn E Wheeler
- Department of Pediatrics, University of Florida, Gainesville, Florida
| | - Gordon H Guyatt
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - Korey Capozza
- Global Parents for Eczema Research, Santa Barbara, California
| | | | - Alexandro W L Chu
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - Irene X Zhao
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - Lina Chen
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - Paul Oykhman
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - Layla Bakaa
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| | - David Golden
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Marcus Shaker
- Dartmouth Geisel School of Medicine and Dartmouth Hitchcock Medical Center, Section of Allergy, Lebanon, New Hampshire
| | | | - Matthew Greenhawt
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado; Section of Allergy and Immunology, Children's Hospital Colorado, Aurora, Colorado
| | - Caroline C Horner
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Jay Lieberman
- University of Tennessee Health Science Center and LeBonheur Children's Hospital, Memphis, Tennessee
| | - David Stukus
- Nationwide Children's Hospital and Ohio State University College of Medicine, Columbus, Ohio
| | - Matthew A Rank
- Mayo Clinic in Arizona and Phoenix Children's Hospital, Scottsdale and Phoenix, Arizona
| | - Julie Wang
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Anne Ellis
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Elissa Abrams
- Section of Allergy and Clinical Immunology, Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Dennis Ledford
- Morsani College of Medicine, University of South Florida and James A. Haley Veterans' Affairs Hospital, Tampa, Florida
| | - Derek K Chu
- Departments of Medicine and Health Research Methods, Evidence & Impact, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, Canada
| |
Collapse
|
20
|
Munera-Campos M, Carrascosa JM. Atopic Dermatitis: Fertility, Pregnancy, and Treatment Perspectives. Am J Clin Dermatol 2024; 25:55-66. [PMID: 37904055 DOI: 10.1007/s40257-023-00821-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/01/2023]
Abstract
Hormonal and immunologic changes during pregnancy can contribute to the development of different dermatoses, the most common of which is atopic eruption of pregnancy (AEP). Of atopic dermatitis (AD) cases during pregnancy, 80% are new-onset presentations, while 20% represent recurrences or exacerbations of preexisting disease. Evidence on the effects of previous AD on fertility is limited. Different factors influence women's desire to conceive in this setting, and it has been hypothesized that barrier defects and systemic inflammation could contribute to biologic infertility, although more data are needed. Clinical practice suggests a tendency toward undertreatment in pregnant woman due to concerns about potential effects on obstetric and fetal outcomes. However, pregnant women should be offered adequate and safe treatments, preferably on an individual basis. The aim of this review was to summarize the evidence on disease course in pregnant women with AD and the challenges associated with its diagnosis and management. We also review the current evidence on the use of conventional and novel systemic therapies for AD in this population.
Collapse
Affiliation(s)
- Mónica Munera-Campos
- Department of Dermatology, Germans Trias i Pujol University Hospital, Autonomous University of Barcelona, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain.
| | - Jose Manuel Carrascosa
- Department of Dermatology, Germans Trias i Pujol University Hospital, Autonomous University of Barcelona, Germans Trias i Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| |
Collapse
|
21
|
Luo M, Su HC, Lin JE, Zhu CH, Lin LH, Han Y. A Retrospective Analysis of Risk Factors for Atopic Dermatitis Severity. Dermatitis 2024; 35:S81-S90. [PMID: 37126941 DOI: 10.1089/derm.2023.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Background: Atopic dermatitis (AD) has the highest burden of any skin disease; however, the severity-associated factors remain unclear. Objective: To evaluate potential severity-associated factors of AD and to design and validate a severity prediction model to inform the management of AD patients. Methods: A cross-sectional study of 900 AD patients was conducted from December 2021 to October 2022 at our hospital. The primary outcome was disease severity, categorized as mild, moderate, or severe using the scoring atopic dermatitis index. Ordinal logistic regression and bootstrapped validation were used to derive and internally validate the model. Results: Increasing age, elevated eosinophil level, higher economic status, and urban residence were associated with severe AD. Breastfeeding, disinfectants and topical emollients use, and short duration of bathing were associated with mild AD. In the prediction model, predictors included age, eosinophil and economic status, residence, feeding, disinfectants and emollients use, and duration of bathing. Prediction models demonstrated good discrimination (bias-corrected concordance index [c-index] = 0.72) and good calibration. Conclusion: Risk factors for the severity of AD were identified that could aid the early prediction of AD progression. The predictive model included variables that are easily evaluated and could inform personalized prevention and therapy.
Collapse
Affiliation(s)
- Min Luo
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Huichun C Su
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Jinger E Lin
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Changhua H Zhu
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Lihang H Lin
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| | - Yue Han
- From the Department of Dermatology, the Union Hospital, Fujian Medical University, Fuzhou, P.R. China
| |
Collapse
|
22
|
Mrkić Kobal I, Plavec D, Vlašić Lončarić Ž, Jerković I, Turkalj M. Atopic March or Atopic Multimorbidity-Overview of Current Research. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:21. [PMID: 38256282 PMCID: PMC10819021 DOI: 10.3390/medicina60010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024]
Abstract
The atopic march encompasses a sequence of allergic conditions, including atopic dermatitis, food allergy, allergic rhinitis, and asthma, that frequently develop in a sequential pattern within the same individual. It was introduced as a conceptual framework aimed at elucidating the developmental trajectory of allergic conditions during childhood. Following the introduction of this concept, it was initially believed that the atopic march represented the sole and definitive trajectory of the development of allergic diseases. However, this perspective evolved with the emergence of new longitudinal studies, which revealed that the evolution of allergic diseases is far more intricate. It involves numerous immunological pathological mechanisms and may not align entirely with the traditional concept of the atopic march. The objective of our review is to portray the atopic march alongside other patterns in the development of childhood allergic diseases, with a specific emphasis on the potential for a personalized approach to the prevention, diagnosis, and treatment of atopic conditions.
Collapse
Affiliation(s)
- Iva Mrkić Kobal
- Clinic for Pediatric Medicine Helena, Ulica kneza Branimira 71, 10000 Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
| | - Davor Plavec
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
- Prima Nova, Zagrebačka cesta 132a, 10000 Zagreb, Croatia
| | - Željka Vlašić Lončarić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
- Children’s Hospital Srebrnjak, Srebrnjak 100, 10000 Zagreb, Croatia
| | - Ivana Jerković
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
- Children’s Hospital Srebrnjak, Srebrnjak 100, 10000 Zagreb, Croatia
| | - Mirjana Turkalj
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Josipa Huttlera 4, 31000 Osijek, Croatia
- Children’s Hospital Srebrnjak, Srebrnjak 100, 10000 Zagreb, Croatia
- Faculty of Medicine, Catholic University of Croatia, Ilica 242, 10000 Zagreb, Croatia
| |
Collapse
|
23
|
Chu AWL, Wong MM, Rayner DG, Guyatt GH, Díaz Martinez JP, Ceccacci R, Zhao IX, McMullen E, Srivastava A, Wang J, Wen A, Wang FC, Brignardello-Petersen R, Izcovich A, Oykhman P, Wheeler KE, Wang J, Spergel JM, Singh JA, Silverberg JI, Ong PY, O'Brien M, Martin SA, Lio PA, Lind ML, LeBovidge J, Kim E, Huynh J, Greenhawt M, Gardner DD, Frazier WT, Ellison K, Chen L, Capozza K, De Benedetto A, Boguniewicz M, Smith Begolka W, Asiniwasis RN, Schneider LC, Chu DK. Systemic treatments for atopic dermatitis (eczema): Systematic review and network meta-analysis of randomized trials. J Allergy Clin Immunol 2023; 152:1470-1492. [PMID: 37678577 DOI: 10.1016/j.jaci.2023.08.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/06/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is an inflammatory skin condition with multiple systemic treatments and uncertainty regarding their comparative impact on AD outcomes. OBJECTIVE We sought to systematically synthesize the benefits and harms of AD systemic treatments. METHODS For the 2023 American Academy of Allergy, Asthma & Immunology and American College of Allergy, Asthma, and Immunology Joint Task Force on Practice Parameters AD guidelines, we searched MEDLINE, EMBASE, CENTRAL, Web of Science, and GREAT databases from inception to November 29, 2022, for randomized trials addressing systemic treatments and phototherapy for AD. Paired reviewers independently screened records, extracted data, and assessed risk of bias. Random-effects network meta-analyses addressed AD severity, itch, sleep, AD-related quality of life, flares, and harms. The Grading of Recommendations Assessment, Development and Evaluation approach informed certainty of evidence ratings. This review is registered in the Open Science Framework (https://osf.io/e5sna). RESULTS The 149 included trials (28,686 patients with moderate-to-severe AD) evaluated 75 interventions. With high-certainty evidence, high-dose upadacitinib was among the most effective for 5 of 6 patient-important outcomes; high-dose abrocitinib and low-dose upadacitinib were among the most effective for 2 outcomes. These Janus kinase inhibitors were among the most harmful in increasing adverse events. With high-certainty evidence, dupilumab, lebrikizumab, and tralokinumab were of intermediate effectiveness and among the safest, modestly increasing conjunctivitis. Low-dose baricitinib was among the least effective. Efficacy and safety of azathioprine, oral corticosteroids, cyclosporine, methotrexate, mycophenolate, phototherapy, and many novel agents are less certain. CONCLUSIONS Among individuals with moderate-to-severe AD, high-certainty evidence demonstrates that high-dose upadacitinib is among the most effective in addressing multiple patient-important outcomes, but also is among the most harmful. High-dose abrocitinib and low-dose upadacitinib are effective, but also among the most harmful. Dupilumab, lebrikizumab, and tralokinumab are of intermediate effectiveness and have favorable safety.
Collapse
Key Words
- Atopic dermatitis (eczema)
- Janus kinase (JAK) inhibitors (upadacitinib, abrocitinib, baricitinib), patient-important outcomes and adverse events or adverse reactions, disease severity, itch, sleep, itch and sleep disturbance quality of life
- network meta-analysis (comparative effectiveness, multiple treatment comparison)
- systemic treatments and phototherapy (light therapy, immunosuppressants, immunomodulators, DMARDs, cyclosporine, methotrexate, azathioprine, mycophenolate, cortiosteroids, narrow-band UVB), biologics (dupilumab, lebrikizumab, tralokinumab, nemolizumab)
Collapse
Affiliation(s)
- Alexandro W L Chu
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Melanie M Wong
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Daniel G Rayner
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Gordon H Guyatt
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Juan Pablo Díaz Martinez
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Renata Ceccacci
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Irene X Zhao
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Eric McMullen
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Archita Srivastava
- Evidence in Allergy Group, McMaster University, Hamilton, Canada; Department of Internal Medicine, Western University, London, Canada
| | - Jason Wang
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Aaron Wen
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | - Fang Chi Wang
- Evidence in Allergy Group, McMaster University, Hamilton, Canada; Schulich School of Medicine & Dentistry, Western University, London, Canada
| | | | - Ariel Izcovich
- Servicio de Clínica Médica, Hospital Aleman, Buenos Aires, Argentina
| | - Paul Oykhman
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada
| | | | - Julie Wang
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jonathan M Spergel
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pa; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jasvinder A Singh
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Ala
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Peck Y Ong
- Division of Clinical Immunology and Allergy, Children's Hospital Los Angeles, Los Angeles, Calif; Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, Calif
| | | | | | - Peter A Lio
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, Ill; Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Mary Laura Lind
- School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, Ariz
| | - Jennifer LeBovidge
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | | | - Joey Huynh
- Sepulveda VA Medical Center, North Hills, Calif
| | - Matthew Greenhawt
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo; Section of Allergy and Immunology, Children's Hospital Colorado, Aurora, Colo
| | | | | | | | - Lina Chen
- Evidence in Allergy Group, McMaster University, Hamilton, Canada; Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Korey Capozza
- Global Parents for Eczema Research, Santa Barbara, Calif
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY
| | - Mark Boguniewicz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colo; Division of Pediatric Allergy and Clinical Immunology, National Jewish Health, Denver, Colo
| | | | - Rachel N Asiniwasis
- Department of Dermatology, University of Saskatchewan, Regina, Saskatchewan, Canada
| | | | - Derek K Chu
- Department of Medicine, McMaster University, Hamilton, Canada; Evidence in Allergy Group, McMaster University, Hamilton, Canada; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada; The Research Institute of St. Joe's Hamilton, Hamilton, Canada.
| |
Collapse
|
24
|
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
|
25
|
Renteria AE, Valera FCP, Maniakas A, Adam D, Filali-Mouhim A, Ruffin M, Mfuna LE, Brochiero E, Desrosiers MY. Azithromycin Mechanisms of Action in CRS Include Epithelial Barrier Restoration and Type 1 Inflammation Reduction. Otolaryngol Head Neck Surg 2023; 169:1055-1063. [PMID: 37125631 DOI: 10.1002/ohn.355] [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: 08/31/2022] [Revised: 03/18/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023]
Abstract
OBJECTIVE Previous in vitro transcriptomic profiling suggests azithromycin exerts its effects in patients with chronic rhinosinusitis (CRS) via modulation of type 1 inflammation and restoration of epithelial barrier function. We wished to verify these postulated effects using in vitro models of epithelial repair and in vivo transcriptional profiling. STUDY DESIGN Functional effects of azithromycin in CRS were verified using in vitro models of wounding. The mechanism of the effect of azithromycin was assessed in vivo using transcriptomic profiling. SETTING Academic medical center. METHODS Effects of azithromycin on the speed of epithelial repair were verified in a wounding model using primary nasal epithelial cells (pNEC) from CRS patients. Nasal brushings collected pre-and posttreatment during a placebo-controlled trial of azithromycin for CRS patients unresponsive to surgery underwent transcriptomic profiling to identify implicated pathways. RESULTS Administration of azithromycin improved the wound healing rates in CRS pNECs and prevented the negative effect of Staphylococcus aureus on epithelial repair. In vivo, response to azithromycin was associated with downregulation in pathways of type 1 inflammation, and upregulation of pathways implicated in the restoration of the cell cycle. CONCLUSION Restoration of healthy epithelial function may represent a major mode of action of azithromycin in CRS. In vitro models show enhanced epithelial repair, while in vivo transcriptomics shows downregulation of pathways type 1 inflammation accompanied by upregulation of DNA repair and cell-cycle pathways. The maximal effect in patients with high levels of type 1-enhanced inflammation suggests that azithromycin may represent a novel therapeutic option for surgery-unresponsive CRS patients.
Collapse
Affiliation(s)
- Axel E Renteria
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département d'Oto-rhino-laryngologie et chirurgie cervico-faciale, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| | - Fabiana C P Valera
- Department of Ophthalmology, Otorhinolaryngology and Head and Neck Surgery, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Damien Adam
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Ali Filali-Mouhim
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Manon Ruffin
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Leandra Endam Mfuna
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
| | - Emmanuelle Brochiero
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Martin Y Desrosiers
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Quebec, Canada
- Département d'Oto-rhino-laryngologie et chirurgie cervico-faciale, Centre hospitalier de l'Université de Montréal (CHUM), Montréal, Quebec, Canada
| |
Collapse
|
26
|
Almoughrabie S, Cau L, Cavagnero K, O’Neill AM, Li F, Roso-Mares A, Mainzer C, Closs B, Kolar MJ, Williams KJ, Bensinger SJ, Gallo RL. Commensal Cutibacterium acnes induce epidermal lipid synthesis important for skin barrier function. SCIENCE ADVANCES 2023; 9:eadg6262. [PMID: 37595033 PMCID: PMC10438445 DOI: 10.1126/sciadv.adg6262] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Lipid synthesis is necessary for formation of epithelial barriers and homeostasis with external microbes. An analysis of the response of human keratinocytes to several different commensal bacteria on the skin revealed that Cutibacterium acnes induced a large increase in essential lipids including triglycerides, ceramides, cholesterol, and free fatty acids. A similar response occurred in mouse epidermis and in human skin affected with acne. Further analysis showed that this increase in lipids was mediated by short-chain fatty acids produced by Cutibacterium acnes and was dependent on increased expression of several lipid synthesis genes including glycerol-3-phosphate-acyltransferase-3. Inhibition or RNA silencing of peroxisome proliferator-activated receptor-α (PPARα), but not PPARβ and PPARγ, blocked this response. The increase in keratinocyte lipid content improved innate barrier functions including antimicrobial activity, paracellular diffusion, and transepidermal water loss. These results reveal that metabolites from a common commensal bacterium have a previously unappreciated influence on the composition of epidermal lipids.
Collapse
Affiliation(s)
- Samia Almoughrabie
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
- SILAB, Brive, France
| | | | - Kellen Cavagnero
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| | - Alan M. O’Neill
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| | - Fengwu Li
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| | - Andrea Roso-Mares
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| | | | | | - Matthew J. Kolar
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| | - Kevin J. Williams
- Department of Biological Chemistry, UCLA, Los Angeles, CA, USA
- UCLA Lipidomics Lab, UCLA, Los Angeles, CA, USA
| | - Steven J. Bensinger
- UCLA Lipidomics Lab, UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Richard L. Gallo
- Department of Dermatology, University of California San Diego, La Jolla CA, USA
| |
Collapse
|
27
|
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
|
28
|
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder with a lifetime prevalence of up to 20% which can occur at any age but is most common among children. There is a significant burden of pediatric AD in the primary care setting; thus, the ability to recognize and manage AD is of utmost importance to pediatricians. Treatment of AD requires a multifaceted approach based on a patient's severity including behavioral modifications, topical and systemic pharmacologic therapies, and phototherapy.
Collapse
Affiliation(s)
- Caitlyn Kellogg
- Department of Internal Medicine, Harbor-UCLA, 1000 West Carson Street, Box 458, Torrance, CA 90509, USA
| | - Jan Smogorzewski
- Department of Internal Medicine, Division of Dermatology, Harbor-UCLA, 1000 West Carson Street, Box 458, Torrance, CA 90502, USA.
| |
Collapse
|
29
|
Grafanaki K, Antonatos C, Maniatis A, Petropoulou A, Vryzaki E, Vasilopoulos Y, Georgiou S, Gregoriou S. Intrinsic Effects of Exposome in Atopic Dermatitis: Genomics, Epigenomics and Regulatory Layers. J Clin Med 2023; 12:4000. [PMID: 37373692 DOI: 10.3390/jcm12124000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Atopic dermatitis (AD) or atopic eczema is an increasingly manifested inflammatory skin disorder of complex etiology which is modulated by both extrinsic and intrinsic factors. The exposome includes a person's lifetime exposures and their effects. We recently reviewed the extrinsic exposome's environmental risk factors that contribute to AD. The periods of pregnancy, infancy, and teenage years are recognized as crucial stages in the formation of AD, where the exposome leads to enduring impacts on the immune system. However, research is now focusing on the interactions between intrinsic pathways that are modulated by the extrinsic exposome, including genetic variation, epigenetic modifications, and signals, such as diet, stress, and microbiome interactions. As a result, immune dysregulation, barrier dysfunction, hormonal fluctuations, and skin microbiome dysbiosis are important factors contributing to AD development, and their in-depth understanding is crucial not only for AD treatment but also for similar inflammatory disorders.
Collapse
Affiliation(s)
- Katerina Grafanaki
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Alexandros Maniatis
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Antonia Petropoulou
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Eleftheria Vryzaki
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Sophia Georgiou
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Stamatis Gregoriou
- Department of Dermatology-Venereology, Faculty of Medicine, Andreas Sygros Hospital, National and Kapodistrian University of Athens, 16121 Athens, Greece
| |
Collapse
|
30
|
Epicutaneous Sensitization and Food Allergy: Preventive Strategies Targeting Skin Barrier Repair-Facts and Challenges. Nutrients 2023; 15:nu15051070. [PMID: 36904070 PMCID: PMC10005101 DOI: 10.3390/nu15051070] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
Food allergy represents a growing public health and socio-economic problem with an increasing prevalence over the last two decades. Despite its substantial impact on the quality of life, current treatment options for food allergy are limited to strict allergen avoidance and emergency management, creating an urgent need for effective preventive strategies. Advances in the understanding of the food allergy pathogenesis allow to develop more precise approaches targeting specific pathophysiological pathways. Recently, the skin has become an important target for food allergy prevention strategies, as it has been hypothesized that allergen exposure through the impaired skin barrier might induce an immune response resulting in subsequent development of food allergy. This review aims to discuss current evidence supporting this complex interplay between the skin barrier dysfunction and food allergy by highlighting the crucial role of epicutaneous sensitization in the causality pathway leading to food allergen sensitization and progression to clinical food allergy. We also summarize recently studied prophylactic and therapeutic interventions targeting the skin barrier repair as an emerging food allergy prevention strategy and discuss current evidence controversies and future challenges. Further studies are needed before these promising strategies can be routinely implemented as prevention advice for the general population.
Collapse
|
31
|
Amin MM, Elkady HM. Prevalence of common sensitizing aeroallergens in patients with atopic dermatitis. Cytokine 2023; 162:156087. [PMID: 36470066 DOI: 10.1016/j.cyto.2022.156087] [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: 06/20/2022] [Revised: 10/26/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Atopic dermatitis (AD), a skin disease burden worldwide, is a complex, multifactorial, chronic inflammatory disease. Prevalence of AD is increasing in developing countries and identifying the causative allergens is a major challenge. Aeroallergens are shown to aggravate atopic dermatitis. PURPOSE Explore the prevalence of aeroallergens sensitization in patients with AD and its possible relation with AD severity. METHODS Cross-sectional study of 132 patients diagnosed to have atopic dermatitis. Atopy was detected by serum specific IgE to a panel of the most encountered aeroallergens. RESULTS From the 132 patients, elevated specific IgE was detected in 72.7 % from which 59.1 % were mild and 86.4 % are moderate/severe as well as 15.9 % are mono-sensitized and 54.5 % are poly-sensitized with poly-sensitization being more in severe cases compared to mild cases (68.2 % vs 4.5 %). Regarding specific IgE to different aeroallergens, the most prevalent were Dermatophagoides pteronyssinus (50 %), followed by Dermatophagoides farinae (34.1 %), Birch pollen (20.5 %), cat epithelium (18.2 %) Regweed (15.9 %), and Cockroach (9.1 %). However, moderate to severe cases were more sensitized to candida (p = 0.012), mix grass pollen (p = 0.002), ragweed (0.00), mite (p = 0.037) and cat epithelium (p = 0.007). CONCLUSION Dermatophagoides pteronyssinus, Dermatophagoides farinae, Birch pollen, cat epithelium Regweed and Cockroach are the most frequent sensitizing aeroallergens in atopic dermatitis.
Collapse
Affiliation(s)
- Mariam M Amin
- Department of Internal Medicine, Allergy and Clinical Immunology, Ain Shams University, Cairo, Egypt.
| | - Hossam M Elkady
- Department of Internal Medicine, Allergy and Clinical Immunology, Ain Shams University, Cairo, Egypt
| |
Collapse
|
32
|
Skin Barrier Function and Infant Tidal Flow-Volume Loops-A Population-Based Observational Study. CHILDREN (BASEL, SWITZERLAND) 2022; 10:children10010088. [PMID: 36670639 PMCID: PMC9856825 DOI: 10.3390/children10010088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023]
Abstract
Background: The relationship between the skin barrier- and lung function in infancy is largely unexplored. We aimed to explore if reduced skin barrier function by high transepidermal water loss (TEWL), or manifestations of eczema or Filaggrin (FLG) mutations, were associated with lower lung function in three-month-old infants. Methods: From the population-based PreventADALL cohort, 899 infants with lung function measurements and information on either TEWL, eczema at three months of age and/or FLG mutations were included. Lower lung function by tidal flow-volume loops was defined as a ratio of time to peak tidal expiratory flow to expiratory time (tPTEF/tE) <0.25 and a tPTEF <0.17 s (<25th percentile). A high TEWL >8.83 g/m2/h (>75th percentile) denoted reduced skin barrier function, and DNA was genotyped for FLG mutations (R501X, 2282del4 and R2447X). Results: Neither a high TEWL, nor eczema or FLG mutations, were associated with a lower tPTEF/tE. While a high TEWL was associated with a lower tPTEF; adjusted OR (95% CI) 1.61 (1.08, 2.42), the presence of eczema or FLG mutations were not. Conclusions: Overall, a high TEWL, eczema or FLG mutations were not associated with lower lung function in healthy three-month-old infants. However, an inverse association between high TEWL and tPTEF was observed, indicating a possible link between the skin barrier- and lung function in early infancy.
Collapse
|
33
|
Radi G, Campanti A, Diotallevi F, Martina E, Marani A, Offidani A. A Systematic Review of Atopic Dermatitis: The Intriguing Journey Starting from Physiopathology to Treatment, from Laboratory Bench to Bedside. Biomedicines 2022; 10:2700. [PMID: 36359220 PMCID: PMC9688004 DOI: 10.3390/biomedicines10112700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/25/2022] [Accepted: 10/13/2022] [Indexed: 09/02/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory and immune-mediated skin disease with a complex pathophysiology and still represents a therapeutic challenge, owing to limited responses to available treatments. However, recent advances in the understanding of AD pathophysiology have led to the discovery of several new potential therapeutic targets, and research in the field of new molecules with therapeutic perspectives is boiling, with more than 70 new promising drugs in development. The aim of this systematic review is to provide the state of the art on the current knowledge concerning the pathophysiology of the disease and on novel agents currently being investigated for AD, and to highlight which type of evolution is going to take place in therapeutic approaches of atopic dermatitis in the coming years.
Collapse
Affiliation(s)
| | | | - Federico Diotallevi
- Dermatological Clinic, Department of Clinical and Molecular Sciences, Polytechnic Marche University, 60126 Ancona, Italy
| | | | | | | |
Collapse
|
34
|
Pesticides and Their Impairing Effects on Epithelial Barrier Integrity, Dysbiosis, Disruption of the AhR Signaling Pathway and Development of Immune-Mediated Inflammatory Diseases. Int J Mol Sci 2022; 23:ijms232012402. [PMID: 36293259 PMCID: PMC9604036 DOI: 10.3390/ijms232012402] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
The environmental and occupational risk we confront from agricultural chemicals increases as their presence in natural habitats rises to hazardous levels, building a major part of the exposome. This is of particular concern in low- and middle-income countries, such as Brazil, known as a leading producer of agricultural commodities and consumer of pesticides. As long as public policies continue to encourage the indiscriminate use of pesticides and governments continue to support this strategy instead of endorsing sustainable agricultural alternatives, the environmental burden that damages epithelial barriers will continue to grow. Chronic exposure to environmental contaminants in early life can affect crucial barrier tissue, such as skin epithelium, airways, and intestine, causing increased permeability, leaking, dysbiosis, and inflammation, with serious implications for metabolism and homeostasis. This vicious cycle of exposure to environmental factors and the consequent damage to the epithelial barrier has been associated with an increase in immune-mediated chronic inflammatory diseases. Understanding how the harmful effects of pesticides on the epithelial barrier impact cellular interactions mediated by endogenous sensors that coordinate a successful immune system represents a crucial challenge. In line with the epithelial barrier hypothesis, this narrative review reports the available evidence on the effects of pesticides on epithelial barrier integrity, dysbiosis, AhR signaling, and the consequent development of immune-mediated inflammatory diseases.
Collapse
|
35
|
Song H, Lee J, Jeong KY, Cheon DS, Park JW. Comparison of sensitization patterns to dust mite allergens between atopic dermatitis patients and dogs, and non-specific reactivity of canine IgE to the storage mite Tyrophagus putrescentiae. EXPERIMENTAL & APPLIED ACAROLOGY 2022; 88:41-55. [PMID: 36190668 DOI: 10.1007/s10493-022-00744-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
House dust mite is a common cause of atopic dermatitis (AD) both in humans and dogs. Detection of serum IgE to allergens is commonly used to diagnose allergic diseases. However, false-positive reactions due to cross-reactivity and non-specific reactivity may lead to misdiagnosis. We compared human and canine IgE reactivities to mite component allergens. Canine IgE-reactive components of Dermatophagoides farinae and Tyrophagus putrescentiae were identified by tandem mass spectrometry. Recombinant proteins were produced and IgE reactivities to component allergens were assessed by ELISA and inhibition assays using sera from AD patients and dogs. Canine IgE-reactive proteins (Der f 1, Der f 11, Tyr p 4, Tyr p 8, Tyr p 11, Tyr p 28) were identified by proteome analysis. Most patients were sensitized to Der f 1 (93.3%) and Der f 2 (86.7%). Dogs showed high sensitization to Der f 2 (94.1%) and Der f 18 (84.6%). Both patients and dogs showed low IgE binding frequency to Tyr p 8, 43.3% and 4%, respectively. The ELISA inhibition study indicated that canine IgE reactivity to T. putrescentiae is mostly due to non-specific reaction and cross-reaction with D. farinae. Different IgE sensitization patterns were shown between allergic humans and dogs with AD, especially to Der f 18, for the first time in Korea. Furthermore, non-specific canine IgE reactivity to storage mite indicates the possibility of misdiagnoses. Standardizations focused on the major canine allergen content of extracts should be developed. This will allow precision diagnosis and individuated treatments for each patient and atopic dog.
Collapse
Affiliation(s)
- Hangyeol Song
- Department of Internal Medicine, Institute of Allergy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jongsun Lee
- Department of Internal Medicine, Institute of Allergy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Kyoung Yong Jeong
- Department of Internal Medicine, Institute of Allergy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| | | | - Jung-Won Park
- Department of Internal Medicine, Institute of Allergy, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| |
Collapse
|
36
|
Xia Y, Cao H, Zheng J, Chen L. Claudin-1 Mediated Tight Junction Dysfunction as a Contributor to Atopic March. Front Immunol 2022; 13:927465. [PMID: 35844593 PMCID: PMC9277052 DOI: 10.3389/fimmu.2022.927465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022] Open
Abstract
Atopic march refers to the phenomenon wherein the occurrence of asthma and food allergy tends to increase after atopic dermatitis. The mechanism underlying the progression of allergic inflammation from the skin to gastrointestinal (GI) tract and airways has still remained elusive. Impaired skin barrier was proposed as a risk factor for allergic sensitization. Claudin-1 protein forms tight junctions and is highly expressed in the epithelium of the skin, airways, and GI tract, thus, the downregulation of claudin-1 expression level caused by CLDN-1 gene polymorphism can mediate common dysregulation of epithelial barrier function in these organs, potentially leading to allergic sensitization at various sites. Importantly, in patients with atopic dermatitis, asthma, and food allergy, claudin-1 expression level was significantly downregulated in the skin, bronchial and intestinal epithelium, respectively. Knockdown of claudin-1 expression level in mouse models of atopic dermatitis and allergic asthma exacerbated allergic inflammation, proving that downregulation of claudin-1 expression level contributes to the pathogenesis of allergic diseases. Therefore, we hypothesized that the tight junction dysfunction mediated by downregulation of claudin-1 expression level contributes to atopic march. Further validation with clinical data from patients with atopic march or mouse models of atopic march is needed. If this hypothesis can be fully confirmed, impaired claudin-1 expression level may be a risk factor and likely a diagnostic marker for atopic march. Claudin-1 may serve as a valuable target to slowdown or block the progression of atopic march.
Collapse
|
37
|
Atopic Dermatitis: Sailing beyond the Sunset with a Multitude of Novel Treatments. J Clin Med 2022; 11:jcm11123475. [PMID: 35743543 PMCID: PMC9225552 DOI: 10.3390/jcm11123475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/17/2022] Open
Abstract
Atopic eczema or dermatitis (AD) is a chronic pruritic inflammatory cutaneous disorder with an incidence up to 20% in children and 10% in adults depending on region and ethnicity [...].
Collapse
|
38
|
Tricarico PM, Mentino D, De Marco A, Del Vecchio C, Garra S, Cazzato G, Foti C, Crovella S, Calamita G. Aquaporins Are One of the Critical Factors in the Disruption of the Skin Barrier in Inflammatory Skin Diseases. Int J Mol Sci 2022; 23:4020. [PMID: 35409378 PMCID: PMC8999368 DOI: 10.3390/ijms23074020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 02/05/2023] Open
Abstract
The skin is the largest organ of the human body, serving as an effective mechanical barrier between the internal milieu and the external environment. The skin is widely considered the first-line defence of the body, with an essential function in rejecting pathogens and preventing mechanical, chemical, and physical damages. Keratinocytes are the predominant cells of the outer skin layer, the epidermis, which acts as a mechanical and water-permeability barrier. The epidermis is a permanently renewed tissue where undifferentiated keratinocytes located at the basal layer proliferate and migrate to the overlying layers. During this migration process, keratinocytes undertake a differentiation program known as keratinization process. Dysregulation of this differentiation process can result in a series of skin disorders. In this context, aquaporins (AQPs), a family of membrane channel proteins allowing the movement of water and small neutral solutes, are emerging as important players in skin physiology and skin diseases. Here, we review the role of AQPs in skin keratinization, hydration, keratinocytes proliferation, water retention, barrier repair, wound healing, and immune response activation. We also discuss the dysregulated involvement of AQPs in some common inflammatory dermatological diseases characterised by skin barrier disruption.
Collapse
Affiliation(s)
- Paola Maura Tricarico
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Via dell’Istria 65/1, 34137 Trieste, Italy;
| | - Donatella Mentino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (D.M.); (S.G.)
| | - Aurora De Marco
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, Piazza Giulio Cesare, 11, 70121 Bari, Italy;
| | - Cecilia Del Vecchio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Sabino Garra
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (D.M.); (S.G.)
| | - Gerardo Cazzato
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70121 Bari, Italy;
| | - Caterina Foti
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari “Aldo Moro”, Piazza Giulio Cesare, 11, 70121 Bari, Italy;
| | - Sergio Crovella
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, University of Qatar, Doha 2713, Qatar;
| | - Giuseppe Calamita
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, Via E. Orabona, 4, 70125 Bari, Italy; (D.M.); (S.G.)
| |
Collapse
|
39
|
Ye L, Lai Q, Wen S, Wang X, Yang B, Man MQ. Correlation of Body Mass Index with Epidermal Biophysical Properties Varies with Gender in Chinese. Skin Pharmacol Physiol 2022; 35:215-223. [PMID: 35378539 PMCID: PMC9254306 DOI: 10.1159/000524295] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/07/2022] [Indexed: 11/19/2022]
Abstract
Background Epidermal function is associated with diabetes and renal disease. Whether obesity can reflect the changes in epidermal function is not clear yet. Objective We assessed here the correlation of epidermal functions with body mass index (BMI) in a large Chinese cohort. Methods and Subjects A total of 1,405 Chinese aged 21–98 years old were enrolled in this study. Epidermal functions, including transepidermal water loss (TEWL), stratum corneum hydration, and skin surface pH, were measured on the flexor forearm and the shin. Subjects' height and body weight were also measured. Results Age positively correlated with both TEWL and skin surface pH, while it negatively correlated with stratum corneum hydration on both the forearm and the shin of females. Similarly, age positively correlated with skin surface pH, while negatively correlating with stratum corneum hydration on both the forearm and the shin of males. In females, BMI positively correlated with skin surface pH, while it negatively correlated with stratum corneum hydration on both the forearm and the shin. However, BMI correlated neither with skin surface pH on both the forearm and the shin nor with stratum corneum hydration on the shin of males. Conclusion These results demonstrate that correlations of BMI with age and epidermal functions vary with gender.
Collapse
Affiliation(s)
- Li Ye
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Qingsong Lai
- Center of Chronic Disease Prevention, Puning, China
| | - Si Wen
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Xiaohua Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Mao-Qiang Man
- Dermatology Hospital, Southern Medical University, Guangzhou, China
- *Mao-Qiang Man,
| |
Collapse
|